Drug (ID: DG00340) and It's Reported Resistant Information
Name
Cetuximab
Synonyms
Erbitux; Cetuximab (genetical recombination); Erbitux (TN); Cetuximab (USAN/INN); Cetuximab (genetical recombination) (JAN); novel EGFR mAb inhibitors
    Click to Show/Hide
Indication
In total 1 Indication(s)
Colorectal cancer [ICD-11: 2B91]
Approved
[1]
Drug Resistance Disease(s)
Disease(s) with Clinically Reported Resistance for This Drug (2 diseases)
Colorectal cancer [ICD-11: 2B91]
[2]
Metastatic colorectal cancer [ICD-11: 2D85]
[3]
Disease(s) with Resistance Information Validated by in-vivo Model for This Drug (1 diseases)
Colorectal cancer [ICD-11: 2B91]
[4]
Disease(s) with Resistance Information Discovered by Cell Line Test for This Drug (3 diseases)
Colon cancer [ICD-11: 2B90]
[5]
Colorectal cancer [ICD-11: 2B91]
[6]
Head and neck cancer [ICD-11: 2D42]
[7]
Target Epidermal growth factor receptor (EGFR) EGFR_HUMAN [1]
Click to Show/Hide the Molecular Information and External Link(s) of This Drug
TTD Drug ID
D0N5OV
DrugBank ID
DB00002
Type(s) of Resistant Mechanism of This Drug
  ADTT: Aberration of the Drug's Therapeutic Target
  EADR: Epigenetic Alteration of DNA, RNA or Protein
  RTDM: Regulation by the Disease Microenvironment
  UAPP: Unusual Activation of Pro-survival Pathway
Drug Resistance Data Categorized by Their Corresponding Diseases
ICD-02: Benign/in-situ/malignant neoplasm
Click to Show/Hide the Resistance Disease of This Class
Oral squamous cell carcinoma [ICD-11: 2B6E]
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_circ_0005379 [1]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Oral squamous cell carcinoma [ICD-11: 2B6E.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
EGFR signaling pathway Inhibition hsa01521
In Vitro Model CAL27 cells Oral Homo sapiens (Human) CVCL_1107
SCC25 cells Oral Homo sapiens (Human) CVCL_1682
In Vivo Model Balb/c athymic nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Upregualtion of hsa_circ_0005379 enhances the sensitivity of OSCC to anticancer drug cetuximab.
Colon cancer [ICD-11: 2B90]
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-199a-5p [5]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Inhibition hsa04151
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model GEO CR cells Colon Homo sapiens (Human) CVCL_0271
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description The ability of miR-199a-5p and miR-375 to target PHLPP1 (PH domain and leucine-rich repeat protein phosphatase 1), a tumor suppressor that negatively regulates the AkT pathway, accounts, at least in part, for their drug-resistance activity. Indeed, restoration of PHLPP1 increases sensitivity of the GEO cells to CTX and reverts the resistance-promoting effect of miR-199a-5p and miR-375.
Key Molecule: hsa-mir-375 [5]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Inhibition hsa04151
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model GEO CR cells Colon Homo sapiens (Human) CVCL_0271
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description The ability of miR-199a-5p and miR-375 to target PHLPP1 (PH domain and leucine-rich repeat protein phosphatase 1), a tumor suppressor that negatively regulates the AkT pathway, accounts, at least in part, for their drug-resistance activity. Indeed, restoration of PHLPP1 increases sensitivity of the GEO cells to CTX and reverts the resistance-promoting effect of miR-199a-5p and miR-375.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: PH domain leucine-rich repeat-containing protein phosphatase 1 (PHLPP1) [5]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Inhibition hsa04151
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model GEO CR cells Colon Homo sapiens (Human) CVCL_0271
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description The ability of miR-199a-5p and miR-375 to target PHLPP1 (PH domain and leucine-rich repeat protein phosphatase 1), a tumor suppressor that negatively regulates the AkT pathway, accounts, at least in part, for their drug-resistance activity. Indeed, restoration of PHLPP1 increases sensitivity of the GEO cells to CTX and reverts the resistance-promoting effect of miR-199a-5p and miR-375.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-143 [8]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
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
EGFR/RAS/MAPK signaling pathway Regulation hsa01521
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Northern blot analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description The extent of caspase and nuclear fragmentation inhibition was higher in cells overexpressing miR-143 or miR-145, which also display reduced Bcl-2 protein steady-state levels. restoration of miR-143 or miR-145 reduces the aggressiveness of mutant kRAS HCT116 cells. In addition, forced expression of these miRNAs in both mutant and wild-type kRAS colon cancer cells increased their sensitivity to cetuximab by increasing cetuximab-mediated ADCC. Moreover, increased levels of effector cell-mediated caspase-dependent apoptosis were observed for mutant kRAS HCT116 miRNAs-overexpressing cells.
Key Molecule: hsa-mir-145 [8]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
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
EGFR/RAS/MAPK signaling pathway Regulation hsa01521
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Northern blot analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description The extent of caspase and nuclear fragmentation inhibition was higher in cells overexpressing miR-143 or miR-145, which also display reduced Bcl-2 protein steady-state levels. restoration of miR-143 or miR-145 reduces the aggressiveness of mutant kRAS HCT116 cells. In addition, forced expression of these miRNAs in both mutant and wild-type kRAS colon cancer cells increased their sensitivity to cetuximab by increasing cetuximab-mediated ADCC. Moreover, increased levels of effector cell-mediated caspase-dependent apoptosis were observed for mutant kRAS HCT116 miRNAs-overexpressing cells.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [8]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
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
EGFR/RAS/MAPK signaling pathway Regulation hsa01521
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description The extent of caspase and nuclear fragmentation inhibition was higher in cells overexpressing miR-143 or miR-145, which also display reduced Bcl-2 protein steady-state levels. restoration of miR-143 or miR-145 reduces the aggressiveness of mutant kRAS HCT116 cells. In addition, forced expression of these miRNAs in both mutant and wild-type kRAS colon cancer cells increased their sensitivity to cetuximab by increasing cetuximab-mediated ADCC. Moreover, increased levels of effector cell-mediated caspase-dependent apoptosis were observed for mutant kRAS HCT116 miRNAs-overexpressing cells.
