Drug (ID: DG00300) and It's Reported Resistant Information
Name
Oxaliplatin
Synonyms
Eloxatin (TN); Medac (TN); Oxaliplatin (TN); Oxaliplatin (JAN/USAN/INN)
    Click to Show/Hide
Indication
In total 1 Indication(s)
Colorectal cancer [ICD-11: 2B91]
Approved
[1]
Structure
Drug Resistance Disease(s)
Disease(s) with Clinically Reported Resistance for This Drug (8 diseases)
Colon cancer [ICD-11: 2B90]
[2]
Colorectal cancer [ICD-11: 2B91]
[3]
Esophageal cancer [ICD-11: 2B70]
[4]
Gastric cancer [ICD-11: 2B72]
[5]
Liver cancer [ICD-11: 2C12]
[6]
Neuroendocrine carcinoma [ICD-11: 2D4Y]
[7]
Oesophagogastric cancer [ICD-11: 2B71]
[8]
Ovarian cancer [ICD-11: 2C73]
[9]
Disease(s) with Resistance Information Discovered by Cell Line Test for This Drug (6 diseases)
Colon cancer [ICD-11: 2B90]
[10]
Colorectal cancer [ICD-11: 2B91]
[11]
Esophageal cancer [ICD-11: 2B70]
[8]
Gastric cancer [ICD-11: 2B72]
[12]
Liver cancer [ICD-11: 2C12]
[13]
Solid tumour/cancer [ICD-11: 2A00-2F9Z]
[14]
Target Human Deoxyribonucleic acid (hDNA) NOUNIPROTAC [1]
Click to Show/Hide the Molecular Information and External Link(s) of This Drug
Formula
C8H14N2O4Pt
IsoSMILES
C1CC[C@H]([C@@H](C1)[NH-])[NH-].C(=O)(C(=O)O)O.[Pt+2]
InChI
1S/C6H12N2.C2H2O4.Pt/c7-5-3-1-2-4-6(5)8;3-1(4)2(5)6;/h5-8H,1-4H2;(H,3,4)(H,5,6);/q-2;;+2/t5-,6-;;/m1../s1
InChIKey
DRMCATBEKSVAPL-BNTLRKBRSA-N
PubChem CID
9887053
TTD Drug ID
D0Y3ME
DrugBank ID
DB00526
Type(s) of Resistant Mechanism of This Drug
  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 Their Corresponding Diseases
ICD-02: Benign/in-situ/malignant neoplasm
Click to Show/Hide the Resistance Disease of This Class
Solid tumour/cancer [ICD-11: 2A00-2F9Z]
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: TP53 target 1 (TP53TG1) [14]
Molecule Alteration Expression
Down-regulation
Resistant Disease Solid tumour/cancer [ICD-11: 2A00-2F9Z]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation p53 signaling pathway Inhibition hsa04115
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
GCIY cells Gastric Homo sapiens (Human) CVCL_1228
KATO-3 cells Gastric Homo sapiens (Human) CVCL_0371
MkN-7 cells Gastric Homo sapiens (Human) CVCL_1417
SNU-1 cells Gastric Homo sapiens (Human) CVCL_0099
TGBC11TkB cells Gastric Homo sapiens (Human) CVCL_1768
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay; TUNEL assay; xCELLigence Real-Time invasion and migration assays
Mechanism Description TP53TG1, a p53-induced LncRNA, binds to the multifaceted RNA/RNA binding protein YBX1 to prevent its nuclear localization and thus the YBX1-mediated activation of oncogenes. The epigenetic silencing of TP53TG1 in cancer cells promotes the YBX1-mediated activation of the PI3k/AkT pathway, which then creates further resistance not only to common chemotherapy RNA-damaging agents but also to small drug-targeted inhibitors.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Y-box-binding protein 1 (YBX1) [14]
Molecule Alteration Expression
Up-regulation
Resistant Disease Solid tumour/cancer [ICD-11: 2A00-2F9Z]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation p53 signaling pathway Inhibition hsa04115
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
GCIY cells Gastric Homo sapiens (Human) CVCL_1228
KATO-3 cells Gastric Homo sapiens (Human) CVCL_0371
MkN-7 cells Gastric Homo sapiens (Human) CVCL_1417
SNU-1 cells Gastric Homo sapiens (Human) CVCL_0099
TGBC11TkB cells Gastric Homo sapiens (Human) CVCL_1768
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; TUNEL assay; xCELLigence Real-Time invasion and migration assays
Mechanism Description TP53TG1, a p53-induced LncRNA, binds to the multifaceted RNA/RNA binding protein YBX1 to prevent its nuclear localization and thus the YBX1-mediated activation of oncogenes. The epigenetic silencing of TP53TG1 in cancer cells promotes the YBX1-mediated activation of the PI3k/AkT pathway, which then creates further resistance not only to common chemotherapy RNA-damaging agents but also to small drug-targeted inhibitors.
