General Information of the Disease (ID: DIS00080)
Name
Melanoma
ICD
ICD-11: 2C30
Resistance Map
Type(s) of Resistant Mechanism of This Disease
  ADTT: Aberration of the Drug's Therapeutic Target
  EADR: Epigenetic Alteration of DNA, RNA or Protein
  IDUE: Irregularity in Drug Uptake and Drug Efflux
  RTDM: Regulation by the Disease Microenvironment
  UAPP: Unusual Activation of Pro-survival Pathway
Drug Resistance Data Categorized by Drug
Approved Drug(s)
16 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
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [1]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L597S (c.1789_1790delCTinsTC)
Sensitive Drug Binimetinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
In Vitro Model Skin sample .
In Vivo Model Mouse PDX model Mus musculus
Experiment for
Drug Resistance
Crystal violet staining assay
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [2]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600X (c.1798_1800)
Sensitive Drug Binimetinib
Experimental Note Identified from the Human Clinical Data
In Vitro Model Cutaneous melanoma tissue .
Mechanism Description The missense mutation p.V600X (c.1798_1800) in gene BRAF cause the sensitivity of Binimetinib by unusual activation of pro-survival pathway
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [2]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Binimetinib
Experimental Note Identified from the Human Clinical Data
In Vitro Model Cutaneous melanoma tissue .
Mechanism Description The missense mutation p.V600E (c.1799T>A) in gene BRAF cause the sensitivity of Binimetinib by unusual activation of pro-survival pathway
Key Molecule: GTPase Nras (NRAS) [2]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Sensitive Drug Binimetinib
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
Key Molecule: GTPase Nras (NRAS) [2]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61R (c.182A>G)
Sensitive Drug Binimetinib
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
Key Molecule: GTPase Nras (NRAS) [2]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Sensitive Drug Binimetinib
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
Key Molecule: GTPase Nras (NRAS) [2]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61R (c.182A>G)
Sensitive Drug Binimetinib
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
Key Molecule: GTPase Nras (NRAS) [2]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Sensitive Drug Binimetinib
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
Cisplatin
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-301 [3]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Resistant Drug Cisplatin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AKT/FAKT signaling pathway Activation hsa04151
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
SkMEL1 cells Skin Homo sapiens (Human) CVCL_0068
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Colony formation assay; Annexin V-fluorescein isothiocyanate (FITC) apoptosis analysis; Wound scratch healing or transwell invasion assay
Mechanism Description PTEN can interact with AkT and FAk and inhibit their activity through their dephosphorylation, Akt and FAk signaling pathways are involved in miR301a/PTEN-promoting malignant phenotypes in MM cells, miR301a promotes MM progression via activation of Akt and FAk signaling pathways by down regulating PTEN.
Key Molecule: hsa-mir-211 [4]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Methylation
Down-regulation
Resistant Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Activation hsa05200
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Sk-Mel28 cells Skin Homo sapiens (Human) CVCL_0526
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Overexpressed 211 could enhance the anticancer effect of cisplatin and restoration of miR-211 rendered susceptibility to cisplatin in cisplatin-resistant cells.miR-211 could be transcriptionally repressed by EZH2 mediated promoter methylation.
Key Molecule: hsa-miR-30a-5p [5]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Resistant Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT/P53 signaling pathway Regulation hsa04151
Cell viability Activation hsa05200
In Vitro Model M8 cells Skin Homo sapiens (Human) N.A.
Sk-Mel-19 cells Skin Homo sapiens (Human) CVCL_6025
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-30a-5p was over-expressed in cisplatin resistant melanoma cells and could influence the activity of PI3k/AkT and the protein level of P53 by targeting IGF1R gene.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Phosphatase and tensin homolog (PTEN) [3]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Resistant Drug Cisplatin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AKT/FAKT signaling pathway Activation hsa04151
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
SkMEL1 cells Skin Homo sapiens (Human) CVCL_0068
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Colony formation assay; Annexin V-fluorescein isothiocyanate (FITC) apoptosis analysis; Wound scratch healing or transwell invasion assay
Mechanism Description PTEN can interact with AkT and FAk and inhibit their activity through their dephosphorylation, Akt and FAk signaling pathways are involved in miR301a/PTEN-promoting malignant phenotypes in MM cells, miR301a promotes MM progression via activation of Akt and FAk signaling pathways by down regulating PTEN.
Key Molecule: Histone-lysine N-methyltransferase EZH2 (EZH2) [4]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Resistant Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Activation hsa05200
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Sk-Mel28 cells Skin Homo sapiens (Human) CVCL_0526
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Overexpressed 211 could enhance the anticancer effect of cisplatin and restoration of miR-211 rendered susceptibility to cisplatin in cisplatin-resistant cells.miR-211 could be transcriptionally repressed by EZH2 mediated promoter methylation.
Key Molecule: Insulin-like growth factor 1 receptor (IGF1R) [5]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Resistant Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT/P53 signaling pathway Regulation hsa04151
Cell viability Activation hsa05200
In Vitro Model M8 cells Skin Homo sapiens (Human) N.A.
Sk-Mel-19 cells Skin Homo sapiens (Human) CVCL_6025
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-30a-5p was over-expressed in cisplatin resistant melanoma cells and could influence the activity of PI3k/AkT and the protein level of P53 by targeting IGF1R gene.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-488-3p [6]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Cisplatin
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
B16 cells Skin Homo sapiens (Human) CVCL_F936
HEMn-LP cells Skin Homo sapiens (Human) N.A.
WM451 cells Skin Homo sapiens (Human) CVCL_6357
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometric analysis
Mechanism Description microRNA-488-3p sensitizes malignant melanoma cells to cisplatin by targeting PRkDC.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: DNA-dependent catalytic protein kinase (PRKDC) [6]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Cisplatin
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
B16 cells Skin Homo sapiens (Human) CVCL_F936
HEMn-LP cells Skin Homo sapiens (Human) N.A.
WM451 cells Skin Homo sapiens (Human) CVCL_6357
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR; Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometric analysis
Mechanism Description microRNA-488-3p sensitizes malignant melanoma cells to cisplatin by targeting PRkDC.
Dabrafenib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: hsa-miR-126-3p [7]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Resistant Drug Dabrafenib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Sk-Mel28 cells Skin Homo sapiens (Human) CVCL_0526
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-126-3p down-regulation contributes to dabrafenib acquired resistance in melanoma by up-regulating ADAM9 and VEGF-A.
Key Molecule: Vascular endothelial growth factor A (VEGFA) [7]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Resistant Drug Dabrafenib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell proliferation Activation hsa05200
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Sk-Mel28 cells Skin Homo sapiens (Human) CVCL_0526
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-126-3p down-regulation contributes to dabrafenib acquired resistance in melanoma by up-regulating ADAM9 and VEGF-A.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Myeloma cell metalloproteinase (ADAM9) [7]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Resistant Drug Dabrafenib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell proliferation Activation hsa05200
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Sk-Mel28 cells Skin Homo sapiens (Human) CVCL_0526
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-126-3p down-regulation contributes to dabrafenib acquired resistance in melanoma by up-regulating ADAM9 and VEGF-A.
Key Molecule: GTPase KRas (KRAS) [8]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61H
Resistant Drug Dabrafenib
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 Melanoma cells Skin Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free and overall survival assay
Mechanism Description K-RAS mutations (G12C, G12R, Q61H) have been detected in resistant melanoma cell lines and in up to 7% of BRAF inhibitor-treated patients, although kRAS mutations are far less common in primary melanomas than NRAS mutations.
Key Molecule: GTPase KRas (KRAS) [8]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G12R
Resistant Drug Dabrafenib
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 Melanoma cells Skin Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free and overall survival assay
Mechanism Description K-RAS mutations (G12C, G12R, Q61H) have been detected in resistant melanoma cell lines and in up to 7% of BRAF inhibitor-treated patients, although kRAS mutations are far less common in primary melanomas than NRAS mutations.
Key Molecule: GTPase KRas (KRAS) [8]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G12C
Resistant Drug Dabrafenib
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 Melanoma cells Skin Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free and overall survival assay
Mechanism Description K-RAS mutations (G12C, G12R, Q61H) have been detected in resistant melanoma cell lines and in up to 7% of BRAF inhibitor-treated patients, although kRAS mutations are far less common in primary melanomas than NRAS mutations.
Key Molecule: GTPase Nras (NRAS) [8], [9], [10]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61K
Resistant Drug Dabrafenib
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.
Key Molecule: GTPase Nras (NRAS) [8], [11]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61R
Resistant Drug Dabrafenib
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.
Key Molecule: MAPK/ERK kinase 2 (MEK2) [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.N126D
Resistant Drug Dabrafenib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Activation hsa04010
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 We identified four mutations involving the MAP2k2 gene (which encodes the MEk2 kinase) in drug-resistant melanoma specimens. Like its homologue MEk1, MEk2 is situated immediately downstream of RAF proteins in the MAPk pathway.
Key Molecule: MAPK/ERK kinase 2 (MEK2) [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L46F
Resistant Drug Dabrafenib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Activation hsa04010
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 We identified four mutations involving the MAP2k2 gene (which encodes the MEk2 kinase) in drug-resistant melanoma specimens. Like its homologue MEk1, MEk2 is situated immediately downstream of RAF proteins in the MAPk pathway.
Key Molecule: MAPK/ERK kinase 2 (MEK2) [12]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.C125S
Resistant Drug Dabrafenib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Activation hsa04010
Experiment for
Molecule Alteration
Whole-exome sequencing assay; Sanger sequencing assay
Experiment for
Drug Resistance
Progression-free survival assay; Overall survival assay
Mechanism Description One portion of the tumour screened by capillary sequencing of reverse transcription PCR (RT-PCR) products contained both the MEk1G128D and MEk2C125S mutations and demonstrated MAPk reactivation.
Key Molecule: MAPK/ERK kinase 2 (MEK2) [13]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q60P
Resistant Drug Dabrafenib
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free survival assay
Mechanism Description Recent whole-exome and RNA sequencing studies have identified a wide array of acquired mutations that confer resistance, including those that reactivate the MAPk pathway (NRAS, kRAS, and MEk1/2 mutations, NF1 loss, BRAF amplification, and BRAF splice variants) and those that activate the PI3k pathway (PIk3CA, PIk3R1, and AkT1/2 mutations and PTEN loss). Of the 6 samples with putative resistance-conferring alterations, 15C harbored an acquired missense PTENR159S mutation in the phosphatase domain, 25C harbored a known acquired MEkQ60L mutation.
Key Molecule: GTPase Nras (NRAS) [8]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61L
Resistant Drug Dabrafenib
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.
Key Molecule: GTPase Nras (NRAS) [8]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G13R
Resistant Drug Dabrafenib
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.
Key Molecule: GTPase Nras (NRAS) [8]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G12R
Resistant Drug Dabrafenib
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.
Key Molecule: GTPase Nras (NRAS) [8], [12]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G12D
Resistant Drug Dabrafenib
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.
Key Molecule: MAPK/ERK kinase 2 (MEK2) [12]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.E207K
Resistant Drug Dabrafenib
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
Whole-exome sequencing assay; Sanger sequencing assay
Experiment for
Drug Resistance
Progression-free survival assay; Overall survival assay
Mechanism Description 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.
Dabrafenib/Trametinib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [14]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Dabrafenib/Trametinib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [15]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600K (c.1798_1799delGTinsAA)
Sensitive Drug Dabrafenib/Trametinib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [15]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Dabrafenib/Trametinib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [16]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600X (c.1798_1799)
Sensitive Drug Dabrafenib/Trametinib
Experimental Note Identified from the Human Clinical Data
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [17]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600X (c.1798_1800)
Sensitive Drug Dabrafenib/Trametinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
Dacarbazine
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-31 [18]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Dacarbazine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell colony Inhibition hsa05200
Cell proliferation Inhibition hsa05200
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
293T cells Breast Homo sapiens (Human) CVCL_0063
HT144 cells Skin Homo sapiens (Human) CVCL_0318
SkMEL5 cells Skin Homo sapiens (Human) CVCL_0527
SkMEL1 cells Skin Homo sapiens (Human) CVCL_0068
A2058 cells Skin Homo sapiens (Human) CVCL_1059
A875 cells Skin Homo sapiens (Human) CVCL_4733
M21 cells Skin Homo sapiens (Human) CVCL_D031
SkMEL13 cells Skin Homo sapiens (Human) CVCL_6022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-31 could suppress tumor growth and enhance sensitivity to dacarbazine (DTIC) by down-regulating SOX10 mainly via inhibiting PI3k/AkT signaling pathway in melanoma.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Transcription factor SOX-10 (SOX10) [18]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Dacarbazine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell colony Inhibition hsa05200
Cell proliferation Inhibition hsa05200
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
293T cells Breast Homo sapiens (Human) CVCL_0063
HT144 cells Skin Homo sapiens (Human) CVCL_0318
SkMEL5 cells Skin Homo sapiens (Human) CVCL_0527
SkMEL1 cells Skin Homo sapiens (Human) CVCL_0068
A2058 cells Skin Homo sapiens (Human) CVCL_1059
A875 cells Skin Homo sapiens (Human) CVCL_4733
M21 cells Skin Homo sapiens (Human) CVCL_D031
SkMEL13 cells Skin Homo sapiens (Human) CVCL_6022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-31 could suppress tumor growth and enhance sensitivity to dacarbazine (DTIC) by down-regulating SOX10 mainly via inhibiting PI3k/AkT signaling pathway in melanoma.
Doxorubicin
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-301 [3]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Resistant Drug Doxorubicin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AKT/FAKT signaling pathway Activation hsa04151
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
SkMEL1 cells Skin Homo sapiens (Human) CVCL_0068
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Colony formation assay; Annexin V-fluorescein isothiocyanate (FITC) apoptosis analysis; Wound scratch healing or transwell invasion assay
Mechanism Description PTEN can interact with AkT and FAk and inhibit their activity through their dephosphorylation, Akt and FAk signaling pathways are involved in miR301a/PTEN-promoting malignant phenotypes in MM cells, miR301a promotes MM progression via activation of Akt and FAk signaling pathways by down regulating PTEN.
Key Molecule: hsa-mir-424 [19]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Resistant Drug Doxorubicin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
A375 cells Skin Homo sapiens (Human) CVCL_0132
U251 cells Brain Homo sapiens (Human) CVCL_0021
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
PARP cells Skin Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Hypoxia induces miR-424 expression and that miR-424 in turn suppresses the level of PDCD4 protein, a tumor suppressor that is involved in apoptosis, by targeting its 3' untranslated region. Functionally, miR-424 overexpression decreases the sensitivity of cancer cells (HCT116 and A375) to doxorubicin (Dox) and etoposide. In contrast, the inhibition of miR-424 (+) apoptosis and increased the sensitivity of cancer cells to Dox. In a xenograft tumor model, miR-424 overexpression promoted tumor growth following Dox treatment, suggesting that miR-424 promotes tumor cell resistance to Dox. Furthermore, miR-424 levels are inversely correlated with PDCD4 expression in clinical breast cancer samples.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Phosphatase and tensin homolog (PTEN) [3]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Resistant Drug Doxorubicin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AKT/FAKT signaling pathway Activation hsa04151
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
SkMEL1 cells Skin Homo sapiens (Human) CVCL_0068
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Colony formation assay; Annexin V-fluorescein isothiocyanate (FITC) apoptosis analysis; Wound scratch healing or transwell invasion assay
Mechanism Description PTEN can interact with AkT and FAk and inhibit their activity through their dephosphorylation, Akt and FAk signaling pathways are involved in miR301a/PTEN-promoting malignant phenotypes in MM cells, miR301a promotes MM progression via activation of Akt and FAk signaling pathways by down regulating PTEN.