Colorectal cancer [ICD-11: 2B91]
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: Epidermal growth factor receptor (EGFR) [9]
Molecule Alteration Missense mutation
p.G465E
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model Colon cells Colon Homo sapiens (Human) N.A.
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Next-generation sequencing assay
Experiment for
Drug Resistance
Liquid biopsy assay
Mechanism Description Mechanisms of resistance to EGFR blockade include the emergence of kRAS, NRAS and EGFR extracellular domain mutations as well as HER2/MET alterations.
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: POU class 5 homeobox 1 pseudogene 4 (POU5F1P4) [10]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model CaCo2 cells Colon Homo sapiens (Human) CVCL_0025
NCI-H508 cells Colon Homo sapiens (Human) CVCL_1564
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Down-regulation of POU5F1P4 decreased the sensitivity of colorectal cancer cells to cetuximab. POU5F1P4 may contribute to cetuximab resistance by interacting with protein coding genes that affect different biological pathways.
Key Molecule: Mir-100-let-7a-2-mir-125b-1 cluster host gene (MIR100HG) [6]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Wnt/Beta-catenin signaling pathway Inhibition hsa04310
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
DLD1 cells Colon Homo sapiens (Human) CVCL_0248
SW620 cells Colon Homo sapiens (Human) CVCL_0547
CaCo2 cells Colon Homo sapiens (Human) CVCL_0025
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
RkO cells Colon Homo sapiens (Human) CVCL_0504
HCT8 cells Colon Homo sapiens (Human) CVCL_2478
NCI-H508 cells Colon Homo sapiens (Human) CVCL_1564
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
COLO 320DM cells Colon Homo sapiens (Human) CVCL_0219
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
LS174T cells Colon Homo sapiens (Human) CVCL_1384
NCI-H716 cells Colon Homo sapiens (Human) CVCL_1581
SW948 cells Colon Homo sapiens (Human) CVCL_0632
SW403 cells Colon Homo sapiens (Human) CVCL_0545
SW48 cells Colon Homo sapiens (Human) CVCL_1724
COLO205 cells Colon Homo sapiens (Human) CVCL_F402
HuTu80 cells Small intestine Homo sapiens (Human) CVCL_1301
LS123 cells Colon Homo sapiens (Human) CVCL_1383
SK-CO-1 cells Colon Homo sapiens (Human) CVCL_0626
SW837 cells Colon Homo sapiens (Human) CVCL_1729
T84 cells Colon Homo sapiens (Human) CVCL_0555
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qPCR; Sequencing assay
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description There is a double-negative feedback loop between MIR100HG and the transcription factor GATA6, whereby GATA6 represses MIR100HG, but this repression is relieved by miR125b targeting of GATA6.
Key Molecule: hsa-mir-100 [6]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Wnt/Beta-catenin signaling pathway Inhibition hsa04310
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
DLD1 cells Colon Homo sapiens (Human) CVCL_0248
SW620 cells Colon Homo sapiens (Human) CVCL_0547
CaCo2 cells Colon Homo sapiens (Human) CVCL_0025
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
RkO cells Colon Homo sapiens (Human) CVCL_0504
HCT8 cells Colon Homo sapiens (Human) CVCL_2478
NCI-H508 cells Colon Homo sapiens (Human) CVCL_1564
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
COLO 320DM cells Colon Homo sapiens (Human) CVCL_0219
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
LS174T cells Colon Homo sapiens (Human) CVCL_1384
NCI-H716 cells Colon Homo sapiens (Human) CVCL_1581
SW948 cells Colon Homo sapiens (Human) CVCL_0632
SW403 cells Colon Homo sapiens (Human) CVCL_0545
SW48 cells Colon Homo sapiens (Human) CVCL_1724
COLO205 cells Colon Homo sapiens (Human) CVCL_F402
HuTu80 cells Small intestine Homo sapiens (Human) CVCL_1301
LS123 cells Colon Homo sapiens (Human) CVCL_1383
SK-CO-1 cells Colon Homo sapiens (Human) CVCL_0626
SW837 cells Colon Homo sapiens (Human) CVCL_1729
T84 cells Colon Homo sapiens (Human) CVCL_0555
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Luciferase reporter assay; qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR100 and miR125b coordinately repressed five Wnt/beta-catenin negative regulators, resulting in increased Wnt signaling, and Wnt inhibition in cetuximab-resistant cells restored cetuximab responsiveness.