Esophageal cancer [ICD-11: 2B70]
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-141-3p [4]
Molecule Alteration Expression
Up-regulation
Resistant Disease Esophageal cancer [ICD-11: 2B70.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model TE-1 cells Esophagus Homo sapiens (Human) CVCL_1759
EC9706 cells Esophagus Homo sapiens (Human) CVCL_E307
KYSE150 cells Esophagus Homo sapiens (Human) CVCL_1348
EC109 cells Esophagus Homo sapiens (Human) CVCL_6898
EC9706-R cells Esophagus Homo sapiens (Human) CVCL_E307
Het-1A cells Esophagus Homo sapiens (Human) CVCL_3702
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Annexin V-FITC Apoptosis Detection assay
Mechanism Description Involvement of microRNA-141-3p in 5-fluorouracil and oxaliplatin chemo-resistance in esophageal cancer cells via down-regulation of PTEN.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Copper-transporting ATPase 1 (ATP7A) [15]
Molecule Alteration Expression
Up-regulation
Resistant Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model EC109 cells Esophagus Homo sapiens (Human) CVCL_6898
Experiment for
Molecule Alteration
qRT-PCR; Western blotting assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description Overexpression of ATP7A in EC109/cisplatin cells might increase pumping platinum out of cells or binding and sequestration of platinum drugs, then decrease cellular platinum concentration or keep them away from accessing their key cytotoxic targets in the nucleus, finally result in cisplatin-resistance.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Phosphatase and tensin homolog (PTEN) [4]
Molecule Alteration Expression
Down-regulation
Resistant Disease Esophageal cancer [ICD-11: 2B70.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model TE-1 cells Esophagus Homo sapiens (Human) CVCL_1759
EC9706 cells Esophagus Homo sapiens (Human) CVCL_E307
KYSE150 cells Esophagus Homo sapiens (Human) CVCL_1348
EC109 cells Esophagus Homo sapiens (Human) CVCL_6898
EC9706-R cells Esophagus Homo sapiens (Human) CVCL_E307
Het-1A cells Esophagus Homo sapiens (Human) CVCL_3702
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Annexin V-FITC Apoptosis Detection assay
Mechanism Description Involvement of microRNA-141-3p in 5-fluorouracil and oxaliplatin chemo-resistance in esophageal cancer cells via down-regulation of PTEN.
Key Molecule: Sphingosine-1-phosphate lyase 1 (SGPL1) [8]
Molecule Alteration Expression
Down-regulation
Resistant Disease Oesophagus adenocarcinoma [ICD-11: 2B70.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model OE33 cells Esophagus Homo sapiens (Human) CVCL_0471
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description S1P could lead to cytotoxic drug resistance in gastroesophegal cancer acting in an autocrine or paracrine manner via cell surface S1P receptors following transportation out of the cytosol. Alternatively S1P may mediate cytotoxic drug resistance acting intracellularly by counteracting apoptosis mediated by its pro-apoptotic precursor ceramide or interaction with known intracellular targets involved in cancer pathogenesis and cytotoxic drug resistance such as Histone deacetylase 1 (HDAC1) and Histone deacetylase 2 (HDAC 2) to which S1P directly binds and inhibits, and TNF Receptor-Associated Factor 2 (TRAF 2), or Protein Kinase C (PKC). S1P production controlled by SPHK1 and SGPL1 are key determinants of cytotoxic drug resistance and that decreasing S1P production in cancer cells could lead to increased cytotoxic sensitivity.