Key Molecule: Programmed cell death protein 4 (PDCD4) [19]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Resistant Drug Doxorubicin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
A375 cells Skin Homo sapiens (Human) CVCL_0132
U251 cells Brain Homo sapiens (Human) CVCL_0021
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
PARP cells Skin Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Hypoxia induces miR-424 expression and that miR-424 in turn suppresses the level of PDCD4 protein, a tumor suppressor that is involved in apoptosis, by targeting its 3' untranslated region. Functionally, miR-424 overexpression decreases the sensitivity of cancer cells (HCT116 and A375) to doxorubicin (Dox) and etoposide. In contrast, the inhibition of miR-424 (+) apoptosis and increased the sensitivity of cancer cells to Dox. In a xenograft tumor model, miR-424 overexpression promoted tumor growth following Dox treatment, suggesting that miR-424 promotes tumor cell resistance to Dox. Furthermore, miR-424 levels are inversely correlated with PDCD4 expression in clinical breast cancer samples.
Etoposide
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-424 [19]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Resistant Drug Etoposide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
A375 cells Skin Homo sapiens (Human) CVCL_0132
U251 cells Brain Homo sapiens (Human) CVCL_0021
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
PARP cells Skin Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Hypoxia induces miR-424 expression and that miR-424 in turn suppresses the level of PDCD4 protein, a tumor suppressor that is involved in apoptosis, by targeting its 3' untranslated region. Functionally, miR-424 overexpression decreases the sensitivity of cancer cells (HCT116 and A375) to doxorubicin (Dox) and etoposide. In contrast, the inhibition of miR-424 (+) apoptosis and increased the sensitivity of cancer cells to Dox. In a xenograft tumor model, miR-424 overexpression promoted tumor growth following Dox treatment, suggesting that miR-424 promotes tumor cell resistance to Dox. Furthermore, miR-424 levels are inversely correlated with PDCD4 expression in clinical breast cancer samples.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Programmed cell death protein 4 (PDCD4) [19]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Resistant Drug Etoposide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
A375 cells Skin Homo sapiens (Human) CVCL_0132
U251 cells Brain Homo sapiens (Human) CVCL_0021
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
PARP cells Skin Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Hypoxia induces miR-424 expression and that miR-424 in turn suppresses the level of PDCD4 protein, a tumor suppressor that is involved in apoptosis, by targeting its 3' untranslated region. Functionally, miR-424 overexpression decreases the sensitivity of cancer cells (HCT116 and A375) to doxorubicin (Dox) and etoposide. In contrast, the inhibition of miR-424 (+) apoptosis and increased the sensitivity of cancer cells to Dox. In a xenograft tumor model, miR-424 overexpression promoted tumor growth following Dox treatment, suggesting that miR-424 promotes tumor cell resistance to Dox. Furthermore, miR-424 levels are inversely correlated with PDCD4 expression in clinical breast cancer samples.
Nivolumab
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [20]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Resistant Drug Nivolumab
Experimental Note Identified from the Human Clinical Data
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [20]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600K (c.1798_1799delGTinsAA)
Sensitive Drug Nivolumab
Experimental Note Identified from the Human Clinical Data
Paclitaxel
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-326 [21]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Resistant Drug Paclitaxel
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
In Vitro Model SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Malme3M cells Skin Homo sapiens (Human) CVCL_1438
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-326, which forms a negative feedback regulatory loop with HDAC3, regulates the invasion and the metastatic potential of cancer cells and tumor-induced angiogenesis in response to anti-cancer drugs. miR-200b, miR-217, and miR-335, which form a positive feedback loop with HDAC3, confer sensitivity to anti-cancer drugs. We show that CAGE, reported to form a feedback loop with miR-200b, serves as a downstream target of HDAC3 and miR-326. In this study, we show that the regulation of the miR-326/HDAC3 axis can be employed for the development of anti-cancer therapeutics.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Histone deacetylase 3 (HDAC3) [21]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Resistant Drug Paclitaxel
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
In Vitro Model SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Malme3M cells Skin Homo sapiens (Human) CVCL_1438
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-326, which forms a negative feedback regulatory loop with HDAC3, regulates the invasion and the metastatic potential of cancer cells and tumor-induced angiogenesis in response to anti-cancer drugs. miR-200b, miR-217, and miR-335, which form a positive feedback loop with HDAC3, confer sensitivity to anti-cancer drugs. We show that CAGE, reported to form a feedback loop with miR-200b, serves as a downstream target of HDAC3 and miR-326. In this study, we show that the regulation of the miR-326/HDAC3 axis can be employed for the development of anti-cancer therapeutics.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-335 [21], [22]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Paclitaxel
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
miR335/SIAH2/HDAC3 signaling pathway Regulation hsa05206
In Vitro Model Malme3M cells Skin Homo sapiens (Human) CVCL_1438
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Trypan blue exclusion assay; Transwell assay
Mechanism Description miR-335-mediated increased sensitivity to anti-cancer drugs was associated with its effect on HDAC3 and SIAH2 expression. miR-335 exerted apoptotic effects and inhibited ubiquitination of HDAC3 in anti-cancer drug-resistant cancer cell lines. miR-335 negatively regulated the invasion, migration, and growth rate of cancer cells. The mouse xenograft model showed that miR-335 negatively regulated the tumorigenic potential of cancer cells. The down-regulation of SIAH2 conferred sensitivity to anti-cancer drugs. The results of the study indicated that the miR-335/SIAH2/HDAC3 axis regulates the response to anti-cancer drugs.
Key Molecule: hsa-mir-217 [21]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Paclitaxel
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Malme3M cells Skin Homo sapiens (Human) CVCL_1438
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-326, which forms a negative feedback regulatory loop with HDAC3, regulates the invasion and the metastatic potential of cancer cells and tumor-induced angiogenesis in response to anti-cancer drugs. miR-200b, miR-217, and miR-335, which form a positive feedback loop with HDAC3, confer sensitivity to anti-cancer drugs. We show that CAGE, reported to form a feedback loop with miR-200b, serves as a downstream target of HDAC3 and miR-326. In this study, we show that the regulation of the miR-326/HDAC3 axis can be employed for the development of anti-cancer therapeutics.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: hsa-mir-200b [21]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Paclitaxel
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Malme3M cells Skin Homo sapiens (Human) CVCL_1438
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-326, which forms a negative feedback regulatory loop with HDAC3, regulates the invasion and the metastatic potential of cancer cells and tumor-induced angiogenesis in response to anti-cancer drugs. miR-200b, miR-217, and miR-335, which form a positive feedback loop with HDAC3, confer sensitivity to anti-cancer drugs. We show that CAGE, reported to form a feedback loop with miR-200b, serves as a downstream target of HDAC3 and miR-326. In this study, we show that the regulation of the miR-326/HDAC3 axis can be employed for the development of anti-cancer therapeutics.
Key Molecule: hsa-miR-326 [21]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Paclitaxel
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Malme3M cells Skin Homo sapiens (Human) CVCL_1438
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-326, which forms a negative feedback regulatory loop with HDAC3, regulates the invasion and the metastatic potential of cancer cells and tumor-induced angiogenesis in response to anti-cancer drugs. miR-200b, miR-217, and miR-335, which form a positive feedback loop with HDAC3, confer sensitivity to anti-cancer drugs. We show that CAGE, reported to form a feedback loop with miR-200b, serves as a downstream target of HDAC3 and miR-326. In this study, we show that the regulation of the miR-326/HDAC3 axis can be employed for the development of anti-cancer therapeutics.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: E3 ubiquitin-protein ligase SIAH2 (SIAH2) [22]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Paclitaxel
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
miR335/SIAH2/HDAC3 signaling pathway Regulation hsa05206
In Vitro Model Malme3M cells Skin Homo sapiens (Human) CVCL_1438
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Trypan blue exclusion assay; Transwell assay
Mechanism Description miR-335-mediated increased sensitivity to anti-cancer drugs was associated with its effect on HDAC3 and SIAH2 expression. miR-335 exerted apoptotic effects and inhibited ubiquitination of HDAC3 in anti-cancer drug-resistant cancer cell lines. miR-335 negatively regulated the invasion, migration, and growth rate of cancer cells. The mouse xenograft model showed that miR-335 negatively regulated the tumorigenic potential of cancer cells. The down-regulation of SIAH2 conferred sensitivity to anti-cancer drugs. The results of the study indicated that the miR-335/SIAH2/HDAC3 axis regulates the response to anti-cancer drugs.
Palbociclib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Cyclin-dependent kinase 4 (CDK4) [23]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.R24C (c.70C>T)
Sensitive Drug Palbociclib
Experimental Note Identified from the Human Clinical Data
In Vitro Model Skin sample .
Experiment for
Molecule Alteration
Western blotting analysis; Immunohistochemistry assay
Experiment for
Drug Resistance
SRB assay
Pembrolizumab
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: GTPase Nras (NRAS) [24]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61R
Resistant Drug Pembrolizumab
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.
Temozolomide
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-203 [25]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HT144 cells Skin Homo sapiens (Human) CVCL_0318
SkMEL5 cells Skin Homo sapiens (Human) CVCL_0527
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Overexpression of miR203 sensitizes MM cells to TMZ by targeting GLS.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: L-glutamine amidohydrolase (GLS) [25]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HT144 cells Skin Homo sapiens (Human) CVCL_0318
SkMEL5 cells Skin Homo sapiens (Human) CVCL_0527
Experiment for
Molecule Alteration
Dual luciferase reporter assay; Western blot analysis; Immunohistochemistry assays
Experiment for
Drug Resistance
MTT assay
Mechanism Description Overexpression of miR203 sensitizes MM cells to TMZ by targeting GLS.
Trametinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: MAPK/ERK kinase 1 (MEK1) [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V60E (c.179T>A)
Resistant Drug Trametinib
Experimental Note Identified from the Human Clinical Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
A2058 cells Skin Homo sapiens (Human) CVCL_1059
WM2664 cells Skin Homo sapiens (Human) CVCL_2765
SkMEL28 cells Skin Homo sapiens (Human) CVCL_0526
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CellTiter-Glo assay
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [26]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.D594G (c.1781A>G)
Sensitive Drug Trametinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation ERK signaling pathway Activation hsa04210
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H1299 cells Lung Homo sapiens (Human) CVCL_0060
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
H1650 cells Pleural effusion Homo sapiens (Human) CVCL_4V01
HTB-56 cells Pleural effusion Homo sapiens (Human) CVCL_0236
HTB-38 cells Colon Homo sapiens (Human) CVCL_0320
HTB-183 cells Lymph node Homo sapiens (Human) CVCL_1577
H661 cells Lymph node Homo sapiens (Human) CVCL_1577
H508 cells Abdominal wall Homo sapiens (Human) CVCL_1564
H2405 cells Lung Homo sapiens (Human) CVCL_1551
H1666 cells Pleural effusion Homo sapiens (Human) CVCL_1485
H1395 cells Lung Homo sapiens (Human) CVCL_1467
CRL-5944 cells Ascites Homo sapiens (Human) CVCL_1551
CRL-5885 cells Pleural effusion Homo sapiens (Human) CVCL_1485
CRL-5883 cells Pleural effusion Homo sapiens (Human) CVCL_1483
CRL-5868 cells Lung Homo sapiens (Human) CVCL_1467
CRL-5803 cells Lymph node Homo sapiens (Human) CVCL_0060
CCL-253 cells Abdominal wall Homo sapiens (Human) CVCL_1564
CCL-185 cells Bowel Homo sapiens (Human) CVCL_0023
Calu-6 cells Lung Homo sapiens (Human) CVCL_0236
In Vivo Model NSG mouse PDX model Mus musculus
Experiment for
Drug Resistance
Promega assay
Mechanism Description Researchers defined three distinct functional classes of BRAF mutants in human tumours. The mutants activate ERK signalling by different mechanisms that dictate their sensitivity to therapeutic inhibitors of the pathway.
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [27]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L597Q (c.1790T>A)
Sensitive Drug Trametinib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [28]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.K601E (c.1801A>G)
Sensitive Drug Trametinib
Experimental Note Identified from the Human Clinical Data
In Vitro Model Melanoma thyroid metastasis .
Experiment for
Molecule Alteration
Western blotting analysis
Mechanism Description The missense mutation p.K601E (c.1801A>G) in gene BRAF cause the sensitivity of Trametinib by unusual activation of pro-survival pathway
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [29]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.K601E (c.1801A>G)
Sensitive Drug Trametinib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [29]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L597R (c.1790T>G)
Sensitive Drug Trametinib
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
Whole-gene resequencing assay
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [29]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600R (c.1798_1799delGTinsAG)
Sensitive Drug Trametinib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [30]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L597V (c.1789C>G)
Sensitive Drug Trametinib
Experimental Note Identified from the Human Clinical Data
In Vitro Model Skin .
Experiment for
Molecule Alteration
Tumour genotyping assay
Mechanism Description The missense mutation p.L597V (c.1789C>G) in gene BRAF cause the sensitivity of Trametinib by unusual activation of pro-survival pathway
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [28]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L597S (c.1789_1790delCTinsTC)
Sensitive Drug Trametinib
Experimental Note Identified from the Human Clinical Data
In Vitro Model Melanoma thyroid metastasis .
Experiment for
Molecule Alteration
Western blotting analysis
Mechanism Description The missense mutation p.L597S (c.1789_1790delCTinsTC) in gene BRAF cause the sensitivity of Trametinib by unusual activation of pro-survival pathway
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [1]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L597S (c.1789_1790delCTinsTC)
Sensitive Drug Trametinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
In Vitro Model Skin sample .
In Vivo Model Mouse PDX model Mus musculus
Experiment for
Drug Resistance
Crystal violet staining assay
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [31]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.D594V (c.1781A>T)
Sensitive Drug Trametinib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Experiment for
Molecule Alteration
Immunoblotting analysis
Mechanism Description The missense mutation p.D594V (c.1781A>T) in gene BRAF cause the sensitivity of Trametinib by unusual activation of pro-survival pathway
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [32]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Synonymous
p.K601K (c.1803A>G)
Sensitive Drug Trametinib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [1]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Synonymous
p.L597L (c.1791A>T)
Sensitive Drug Trametinib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [29]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.K601R (c.1802A>G)
Sensitive Drug Trametinib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [30]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L597R (c.1790T>G)
Sensitive Drug Trametinib
Experimental Note Identified from the Human Clinical Data
In Vitro Model 293H cells Fetal kidney Homo sapiens (Human) CVCL_ZK99
Experiment for
Molecule Alteration
Whole genome sequencing assay
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [14]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600K (c.1798_1799delGTinsAA)
Sensitive Drug Trametinib
Experimental Note Identified from the Human Clinical Data
Trametinib/Dabrafenib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [33]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600X (c.1798_1799)
Sensitive Drug Trametinib/Dabrafenib
Experimental Note Identified from the Human Clinical Data
In Vitro Model Skin .
Experiment for
Drug Resistance
Tumor evaluation assay
Tryptophan
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Mitogen-activated protein kinase 1 (MAPK1) [34]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Resistant Drug Tryptophan
Experimental Note Discovered Using In-vivo Testing Model
Cell Pathway Regulation MAPK/RAS signaling pathway Activation hsa04010
In Vivo Model VillinCreErt2 and VillinCreErt2 APCfl/fl KRASG12D/+ C57BL/6J mouse model Mus musculus
Experiment for
Molecule Alteration
Amino acid mass spectrometry assay
Experiment for
Drug Resistance
Flow cytometry (SIINFEKL assays); T cell killing assay and clonogenic assay
Mechanism Description Sloppiness is defined by ribosomal frameshifting upon tryptophan shortage. MAPK pathway hyperactivation links sloppiness to cancer. Drug-resistant cancer cells remain sloppy and are targeted by T cells.