Key Molecule: hsa-mir-125b [6]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Wnt/Beta-catenin signaling pathway Inhibition hsa04310
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
DLD1 cells Colon Homo sapiens (Human) CVCL_0248
SW620 cells Colon Homo sapiens (Human) CVCL_0547
CaCo2 cells Colon Homo sapiens (Human) CVCL_0025
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
RkO cells Colon Homo sapiens (Human) CVCL_0504
HCT8 cells Colon Homo sapiens (Human) CVCL_2478
NCI-H508 cells Colon Homo sapiens (Human) CVCL_1564
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
COLO 320DM cells Colon Homo sapiens (Human) CVCL_0219
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
LS174T cells Colon Homo sapiens (Human) CVCL_1384
NCI-H716 cells Colon Homo sapiens (Human) CVCL_1581
SW948 cells Colon Homo sapiens (Human) CVCL_0632
SW403 cells Colon Homo sapiens (Human) CVCL_0545
SW48 cells Colon Homo sapiens (Human) CVCL_1724
COLO205 cells Colon Homo sapiens (Human) CVCL_F402
HuTu80 cells Small intestine Homo sapiens (Human) CVCL_1301
LS123 cells Colon Homo sapiens (Human) CVCL_1383
SK-CO-1 cells Colon Homo sapiens (Human) CVCL_0626
SW837 cells Colon Homo sapiens (Human) CVCL_1729
T84 cells Colon Homo sapiens (Human) CVCL_0555
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Luciferase reporter assay; qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR100 and miR125b coordinately repressed five Wnt/beta-catenin negative regulators, resulting in increased Wnt signaling, and Wnt inhibition in cetuximab-resistant cells restored cetuximab responsiveness.
Key Molecule: Mir-100-let-7a-2-mir-125b-1 cluster host gene (MIR100HG) [6]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Wnt/Beta-catenin signaling pathway Activation hsa04310
In Vitro Model MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
GIST-T1 cells Gastric Homo sapiens (Human) CVCL_4976
CAL62 cells Thyroid gland Homo sapiens (Human) CVCL_1112
CAL-62 cells Thyroid gland Homo sapiens (Human) CVCL_1112
CCL-131 cells Brain Mus musculus (Mouse) CVCL_0470
COLO320DM cells Colon Homo sapiens (Human) CVCL_0219
CT26 WT cells Colon Mus musculus (Mouse) CVCL_7256
Detroit562 cells Pleural effusion Homo sapiens (Human) CVCL_1171
DIPG 007 cells Brain Homo sapiens (Human) CVCL_VU70
DLD-1 cells Colon Homo sapiens (Human) CVCL_0248
DU145 cells Prostate Homo sapiens (Human) CVCL_0105
FL83B cells Liver Mus musculus (Mouse) CVCL_4691
GH3 cells Pituitary gland Rattus norvegicus (Rat) CVCL_0273
GH4C1 cells pituitary gland Rattus norvegicus (Rat) CVCL_0276
H1650 cells Pleural effusion Homo sapiens (Human) CVCL_4V01
H9 cells Peripheral blood Homo sapiens (Human) CVCL_1240
H9/HTLV cells Peripheral blood Homo sapiens (Human) CVCL_3514
HEK 293T cells Kidney Homo sapiens (Human) CVCL_0063
HeLa S cells Uterus Homo sapiens (Human) CVCL_0058
HeLa229 cells Uterus Homo sapiens (Human) CVCL_1276
HH cells Peripheral blood Homo sapiens (Human) CVCL_1280
HPrEC cells Prostate Homo sapiens (Human) CVCL_A2EM
Human RPMI8226 myeloma cells Peripheral blood Homo sapiens (Human) CVCL_0014
KB-C2 cells Uterus Homo sapiens (Human) CVCL_D600
Experiment for
Molecule Alteration
RT-PCR
Mechanism Description miR-100HG, miR-100 and miR-125b overexpression was also observed in cetuximab-resistant colorectal cancer and head and neck squamous cell cancer cell lines and in tumors from colorectal cancer patients that progressed on cetuximab. miR-100 and miR-125b coordinately repressed five Wnt/beta-catenin negative regulators, resulting in increased Wnt signaling, and Wnt inhibition in cetuximab-resistant cells restored cetuximab responsiveness.
Key Molecule: hsa-mir-100 [6]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Wnt/Beta-catenin signaling pathway Activation hsa04310
In Vitro Model MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
GIST-T1 cells Gastric Homo sapiens (Human) CVCL_4976
CAL62 cells Thyroid gland Homo sapiens (Human) CVCL_1112
CAL-62 cells Thyroid gland Homo sapiens (Human) CVCL_1112
CCL-131 cells Brain Mus musculus (Mouse) CVCL_0470
COLO320DM cells Colon Homo sapiens (Human) CVCL_0219
CT26 WT cells Colon Mus musculus (Mouse) CVCL_7256
Detroit562 cells Pleural effusion Homo sapiens (Human) CVCL_1171
DIPG 007 cells Brain Homo sapiens (Human) CVCL_VU70
DLD-1 cells Colon Homo sapiens (Human) CVCL_0248
DU145 cells Prostate Homo sapiens (Human) CVCL_0105
FL83B cells Liver Mus musculus (Mouse) CVCL_4691
GH3 cells Pituitary gland Rattus norvegicus (Rat) CVCL_0273
GH4C1 cells pituitary gland Rattus norvegicus (Rat) CVCL_0276
H1650 cells Pleural effusion Homo sapiens (Human) CVCL_4V01
H9 cells Peripheral blood Homo sapiens (Human) CVCL_1240
H9/HTLV cells Peripheral blood Homo sapiens (Human) CVCL_3514
HEK 293T cells Kidney Homo sapiens (Human) CVCL_0063
HeLa S cells Uterus Homo sapiens (Human) CVCL_0058
HeLa229 cells Uterus Homo sapiens (Human) CVCL_1276
HH cells Peripheral blood Homo sapiens (Human) CVCL_1280
HPrEC cells Prostate Homo sapiens (Human) CVCL_A2EM
Human RPMI8226 myeloma cells Peripheral blood Homo sapiens (Human) CVCL_0014
KB-C2 cells Uterus Homo sapiens (Human) CVCL_D600
Experiment for
Molecule Alteration
RT-PCR
Mechanism Description miR-100HG, miR-100 and miR-125b overexpression was also observed in cetuximab-resistant colorectal cancer and head and neck squamous cell cancer cell lines and in tumors from colorectal cancer patients that progressed on cetuximab. miR-100 and miR-125b coordinately repressed five Wnt/beta-catenin negative regulators, resulting in increased Wnt signaling, and Wnt inhibition in cetuximab-resistant cells restored cetuximab responsiveness.