Key Molecule: Sphingosine kinase 1 (SPHK1) [8]
Molecule Alteration Expression
Up-regulation
Resistant Disease Oesophagus adenocarcinoma [ICD-11: 2B70.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model OE33 cells Esophagus Homo sapiens (Human) CVCL_0471
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description S1P could lead to cytotoxic drug resistance in gastroesophegal cancer acting in an autocrine or paracrine manner via cell surface S1P receptors following transportation out of the cytosol. Alternatively S1P may mediate cytotoxic drug resistance acting intracellularly by counteracting apoptosis mediated by its pro-apoptotic precursor ceramide or interaction with known intracellular targets involved in cancer pathogenesis and cytotoxic drug resistance such as Histone deacetylase 1 (HDAC1) and Histone deacetylase 2 (HDAC 2) to which S1P directly binds and inhibits, and TNF Receptor-Associated Factor 2 (TRAF 2), or Protein Kinase C (PKC). S1P production controlled by SPHK1 and SGPL1 are key determinants of cytotoxic drug resistance and that decreasing S1P production in cancer cells could lead to increased cytotoxic sensitivity.
Key Molecule: Sphingosine-1-phosphate lyase 1 (SGPL1) [8]
Molecule Alteration Expression
Down-regulation
Resistant Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model OE21 cells Esophagus Homo sapiens (Human) CVCL_2661
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description S1P could lead to cytotoxic drug resistance in gastroesophegal cancer acting in an autocrine or paracrine manner via cell surface S1P receptors following transportation out of the cytosol. Alternatively S1P may mediate cytotoxic drug resistance acting intracellularly by counteracting apoptosis mediated by its pro-apoptotic precursor ceramide or interaction with known intracellular targets involved in cancer pathogenesis and cytotoxic drug resistance such as Histone deacetylase 1 (HDAC1) and Histone deacetylase 2 (HDAC 2) to which S1P directly binds and inhibits, and TNF Receptor-Associated Factor 2 (TRAF 2), or Protein Kinase C (PKC). S1P production controlled by SPHK1 and SGPL1 are key determinants of cytotoxic drug resistance and that decreasing S1P production in cancer cells could lead to increased cytotoxic sensitivity.
Key Molecule: Sphingosine kinase 1 (SPHK1) [8]
Molecule Alteration Expression
Up-regulation
Resistant Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model OE21 cells Esophagus Homo sapiens (Human) CVCL_2661
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description S1P could lead to cytotoxic drug resistance in gastroesophegal cancer acting in an autocrine or paracrine manner via cell surface S1P receptors following transportation out of the cytosol. Alternatively S1P may mediate cytotoxic drug resistance acting intracellularly by counteracting apoptosis mediated by its pro-apoptotic precursor ceramide or interaction with known intracellular targets involved in cancer pathogenesis and cytotoxic drug resistance such as Histone deacetylase 1 (HDAC1) and Histone deacetylase 2 (HDAC 2) to which S1P directly binds and inhibits, and TNF Receptor-Associated Factor 2 (TRAF 2), or Protein Kinase C (PKC). S1P production controlled by SPHK1 and SGPL1 are key determinants of cytotoxic drug resistance and that decreasing S1P production in cancer cells could lead to increased cytotoxic sensitivity.
Oesophagogastric cancer [ICD-11: 2B71]
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: Sphingosine-1-phosphate lyase 1 (SGPL1) [8]
Molecule Alteration Expression
Down-regulation
Resistant Disease Gastroesophageal cancer [ICD-11: 2B71.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model Gastroesophageal cancer tissue N.A.
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description S1P could lead to cytotoxic drug resistance in gastroesophegal cancer acting in an autocrine or paracrine manner via cell surface S1P receptors following transportation out of the cytosol. Alternatively S1P may mediate cytotoxic drug resistance acting intracellularly by counteracting apoptosis mediated by its pro-apoptotic precursor ceramide or interaction with known intracellular targets involved in cancer pathogenesis and cytotoxic drug resistance such as Histone deacetylase 1 (HDAC1) and Histone deacetylase 2 (HDAC 2) to which S1P directly binds and inhibits, and TNF Receptor-Associated Factor 2 (TRAF 2), or Protein Kinase C (PKC). S1P production controlled by SPHK1 and SGPL1 are key determinants of cytotoxic drug resistance and that decreasing S1P production in cancer cells could lead to increased cytotoxic sensitivity.
Key Molecule: Sphingosine kinase 1 (SPHK1) [8]
Molecule Alteration Expression
Up-regulation
Resistant Disease Gastroesophageal cancer [ICD-11: 2B71.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model Gastroesophageal cancer tissue N.A.