Key Molecule: Ras-specific guanine nucleotide-releasing factor 2 (RGRF2) [34]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Resistant Drug Tryptophan
Experimental Note Discovered Using In-vivo Testing Model
Cell Pathway Regulation MAPK/RAS signaling pathway Activation hsa04010
In Vivo Model VillinCreErt2 and VillinCreErt2 APCfl/fl KRASG12D/+ C57BL/6J mouse model Mus musculus
Experiment for
Molecule Alteration
Amino acid mass spectrometry assay
Experiment for
Drug Resistance
Flow cytometry (SIINFEKL assays); T cell killing assay and clonogenic assay
Mechanism Description Sloppiness is defined by ribosomal frameshifting upon tryptophan shortage. MAPK pathway hyperactivation links sloppiness to cancer. Drug-resistant cancer cells remain sloppy and are targeted by T cells.
Vemurafenib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [35]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Structural variation
Copy number gain
Resistant Drug Vemurafenib
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Multivariate analysis of overall or disease-free survival assay
Mechanism Description Melanoma whole-exome sequencing identifies (V600E)B-RAF amplification-mediated acquired B-RAF inhibitor resistance.
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-204-5p [36]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Resistant Drug Vemurafenib
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation ERK1/2/MEK activation signaling pathway|hsa04210) Regulation
MAPK signaling pathway Activation hsa04010
PI3K signaling pathway Activation hsa04151
RAS signaling pathway Activation hsa04014
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR204-5p and miR211-5p contribute to BRAF inhibitor resistance in melanoma. MTT assays revealed a moderate but consistent increase in resistance to VMF in cells overexpressing miR211-5p or miR204-5p. Joint overexpression of miR204-5p and miR211-5p durably stimulated Ras and MAPk upregulation. Resistance to BRAFi in melanoma involves genetic alterations that lead to reactivation of the MAPk pathway or activation of PI3-k/AkT signalling.
Key Molecule: hsa-miR-211-5p [36]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Resistant Drug Vemurafenib
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation ERK1/2/MEK activation signaling pathway|hsa04210) Regulation
MAPK signaling pathway Activation hsa04010
PI3K signaling pathway Activation hsa04151
RAS signaling pathway Activation hsa04014
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR204-5p and miR211-5p contribute to BRAF inhibitor resistance in melanoma. MTT assays revealed a moderate but consistent increase in resistance to VMF in cells overexpressing miR211-5p or miR204-5p. Joint overexpression of miR204-5p and miR211-5p durably stimulated Ras and MAPk upregulation. Resistance to BRAFi in melanoma involves genetic alterations that lead to reactivation of the MAPk pathway or activation of PI3-k/AkT signalling.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [36]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Resistant Drug Vemurafenib
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation ERK1/2/MEK activation signaling pathway|hsa04210) Regulation
MAPK signaling pathway Activation hsa04010
PI3K signaling pathway Activation hsa04151
RAS signaling pathway Activation hsa04014
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Experiment for
Molecule Alteration
Western blotting analysis; GTPase assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR204-5p and miR211-5p contribute to BRAF inhibitor resistance in melanoma. MTT assays revealed a moderate but consistent increase in resistance to VMF in cells overexpressing miR211-5p or miR204-5p. Joint overexpression of miR204-5p and miR211-5p durably stimulated Ras and MAPk upregulation. Resistance to BRAFi in melanoma involves genetic alterations that lead to reactivation of the MAPk pathway or activation of PI3-k/AkT signalling.
Key Molecule: GTPase KRas (KRAS) [8]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61H
Resistant Drug Vemurafenib
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 Melanoma cells Skin Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free and overall survival assay
Mechanism Description K-RAS mutations (G12C, G12R, Q61H) have been detected in resistant melanoma cell lines and in up to 7% of BRAF inhibitor-treated patients, although kRAS mutations are far less common in primary melanomas than NRAS mutations.
Key Molecule: GTPase KRas (KRAS) [8]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G12R
Resistant Drug Vemurafenib
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 Melanoma cells Skin Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free and overall survival assay
Mechanism Description K-RAS mutations (G12C, G12R, Q61H) have been detected in resistant melanoma cell lines and in up to 7% of BRAF inhibitor-treated patients, although kRAS mutations are far less common in primary melanomas than NRAS mutations.
Key Molecule: GTPase KRas (KRAS) [8]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G12C
Resistant Drug Vemurafenib
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 Melanoma cells Skin Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free and overall survival assay
Mechanism Description K-RAS mutations (G12C, G12R, Q61H) have been detected in resistant melanoma cell lines and in up to 7% of BRAF inhibitor-treated patients, although kRAS mutations are far less common in primary melanomas than NRAS mutations.
Key Molecule: GTPase Nras (NRAS) [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61H
Resistant Drug Vemurafenib
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.
Key Molecule: MAPK/ERK kinase 2 (MEK2) [10]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.F57C
Resistant Drug Vemurafenib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation ERK signaling pathway Activation hsa04210
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Sanger sequencing assay; Capillary sequencing assay
Experiment for
Drug Resistance
Progression-free survival assay; Overall survival assay
Mechanism Description Selecting sequential drugs based on the molecular characteristics of a single progressing biopsy is unlikely to provide improved responses, and first-line therapies targeting multiple pathways will be required. Functional analyses confirmed that MEk1k57E and MEk2F57C mutants restored extracellular signal-regulated kinase (ERk) activation in the presence of dabrafenib, whereas MEk1G176S did not alter melanoma cell sensitivity to dabrafenib.
Key Molecule: MAPK/ERK kinase 2 (MEK2) [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V35M
Resistant Drug Vemurafenib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Activation hsa04010
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 We identified four mutations involving the MAP2k2 gene (which encodes the MEk2 kinase) in drug-resistant melanoma specimens. Like its homologue MEk1, MEk2 is situated immediately downstream of RAF proteins in the MAPk pathway.
Key Molecule: MAPK/ERK kinase 2 (MEK2) [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.C125S
Resistant Drug Vemurafenib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Activation hsa04010
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 We identified four mutations involving the MAP2k2 gene (which encodes the MEk2 kinase) in drug-resistant melanoma specimens. Like its homologue MEk1, MEk2 is situated immediately downstream of RAF proteins in the MAPk pathway.
Key Molecule: PI3-kinase alpha (PIK3CA) [37]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.E545K
Resistant Drug Vemurafenib
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
Next-generation sequencing assay
Experiment for
Drug Resistance
Computerized tomography assay
Mechanism Description In patient #11, sequential biopsies showed three mutations that were not detected in the pretreatment biopsy, including an activating mutation in PIk3CA E545k readily explaining the resistance.
Key Molecule: Phosphatase and tensin homolog (PTEN) [13]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.R159S
Resistant Drug Vemurafenib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PI3K signaling pathway Activation hsa04151
Experiment for
Molecule Alteration
Whole-exome sequencing assay
Experiment for
Drug Resistance
Progression-free survival assay
Mechanism Description Recent whole-exome and RNA sequencing studies have identified a wide array of acquired mutations that confer resistance, including those that reactivate the MAPk pathway (NRAS, kRAS, and MEk1/2 mutations, NF1 loss, BRAF amplification, and BRAF splice variants) and those that activate the PI3k pathway (PIk3CA, PIk3R1, and AkT1/2 mutations and PTEN loss). Of the 6 samples with putative resistance-conferring alterations, 15C harbored an acquired missense PTENR159S mutation in the phosphatase domain, 25C harbored a known acquired MEkQ60L mutation.
Key Molecule: GTPase Nras (NRAS) [8], [9], [38]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61R
Resistant Drug Vemurafenib
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.
Key Molecule: GTPase Nras (NRAS) [8]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61L
Resistant Drug Vemurafenib
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.
Key Molecule: GTPase Nras (NRAS) [8], [9], [38]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61K
Resistant Drug Vemurafenib
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.
Key Molecule: GTPase Nras (NRAS) [8]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G13R
Resistant Drug Vemurafenib
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.
Key Molecule: GTPase Nras (NRAS) [8]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G12R
Resistant Drug Vemurafenib
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.
Key Molecule: GTPase Nras (NRAS) [8]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G12D
Resistant Drug Vemurafenib
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.
Key Molecule: GTPase Nras (NRAS) [39], [35]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61K
Resistant Drug Vemurafenib
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.
Key Molecule: GTPase Nras (NRAS) [40]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61K
Resistant Drug Vemurafenib
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.
Key Molecule: GTPase Nras (NRAS) [40]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61K
Resistant Drug Vemurafenib
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.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: Autophagy protein 5 (ATG5) [41]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Vemurafenib
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell autophagy Inhibition hsa04140
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
A375-R cells Skin Homo sapiens (Human) CVCL_6234
G-361 cells Skin Homo sapiens (Human) CVCL_1220
G361/R cells Skin Homo sapiens (Human) CVCL_IW13
MeWo cells Skin Homo sapiens (Human) CVCL_0445
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description miR216b enhances the efficacy of vemurafenib by targeting Beclin-1, UVRAG and ATG5 in melanoma. miR216b suppresses autophagy in both BRAFi-sensitive and -resistant melanoma cells.
Key Molecule: Beclin-1 (BECN1) [41]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Vemurafenib
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell autophagy Inhibition hsa04140
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
A375-R cells Skin Homo sapiens (Human) CVCL_6234
G-361 cells Skin Homo sapiens (Human) CVCL_1220
G361/R cells Skin Homo sapiens (Human) CVCL_IW13
MeWo cells Skin Homo sapiens (Human) CVCL_0445
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description miR216b enhances the efficacy of vemurafenib by targeting Beclin-1, UVRAG and ATG5 in melanoma. miR216b suppresses autophagy in both BRAFi-sensitive and -resistant melanoma cells.
Key Molecule: hsa-mir-216b [41]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Vemurafenib
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell autophagy Inhibition hsa04140
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
A375-R cells Skin Homo sapiens (Human) CVCL_6234
G-361 cells Skin Homo sapiens (Human) CVCL_1220
G361/R cells Skin Homo sapiens (Human) CVCL_IW13
MeWo cells Skin Homo sapiens (Human) CVCL_0445
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description miR216b enhances the efficacy of vemurafenib by targeting Beclin-1, UVRAG and ATG5 in melanoma. miR216b suppresses autophagy in both BRAFi-sensitive and -resistant melanoma cells.
Key Molecule: UV radiation resistance-associated gene protein (UVRAG) [41]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Vemurafenib
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell autophagy Inhibition hsa04140
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
A375-R cells Skin Homo sapiens (Human) CVCL_6234
G-361 cells Skin Homo sapiens (Human) CVCL_1220
G361/R cells Skin Homo sapiens (Human) CVCL_IW13
MeWo cells Skin Homo sapiens (Human) CVCL_0445
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description miR216b enhances the efficacy of vemurafenib by targeting Beclin-1, UVRAG and ATG5 in melanoma. miR216b suppresses autophagy in both BRAFi-sensitive and -resistant melanoma cells.
Key Molecule: hsa-mir-7 [42]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Vemurafenib
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
qRT-PCR
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.
Key Molecule: hsa-mir-100 [43]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Vemurafenib
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model PLX4032-resistant cells Skin Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description CCL2 and miR-125b, miR-34a and miR-100 are potential targets for overcoming the miR-34a and miR-100 are potential targets for overcoming the resistance to BRAFi in melanoma.
Key Molecule: hsa-mir-125b [43]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Vemurafenib
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model PLX4032-resistant cells Skin Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description CCL2 and miR-125b, miR-34a and miR-100 are potential targets for overcoming the miR-34a and miR-100 are potential targets for overcoming the resistance to BRAFi in melanoma.
Key Molecule: hsa-mir-34 [43]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Vemurafenib
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model PLX4032-resistant cells Skin Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description CCL2 and miR-125b, miR-34a and miR-100 are potential targets for overcoming the miR-34a and miR-100 are potential targets for overcoming the resistance to BRAFi in melanoma.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: RAF proto-oncogene serine/threonine-protein kinase (RAF1) [42]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Vemurafenib
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.
Key Molecule: Epidermal growth factor receptor (EGFR) [42]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Vemurafenib
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.
Key Molecule: Insulin-like growth factor 1 receptor (IGF1R) [42]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Vemurafenib
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.
Key Molecule: Monocyte chemotactic and activating factor (CCL2) [43]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Vemurafenib
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model PLX4032-resistant cells Skin Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description CCL2 and miR-125b, miR-34a and miR-100 are potential targets for overcoming the miR-34a and miR-100 are potential targets for overcoming the resistance to BRAFi in melanoma.
Clinical Trial Drug(s)
22 drug(s) in total
Click to Show/Hide the Full List of Drugs
Capivasertib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase mTOR (mTOR) [44]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.H1968Y (c.5902C>T)
Sensitive Drug Capivasertib
Experimental Note Identified from the Human Clinical Data
In Vitro Model HEK 292T cells Kidney Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Cobimetinib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [1]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L597S (c.1789_1790delCTinsTC)
Sensitive Drug Cobimetinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
In Vitro Model Skin sample .