Key Molecule: hsa-mir-125b [6]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Wnt/Beta-catenin signaling pathway Activation hsa04310
In Vitro Model MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
GIST-T1 cells Gastric Homo sapiens (Human) CVCL_4976
CAL62 cells Thyroid gland Homo sapiens (Human) CVCL_1112
CAL-62 cells Thyroid gland Homo sapiens (Human) CVCL_1112
CCL-131 cells Brain Mus musculus (Mouse) CVCL_0470
COLO320DM cells Colon Homo sapiens (Human) CVCL_0219
CT26 WT cells Colon Mus musculus (Mouse) CVCL_7256
Detroit562 cells Pleural effusion Homo sapiens (Human) CVCL_1171
DIPG 007 cells Brain Homo sapiens (Human) CVCL_VU70
DLD-1 cells Colon Homo sapiens (Human) CVCL_0248
DU145 cells Prostate Homo sapiens (Human) CVCL_0105
FL83B cells Liver Mus musculus (Mouse) CVCL_4691
GH3 cells Pituitary gland Rattus norvegicus (Rat) CVCL_0273
GH4C1 cells pituitary gland Rattus norvegicus (Rat) CVCL_0276
H1650 cells Pleural effusion Homo sapiens (Human) CVCL_4V01
H9 cells Peripheral blood Homo sapiens (Human) CVCL_1240
H9/HTLV cells Peripheral blood Homo sapiens (Human) CVCL_3514
HEK 293T cells Kidney Homo sapiens (Human) CVCL_0063
HeLa S cells Uterus Homo sapiens (Human) CVCL_0058
HeLa229 cells Uterus Homo sapiens (Human) CVCL_1276
HH cells Peripheral blood Homo sapiens (Human) CVCL_1280
HPrEC cells Prostate Homo sapiens (Human) CVCL_A2EM
Human RPMI8226 myeloma cells Peripheral blood Homo sapiens (Human) CVCL_0014
KB-C2 cells Uterus Homo sapiens (Human) CVCL_D600
Experiment for
Molecule Alteration
RT-PCR
Mechanism Description miR-100HG, miR-100 and miR-125b overexpression was also observed in cetuximab-resistant colorectal cancer and head and neck squamous cell cancer cell lines and in tumors from colorectal cancer patients that progressed on cetuximab. miR-100 and miR-125b coordinately repressed five Wnt/beta-catenin negative regulators, resulting in increased Wnt signaling, and Wnt inhibition in cetuximab-resistant cells restored cetuximab responsiveness.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: Programmed cell death 6-interacting protein (PDCD6IP) [11]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell colony Activation hsa05200
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model CaCo2 cells Colon Homo sapiens (Human) CVCL_0025
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description UCA1 expression was markedly higher in cetuximab-resistant cancer cells and their exosomes and the expression of TSG101, Alix, and CD81, which are all exosome markers and are associated with exosome formation, in both exosomes and cells.
Key Molecule: Urothelial cancer associated 1 (UCA1) [11]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell colony Activation hsa05200
Cell proliferation Activation hsa05200
In Vitro Model CaCo2 cells Colon Homo sapiens (Human) CVCL_0025
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description UCA1 expression was markedly higher in cetuximab-resistant cancer cells and their exosomes and the expression of TSG101, Alix, and CD81, which are all exosome markers and are associated with exosome formation, in both exosomes and cells.
Key Molecule: GDH/6PGL endoplasmic bifunctional protein (H6PD) [4]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Discovered Using In-vivo Testing Model
Cell Pathway Regulation Pentose phosphate signaling pathway Activation hsa00030
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
GEO cells Colon Homo sapiens (Human) CVCL_0271
In Vivo Model Xenografts mouse model Mus musculus
Experiment for
Molecule Alteration
2D DIGE assay
Mechanism Description LDHB and PDHA1 were downregulated in GEO-CR tumor xenografts, similarly to the corresponding deregulations observed in the derived cell lines. Upregulation of G6PDH and transketolase (TkT) was also actually maintained in tumor xenografts. Indeed, PPP2CA expression in xenografted samples was similarly evaluated, demonstrating that protein downregulation in vivo was even more pronounced than that measured in GEO-CR cells.
Key Molecule: L-lactate dehydrogenase B chain (LDHB) [4]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Discovered Using In-vivo Testing Model
Cell Pathway Regulation Pentose phosphate signaling pathway Activation hsa00030
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
GEO cells Colon Homo sapiens (Human) CVCL_0271
In Vivo Model Xenografts mouse model Mus musculus
Experiment for
Molecule Alteration
2D DIGE assay
Mechanism Description LDHB and PDHA1 were downregulated in GEO-CR tumor xenografts, similarly to the corresponding deregulations observed in the derived cell lines. Upregulation of G6PDH and transketolase (TkT) was also actually maintained in tumor xenografts. Indeed, PPP2CA expression in xenografted samples was similarly evaluated, demonstrating that protein downregulation in vivo was even more pronounced than that measured in GEO-CR cells.