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description S1P could lead to cytotoxic drug resistance in gastroesophegal cancer acting in an autocrine or paracrine manner via cell surface S1P receptors following transportation out of the cytosol. Alternatively S1P may mediate cytotoxic drug resistance acting intracellularly by counteracting apoptosis mediated by its pro-apoptotic precursor ceramide or interaction with known intracellular targets involved in cancer pathogenesis and cytotoxic drug resistance such as Histone deacetylase 1 (HDAC1) and Histone deacetylase 2 (HDAC 2) to which S1P directly binds and inhibits, and TNF Receptor-Associated Factor 2 (TRAF 2), or Protein Kinase C (PKC). S1P production controlled by SPHK1 and SGPL1 are key determinants of cytotoxic drug resistance and that decreasing S1P production in cancer cells could lead to increased cytotoxic sensitivity.
Gastric cancer [ICD-11: 2B72]
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-135a [5]
Molecule Alteration Expression
Up-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Sp1/DAPK2 signaling signaling pathway Inhibition hsa05231
In Vitro Model MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
NCI-N87 cells Gastric Homo sapiens (Human) CVCL_1603
MGC-803/OXA cells Gastric Homo sapiens (Human) CVCL_5334
SGC7901/OXA cells Gastric Homo sapiens (Human) CVCL_B0A1
SNU-5 cells Gastric Homo sapiens (Human) CVCL_0078
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR135a promotes gastric cancer progression and resistance to oxaliplatin. The mechanism whereby miR135a promotes GC pathogenesis appears to be the suppression of E2F1 expression and Sp1/DAPk2 pathway signaling.
Key Molecule: BLACAT1 overlapping LEMD1 locus (BLACAT1) [16]
Molecule Alteration Expression
Up-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation BLACAT1/miR361/ABCB1 signaling pathway Regulation hsa05206
Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell viability Activation hsa05200
In Vitro Model BGC-823 cells Gastric Homo sapiens (Human) CVCL_3360
MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Transwell assay
Mechanism Description BLACAT1 accelerates the oxaliplatin-resistance of gastric cancer via promoting ABCB1 protein expression by targeting miR-361.
Key Molecule: BLACAT1 overlapping LEMD1 locus (BLACAT1) [16]
Molecule Alteration Expression
Up-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell viability Activation hsa05200
miR125a/hexokinase 2 pathway Regulation hsa05206
In Vitro Model BGC-823 cells Gastric Homo sapiens (Human) CVCL_3360
MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Transwell assay
Mechanism Description BLACAT1 accelerates the oxaliplatin-resistance of gastric cancer via promoting ABCB1 protein expression by targeting miR-361.
Key Molecule: hsa-mir-361 [16]
Molecule Alteration Expression
Down-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation BLACAT1/miR361/ABCB1 signaling pathway Regulation hsa05206
Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell viability Activation hsa05200
In Vitro Model BGC-823 cells Gastric Homo sapiens (Human) CVCL_3360
MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Transwell assay
Mechanism Description BLACAT1 accelerates the oxaliplatin-resistance of gastric cancer via promoting ABCB1 protein expression by targeting miR-361.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [16]
Molecule Alteration Expression
Up-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell viability Activation hsa05200
miR125a/hexokinase 2 pathway Regulation hsa05206
In Vitro Model BGC-823 cells Gastric Homo sapiens (Human) CVCL_3360
MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Transwell assay
Mechanism Description BLACAT1 accelerates the oxaliplatin-resistance of gastric cancer via promoting ABCB1 protein expression by targeting miR-361.
Key Molecule: Multidrug resistance protein 1 (ABCB1) [16]
Molecule Alteration Expression
Up-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell viability Activation hsa05200
miR125a/hexokinase 2 pathway Regulation hsa05206
In Vitro Model BGC-823 cells Gastric Homo sapiens (Human) CVCL_3360
MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Transwell assay
Mechanism Description BLACAT1 accelerates the oxaliplatin-resistance of gastric cancer via promoting ABCB1 protein expression by targeting miR-361.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: Transforming growth factor beta 1 (TGFB1) [12]
Molecule Alteration Expression
Up-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation FAO signaling pathway Activation hsa04550
In Vitro Model AGS cells Gastric Homo sapiens (Human) CVCL_0139
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Enzyme-linked immunosorbent assay
Experiment for
Drug Resistance
MTT assay; Colony formation assays
Mechanism Description Transforming growth factor beta1 (TGF-beta1) secretion by MSCs activated SMAD2/3 through TGF-beta receptors and induced long non-coding RNA (LncRNA) MACC1-AS1 expression in GC cells, which promoted FAO-dependent stemness and chemoresistance through antagonizing miR-145-5p.