In Vivo Model Mouse PDX model Mus musculus
Experiment for
Drug Resistance
Crystal violet staining assay
Crenolanib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Platelet-derived growth factor receptor alpha (PDGFRA) [45]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.P577S (c.1729C>T)
Sensitive Drug Crenolanib
Experimental Note Identified from the Human Clinical Data
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
Ba/F3 cells Colon Homo sapiens (Human) CVCL_0161
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Mechanism Description The missense mutation p.P577S (c.1729C>T) in gene PDGFRA cause the sensitivity of Crenolanib by aberration of the drug's therapeutic target
Key Molecule: Platelet-derived growth factor receptor alpha (PDGFRA) [45]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V658A (c.1973T>C)
Sensitive Drug Crenolanib
Experimental Note Identified from the Human Clinical Data
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
Ba/F3 cells Colon Homo sapiens (Human) CVCL_0161
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Mechanism Description The missense mutation p.V658A (c.1973T>C) in gene PDGFRA cause the sensitivity of Crenolanib by aberration of the drug's therapeutic target
Key Molecule: Platelet-derived growth factor receptor alpha (PDGFRA) [45]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.R841K (c.2522G>A)
Sensitive Drug Crenolanib
Experimental Note Identified from the Human Clinical Data
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
Ba/F3 cells Colon Homo sapiens (Human) CVCL_0161
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Mechanism Description The missense mutation p.R841K (c.2522G>A) in gene PDGFRA cause the sensitivity of Crenolanib by aberration of the drug's therapeutic target
Key Molecule: Platelet-derived growth factor receptor alpha (PDGFRA) [45]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.H845Y (c.2533C>T)
Sensitive Drug Crenolanib
Experimental Note Identified from the Human Clinical Data
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
Ba/F3 cells Colon Homo sapiens (Human) CVCL_0161
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Mechanism Description The missense mutation p.H845Y (c.2533C>T) in gene PDGFRA cause the sensitivity of Crenolanib by aberration of the drug's therapeutic target
Key Molecule: Platelet-derived growth factor receptor alpha (PDGFRA) [45]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G853D (c.2558G>A)
Sensitive Drug Crenolanib
Experimental Note Identified from the Human Clinical Data
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
Ba/F3 cells Colon Homo sapiens (Human) CVCL_0161
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Mechanism Description The missense mutation p.G853D (c.2558G>A) in gene PDGFRA cause the sensitivity of Crenolanib by aberration of the drug's therapeutic target
Key Molecule: Platelet-derived growth factor receptor alpha (PDGFRA) [45]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.P577S (c.1729C>T)
Sensitive Drug Crenolanib
Experimental Note Identified from the Human Clinical Data
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
Ba/F3 cells Colon Homo sapiens (Human) CVCL_0161
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Mechanism Description The missense mutation p.P577S (c.1729C>T) in gene PDGFRA cause the sensitivity of Crenolanib by aberration of the drug's therapeutic target
Key Molecule: Platelet-derived growth factor receptor alpha (PDGFRA) [45]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V658A (c.1973T>C)
Sensitive Drug Crenolanib
Experimental Note Identified from the Human Clinical Data
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
Ba/F3 cells Colon Homo sapiens (Human) CVCL_0161
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Mechanism Description The missense mutation p.V658A (c.1973T>C) in gene PDGFRA cause the sensitivity of Crenolanib by aberration of the drug's therapeutic target
Key Molecule: Platelet-derived growth factor receptor alpha (PDGFRA) [45]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.R841K (c.2522G>A)
Sensitive Drug Crenolanib
Experimental Note Identified from the Human Clinical Data
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
Ba/F3 cells Colon Homo sapiens (Human) CVCL_0161
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Mechanism Description The missense mutation p.R841K (c.2522G>A) in gene PDGFRA cause the sensitivity of Crenolanib by aberration of the drug's therapeutic target
Key Molecule: Platelet-derived growth factor receptor alpha (PDGFRA) [45]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.H845Y (c.2533C>T)
Sensitive Drug Crenolanib
Experimental Note Identified from the Human Clinical Data
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
Ba/F3 cells Colon Homo sapiens (Human) CVCL_0161
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Mechanism Description The missense mutation p.H845Y (c.2533C>T) in gene PDGFRA cause the sensitivity of Crenolanib by aberration of the drug's therapeutic target
Key Molecule: Platelet-derived growth factor receptor alpha (PDGFRA) [45]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G853D (c.2558G>A)
Sensitive Drug Crenolanib
Experimental Note Identified from the Human Clinical Data
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
Ba/F3 cells Colon Homo sapiens (Human) CVCL_0161
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Mechanism Description The missense mutation p.G853D (c.2558G>A) in gene PDGFRA cause the sensitivity of Crenolanib by aberration of the drug's therapeutic target
Ganetespib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [46]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Ganetespib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model Ba/F3 cells Colon Homo sapiens (Human) CVCL_0161
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
WST-1 cell proliferation assay
Refametinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [47]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600X (c.1798_1799)
Resistant Drug Refametinib
Experimental Note Identified from the Human Clinical Data
Mechanism Description The missense mutation p.V600X (c.1798_1799) in gene BRAF cause the resistance of Refametinib by aberration of the drug's therapeutic target
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [48]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Refametinib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
A375 cells Skin Homo sapiens (Human) CVCL_0132
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
A431 cells Skin Homo sapiens (Human) CVCL_0037
COLO205 cells Colon Homo sapiens (Human) CVCL_F402
BxPc3 cells Pancreas Homo sapiens (Human) CVCL_0186
SkMEL28 cells Skin Homo sapiens (Human) CVCL_0526
In Vivo Model Female athymic nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Biochemical kinase assays
Experiment for
Drug Resistance
CellTiter 96 Aqueous One assay
Mechanism Description The missense mutation p.V600E (c.1799T>A) in gene BRAF cause the sensitivity of Refametinib by unusual activation of pro-survival pathway
Selumetinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: MAPK/ERK kinase 1 (MEK1) [49]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q56P (c.167A>C)
Resistant Drug Selumetinib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
293T cells Breast Homo sapiens (Human) CVCL_0063
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Colony formation assay
Mechanism Description The missense mutation p.Q56P (c.167A>C) in gene MAP2K1 cause the resistance of Selumetinib by aberration of the drug's therapeutic target
Key Molecule: MAPK/ERK kinase 1 (MEK1) [49]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.I103N (c.308T>A)
Resistant Drug Selumetinib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
293T cells Breast Homo sapiens (Human) CVCL_0063
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Colony formation assay
Mechanism Description The missense mutation p.I103N (c.308T>A) in gene MAP2K1 cause the resistance of Selumetinib by aberration of the drug's therapeutic target
Key Molecule: MAPK/ERK kinase 1 (MEK1) [49]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L115P (c.344T>C)
Resistant Drug Selumetinib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
293T cells Breast Homo sapiens (Human) CVCL_0063
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Colony formation assay
Mechanism Description The missense mutation p.L115P (c.344T>C) in gene MAP2K1 cause the resistance of Selumetinib by aberration of the drug's therapeutic target
Key Molecule: MAPK/ERK kinase 1 (MEK1) [49]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.P124S (c.370C>T)
Resistant Drug Selumetinib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
293T cells Breast Homo sapiens (Human) CVCL_0063
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Colony formation assay
Mechanism Description The missense mutation p.P124S (c.370C>T) in gene MAP2K1 cause the resistance of Selumetinib by aberration of the drug's therapeutic target
Key Molecule: MAPK/ERK kinase 1 (MEK1) [49]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.P124L (c.371C>T)
Resistant Drug Selumetinib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
293T cells Breast Homo sapiens (Human) CVCL_0063
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Colony formation assay
Mechanism Description The missense mutation p.P124L (c.371C>T) in gene MAP2K1 cause the resistance of Selumetinib by aberration of the drug's therapeutic target
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: GTPase Nras (NRAS) [50]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Resistant Drug Selumetinib
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
Key Molecule: Guanine nucleotide-binding protein subunit alpha-11 (GNA11) [51]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q209P (c.626A>C)
Sensitive Drug Selumetinib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Guanine nucleotide-binding protein subunit alpha-11 (GNA11) [51]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q209L (c.626A>T)
Sensitive Drug Selumetinib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Guanine nucleotide-binding protein subunit alpha-11 (GNA11) [51]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q209P (c.626A>C)
Sensitive Drug Selumetinib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Guanine nucleotide-binding protein subunit alpha-11 (GNA11) [51]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q209L (c.626A>T)
Sensitive Drug Selumetinib
Experimental Note Identified from the Human Clinical Data
Key Molecule: GTPase Nras (NRAS) [52]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Sensitive Drug Selumetinib
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
Key Molecule: GTPase Nras (NRAS) [50]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Sensitive Drug Selumetinib
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
Key Molecule: GTPase Nras (NRAS) [52]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61R (c.182A>G)
Sensitive Drug Selumetinib
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
Key Molecule: GTPase Nras (NRAS) [53]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Sensitive Drug Selumetinib
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
Key Molecule: GTPase Nras (NRAS) [50]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G13D (c.38G>A)
Sensitive Drug Selumetinib
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
Key Molecule: Cellular tumor antigen p53 (TP53) [50]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.D259Y (c.775G>T)
Sensitive Drug Selumetinib
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
Vemurafenib/Cobimetinib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [54]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600X (c.1798_1799)
Sensitive Drug Vemurafenib/Cobimetinib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [14]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600K (c.1798_1799delGTinsAA)
Sensitive Drug Vemurafenib/Cobimetinib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [14]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Vemurafenib/Cobimetinib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [15]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Vemurafenib/Cobimetinib
Experimental Note Identified from the Human Clinical Data
E6201
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [55]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug E6201
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
G-361 cells Skin Homo sapiens (Human) CVCL_1220
MDA-MB-435s cells Breast Homo sapiens (Human) CVCL_0622
SEKI cells Skin Homo sapiens (Human) CVCL_3162
HMV-1 cells Uterus Homo sapiens (Human) CVCL_8233
HMCB cells Skin Homo sapiens (Human) CVCL_3317
CHL-1 cells Pleural effusion Homo sapiens (Human) CVCL_1122
Experiment for
Molecule Alteration
Western blotting analysis; Immunohistochemistry assay
Experiment for
Drug Resistance
WST-8 assay
Pictilisib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [56]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Pictilisib
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
ICH assay
Experiment for
Drug Resistance
Plasma level assay; Electrochemiluminescense assay
RAF-265
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [57]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug RAF-265
Experimental Note Identified from the Human Clinical Data
In Vitro Model Skin sample .
RO-5126766 free base
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [58]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug RO-5126766 free base
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
NCI-N87 cells Gastric Homo sapiens (Human) CVCL_1603
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
NCI-H716 cells Colon Homo sapiens (Human) CVCL_1581
SNU-16 cells Gastric Homo sapiens (Human) CVCL_0076
NCI-H520 cells Lung Homo sapiens (Human) CVCL_1566
RT-4 cells Urinary bladder Homo sapiens (Human) CVCL_0036
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
NCI-N87 cells Gastric Homo sapiens (Human) CVCL_1603
NCI-H716 cells Colon Homo sapiens (Human) CVCL_1581
KATO-3 cells Gastric Homo sapiens (Human) CVCL_0371
SNU-16 cells Gastric Homo sapiens (Human) CVCL_0076
NCI-H520 cells Lung Homo sapiens (Human) CVCL_1566
RT-4 cells Urinary bladder Homo sapiens (Human) CVCL_0036
UM-UC-14 cells Kidney Homo sapiens (Human) CVCL_2747
SUM-52PE cells Pleural effusion Homo sapiens (Human) CVCL_3425
NCI-H1581 cells Lung Homo sapiens (Human) CVCL_1479
MFE296 cells Endometrium Homo sapiens (Human) CVCL_1406
MFE280 cells Endometrium Homo sapiens (Human) CVCL_1405
KMS-11 cells Pleural effusion Homo sapiens (Human) CVCL_2989
HSC-39 cells Ascites Homo sapiens (Human) CVCL_A385
DMS-114 cells Lung Homo sapiens (Human) CVCL_1174
AN3 CA cells Endometrium Homo sapiens (Human) CVCL_0028
UM-UC-14 cells Kidney Homo sapiens (Human) CVCL_2747
KATO-III cells Pleural effusion Homo sapiens (Human) CVCL_0371
AN3 CA cells Endometrium Homo sapiens (Human) CVCL_0028
Experiment for
Molecule Alteration
Microarray assay; Western blotting analysis
Experiment for
Drug Resistance
CCK-8 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
Key Molecule: GTPase Nras (NRAS) [59]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G13D (c.38G>A)
Sensitive Drug Tanespimycin
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
Ulixertinib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [60]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Ulixertinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
A375 cells Skin Homo sapiens (Human) CVCL_0132
RkO cells Colon Homo sapiens (Human) CVCL_0504
G-361 cells Skin Homo sapiens (Human) CVCL_1220
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
SW48 cells Colon Homo sapiens (Human) CVCL_1724
COLO205 cells Colon Homo sapiens (Human) CVCL_F402
AN3CA cells Ovary Homo sapiens (Human) CVCL_0028
MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
In Vivo Model Athymic nude mouse PDX model Mus musculus
Experiment for
Drug Resistance
Standard coupled-enzyme assay
Uprosertib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: RAC-alpha serine/threonine-protein kinase (AKT1) [61]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Sensitive Drug Uprosertib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model 19 drug na?ve cell lines and four sub-lines N.A. . N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
ATP-based luminescent assay; Flow cytometry assay
Agerafenib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [62]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Agerafenib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Mechanism Description The missense mutation p.V600E (c.1799T>A) in gene BRAF cause the sensitivity of Agerafenib by aberration of the drug's therapeutic target
Lifirafenib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [63]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Lifirafenib
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
BI-847325
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [64]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug BI-847325
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
WM793 cells N.A. Homo sapiens (Human) CVCL_8787/CVCL_5414
WM39 cells Skin Homo sapiens (Human) CVCL_2240
WM164 cells Skin Homo sapiens (Human) CVCL_7928
RPMI-7951 cells Lymph node Homo sapiens (Human) CVCL_1666
1205Lu cells Skin Homo sapiens (Human) CVCL_5239
In Vivo Model BALB SCID mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis; qPCR; Proteasome-Glo Chymotrypsin-like cell-based assay
Experiment for
Drug Resistance
Alamar blue assay; Colony formation assay
GSK126
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: Histone-lysine N-methyltransferase EZH2 (EZH2) [65]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Y641S (c.1922A>C)
Sensitive Drug GSK126
Experimental Note Identified from the Human Clinical Data
In Vitro Model Skin sample .
Experiment for
Molecule Alteration
Western blotting analysis; BCA assay
Mechanism Description EZH2 activation by mutations, gene amplification and increased transcription occurred in about 20% of the cohort. These alterations were associated with significant hypermethylation of DNA and significant downregulation of 11% of transcripts in patient RNASeq data.
Key Molecule: Histone-lysine N-methyltransferase EZH2 (EZH2) [65]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Y641N (c.1921T>A)
Sensitive Drug GSK126
Experimental Note Identified from the Human Clinical Data
In Vitro Model Skin sample .
Experiment for
Molecule Alteration
Western blotting analysis; BCA assay
Mechanism Description EZH2 activation by mutations, gene amplification and increased transcription occurred in about 20% of the cohort. These alterations were associated with significant hypermethylation of DNA and significant downregulation of 11% of transcripts in patient RNASeq data.
Key Molecule: Histone-lysine N-methyltransferase EZH2 (EZH2) [65]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Y641H (c.1921T>C)
Sensitive Drug GSK126
Experimental Note Identified from the Human Clinical Data
In Vitro Model Skin sample .
Experiment for
Molecule Alteration
Western blotting analysis; BCA assay
Mechanism Description EZH2 activation by mutations, gene amplification and increased transcription occurred in about 20% of the cohort. These alterations were associated with significant hypermethylation of DNA and significant downregulation of 11% of transcripts in patient RNASeq data.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Histone-lysine N-methyltransferase EZH2 (EZH2) [66]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Y646N (c.1936T>A)
Sensitive Drug GSK126
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SKMEL-28 cells Skin Homo sapiens (Human) CVCL_0526
MM386 cells Lymph node Homo sapiens (Human) CVCL_2607
MM200 cells Skin Homo sapiens (Human) CVCL_C836
MEL-RM cells Lymph node Homo sapiens (Human) CVCL_D548
MEL-JD cells Lymph node Homo sapiens (Human) CVCL_BS80
ME4405 cells Skin Homo sapiens (Human) CVCL_C680
ME1007 cells Skin Homo sapiens (Human) CVCL_C668
IGR1 Mel-RMU cells Lymph node Homo sapiens (Human) CVCL_S994
HEM cells N.A. . N.A.
HDF cells N.A. . N.A.
C001 cells Skin Homo sapiens (Human) CVCL_B4K8
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CellTiter-Glo assay
Key Molecule: Histone-lysine N-methyltransferase EZH2 (EZH2) [66]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Y646N (c.1936T>A)
Sensitive Drug GSK126
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SKMEL-28 cells Skin Homo sapiens (Human) CVCL_0526
MM386 cells Lymph node Homo sapiens (Human) CVCL_2607
MM200 cells Skin Homo sapiens (Human) CVCL_C836
MEL-RM cells Lymph node Homo sapiens (Human) CVCL_D548
MEL-JD cells Lymph node Homo sapiens (Human) CVCL_BS80
ME4405 cells Skin Homo sapiens (Human) CVCL_C680
ME1007 cells Skin Homo sapiens (Human) CVCL_C668
IGR1 Mel-RMU cells Lymph node Homo sapiens (Human) CVCL_S994
HEM cells N.A. . N.A.
HDF cells N.A. . N.A.
C001 cells Skin Homo sapiens (Human) CVCL_B4K8
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CellTiter-Glo assay
LY-294002
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase mTOR (mTOR) [44]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.H1968Y (c.5902C>T)
Sensitive Drug LY-294002
Experimental Note Identified from the Human Clinical Data
In Vitro Model HEK 292T cells Kidney Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
LY3009120
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: GTPase Nras (NRAS) [50]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Resistant Drug LY3009120
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
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [1]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L597S (c.1789_1790delCTinsTC)
Sensitive Drug LY3009120
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
In Vitro Model Skin sample .