Key Molecule: Pyruvate dehydrogenase E1 component subunit alpha (PDHA1) [4]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Discovered Using In-vivo Testing Model
Cell Pathway Regulation Pentose phosphate signaling pathway Activation hsa00030
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
GEO cells Colon Homo sapiens (Human) CVCL_0271
In Vivo Model Xenografts mouse model Mus musculus
Experiment for
Molecule Alteration
2D DIGE assay
Mechanism Description LDHB and PDHA1 were downregulated in GEO-CR tumor xenografts, similarly to the corresponding deregulations observed in the derived cell lines. Upregulation of G6PDH and transketolase (TkT) was also actually maintained in tumor xenografts. Indeed, PPP2CA expression in xenografted samples was similarly evaluated, demonstrating that protein downregulation in vivo was even more pronounced than that measured in GEO-CR cells.
Key Molecule: Transketolase (TKT) [4]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Discovered Using In-vivo Testing Model
Cell Pathway Regulation Pentose phosphate signaling pathway Activation hsa00030
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
GEO cells Colon Homo sapiens (Human) CVCL_0271
In Vivo Model Xenografts mouse model Mus musculus
Experiment for
Molecule Alteration
2D DIGE assay
Mechanism Description LDHB and PDHA1 were downregulated in GEO-CR tumor xenografts, similarly to the corresponding deregulations observed in the derived cell lines. Upregulation of G6PDH and transketolase (TkT) was also actually maintained in tumor xenografts. Indeed, PPP2CA expression in xenografted samples was similarly evaluated, demonstrating that protein downregulation in vivo was even more pronounced than that measured in GEO-CR cells.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Transcription factor GATA6 (GATA6) [6]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Chemoresistance Activation hsa05207
Wnt/Beta-catenin signaling pathway Inhibition hsa04310
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
DLD1 cells Colon Homo sapiens (Human) CVCL_0248
SW620 cells Colon Homo sapiens (Human) CVCL_0547
CaCo2 cells Colon Homo sapiens (Human) CVCL_0025
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
RkO cells Colon Homo sapiens (Human) CVCL_0504
HCT8 cells Colon Homo sapiens (Human) CVCL_2478
NCI-H508 cells Colon Homo sapiens (Human) CVCL_1564
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
COLO 320DM cells Colon Homo sapiens (Human) CVCL_0219
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
LS174T cells Colon Homo sapiens (Human) CVCL_1384
NCI-H716 cells Colon Homo sapiens (Human) CVCL_1581
SW948 cells Colon Homo sapiens (Human) CVCL_0632
SW403 cells Colon Homo sapiens (Human) CVCL_0545
SW48 cells Colon Homo sapiens (Human) CVCL_1724
COLO205 cells Colon Homo sapiens (Human) CVCL_F402
HuTu80 cells Small intestine Homo sapiens (Human) CVCL_1301
LS123 cells Colon Homo sapiens (Human) CVCL_1383
SK-CO-1 cells Colon Homo sapiens (Human) CVCL_0626
SW837 cells Colon Homo sapiens (Human) CVCL_1729
T84 cells Colon Homo sapiens (Human) CVCL_0555
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qPCR; Sequencing assay; Western blot analysis; Immunofluorescent staining assay
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description There is a double-negative feedback loop between MIR100HG and the transcription factor GATA6, whereby GATA6 represses MIR100HG, but this repression is relieved by miR125b targeting of GATA6.
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [9]
Molecule Alteration Missense mutation
p.V600E
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model Colon cells Colon Homo sapiens (Human) N.A.
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Next-generation sequencing assay
Experiment for
Drug Resistance
Liquid biopsy assay
Mechanism Description Mechanisms of resistance to EGFR blockade include the emergence of kRAS, NRAS and EGFR extracellular domain mutations as well as HER2/MET alterations.
Key Molecule: Hepatocyte growth factor receptor (MET) [9]
Molecule Alteration Structural variation
Amplification
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Next-generation sequencing assay
Experiment for
Drug Resistance
Liquid biopsy assay
Mechanism Description Mechanisms of resistance to EGFR blockade include the emergence of kRAS, NRAS and EGFR extracellular domain mutations as well as HER2/MET alterations.
Key Molecule: GTPase KRas (KRAS) [9]
Molecule Alteration Structural variation
Amplification
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Next-generation sequencing assay
Experiment for
Drug Resistance
Liquid biopsy assay
Mechanism Description Mechanisms of resistance to EGFR blockade include the emergence of kRAS, NRAS and EGFR extracellular domain mutations as well as HER2/MET alterations.
Key Molecule: Receptor tyrosine-protein kinase erbB-2 (ERBB2) [9]
Molecule Alteration Structural variation
Amplification
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Next-generation sequencing assay
Experiment for
Drug Resistance
Liquid biopsy assay
Mechanism Description Mechanisms of resistance to EGFR blockade include the emergence of kRAS, NRAS and EGFR extracellular domain mutations as well as HER2/MET alterations.
Key Molecule: GTPase KRas (KRAS) [9]
Molecule Alteration Missense mutation
p.Q61H
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Next-generation sequencing assay
Experiment for
Drug Resistance
Liquid biopsy assay
Mechanism Description Mechanisms of resistance to EGFR blockade include the emergence of kRAS, NRAS and EGFR extracellular domain mutations as well as HER2/MET alterations.