Key Molecule: hsa-miR-145-5p [12]
Molecule Alteration Expression
Down-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation FAO signaling pathway Activation hsa04550
In Vitro Model AGS cells Gastric Homo sapiens (Human) CVCL_0139
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Colony formation assays
Mechanism Description Transforming growth factor beta1 (TGF-beta1) secretion by MSCs activated SMAD2/3 through TGF-beta receptors and induced long non-coding RNA (LncRNA) MACC1-AS1 expression in GC cells, which promoted FAO-dependent stemness and chemoresistance through antagonizing miR-145-5p.
Key Molecule: MACC1 antisense RNA 1 (MACC1-AS1) [12]
Molecule Alteration Expression
Up-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation FAO signaling pathway Activation hsa04550
In Vitro Model AGS cells Gastric Homo sapiens (Human) CVCL_0139
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Colony formation assays
Mechanism Description Transforming growth factor beta1 (TGF-beta1) secretion by MSCs activated SMAD2/3 through TGF-beta receptors and induced long non-coding RNA (LncRNA) MACC1-AS1 expression in GC cells, which promoted FAO-dependent stemness and chemoresistance through antagonizing miR-145-5p.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Transcription factor E2F1 (E2F1) [5]
Molecule Alteration Expression
Down-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Sp1/DAPK2 signaling signaling pathway Inhibition hsa05231
In Vitro Model MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
NCI-N87 cells Gastric Homo sapiens (Human) CVCL_1603
MGC-803/OXA cells Gastric Homo sapiens (Human) CVCL_5334
SGC7901/OXA cells Gastric Homo sapiens (Human) CVCL_B0A1
SNU-5 cells Gastric Homo sapiens (Human) CVCL_0078
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR135a promotes gastric cancer progression and resistance to oxaliplatin. The mechanism whereby miR135a promotes GC pathogenesis appears to be the suppression of E2F1 expression and Sp1/DAPk2 pathway signaling.
Key Molecule: Sphingosine-1-phosphate lyase 1 (SGPL1) [8]
Molecule Alteration Expression
Down-regulation
Resistant Disease Gastric cardia adenocarcinoma [ICD-11: 2B72.2]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model AGS cells Gastric Homo sapiens (Human) CVCL_0139
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description S1P could lead to cytotoxic drug resistance in gastroesophegal cancer acting in an autocrine or paracrine manner via cell surface S1P receptors following transportation out of the cytosol. Alternatively S1P may mediate cytotoxic drug resistance acting intracellularly by counteracting apoptosis mediated by its pro-apoptotic precursor ceramide or interaction with known intracellular targets involved in cancer pathogenesis and cytotoxic drug resistance such as Histone deacetylase 1 (HDAC1) and Histone deacetylase 2 (HDAC 2) to which S1P directly binds and inhibits, and TNF Receptor-Associated Factor 2 (TRAF 2), or Protein Kinase C (PKC). S1P production controlled by SPHK1 and SGPL1 are key determinants of cytotoxic drug resistance and that decreasing S1P production in cancer cells could lead to increased cytotoxic sensitivity.
Key Molecule: Sphingosine kinase 1 (SPHK1) [8]
Molecule Alteration Expression
Up-regulation
Resistant Disease Gastric cardia adenocarcinoma [ICD-11: 2B72.2]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model AGS cells Gastric Homo sapiens (Human) CVCL_0139
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description S1P could lead to cytotoxic drug resistance in gastroesophegal cancer acting in an autocrine or paracrine manner via cell surface S1P receptors following transportation out of the cytosol. Alternatively S1P may mediate cytotoxic drug resistance acting intracellularly by counteracting apoptosis mediated by its pro-apoptotic precursor ceramide or interaction with known intracellular targets involved in cancer pathogenesis and cytotoxic drug resistance such as Histone deacetylase 1 (HDAC1) and Histone deacetylase 2 (HDAC 2) to which S1P directly binds and inhibits, and TNF Receptor-Associated Factor 2 (TRAF 2), or Protein Kinase C (PKC). S1P production controlled by SPHK1 and SGPL1 are key determinants of cytotoxic drug resistance and that decreasing S1P production in cancer cells could lead to increased cytotoxic sensitivity.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-195-5p [17]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Inhibition hsa05200
In Vitro Model MkN28 cells Gastric Homo sapiens (Human) CVCL_1416
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Overexpression of miR 195 5p inhibit multi drug resistance of gastric cancer cells via downregulating ZNF139.