In Vivo Model Mouse PDX model Mus musculus
Experiment for
Drug Resistance
Crystal violet staining assay
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [50]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug LY3009120
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
Key Molecule: GTPase Nras (NRAS) [50]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Sensitive Drug LY3009120
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
Key Molecule: GTPase Nras (NRAS) [50]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G13D (c.38G>A)
Sensitive Drug LY3009120
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
Key Molecule: Cellular tumor antigen p53 (TP53) [50]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.D259Y (c.775G>T)
Sensitive Drug LY3009120
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
Omipalisib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [67]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600K (c.1798_1799delGTinsAA)
Sensitive Drug Omipalisib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CellTiter-Glo assay
Mechanism Description The missense mutation p.V600K (c.1798_1799delGTinsAA) in gene BRAF cause the sensitivity of Omipalisib by unusual activation of pro-survival pathway
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [67]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Omipalisib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CellTiter-Glo assay
Mechanism Description The missense mutation p.V600E (c.1799T>A) in gene BRAF cause the sensitivity of Omipalisib by unusual activation of pro-survival pathway
PLX4720
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Guanine nucleotide-binding protein alpha-q (GNAQ) [68]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q209P (c.626A>C)
Resistant Drug PLX4720
Experimental Note Identified from the Human Clinical Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Experiment for
Molecule Alteration
Sanger sequencing assay; SNP array; qPCR
Experiment for
Drug Resistance
CellTiter-Glo assay
Key Molecule: MAPK/ERK kinase 1 (MEK1) [49]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q56P (c.167A>C)
Resistant Drug PLX4720
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
293T cells Breast Homo sapiens (Human) CVCL_0063
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Colony formation assay
Mechanism Description The missense mutation p.Q56P (c.167A>C) in gene MAP2K1 cause the resistance of PLX4720 by unusual activation of pro-survival pathway
Key Molecule: GTPase Nras (NRAS) [69]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Resistant Drug PLX4720
Experimental Note Identified from the Human Clinical Data
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [70]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug PLX4720
Experimental Note Revealed Based on the Cell Line Data
Experiment for
Drug Resistance
CellTiter-Glo luminescent cell viability assay
Mechanism Description The missense mutation p.V600E (c.1799T>A) in gene BRAF cause the sensitivity of PLX4720 by aberration of the drug's therapeutic target
Discontinued Drug(s)
1 drug(s) in total
Click to Show/Hide the Full List of Drugs
RO4987655
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [71]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600X (c.1798_1799)
Sensitive Drug RO4987655
Experimental Note Identified from the Human Clinical Data
In Vitro Model Skin sample .
Experiment for
Molecule Alteration
IHC assay; Tumor-DNA mutation analysis
Experiment for
Drug Resistance
Pharmacokinetics analysis; Tumor biopsies analysis
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [58]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug RO4987655
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
NCI-N87 cells Gastric Homo sapiens (Human) CVCL_1603
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
NCI-H716 cells Colon Homo sapiens (Human) CVCL_1581
SNU-16 cells Gastric Homo sapiens (Human) CVCL_0076
NCI-H520 cells Lung Homo sapiens (Human) CVCL_1566
RT-4 cells Urinary bladder Homo sapiens (Human) CVCL_0036
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
NCI-N87 cells Gastric Homo sapiens (Human) CVCL_1603
NCI-H716 cells Colon Homo sapiens (Human) CVCL_1581
KATO-3 cells Gastric Homo sapiens (Human) CVCL_0371
SNU-16 cells Gastric Homo sapiens (Human) CVCL_0076
NCI-H520 cells Lung Homo sapiens (Human) CVCL_1566
RT-4 cells Urinary bladder Homo sapiens (Human) CVCL_0036
UM-UC-14 cells Kidney Homo sapiens (Human) CVCL_2747
SUM-52PE cells Pleural effusion Homo sapiens (Human) CVCL_3425
NCI-H1581 cells Lung Homo sapiens (Human) CVCL_1479
MFE296 cells Endometrium Homo sapiens (Human) CVCL_1406
MFE280 cells Endometrium Homo sapiens (Human) CVCL_1405
KMS-11 cells Pleural effusion Homo sapiens (Human) CVCL_2989
HSC-39 cells Ascites Homo sapiens (Human) CVCL_A385
DMS-114 cells Lung Homo sapiens (Human) CVCL_1174
AN3 CA cells Endometrium Homo sapiens (Human) CVCL_0028
UM-UC-14 cells Kidney Homo sapiens (Human) CVCL_2747
KATO-III cells Pleural effusion Homo sapiens (Human) CVCL_0371
AN3 CA cells Endometrium Homo sapiens (Human) CVCL_0028
Experiment for
Molecule Alteration
Microarray assay; Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Preclinical Drug(s)
28 drug(s) in total
Click to Show/Hide the Full List of Drugs
BI-69A11
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [72]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug BI-69A11
Experimental Note Identified from the Human Clinical Data
Buparlisib/Vemurafenib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [73]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600X (c.1798_1800)
Sensitive Drug Buparlisib/Vemurafenib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Melanoma cells Skin Homo sapiens (Human) N.A.
WM cells N.A. Homo sapiens (Human) N.A.
SK cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CellTiter-Glo assay; Fluorescent microscopy assay; Apoptosis analysis
CCT196969
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [74]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600D (c.1799_1800delTGinsAC)
Sensitive Drug CCT196969
Experimental Note Identified from the Human Clinical Data
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [69]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug CCT196969
Experimental Note Identified from the Human Clinical Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
In Vivo Model Female xenograft nude mouse model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis; Sanger Sequencing
Experiment for
Drug Resistance
CellTiter-Glo assay; Tumor volume measurement assay
Key Molecule: GTPase Nras (NRAS) [69]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Sensitive Drug CCT196969
Experimental Note Identified from the Human Clinical Data
CCT241161
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [74]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600D (c.1799_1800delTGinsAC)
Sensitive Drug CCT241161
Experimental Note Identified from the Human Clinical Data
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [69]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug CCT241161
Experimental Note Identified from the Human Clinical Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
In Vivo Model Female xenograft nude mouse model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis; Sanger Sequencing
Experiment for
Drug Resistance
CellTiter-Glo assay; Tumor volume measurement assay
Key Molecule: GTPase Nras (NRAS) [69]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Sensitive Drug CCT241161
Experimental Note Identified from the Human Clinical Data
Cediranib/PLX4720
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [75]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Cediranib/PLX4720
Experimental Note Identified from the Human Clinical Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
WM451 cells Skin Homo sapiens (Human) CVCL_6357
In Vivo Model Nu/Nu(ISTMel1) mouse xenograft model; NSG (RPMI7951) female mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis; Immunohistochemistry assay
Experiment for
Drug Resistance
CellTiter-Glo assay
DEL-22379
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [76]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug DEL-22379
Experimental Note Identified from the Human Clinical Data
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
A375 cells Skin Homo sapiens (Human) CVCL_0132
In Vivo Model Female athymic nu/nu mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Caspase-Glo 3/7 luminogenic assay
DETD-35
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [77]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug DETD-35
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation MEK/ERK signaling pathway Inhibition hsa04011
AKT signaling pathway Inhibition hsa04151
STAT3 signaling pathway Inhibition hsa04550
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
MeWo cells Skin Homo sapiens (Human) CVCL_0445
A2058 cells Skin Homo sapiens (Human) CVCL_1059
B16-F10 cells Skin Mus musculus (Mouse) CVCL_0159
SkMEL2 cells Skin Homo sapiens (Human) CVCL_0069
BRAF cells N.A. Homo sapiens (Human) N.A.
In Vivo Model NSG mouse PDX model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis; Immunohistochemistry analysis
Experiment for
Drug Resistance
MTT assay; Crystal violet staining assay; FACS assay
Mechanism Description DETD-35 overcame acquired vemurafenib resistance at least in part through deregulating MEK-ERK, Akt, and STAT3 signaling pathways and promoting apoptosis of cancer cells.
EBI-907
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [78]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug EBI-907
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
SW-480 cells Colon Homo sapiens (Human) CVCL_0546
Colo-205 cells Ascites Homo sapiens (Human) CVCL_0218
Calu-6 cells Lung Homo sapiens (Human) CVCL_0236
In Vivo Model Nu/Nu Colo-205 xenograft mouse model; Nu/Nu A357 xenograft mouse model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CellTiter-Glo luminescent cell viability assay
Ganetespib/TAK-733
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [46]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Ganetespib/TAK-733
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model Ba/F3 cells Colon Homo sapiens (Human) CVCL_0161
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
WST-1 cell proliferation assay
GDC0879
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [79]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug GDC0879
Experimental Note Revealed Based on the Cell Line Data
In Vivo Model Female nu/nu mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Immunoblotting assay
Experiment for
Drug Resistance
CellTiter-Glo assay
Mechanism Description The missense mutation p.V600E (c.1799T>A) in gene BRAF cause the sensitivity of GDC0879 by aberration of the drug's therapeutic target
INU-152
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [80]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug INU-152
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
In Vitro Model HT-29 cells Colon Homo sapiens (Human) CVCL_0320
A375 cells Skin Homo sapiens (Human) CVCL_0132
HEK 293 cells Kidney Homo sapiens (Human) CVCL_0045
SkMEL2 cells Skin Homo sapiens (Human) CVCL_0069
Colo-205 cells Ascites Homo sapiens (Human) CVCL_0218
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 INU-152 inhibits all RAF isoforms and inhibits MAPK pathways in mutant BRAF cells. More importantly, INU-152 exhibits minimal paradoxical pathway activation in melanoma cells with mutant RAS. INU-152 exhibits anti-tumor activities in xenograft models carrying BRAF mutations.
LY-294002/Capivasertib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase mTOR (mTOR) [44]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.P2213S (c.6637C>T)
Sensitive Drug LY-294002/Capivasertib
Experimental Note Identified from the Human Clinical Data
In Vitro Model HEK 292T cells Kidney Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Key Molecule: Serine/threonine-protein kinase mTOR (mTOR) [44]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.P2213S (c.6637C>T)
Sensitive Drug LY-294002/Capivasertib
Experimental Note Identified from the Human Clinical Data
In Vitro Model HEK 292T cells Kidney Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
MK-2206/PD184352
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Phosphatase and tensin homolog (PTEN) [68]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration FS-deletion
p.V317fs (c.950_953)
Sensitive Drug MK-2206/PD184352
Experimental Note Identified from the Human Clinical Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Experiment for
Molecule Alteration
Sanger sequencing assay; SNP array; qPCR
Experiment for
Drug Resistance
CellTiter-Glo assay
MK2206/Vemurafenib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [73]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600X (c.1798_1800)
Sensitive Drug MK2206/Vemurafenib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Melanoma cells Skin Homo sapiens (Human) N.A.
WM cells N.A. Homo sapiens (Human) N.A.
SK cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CellTiter-Glo assay; Fluorescent microscopy assay; Apoptosis analysis
Palbociclib/PD-0325901
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [81]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Palbociclib/PD-0325901
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation CDK4/6 signaling pathway Regulation hsa04218
MEK signaling pathway Inhibition hsa04011
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
WM cells N.A. Homo sapiens (Human) N.A.
SKMEL207 cells Skin Homo sapiens (Human) CVCL_6108
SBCL2 cells Skin Homo sapiens (Human) CVCL_D732
CHL-1 cells Pleural effusion Homo sapiens (Human) CVCL_1122
BOWES cells N.A. Homo sapiens (Human) N.A.
1205Lu cells Skin Homo sapiens (Human) CVCL_5239
In Vivo Model Female athymic mouse PDX model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay; Crystal violet staining assay
Mechanism Description In cutaneous melanoma, driver mutations in NRAS and BRAF promote CDK4/6 activation suggesting that inhibitors such as palbociclib are likely to provide therapeutic benefit in combination with BRAF inhibitors and/or MEK inhibitors that are FDA-approved. However, the determinants of the response to CDK4/6 inhibitors alone and in combination with other targeted inhibitors are poorly defined. Furthermore, in vivo systems to quantitatively and temporally measure the efficacy of CDK4/6 inhibitors and determine the extent that CDK activity is reactivated during acquired resistance are lacking.
PLX7904
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [82]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G466E (c.1397G>A)
Resistant Drug PLX7904
Experimental Note Identified from the Human Clinical Data
In Vitro Model SW1736 cells Thyroid Homo sapiens (Human) CVCL_3883
8505C cells Thyroid Homo sapiens (Human) CVCL_1054
Hth104 cells Thyroid gland Homo sapiens (Human) CVCL_A427
In Vivo Model mouse xenograft model Mus musculus
Mechanism Description The missense mutation p.G466E (c.1397G>A) in gene BRAF cause the resistance of PLX7904 by aberration of the drug's therapeutic target
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [82]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600R (c.1798_1799delGTinsAG)
Sensitive Drug PLX7904
Experimental Note Identified from the Human Clinical Data
In Vitro Model SW1736 cells Thyroid Homo sapiens (Human) CVCL_3883
8505C cells Thyroid Homo sapiens (Human) CVCL_1054
Hth104 cells Thyroid gland Homo sapiens (Human) CVCL_A427
In Vivo Model mouse xenograft model Mus musculus
Mechanism Description The missense mutation p.V600R (c.1798_1799delGTinsAG) in gene BRAF cause the sensitivity of PLX7904 by aberration of the drug's therapeutic target
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [82]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600D (c.1799_1800delTGinsAC)
Sensitive Drug PLX7904
Experimental Note Identified from the Human Clinical Data
In Vitro Model SW1736 cells Thyroid Homo sapiens (Human) CVCL_3883
8505C cells Thyroid Homo sapiens (Human) CVCL_1054
Hth104 cells Thyroid gland Homo sapiens (Human) CVCL_A427
In Vivo Model mouse xenograft model Mus musculus
Mechanism Description The missense mutation p.V600D (c.1799_1800delTGinsAC) in gene BRAF cause the sensitivity of PLX7904 by aberration of the drug's therapeutic target
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [83]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug PLX7904
Experimental Note Identified from the Human Clinical Data
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
A375 cells Skin Homo sapiens (Human) CVCL_0132
A431 cells Skin Homo sapiens (Human) CVCL_0037
SkBR3 cells Breast Homo sapiens (Human) CVCL_0033
SkMEL239-C3 cells Skin Homo sapiens (Human) CVCL_6122
SkMEL239 cells Skin Homo sapiens (Human) CVCL_6122
IPC-298 cells Skin Homo sapiens (Human) CVCL_1307
Colo829 cells Skin Homo sapiens (Human) CVCL_1137
B9 cells N.A. Mus musculus (Mouse) CVCL_1952
In Vivo Model mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis; Microarray gene expression analysis; Crystallization and structure determination assay
Experiment for
Drug Resistance
CellTiter-Glo assay; Anchorage-independent growth assay
RAF709
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [84]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug RAF709
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.
Key Molecule: GTPase Nras (NRAS) [84]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Sensitive Drug RAF709
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.
Key Molecule: GTPase Nras (NRAS) [84]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q61L (c.182A>T)
Sensitive Drug RAF709
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.
RMC-4550
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [85]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Resistant Drug RMC-4550
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation RAS/RAF/MEK/ERK signaling pathway Activation hsa01521
In Vitro Model NCI-H358 cells Lung Homo sapiens (Human) CVCL_1559
NCI-H508 cells Colon Homo sapiens (Human) CVCL_1564
HEK 293 cells Kidney Homo sapiens (Human) CVCL_0045
Nras cells N.A. . N.A.
NCI-H1838 cells Lung Homo sapiens (Human) CVCL_1499
KRAS cells N.A. . N.A.
Hras cells N.A. . N.A.