Key Molecule: GTPase KRas (KRAS) [9], [12], [13]
Molecule Alteration Missense mutation
p.G12V
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation EGFR/RAS signaling pathway Activation hsa01521
In Vitro Model LIM1215 cells Colon Homo sapiens (Human) CVCL_2574
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Next-generation sequencing assay
Experiment for
Drug Resistance
Liquid biopsy assay
Mechanism Description Mechanisms of resistance to EGFR blockade include the emergence of kRAS, NRAS and EGFR extracellular domain mutations as well as HER2/MET alterations (27780856). kRAS and EGFR ectodomain-acquired mutations in patients with metastatic colorectal cancer (mCRC) have been correlated with acquired resistance to anti-EGFR monoclonal antibodies (mAbs).
Key Molecule: GTPase KRas (KRAS) [9]
Molecule Alteration Missense mutation
p.G12D
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Next-generation sequencing assay
Experiment for
Drug Resistance
Liquid biopsy assay
Mechanism Description Mechanisms of resistance to EGFR blockade include the emergence of kRAS, NRAS and EGFR extracellular domain mutations as well as HER2/MET alterations.
Key Molecule: Hepatocyte growth factor receptor (MET) [2]
Molecule Alteration Structural variation
Copy number gain
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Next-generation sequencing analysis; Gene copy number analysis
Mechanism Description As amplification of the MET gene has recently been shown to drive resistance to anti-EGFR therapies, this copy number change is the best candidate to explain the poor treatment response.
Key Molecule: Receptor tyrosine-protein kinase erbB-2 (ERBB2) [14]
Molecule Alteration Structural variation
Amplification
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
Sanger sequencing assay; Next-generation sequencing assay
Mechanism Description Mutations in kRAS, NRAS, and BRAF and amplification of ERBB2 and MET drive primary (de novo) resistance to anti-EGFR treatment.
Key Molecule: GTPase KRas (KRAS) [15]
Molecule Alteration Mutation
Mutations in codons 12, 13 and 61
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation RAS/RAF/Mek/ERK signaling pathway Activation hsa04010
In Vitro Model Colorectal cancer cells Colon Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
High throughout experiment assay
Experiment for
Drug Resistance
Circulating tumor DNA analysis
Mechanism Description The identification of kRAS mutations as a cause for intrinsic resistance of colorectal cancers also contributed to the identification of a mechanism for the acquired resistance. Establishment and analysis of cetuximabresistant colorectal cancer cell lines revealed that the resistant variants harbored kRAS point mutations or amplification, and the findings were confirmed in clinical specimens.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-7 [16]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description microRNA-7 expression in colorectal cancer is associated with poor prognosis and regulates cetuximab sensitivity via EGFR regulation.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Epidermal growth factor receptor (EGFR) [16]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description microRNA-7 expression in colorectal cancer is associated with poor prognosis and regulates cetuximab sensitivity via EGFR regulation.
Key Molecule: RAF proto-oncogene serine/threonine-protein kinase (RAF1) [16]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description microRNA-7 expression in colorectal cancer is associated with poor prognosis and regulates cetuximab sensitivity via EGFR regulation.
Liver cancer [ICD-11: 2C12]
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-let-7a [17]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
SNU449 cells Liver Homo sapiens (Human) CVCL_0454
SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Let-7a enhances the sensitivity of hepatocellular carcinoma cells to cetuximab by negatively regulating STAT3 expression.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Signal transducer activator transcription 3 (STAT3) [17]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
SNU449 cells Liver Homo sapiens (Human) CVCL_0454
SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Let-7a enhances the sensitivity of hepatocellular carcinoma cells to cetuximab by negatively regulating STAT3 expression.
Lung cancer [ICD-11: 2C25]
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-200c [18]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model Calu3 cells Lung Homo sapiens (Human) CVCL_0609
H1299 cells Lung Homo sapiens (Human) CVCL_0060
Sk-MES-1 cells Lung Homo sapiens (Human) CVCL_0630
NCI-H460 cells Lung Homo sapiens (Human) CVCL_0459
NCI-H522 cells Lung Homo sapiens (Human) CVCL_1567
NCl-H596 cells Lung Homo sapiens (Human) CVCL_1571
NCI-H520 cells Lung Homo sapiens (Human) CVCL_1566
Calu1 cells Lung Homo sapiens (Human) CVCL_0608
NCI-H1395 cells Lung Homo sapiens (Human) CVCL_1467
Experiment for
Molecule Alteration
Methylation-specific PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Reintroduction of miR-200c into highly invasive/aggressive NSCLC cells induced a loss of the mesenchymal phenotype by restoring E-cadherin and reducing N-cadherin expression, and inhibited in vitro cell invasion as well as in vivo metastasis formation.
Bladder cancer [ICD-11: 2C94]
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-200b [19]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Bladder cancer [ICD-11: 2C94.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation EGFR signaling pathway Regulation hsa01521
In Vitro Model 253J BV cells Bladder Homo sapiens (Human) CVCL_7937
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Pulse-labeling cells with [3H]thymidine
Mechanism Description Members of the miR-200 family appear to control the EMT process and sensitivity to EGFR therapy, in bladder cancer cells and that expression of miR-200 is sufficient to restore EGFR dependency, at least in some of the mesenchymal bladder cancer cells. The targets of miR-200 include ERRFI-1, which is a novel regulator of EGFR-independent growth.
Key Molecule: hsa-mir-200c [19]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Bladder cancer [ICD-11: 2C94.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation EGFR signaling pathway Regulation hsa01521
In Vitro Model 253J BV cells Bladder Homo sapiens (Human) CVCL_7937
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Pulse-labeling cells with [3H]thymidine
Mechanism Description Members of the miR-200 family appear to control the EMT process and sensitivity to EGFR therapy, in bladder cancer cells and that expression of miR-200 is sufficient to restore EGFR dependency, at least in some of the mesenchymal bladder cancer cells. The targets of miR-200 include ERRFI-1, which is a novel regulator of EGFR-independent growth.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: ERBB receptor feedback inhibitor 1 (ERRFI1) [19]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Bladder cancer [ICD-11: 2C94.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation EGFR signaling pathway Regulation hsa01521
In Vitro Model 253J BV cells Bladder Homo sapiens (Human) CVCL_7937
Experiment for
Molecule Alteration
Immunoblotting analysis
Experiment for
Drug Resistance
Pulse-labeling cells with [3H]thymidine
Mechanism Description Members of the miR-200 family appear to control the EMT process and sensitivity to EGFR therapy, in bladder cancer cells and that expression of miR-200 is sufficient to restore EGFR dependency, at least in some of the mesenchymal bladder cancer cells. The targets of miR-200 include ERRFI-1, which is a novel regulator of EGFR-independent growth.
Head and neck cancer [ICD-11: 2D42]
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-212 [7]
Molecule Alteration Expression
Down-regulation
Resistant Disease Head and neck squamous cell carcinoma [ICD-11: 2D42.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
In Vitro Model SCC1 cells Tongue Homo sapiens (Human) CVCL_A5SA
1Cc8 cells Epithelium Homo sapiens (Human) CVCL_L893
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description HB-EGF can induce EMT, enhance metastasis, and modulate chemotherapy resistance. Increased expression of HB-EGF due to down-regulation of miR-212 is a possible mechanism of cetuximab resistance.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Proheparin-binding EGF-like growth factor (HBEGF) [7]
Molecule Alteration Expression
Up-regulation
Resistant Disease Head and neck squamous cell carcinoma [ICD-11: 2D42.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
In Vitro Model SCC1 cells Tongue Homo sapiens (Human) CVCL_A5SA
1Cc8 cells Epithelium Homo sapiens (Human) CVCL_L893
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Immunoblotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description HB-EGF can induce EMT, enhance metastasis, and modulate chemotherapy resistance. Increased expression of HB-EGF due to down-regulation of miR-212 is a possible mechanism of cetuximab resistance.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-204 [20]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Head and neck squamous cell carcinoma [ICD-11: 2D42.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation JAKT2/STAT3 signaling pathway Inhibition hsa04030
In Vitro Model 5-8F cells Nasopharynx Homo sapiens (Human) CVCL_C528
CNE2 cells Nasopharynx Homo sapiens (Human) CVCL_6889
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR204 inhibits angiogenesis and promotes sensitivity to cetuximab in head and neck squamous cell carcinoma cells by blocking JAk2-STAT3 signaling.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Tyrosine-protein kinase JAK2 (JAK3) [20]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Head and neck squamous cell carcinoma [ICD-11: 2D42.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation JAKT2/STAT3 signaling pathway Inhibition hsa04030
In Vitro Model 5-8F cells Nasopharynx Homo sapiens (Human) CVCL_C528
CNE2 cells Nasopharynx Homo sapiens (Human) CVCL_6889
Experiment for
Molecule Alteration
qRT-PCR; Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR204 inhibits angiogenesis and promotes sensitivity to cetuximab in head and neck squamous cell carcinoma cells by blocking JAk2-STAT3 signaling.
Metastatic colorectal cancer [ICD-11: 2D85]
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: Epidermal growth factor receptor (EGFR) [13]
Molecule Alteration Missense mutation
p.S492R
Resistant Disease Metastatic colorectal cancer [ICD-11: 2D85.0]
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
Circulating-free DNA assay; Standard-of-care sequencing assay
Mechanism Description K-RAS and EGFR ectodomain-acquired mutations in patients with metastatic colorectal cancer (mCRC) have been correlated with acquired resistance to anti-EGFR monoclonal antibodies (mAbs).
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: GTPase KRas (KRAS) [3]
Molecule Alteration Missense mutation
p.Q61H
Resistant Disease Metastatic colorectal cancer [ICD-11: 2D85.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model DiFi cells Colon Homo sapiens (Human) CVCL_6895
DiFi-R cells Colon Homo sapiens (Human) CVCL_A2BW
Lim1215-R cells Colon Homo sapiens (Human) CVCL_1736
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
FISH analysis; Sanger sequencing assay
Experiment for
Drug Resistance
Cell viability assay
Mechanism Description Nevertheless, our functional analysis in cell models show that kRAS mutations are causally responsible for acquired resistance to cetuximab.
Key Molecule: GTPase Nras (NRAS) [21]
Molecule Alteration Missense mutation
p.G12C
Resistant Disease Metastatic colorectal cancer [ICD-11: 2D85.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation EGFR/RAS signaling pathway Inhibition hsa01521
In Vitro Model LIM1215 cells Colon Homo sapiens (Human) CVCL_2574
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Next-generation sequencing assay
Experiment for
Drug Resistance
Liquid biopsies assay; Functional analyses of cell populations assay
Mechanism Description Acquired resistance to EGFR blockade is driven by the emergence of kRAS/NRAS mutations or the development of EGFR extracellular domain (ECD) variants, which impair antibody binding.
Key Molecule: Hepatocyte growth factor receptor (MET) [14]
Molecule Alteration Structural variation
Copy number gain
Resistant Disease Metastatic colorectal cancer [ICD-11: 2D85.0]
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
Sanger sequencing assay; Next-generation sequencing assay
Mechanism Description Mutations in kRAS, NRAS, and BRAF and amplification of ERBB2 and MET drive primary (de novo) resistance to anti-EGFR treatment.
Key Molecule: GTPase KRas (KRAS) [14]
Molecule Alteration Mutation
.