In Vivo Model Athymic Balb/C nude mouse model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Promega assay
Mechanism Description SHP2 inhibitor treatment decreases oncogenic RAS-RAF-MEK-ERK signaling and cancer growth by disrupting SOS1-mediated RAS-GTP loading.
S3I-201
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [86]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug S3I-201
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Colo829 cells Skin Homo sapiens (Human) CVCL_1137
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Drug Resistance
CellTiter-Glo assay
Mechanism Description The missense mutation p.V600E (c.1799T>A) in gene BRAF cause the sensitivity of S3I-201 by unusual activation of pro-survival pathway
SB590885
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [82]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug SB590885
Experimental Note Identified from the Human Clinical Data
In Vitro Model SW1736 cells Thyroid Homo sapiens (Human) CVCL_3883
8505C cells Thyroid Homo sapiens (Human) CVCL_1054
Hth104 cells Thyroid gland Homo sapiens (Human) CVCL_A427
In Vivo Model mouse xenograft model Mus musculus
Mechanism Description The missense mutation p.V600E (c.1799T>A) in gene BRAF cause the sensitivity of SB590885 by aberration of the drug's therapeutic target
SBI-0640726
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [72]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug SBI-0640726
Experimental Note Identified from the Human Clinical Data
SBI-0640756
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [72]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug SBI-0640756
Experimental Note Identified from the Human Clinical Data
SBI-755199
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [72]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug SBI-755199
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model Melanoma cells Skin Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Crystal violet staining assay
SEL201
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: Mast/stem cell growth factor receptor Kit (KIT) [87]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L576P (c.1727T>C)
Sensitive Drug SEL201
Experimental Note Identified from the Human Clinical Data
In Vitro Model MM61 cells N.A. . N.A.
MM111 cells N.A. . N.A.
M230 cells Skin Homo sapiens (Human) CVCL_D749
LND1 cells Skin Homo sapiens (Human) CVCL_J076
HBL cells Skin Homo sapiens (Human) CVCL_J075
In Vivo Model CD-1 mouse PDX model Mus musculus
Experiment for
Drug Resistance
SRB assay; Crystal violet staining assay; Promega assay
Mechanism Description c-KIT stimulates MAP kinase-interacting serine/threonine kinases 1 and 2 (MNK1/2), which phosphorylate eukaryotic translation initiation factor 4E (eIF4E) and render it oncogenic. Depletion of MNK1/2 in melanoma cells with oncogenic C-KIT inhibited cell migration and mRNA translation of the transcriptional repressor SNAI1 and the cell cycle gene CCNE1. This suggested that blocking MNK1/2 activity may inhibit tumor progression, at least in part, by blocking translation initiation of mRNAs encoding cell migration proteins.
Key Molecule: Mast/stem cell growth factor receptor Kit (KIT) [87]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.D820Y (c.2458G>T)
Sensitive Drug SEL201
Experimental Note Identified from the Human Clinical Data
In Vitro Model MM61 cells N.A. . N.A.
MM111 cells N.A. . N.A.
M230 cells Skin Homo sapiens (Human) CVCL_D749
LND1 cells Skin Homo sapiens (Human) CVCL_J076
HBL cells Skin Homo sapiens (Human) CVCL_J075
In Vivo Model CD-1 mouse PDX model Mus musculus
Experiment for
Drug Resistance
SRB assay; Crystal violet staining assay; Promega assay
Mechanism Description c-KIT stimulates MAP kinase-interacting serine/threonine kinases 1 and 2 (MNK1/2), which phosphorylate eukaryotic translation initiation factor 4E (eIF4E) and render it oncogenic. Depletion of MNK1/2 in melanoma cells with oncogenic C-KIT inhibited cell migration and mRNA translation of the transcriptional repressor SNAI1 and the cell cycle gene CCNE1. This suggested that blocking MNK1/2 activity may inhibit tumor progression, at least in part, by blocking translation initiation of mRNAs encoding cell migration proteins.
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
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [88]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Selumetinib/Dactolisib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model BRAF-mutant melanoma cells N.A. Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Selumetinib/Vemurafenib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [73]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600X (c.1798_1800)
Sensitive Drug Selumetinib/Vemurafenib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Melanoma cells Skin Homo sapiens (Human) N.A.
WM cells N.A. Homo sapiens (Human) N.A.
SK cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CellTiter-Glo assay; Fluorescent microscopy assay; Apoptosis analysis
TAK-632
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [82]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G466E (c.1397G>A)
Sensitive Drug TAK-632
Experimental Note Identified from the Human Clinical Data
In Vitro Model SW1736 cells Thyroid Homo sapiens (Human) CVCL_3883
8505C cells Thyroid Homo sapiens (Human) CVCL_1054
Hth104 cells Thyroid gland Homo sapiens (Human) CVCL_A427
In Vivo Model mouse xenograft model Mus musculus
Mechanism Description The missense mutation p.G466E (c.1397G>A) in gene BRAF cause the sensitivity of TAK-632 by aberration of the drug's therapeutic target
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [82]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600R (c.1798_1799delGTinsAG)
Sensitive Drug TAK-632
Experimental Note Identified from the Human Clinical Data
In Vitro Model SW1736 cells Thyroid Homo sapiens (Human) CVCL_3883
8505C cells Thyroid Homo sapiens (Human) CVCL_1054
Hth104 cells Thyroid gland Homo sapiens (Human) CVCL_A427
In Vivo Model mouse xenograft model Mus musculus
Mechanism Description The missense mutation p.V600R (c.1798_1799delGTinsAG) in gene BRAF cause the sensitivity of TAK-632 by aberration of the drug's therapeutic target
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [82]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug TAK-632
Experimental Note Identified from the Human Clinical Data
In Vitro Model SW1736 cells Thyroid Homo sapiens (Human) CVCL_3883
8505C cells Thyroid Homo sapiens (Human) CVCL_1054
Hth104 cells Thyroid gland Homo sapiens (Human) CVCL_A427
In Vivo Model mouse xenograft model Mus musculus
Mechanism Description The missense mutation p.V600E (c.1799T>A) in gene BRAF cause the sensitivity of TAK-632 by aberration of the drug's therapeutic target
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [82]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600D (c.1799_1800delTGinsAC)
Sensitive Drug TAK-632
Experimental Note Identified from the Human Clinical Data
In Vitro Model SW1736 cells Thyroid Homo sapiens (Human) CVCL_3883
8505C cells Thyroid Homo sapiens (Human) CVCL_1054
Hth104 cells Thyroid gland Homo sapiens (Human) CVCL_A427
In Vivo Model mouse xenograft model Mus musculus
Mechanism Description The missense mutation p.V600D (c.1799_1800delTGinsAC) in gene BRAF cause the sensitivity of TAK-632 by aberration of the drug's therapeutic target
Unspecified VEGFR2 antibody
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: VEGF-2 receptor (KDR) [89]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q472H (c.1416A>T)
Sensitive Drug Unspecified VEGFR2 antibody
Experimental Note Identified from the Human Clinical Data
In Vitro Model WM3248 cells Skin Homo sapiens (Human) CVCL_6798
WM1575 cells Skin Homo sapiens (Human) CVCL_L027
Experiment for
Molecule Alteration
Targeted NGS assay
Experiment for
Drug Resistance
Proliferation assay; Invasion assay
Investigative Drug(s)
17 drug(s) in total
Click to Show/Hide the Full List of Drugs
AZ628
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [82]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G466E (c.1397G>A)
Sensitive Drug AZ628
Experimental Note Identified from the Human Clinical Data
In Vitro Model SW1736 cells Thyroid Homo sapiens (Human) CVCL_3883
8505C cells Thyroid Homo sapiens (Human) CVCL_1054
Hth104 cells Thyroid gland Homo sapiens (Human) CVCL_A427
In Vivo Model mouse xenograft model Mus musculus
Mechanism Description The missense mutation p.G466E (c.1397G>A) in gene BRAF cause the sensitivity of AZ628 by aberration of the drug's therapeutic target
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [82]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600R (c.1798_1799delGTinsAG)
Sensitive Drug AZ628
Experimental Note Identified from the Human Clinical Data
In Vitro Model SW1736 cells Thyroid Homo sapiens (Human) CVCL_3883
8505C cells Thyroid Homo sapiens (Human) CVCL_1054
Hth104 cells Thyroid gland Homo sapiens (Human) CVCL_A427
In Vivo Model mouse xenograft model Mus musculus
Mechanism Description The missense mutation p.V600R (c.1798_1799delGTinsAG) in gene BRAF cause the sensitivity of AZ628 by aberration of the drug's therapeutic target
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [82]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug AZ628
Experimental Note Identified from the Human Clinical Data
In Vitro Model SW1736 cells Thyroid Homo sapiens (Human) CVCL_3883
8505C cells Thyroid Homo sapiens (Human) CVCL_1054
Hth104 cells Thyroid gland Homo sapiens (Human) CVCL_A427
In Vivo Model mouse xenograft model Mus musculus
Mechanism Description The missense mutation p.V600E (c.1799T>A) in gene BRAF cause the sensitivity of AZ628 by aberration of the drug's therapeutic target
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [82]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600D (c.1799_1800delTGinsAC)
Sensitive Drug AZ628
Experimental Note Identified from the Human Clinical Data
In Vitro Model SW1736 cells Thyroid Homo sapiens (Human) CVCL_3883
8505C cells Thyroid Homo sapiens (Human) CVCL_1054
Hth104 cells Thyroid gland Homo sapiens (Human) CVCL_A427
In Vivo Model mouse xenograft model Mus musculus
Mechanism Description The missense mutation p.V600D (c.1799_1800delTGinsAC) in gene BRAF cause the sensitivity of AZ628 by aberration of the drug's therapeutic target
Bevacizumab/Sorafenib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Mast/stem cell growth factor receptor Kit (KIT) [90]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L576P (c.1727T>C)
Sensitive Drug Bevacizumab/Sorafenib
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
ICH assay
Experiment for
Drug Resistance
Radiologic assessment assay
Binimetinib/Encorafenib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [91]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Binimetinib/Encorafenib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [91]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600K (c.1798_1799delGTinsAA)
Sensitive Drug Binimetinib/Encorafenib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [91]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600K (c.1798_1799delGTinsAA)
Sensitive Drug Binimetinib/Encorafenib
Experimental Note Identified from the Human Clinical Data
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [1]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L597S (c.1789_1790delCTinsTC)
Sensitive Drug Binimetinib/Encorafenib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
In Vitro Model Skin sample .
In Vivo Model Mouse PDX model Mus musculus
Experiment for
Drug Resistance
Crystal violet staining assay
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [92]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600X (c.1798_1799)
Sensitive Drug Binimetinib/Encorafenib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
In Vitro Model Skin sample .
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [92]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600X (c.1798_1799)
Sensitive Drug Binimetinib/Encorafenib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
In Vitro Model Skin sample .
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
Key Molecule: GTPase Nras (NRAS) [93]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Mutation
.
Resistant Drug Braf inhibitor
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.
Key Molecule: Homeobox protein Hox-D8 (HOXD8) [93]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Mutation
.
Resistant Drug Braf inhibitor
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 Earlier, it was demonstrated that RAC1codon29 mutant melanomas are resistant to this therapy. Later, it was found that a rare genetic alteration, the mutation of HOXD8, can also be the cause of primary resistance to BRAF inhibitors.
BRAF/MEK inhibitors
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: MAPK/ERK kinase 2 (MEK2) [94]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.Q60P (c.179A>C)
Resistant Drug BRAF/MEK inhibitors
Experimental Note Identified from the Human Clinical Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CellTiter96 AQueous assay
Celastrol
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-217 [21]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Celastrol
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Malme3M cells Skin Homo sapiens (Human) CVCL_1438
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-326, which forms a negative feedback regulatory loop with HDAC3, regulates the invasion and the metastatic potential of cancer cells and tumor-induced angiogenesis in response to anti-cancer drugs. miR-200b, miR-217, and miR-335, which form a positive feedback loop with HDAC3, confer sensitivity to anti-cancer drugs. We show that CAGE, reported to form a feedback loop with miR-200b, serves as a downstream target of HDAC3 and miR-326. In this study, we show that the regulation of the miR-326/HDAC3 axis can be employed for the development of anti-cancer therapeutics.
Key Molecule: hsa-mir-335 [21]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Celastrol
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Malme3M cells Skin Homo sapiens (Human) CVCL_1438
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-326, which forms a negative feedback regulatory loop with HDAC3, regulates the invasion and the metastatic potential of cancer cells and tumor-induced angiogenesis in response to anti-cancer drugs. miR-200b, miR-217, and miR-335, which form a positive feedback loop with HDAC3, confer sensitivity to anti-cancer drugs. We show that CAGE, reported to form a feedback loop with miR-200b, serves as a downstream target of HDAC3 and miR-326. In this study, we show that the regulation of the miR-326/HDAC3 axis can be employed for the development of anti-cancer therapeutics.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: hsa-mir-200b [21]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Celastrol
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Malme3M cells Skin Homo sapiens (Human) CVCL_1438
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-326, which forms a negative feedback regulatory loop with HDAC3, regulates the invasion and the metastatic potential of cancer cells and tumor-induced angiogenesis in response to anti-cancer drugs. miR-200b, miR-217, and miR-335, which form a positive feedback loop with HDAC3, confer sensitivity to anti-cancer drugs. We show that CAGE, reported to form a feedback loop with miR-200b, serves as a downstream target of HDAC3 and miR-326. In this study, we show that the regulation of the miR-326/HDAC3 axis can be employed for the development of anti-cancer therapeutics.
Key Molecule: hsa-miR-326 [21]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Celastrol
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Malme3M cells Skin Homo sapiens (Human) CVCL_1438
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-326, which forms a negative feedback regulatory loop with HDAC3, regulates the invasion and the metastatic potential of cancer cells and tumor-induced angiogenesis in response to anti-cancer drugs. miR-200b, miR-217, and miR-335, which form a positive feedback loop with HDAC3, confer sensitivity to anti-cancer drugs. We show that CAGE, reported to form a feedback loop with miR-200b, serves as a downstream target of HDAC3 and miR-326. In this study, we show that the regulation of the miR-326/HDAC3 axis can be employed for the development of anti-cancer therapeutics.
CI-1040
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: MAPK/ERK kinase 1 (MEK1) [49]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.I103N (c.308T>A)
Resistant Drug CI-1040
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
293T cells Breast Homo sapiens (Human) CVCL_0063
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Colony formation assay
Mechanism Description The missense mutation p.I103N (c.308T>A) in gene MAP2K1 cause the resistance of CI-1040 by aberration of the drug's therapeutic target
Key Molecule: MAPK/ERK kinase 1 (MEK1) [49]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L115P (c.344T>C)
Resistant Drug CI-1040
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
293T cells Breast Homo sapiens (Human) CVCL_0063
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Colony formation assay
Mechanism Description The missense mutation p.L115P (c.344T>C) in gene MAP2K1 cause the resistance of CI-1040 by aberration of the drug's therapeutic target
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: GTPase Hras (HRAS) [76]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G12V (c.35G>T)
Sensitive Drug CI-1040
Experimental Note Identified from the Human Clinical Data
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
A375 cells Skin Homo sapiens (Human) CVCL_0132
In Vivo Model Female athymic nu/nu mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Caspase-Glo 3/7 luminogenic assay
Cisplatinum
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: H19, imprinted maternally expressed transcript (H19) [95]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Up-regulation
Interaction
Resistant Drug Cisplatinum
Experimental Note Identified from the Human Clinical Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
M8 cells Skin Homo sapiens (Human) N.A.
WM35 cells Skin Homo sapiens (Human) CVCL_0580
SK-MEL-2 cells Skin Homo sapiens (Human) CVCL_0069
A2508 cells N.A. . N.A.
Experiment for
Molecule Alteration
Knockdown assay; qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Down-regulation of LncRNA H19 sensitizes melanoma cells to cisplatin by regulating the miR-18b/IGF1 axis.
Cobimetinib/Vemurafenib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [96]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600X (c.1798_1800)
Sensitive Drug Cobimetinib/Vemurafenib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
Gardiquimod
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [97]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Resistant Drug Gardiquimod
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Resazurin Cell Viability Assay
Mechanism Description Imidazoquinolines IMQ, RSQ, and GDQ are substrates for P-gp and begins to elucidate differences in their trafficking in cancer cells as a consequence of acquired drug resistance. We believe this work that begins to examine imidazoquinoline trafficking will prove useful in the future rational design of immunotherapeutics with enhanced susceptibility to P-gp efflux that enable increased bioavailability, in MDR cancers.
Imiquimod
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [97]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Resistant Drug Imiquimod
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Resazurin Cell Viability Assay
Mechanism Description Imidazoquinolines IMQ, RSQ, and GDQ are substrates for P-gp and begins to elucidate differences in their trafficking in cancer cells as a consequence of acquired drug resistance. We believe this work that begins to examine imidazoquinoline trafficking will prove useful in the future rational design of immunotherapeutics with enhanced susceptibility to P-gp efflux that enable increased bioavailability, in MDR cancers.
MEK inhibitors
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: MAPK/ERK kinase 1 (MEK1) [49]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.P124L (c.371C>T)
Resistant Drug MEK inhibitors
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
293T cells Breast Homo sapiens (Human) CVCL_0063
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Colony formation assay
Mechanism Description The missense mutation p.P124L (c.371C>T) in gene MAP2K1 cause the resistance of MEK inhibitors by aberration of the drug's therapeutic target
Key Molecule: MAPK/ERK kinase 2 (MEK2) [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.N126D (c.376A>G)
Resistant Drug MEK inhibitors
Experimental Note Identified from the Human Clinical Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
A2058 cells Skin Homo sapiens (Human) CVCL_1059
WM2664 cells Skin Homo sapiens (Human) CVCL_2765
SkMEL28 cells Skin Homo sapiens (Human) CVCL_0526
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CellTiter-Glo assay
Key Molecule: MAPK/ERK kinase 2 (MEK2) [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V35M (c.103G>A)
Resistant Drug MEK inhibitors
Experimental Note Identified from the Human Clinical Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
A2058 cells Skin Homo sapiens (Human) CVCL_1059
WM2664 cells Skin Homo sapiens (Human) CVCL_2765
SkMEL28 cells Skin Homo sapiens (Human) CVCL_0526
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CellTiter-Glo assay
Key Molecule: MAPK/ERK kinase 2 (MEK2) [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L46F (c.136C>T)
Resistant Drug MEK inhibitors
Experimental Note Identified from the Human Clinical Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
A2058 cells Skin Homo sapiens (Human) CVCL_1059
WM2664 cells Skin Homo sapiens (Human) CVCL_2765
SkMEL28 cells Skin Homo sapiens (Human) CVCL_0526
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CellTiter-Glo assay
Key Molecule: MAPK/ERK kinase 2 (MEK2) [9]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.C125S (c.373T>A)
Resistant Drug MEK inhibitors
Experimental Note Identified from the Human Clinical Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
A2058 cells Skin Homo sapiens (Human) CVCL_1059
WM2664 cells Skin Homo sapiens (Human) CVCL_2765
SkMEL28 cells Skin Homo sapiens (Human) CVCL_0526
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CellTiter-Glo assay
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: MAPK/ERK kinase 1 (MEK1) [98]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.K57N (c.171G>C)
Resistant Drug MEK inhibitors
Experimental Note Identified from the Human Clinical Data
In Vitro Model Skin .
Experiment for
Molecule Alteration
Immunoblotting analysis
Mechanism Description The missense mutation p.K57N (c.171G>C) in gene MAP2K1 cause the resistance of MEK inhibitors by unusual activation of pro-survival pathway
Key Molecule: MAPK/ERK kinase 1 (MEK1) [99]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.C121S (c.361T>A)
Resistant Drug MEK inhibitors
Experimental Note Identified from the Human Clinical Data
In Vitro Model Human melanoma tissue .
Mechanism Description The missense mutation p.C121S (c.361T>A) in gene MAP2K1 cause the resistance of MEK inhibitors by unusual activation of pro-survival pathway
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [30]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L597R (c.1790T>G)
Sensitive Drug MEK inhibitors
Experimental Note Identified from the Human Clinical Data
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [31]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.D594A (c.1781A>C)
Sensitive Drug MEK inhibitors
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Experiment for
Molecule Alteration
Immunoblotting analysis
Mechanism Description The missense mutation p.D594A (c.1781A>C) in gene BRAF cause the sensitivity of MEK inhibitors by unusual activation of pro-survival pathway
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [31]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.K483M (c.1448_1449delAAinsTG)
Sensitive Drug MEK inhibitors
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
Experiment for
Molecule Alteration
Immunoblotting analysis
Mechanism Description The missense mutation p.K483M (c.1448_1449delAAinsTG) in gene BRAF cause the sensitivity of MEK inhibitors by unusual activation of pro-survival pathway
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [29]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.K601R (c.1802A>G)
Sensitive Drug MEK inhibitors
Experimental Note Identified from the Human Clinical Data
In Vitro Model Skin .
Mechanism Description The missense mutation p.K601R (c.1802A>G) in gene BRAF cause the sensitivity of MEK inhibitors by unusual activation of pro-survival pathway
Orthocresol
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: Growth arrest specific 5 (GAS5) [100]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Orthocresol
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model A375 cells Skin Homo sapiens (Human) CVCL_0132
A431 cells Skin Homo sapiens (Human) CVCL_0037
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description 2-O-Methylmagnolol upregulates the long non-coding RNA, GAS5, and enhances apoptosis in skin cancer cells. Overexpression of LncRNA GAS5 inhibited cell proliferation and promoted cell apoptosis in skin cancer cells.
Resiquimod
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [97]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Expression
Up-regulation
Resistant Drug Resiquimod
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Resazurin Cell Viability Assay
Mechanism Description Imidazoquinolines IMQ, RSQ, and GDQ are substrates for P-gp and begins to elucidate differences in their trafficking in cancer cells as a consequence of acquired drug resistance. We believe this work that begins to examine imidazoquinoline trafficking will prove useful in the future rational design of immunotherapeutics with enhanced susceptibility to P-gp efflux that enable increased bioavailability, in MDR cancers.
SCH772984
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [76]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug SCH772984
Experimental Note Identified from the Human Clinical Data
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
A375 cells Skin Homo sapiens (Human) CVCL_0132
In Vivo Model Female athymic nu/nu mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Caspase-Glo 3/7 luminogenic assay
TAK-733
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [28]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L597S (c.1789_1790delCTinsTC)
Sensitive Drug TAK-733
Experimental Note Identified from the Human Clinical Data
In Vitro Model Melanoma thyroid metastasis .
Experiment for
Molecule Alteration
Western blotting analysis
Mechanism Description The missense mutation p.L597S (c.1789_1790delCTinsTC) in gene BRAF cause the sensitivity of TAK-733 by unusual activation of pro-survival pathway
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [28]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.L597S (c.1789_1790delCTinsTC)
Sensitive Drug TAK-733
Experimental Note Identified from the Human Clinical Data
In Vitro Model Melanoma thyroid metastasis .
Experiment for
Molecule Alteration
Western blotting analysis
Mechanism Description The missense mutation p.L597S (c.1789_1790delCTinsTC) in gene BRAF cause the sensitivity of TAK-733 by unusual activation of pro-survival pathway
U0126
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [101]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.G469E (c.1406G>A)
Resistant Drug U0126
Experimental Note Identified from the Human Clinical Data
In Vitro Model Human melanoma tissue .
In Vivo Model (SCID) CB-17 mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The missense mutation p.G469E (c.1406G>A) in gene BRAF cause the resistance of U0126 by unusual activation of pro-survival pathway
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [101]
Resistant Disease Melanoma [ICD-11: 2C30.0]
Molecule Alteration Missense mutation
p.D594G (c.1781A>G)
Resistant Drug U0126
Experimental Note Identified from the Human Clinical Data
In Vitro Model Human melanoma tissue .
In Vivo Model (SCID) CB-17 mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The missense mutation p.D594G (c.1781A>G) in gene BRAF cause the resistance of U0126 by unusual activation of pro-survival pathway
References
Ref 1 Dual MAPK Inhibition Is an Effective Therapeutic Strategy for a Subset of Class II BRAF Mutant MelanomasClin Cancer Res. 2018 Dec 15;24(24):6483-6494. doi: 10.1158/1078-0432.CCR-17-3384. Epub 2018 Jun 14.
Ref 2 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 3 Expression of MicroRNA-301a and its Functional Roles in Malignant Melanoma. Cell Physiol Biochem. 2016;40(1-2):230-244. doi: 10.1159/000452540. Epub 2016 Nov 18.
Ref 4 Methylation-Mediated Silencing of MicroRNA-211 Decreases the Sensitivity of Melanoma Cells to Cisplatin. Med Sci Monit. 2019 Mar 1;25:1590-1599. doi: 10.12659/MSM.911862.
Ref 5 MiR-30a-5p confers cisplatin resistance by regulating IGF1R expression in melanoma cells. BMC Cancer. 2018 Apr 11;18(1):404. doi: 10.1186/s12885-018-4233-9.
Ref 6 MicroRNA-488-3p sensitizes malignant melanoma cells to cisplatin by targeting PRKDC. Cell Biol Int. 2017 Jun;41(6):622-629. doi: 10.1002/cbin.10765. Epub 2017 Apr 7.
Ref 7 miR-126-3p down-regulation contributes to dabrafenib acquired resistance in melanoma by up-regulating ADAM9 and VEGF-A. J Exp Clin Cancer Res. 2019 Jun 21;38(1):272. doi: 10.1186/s13046-019-1238-4.
Ref 8 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 9 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 10 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 11 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 12 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 13 Co-clinical assessment identifies patterns of BRAF inhibitor resistance in melanoma. J Clin Invest. 2015 Apr;125(4):1459-70. doi: 10.1172/JCI78954. Epub 2015 Feb 23.
Ref 14 Improved survival with MEK inhibition in BRAF-mutated melanomaN Engl J Med. 2012 Jul 12;367(2):107-14. doi: 10.1056/NEJMoa1203421. Epub 2012 Jun 4.
Ref 15 U.S. Food and Drug Administration.
Ref 16 Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutationsN Engl J Med. 2012 Nov 1;367(18):1694-703. doi: 10.1056/NEJMoa1210093. Epub 2012 Sep 29.
Ref 17 Overall Survival and Durable Responses in Patients With BRAF V600-Mutant Metastatic Melanoma Receiving Dabrafenib Combined With TrametinibJ Clin Oncol. 2016 Mar 10;34(8):871-8. doi: 10.1200/JCO.2015.62.9345. Epub 2016 Jan 25.
Ref 18 The miR-31-SOX10 axis regulates tumor growth and chemotherapy resistance of melanoma via PI3K/AKT pathway. Biochem Biophys Res Commun. 2018 Sep 18;503(4):2451-2458. doi: 10.1016/j.bbrc.2018.06.175. Epub 2018 Jul 3.
Ref 19 Hypoxia-induced miR-424 decreases tumor sensitivity to chemotherapy by inhibiting apoptosis. Cell Death Dis. 2014 Jun 26;5(6):e1301. doi: 10.1038/cddis.2014.240.
Ref 20 Distinct Molecular Profiles and Immunotherapy Treatment Outcomes of V600E and V600K BRAF-Mutant MelanomaClin Cancer Res. 2019 Feb 15;25(4):1272-1279. doi: 10.1158/1078-0432.CCR-18-1680. Epub 2019 Jan 10.
Ref 21 miR-326-histone deacetylase-3 feedback loop regulates the invasion and tumorigenic and angiogenic response to anti-cancer drugs. J Biol Chem. 2014 Oct 3;289(40):28019-39. doi: 10.1074/jbc.M114.578229. Epub 2014 Aug 19.
Ref 22 miR-335 Targets SIAH2 and Confers Sensitivity to Anti-Cancer Drugs by Increasing the Expression of HDAC3. Mol Cells. 2015 Jun;38(6):562-72. doi: 10.14348/molcells.2015.0051. Epub 2015 May 22.
Ref 23 Loss of CDKN2A expression is a frequent event in primary invasive melanoma and correlates with sensitivity to the CDK4/6 inhibitor PD0332991 in melanoma cell linesPigment Cell Melanoma Res. 2014 Jul;27(4):590-600. doi: 10.1111/pcmr.12228. Epub 2014 Mar 6.
Ref 24 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 25 Sensitization of melanoma cells to temozolomide by overexpression of microRNA 203 through direct targeting of glutaminase-mediated glutamine metabolism. Clin Exp Dermatol. 2017 Aug;42(6):614-621. doi: 10.1111/ced.13119. Epub 2017 Jun 9.
Ref 26 Tumours with class 3 BRAF mutants are sensitive to the inhibition of activated RASNature. 2017 Aug 10;548(7666):234-238. doi: 10.1038/nature23291. Epub 2017 Aug 2.
Ref 27 Activity of trametinib in K601E and L597Q BRAF mutation-positive metastatic melanomaMelanoma Res. 2014 Oct;24(5):504-8. doi: 10.1097/CMR.0000000000000099.
Ref 28 BRAF(L597) mutations in melanoma are associated with sensitivity to MEK inhibitorsCancer Discov. 2012 Sep;2(9):791-7. doi: 10.1158/2159-8290.CD-12-0097. Epub 2012 Jul 13.
Ref 29 Phase II study of the MEK1/MEK2 inhibitor Trametinib in patients with metastatic BRAF-mutant cutaneous melanoma previously treated with or without a BRAF inhibitorJ Clin Oncol. 2013 Feb 1;31(4):482-9. doi: 10.1200/JCO.2012.43.5966. Epub 2012 Dec 17.
Ref 30 Activity of the oral MEK inhibitor trametinib in patients with advanced melanoma: a phase 1 dose-escalation trialLancet Oncol. 2012 Aug;13(8):782-9. doi: 10.1016/S1470-2045(12)70269-3. Epub 2012 Jul 16.
Ref 31 Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAFCell. 2010 Jan 22;140(2):209-21. doi: 10.1016/j.cell.2009.12.040.
Ref 32 Metastatic BRAF K601E-mutated melanoma reaches complete response to MEK inhibitor trametinib administered for over 36 monthsExp Hematol Oncol. 2017 Mar 21;6:6. doi: 10.1186/s40164-017-0067-4. eCollection 2017.
Ref 33 Improved overall survival in melanoma with combined dabrafenib and trametinibN Engl J Med. 2015 Jan 1;372(1):30-9. doi: 10.1056/NEJMoa1412690. Epub 2014 Nov 16.
Ref 34 Oncogene-dependent sloppiness in mRNA translation .Mol Cell. 2021 Nov 18;81(22):4709-4721.e9. doi: 10.1016/j.molcel.2021.09.002. Epub 2021 Sep 24. 10.1016/j.molcel.2021.09.002
Ref 35 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 36 miR-204-5p and miR-211-5p Contribute to BRAF Inhibitor Resistance in Melanoma. Cancer Res. 2018 Feb 15;78(4):1017-1030. doi: 10.1158/0008-5472.CAN-17-1318. Epub 2017 Dec 11.
Ref 37 Detailed imaging and genetic analysis reveal a secondary BRAF(L505H) resistance mutation and extensive intrapatient heterogeneity in metastatic BRAF mutant melanoma patients treated with vemurafenib. Pigment Cell Melanoma Res. 2015 May;28(3):318-23. doi: 10.1111/pcmr.12347. Epub 2015 Jan 7.