Resistant Disease Metastatic colorectal cancer [ICD-11: 2D85.0]
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
Sanger sequencing assay; Next-generation sequencing assay
Mechanism Description Mutations in kRAS, NRAS, and BRAF and amplification of ERBB2 and MET drive primary (de novo) resistance to anti-EGFR treatment.
References
Ref 1 Hsa_circ_0005379 regulates malignant behavior of oral squamous cell carcinoma through the EGFR pathway. BMC Cancer. 2019 Apr 29;19(1):400. doi: 10.1186/s12885-019-5593-5.
Ref 2 Amplification of the MET receptor drives resistance to anti-EGFR therapies in colorectal cancer. Cancer Discov. 2013 Jun;3(6):658-73. doi: 10.1158/2159-8290.CD-12-0558. Epub 2013 Jun 2.
Ref 3 Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature. 2012 Jun 28;486(7404):532-6. doi: 10.1038/nature11156.
Ref 4 Increased anaerobic metabolism is a distinctive signature in a colorectal cancer cellular model of resistance to antiepidermal growth factor receptor antibody. Proteomics. 2013 Mar;13(5):866-77. doi: 10.1002/pmic.201200303. Epub 2013 Jan 24.
Ref 5 MiR-199a-5p and miR-375 affect colon cancer cell sensitivity to cetuximab by targeting PHLPP1. Expert Opin Ther Targets. 2015;19(8):1017-26. doi: 10.1517/14728222.2015.1057569. Epub 2015 Jun 24.
Ref 6 lncRNA MIR100HG-derived miR-100 and miR-125b mediate cetuximab resistance via Wnt/Beta-catenin signaling. Nat Med. 2017 Nov;23(11):1331-1341. doi: 10.1038/nm.4424. Epub 2017 Oct 16.
Ref 7 Regulation of heparin-binding EGF-like growth factor by miR-212 and acquired cetuximab-resistance in head and neck squamous cell carcinoma. PLoS One. 2010 Sep 13;5(9):e12702. doi: 10.1371/journal.pone.0012702.
Ref 8 miR-143 or miR-145 overexpression increases cetuximab-mediated antibody-dependent cellular cytotoxicity in human colon cancer cells. Oncotarget. 2016 Feb 23;7(8):9368-87. doi: 10.18632/oncotarget.7010.
Ref 9 Heterogeneity of Acquired Resistance to Anti-EGFR Monoclonal Antibodies in Patients with Metastatic Colorectal Cancer. Clin Cancer Res. 2017 May 15;23(10):2414-2422. doi: 10.1158/1078-0432.CCR-16-1863. Epub 2016 Oct 25.
Ref 10 Identification and validation of cetuximab resistance associated long noncoding RNA biomarkers in metastatic colorectal cancer. Biomed Pharmacother. 2018 Jan;97:1138-1146. doi: 10.1016/j.biopha.2017.11.031. Epub 2017 Nov 10.
Ref 11 Predictive role of UCA1-containing exosomes in cetuximab-resistant colorectal cancer. Cancer Cell Int. 2018 Oct 22;18:164. doi: 10.1186/s12935-018-0660-6. eCollection 2018.
Ref 12 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 13 Characterizing the patterns of clonal selection in circulating tumor DNA from patients with colorectal cancer refractory to anti-EGFR treatment. Ann Oncol. 2015 Apr;26(4):731-736. doi: 10.1093/annonc/mdv005. Epub 2015 Jan 26.
Ref 14 Resistance to anti-EGFR therapy in colorectal cancer: from heterogeneity to convergent evolution. Cancer Discov. 2014 Nov;4(11):1269-80. doi: 10.1158/2159-8290.CD-14-0462. Epub 2014 Oct 7.
Ref 15 The Moment that KRAS Mutation Started to Evolve into Precision Medicine in Metastatic Colorectal Cancer. Cancer Res. 2016 Nov 15;76(22):6443-6444. doi: 10.1158/0008-5472.CAN-16-2867.
Ref 16 MicroRNA-7 expression in colorectal cancer is associated with poor prognosis and regulates cetuximab sensitivity via EGFR regulation. Carcinogenesis. 2015 Mar;36(3):338-45. doi: 10.1093/carcin/bgu242. Epub 2014 Dec 10.
Ref 17 Let-7a enhances the sensitivity of hepatocellular carcinoma cells to cetuximab by regulating STAT3 expression. Onco Targets Ther. 2016 Nov 28;9:7253-7261. doi: 10.2147/OTT.S116127. eCollection 2016.
Ref 18 Loss of miR-200c expression induces an aggressive, invasive, and chemoresistant phenotype in non-small cell lung cancer. Mol Cancer Res. 2010 Sep;8(9):1207-16. doi: 10.1158/1541-7786.MCR-10-0052. Epub 2010 Aug 9.
Ref 19 miR-200 expression regulates epithelial-to-mesenchymal transition in bladder cancer cells and reverses resistance to epidermal growth factor receptor therapy. Clin Cancer Res. 2009 Aug 15;15(16):5060-72. doi: 10.1158/1078-0432.CCR-08-2245. Epub 2009 Aug 11.
Ref 20 miR-204 inhibits angiogenesis and promotes sensitivity to cetuximab in head and neck squamous cell carcinoma cells by blocking JAK2-STAT3 signaling. Biomed Pharmacother. 2018 Mar;99:278-285. doi: 10.1016/j.biopha.2018.01.055.
Ref 21 Acquired RAS or EGFR mutations and duration of response to EGFR blockade in colorectal cancer. Nat Commun. 2016 Dec 8;7:13665. doi: 10.1038/ncomms13665.

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