Ref 38 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 39 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 40 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 41 miR-216b enhances the efficacy of vemurafenib by targeting Beclin-1, UVRAG and ATG5 in melanoma. Cell Signal. 2018 Jan;42:30-43. doi: 10.1016/j.cellsig.2017.09.024. Epub 2017 Oct 2.
Ref 42 miR-7 reverses the resistance to BRAFi in melanoma by targeting EGFR/IGF-1R/CRAF and inhibiting the MAPK and PI3K/AKT signaling pathways. Oncotarget. 2016 Aug 16;7(33):53558-53570. doi: 10.18632/oncotarget.10669.
Ref 43 Overcoming melanoma resistance to vemurafenib by targeting CCL2-induced miR-34a, miR-100 and miR-125b. Oncotarget. 2016 Jan 26;7(4):4428-41. doi: 10.18632/oncotarget.6599.
Ref 44 Analysis of mTOR Gene Aberrations in Melanoma Patients and Evaluation of Their Sensitivity to PI3K-AKT-mTOR Pathway InhibitorsClin Cancer Res. 2016 Feb 15;22(4):1018-27. doi: 10.1158/1078-0432.CCR-15-1110. Epub 2015 Oct 21.
Ref 45 Large-scale analysis of PDGFRA mutations in melanomas and evaluation of their sensitivity to tyrosine kinase inhibitors imatinib and crenolanibClin Cancer Res. 2013 Dec 15;19(24):6935-42. doi: 10.1158/1078-0432.CCR-13-1266. Epub 2013 Oct 16.
Ref 46 Overcoming acquired BRAF inhibitor resistance in melanoma via targeted inhibition of Hsp90 with ganetespibMol Cancer Ther. 2014 Feb;13(2):353-63. doi: 10.1158/1535-7163.MCT-13-0481. Epub 2014 Jan 7.
Ref 47 Multicenter phase I trial of the mitogen-activated protein kinase 1/2 inhibitor BAY 86-9766 in patients with advanced cancerClin Cancer Res. 2013 Mar 1;19(5):1232-43. doi: 10.1158/1078-0432.CCR-12-3529. Epub 2013 Feb 22.
Ref 48 RDEA119/BAY 869766: a potent, selective, allosteric inhibitor of MEK1/2 for the treatment of cancerCancer Res. 2009 Sep 1;69(17):6839-47. doi: 10.1158/0008-5472.CAN-09-0679. Epub 2009 Aug 25.
Ref 49 MEK1 mutations confer resistance to MEK and B-RAF inhibitionProc Natl Acad Sci U S A. 2009 Dec 1;106(48):20411-6. doi: 10.1073/pnas.0905833106. Epub 2009 Nov 13.
Ref 50 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 51 Turk J Haematol. 2016 Mar 5;33(1):41-7. doi: 10.4274/tjh.2014.0010. Epub 2014 May 21.
Ref 52 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 53 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 54 Combination of vemurafenib and cobimetinib in patients with advanced BRAF(V600)-mutated melanoma: a phase 1b studyLancet Oncol. 2014 Aug;15(9):954-65. doi: 10.1016/S1470-2045(14)70301-8. Epub 2014 Jul 15.
Ref 55 Novel ATP-competitive MEK inhibitor E6201 is effective against vemurafenib-resistant melanoma harboring the MEK1-C121S mutation in a preclinical modelMol Cancer Ther. 2014 Apr;13(4):823-32. doi: 10.1158/1535-7163.MCT-13-0667. Epub 2014 Jan 21.
Ref 56 First-in-human phase I study of pictilisib (GDC-0941), a potent pan-class I phosphatidylinositol-3-kinase (PI3K) inhibitor, in patients with advanced solid tumorsClin Cancer Res. 2015 Jan 1;21(1):77-86. doi: 10.1158/1078-0432.CCR-14-0947. Epub 2014 Nov 4.
Ref 57 A first-in-human phase I, multicenter, open-label, dose-escalation study of the oral RAF/VEGFR-2 inhibitor (RAF265) in locally advanced or metastatic melanoma independent from BRAF mutation statusCancer Med. 2017 Aug;6(8):1904-1914. doi: 10.1002/cam4.1140. Epub 2017 Jul 18.
Ref 58 ERK Signal Suppression and Sensitivity to CH5183284/Debio 1347, a Selective FGFR InhibitorMol Cancer Ther. 2015 Dec;14(12):2831-9. doi: 10.1158/1535-7163.MCT-15-0497. Epub 2015 Oct 5.
Ref 59 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 60 Targeting the MAPK Signaling Pathway in Cancer: Promising Preclinical Activity with the Novel Selective ERK1/2 Inhibitor BVD-523 (Ulixertinib)Mol Cancer Ther. 2017 Nov;16(11):2351-2363. doi: 10.1158/1535-7163.MCT-17-0456. Epub 2017 Sep 22.
Ref 61 Effects of AKT inhibitor therapy in response and resistance to BRAF inhibition in melanomaMol Cancer. 2014 Apr 16;13:83. doi: 10.1186/1476-4598-13-83.
Ref 62 CEP-32496: a novel orally active BRAF(V600E) inhibitor with selective cellular and in vivo antitumor activityMol Cancer Ther. 2012 Apr;11(4):930-41. doi: 10.1158/1535-7163.MCT-11-0645. Epub 2012 Feb 7.
Ref 63 BGB-283, a Novel RAF Kinase and EGFR Inhibitor, Displays Potent Antitumor Activity in BRAF-Mutated Colorectal CancersMol Cancer Ther. 2015 Oct;14(10):2187-97. doi: 10.1158/1535-7163.MCT-15-0262. Epub 2015 Jul 24.
Ref 64 The Novel ATP-Competitive MEK/Aurora Kinase Inhibitor BI-847325 Overcomes Acquired BRAF Inhibitor Resistance through Suppression of Mcl-1 and MEK ExpressionMol Cancer Ther. 2015 Jun;14(6):1354-64. doi: 10.1158/1535-7163.MCT-14-0832. Epub 2015 Apr 14.
Ref 65 Somatic Copy Number Amplification and Hyperactivating Somatic Mutations of EZH2 Correlate With DNA Methylation and Drive Epigenetic Silencing of Genes Involved in Tumor Suppression and Immune Responses in MelanomaNeoplasia. 2016 Feb;18(2):121-32. doi: 10.1016/j.neo.2016.01.003.
Ref 66 Targeting activating mutations of EZH2 leads to potent cell growth inhibition in human melanoma by derepression of tumor suppressor genesOncotarget. 2015 Sep 29;6(29):27023-36. doi: 10.18632/oncotarget.4809.
Ref 67 Combinations of BRAF, MEK, and PI3K/mTOR inhibitors overcome acquired resistance to the BRAF inhibitor GSK2118436 dabrafenib, mediated by NRAS or MEK mutationsMol Cancer Ther. 2012 Apr;11(4):909-20. doi: 10.1158/1535-7163.MCT-11-0989. Epub 2012 Mar 2.
Ref 68 Whole-genome sequencing reveals complex mechanisms of intrinsic resistance to BRAF inhibitionAnn Oncol. 2014 May;25(5):959-67. doi: 10.1093/annonc/mdu049. Epub 2014 Feb 6.
Ref 69 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 70 Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activityProc Natl Acad Sci U S A. 2008 Feb 26;105(8):3041-6. doi: 10.1073/pnas.0711741105. Epub 2008 Feb 19.
Ref 71 Phase I expansion and pharmacodynamic study of the oral MEK inhibitor RO4987655 (CH4987655) in selected patients with advanced cancer with RAS-RAF mutationsClin Cancer Res. 2014 Aug 15;20(16):4251-61. doi: 10.1158/1078-0432.CCR-14-0341. Epub 2014 Jun 19.
Ref 72 SBI-0640756 Attenuates the Growth of Clinically Unresponsive Melanomas by Disrupting the eIF4F Translation Initiation ComplexCancer Res. 2015 Dec 15;75(24):5211-8. doi: 10.1158/0008-5472.CAN-15-0885. Epub 2015 Nov 24.
Ref 73 A novel AKT1 mutant amplifies an adaptive melanoma response to BRAF inhibitionCancer Discov. 2014 Jan;4(1):69-79. doi: 10.1158/2159-8290.CD-13-0279. Epub 2013 Nov 21.
Ref 74 Tumor necrosis factor-alpha blocks apoptosis in melanoma cells when BRAF signaling is inhibitedCancer Res. 2007 Jan 1;67(1):122-9. doi: 10.1158/0008-5472.CAN-06-1880.
Ref 75 Landscape of Targeted Anti-Cancer Drug Synergies in Melanoma Identifies a Novel BRAF-VEGFR/PDGFR Combination TreatmentPLoS One. 2015 Oct 13;10(10):e0140310. doi: 10.1371/journal.pone.0140310. eCollection 2015.
Ref 76 Small Molecule Inhibition of ERK Dimerization Prevents Tumorigenesis by RAS-ERK Pathway OncogenesCancer Cell. 2015 Aug 10;28(2):170-82. doi: 10.1016/j.ccell.2015.07.001.
Ref 77 A Novel Plant Sesquiterpene Lactone Derivative, DETD-35, Suppresses BRAFV600E Mutant Melanoma Growth and Overcomes Acquired Vemurafenib Resistance in MiceMol Cancer Ther. 2016 Jun;15(6):1163-76. doi: 10.1158/1535-7163.MCT-15-0973. Epub 2016 Apr 5.
Ref 78 EBI-907, a novel BRAF(V600E) inhibitor, has potent oral anti-tumor activity and a broad kinase selectivity profileCancer Biol Ther. 2016;17(2):199-207. doi: 10.1080/15384047.2016.1139231. Epub 2016 Jan 25.
Ref 79 Antitumor efficacy of the novel RAF inhibitor GDC-0879 is predicted by BRAFV600E mutational status and sustained extracellular signal-regulated kinase/mitogen-activated protein kinase pathway suppressionCancer Res. 2009 Apr 1;69(7):3042-51. doi: 10.1158/0008-5472.CAN-08-3563. Epub 2009 Mar 10.
Ref 80 Discovery of a novel pan-RAF inhibitor with potent anti-tumor activity in preclinical models of BRAF(V600E) mutant cancerLife Sci. 2017 Aug 15;183:37-44. doi: 10.1016/j.lfs.2017.06.021. Epub 2017 Jun 21.
Ref 81 An In Vivo Reporter to Quantitatively and Temporally Analyze the Effects of CDK4/6 Inhibitor-Based Therapies in MelanomaCancer Res. 2016 Sep 15;76(18):5455-66. doi: 10.1158/0008-5472.CAN-15-3384. Epub 2016 Aug 3.
Ref 82 An Integrated Model of RAF Inhibitor Action Predicts Inhibitor Activity against Oncogenic BRAF SignalingCancer Cell. 2016 Sep 12;30(3):485-498. doi: 10.1016/j.ccell.2016.06.024. Epub 2016 Aug 11.
Ref 83 RAF inhibitors that evade paradoxical MAPK pathway activationNature. 2015 Oct 22;526(7574):583-6. doi: 10.1038/nature14982. Epub 2015 Oct 14.
Ref 84 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 85 RAS nucleotide cycling underlies the SHP2 phosphatase dependence of mutant BRAF-, NF1- and RAS-driven cancersNat Cell Biol. 2018 Sep;20(9):1064-1073. doi: 10.1038/s41556-018-0169-1. Epub 2018 Aug 13.
Ref 86 Inhibiting EGF receptor or SRC family kinase signaling overcomes BRAF inhibitor resistance in melanomaCancer Discov. 2013 Feb;3(2):158-67. doi: 10.1158/2159-8290.CD-12-0386. Epub 2012 Dec 14.
Ref 87 MNK1/2 inhibition limits oncogenicity and metastasis of KIT-mutant melanomaJ Clin Invest. 2017 Nov 1;127(11):4179-4192. doi: 10.1172/JCI91258. Epub 2017 Oct 16.
Ref 88 Primary cross-resistance to BRAFV600E-, MEK1/2- and PI3K/mTOR-specific inhibitors in BRAF-mutant melanoma cells counteracted by dual pathway blockadeOncotarget. 2016 Jan 26;7(4):3947-65. doi: 10.18632/oncotarget.6600.
Ref 89 Identification of a Novel Pathogenic Germline KDR Variant in MelanomaClin Cancer Res. 2016 May 15;22(10):2377-85. doi: 10.1158/1078-0432.CCR-15-1811. Epub 2015 Dec 2.
Ref 90 Dual antiangiogenic inhibition: a phase I dose escalation and expansion trial targeting VEGF-A and VEGFR in patients with advanced solid tumorsInvest New Drugs. 2015 Feb;33(1):215-24. doi: 10.1007/s10637-014-0176-4. Epub 2014 Nov 4.
Ref 91 Cutaneous Melanoma, Version 2.2019, NCCN Clinical Practice Guidelines in OncologyJ Natl Compr Canc Netw. 2019 Apr 1;17(4):367-402. doi: 10.6004/jnccn.2019.0018.
Ref 92 Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): a multicentre, open-label, randomised phase 3 trialLancet Oncol. 2018 May;19(5):603-615. doi: 10.1016/S1470-2045(18)30142-6. Epub 2018 Mar 21.
Ref 93 Genetic progression of malignant melanoma. Cancer Metastasis Rev. 2016 Mar;35(1):93-107. doi: 10.1007/s10555-016-9613-5.
Ref 94 MAP kinase pathway alterations in BRAF-mutant melanoma patients with acquired resistance to combined RAF/MEK inhibitionCancer Discov. 2014 Jan;4(1):61-8. doi: 10.1158/2159-8290.CD-13-0631. Epub 2013 Nov 21.
Ref 95 Downregulation of lncRNA H19 sensitizes melanoma cells to cisplatin by regulating the miR-18b/IGF1 axisAnticancer Drugs. 2020 Jun;31(5):473-482. doi: 10.1097/CAD.0000000000000888.
Ref 96 Cobimetinib combined with vemurafenib in advanced BRAF(V600)-mutant melanoma (coBRIM): updated efficacy results from a randomised, double-blind, phase 3 trialLancet Oncol. 2016 Sep;17(9):1248-60. doi: 10.1016/S1470-2045(16)30122-X. Epub 2016 Jul 30.
Ref 97 Acquired Drug Resistance Enhances Imidazoquinoline Efflux by P-Glycoprotein .Pharmaceuticals (Basel). 2021 Dec 10;14(12):1292. doi: 10.3390/ph14121292. 10.3390/ph14121292
Ref 98 Phase II trial of MEK inhibitor selumetinib (AZD6244, ARRY-142886) in patients with BRAFV600E/K-mutated melanomaClin Cancer Res. 2013 Apr 15;19(8):2257-64. doi: 10.1158/1078-0432.CCR-12-3476. Epub 2013 Feb 26.
Ref 99 Dissecting therapeutic resistance to RAF inhibition in melanoma by tumor genomic profilingJ Clin Oncol. 2011 Aug 1;29(22):3085-96. doi: 10.1200/JCO.2010.33.2312. Epub 2011 Mar 7.
Ref 100 2-O-Methylmagnolol upregulates the long non-coding RNA, GAS5, and enhances apoptosis in skin cancer cells. Cell Death Dis. 2017 Mar 2;8(3):e2638. doi: 10.1038/cddis.2017.66.
Ref 101 CRAF inhibition induces apoptosis in melanoma cells with non-V600E BRAF mutationsOncogene. 2009 Jan 8;28(1):85-94. doi: 10.1038/onc.2008.362. Epub 2008 Sep 15.

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