Key Molecule: Protein lin-28 homolog A (CSDD1) [18]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
Lin28/miR107 pathway Regulation hsa05206
In Vitro Model MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
MkN28 cells Gastric Homo sapiens (Human) CVCL_1416
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Lin28 could inhibit the expression of miR-107, thereby up-regulating C-myc, P-gp and down-regulating Cyclin D1, subsequently result in chemo-resistance of gastric cancer cells. The Lin28/miR-107 pathway might be served as one of many signaling pathways that is associated with gastric cancer chemo-resistance.
Key Molecule: Protein lin-28 homolog B (CSDD2) [18]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
Lin28/miR107 pathway Regulation hsa05206
In Vitro Model MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
MkN28 cells Gastric Homo sapiens (Human) CVCL_1416
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Lin28 could inhibit the expression of miR-107, thereby up-regulating C-myc, P-gp and down-regulating Cyclin D1, subsequently result in chemo-resistance of gastric cancer cells. The Lin28/miR-107 pathway might be served as one of many signaling pathways that is associated with gastric cancer chemo-resistance.
Key Molecule: hsa-miR-107 [18]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
Lin28/miR107 pathway Regulation hsa05206
In Vitro Model MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
MkN28 cells Gastric Homo sapiens (Human) CVCL_1416
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Lin28 could inhibit the expression of miR-107, thereby up-regulating C-myc, P-gp and down-regulating Cyclin D1, subsequently result in chemo-resistance of gastric cancer cells. The Lin28/miR-107 pathway might be served as one of many signaling pathways that is associated with gastric cancer chemo-resistance.
Key Molecule: hsa-mir-218 [19]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-218 may inhibit efflux of ADM and oxaliplatin by down-regulating P-gp expression.
Key Molecule: hsa-mir-204 [20]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model NCI-N87 cells Gastric Homo sapiens (Human) CVCL_1603
GTL-16 cells Gastric Homo sapiens (Human) CVCL_7668
In Vivo Model CD1 nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-204 targeted Bcl-2 messenger RNA and increased responsiveness of GC cells to 5-fluorouracil and oxaliplatin treatment. Ectopic expression of Bcl-2 protein counteracted miR-204 pro-apoptotic activity in response to 5-fluorouracil.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: ATP-binding cassette sub-family B5 (ABCB5) [19]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-218 may inhibit efflux of ADM and oxaliplatin by down-regulating P-gp expression.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Zinc finger protein with KRAB and SCAN domains 1 (ZKSCAN1) [17]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Inhibition hsa05200
In Vitro Model MkN28 cells Gastric Homo sapiens (Human) CVCL_1416
Experiment for
Molecule Alteration
Western blot analysis; RIP assay; Luciferase reporter assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description Overexpression of miR 195 5p inhibit multi drug resistance of gastric cancer cells via downregulating ZNF139.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [19]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Higher miR-218 levels increased the level of Bax and reduced the level of Bcl-2 and miR-218 inhibits multidrug resistance (MDR) of gastric cancer cells by targeting Hedgehog/smoothened.
Key Molecule: Smoothened homolog (SMO) [19]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Higher miR-218 levels increased the level of Bax and reduced the level of Bcl-2 and miR-218 inhibits multidrug resistance (MDR) of gastric cancer cells by targeting Hedgehog/smoothened.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [20]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model NCI-N87 cells Gastric Homo sapiens (Human) CVCL_1603
GTL-16 cells Gastric Homo sapiens (Human) CVCL_7668
In Vivo Model CD1 nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-204 targeted Bcl-2 messenger RNA and increased responsiveness of GC cells to 5-fluorouracil and oxaliplatin treatment. Ectopic expression of Bcl-2 protein counteracted miR-204 pro-apoptotic activity in response to 5-fluorouracil.
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-19b-3p [21]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
In Vitro Model 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
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
NCM460 cells Colon Homo sapiens (Human) CVCL_0460
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Annexin V-PE and 7-AAD double staining method to examine cell viabilityNA; CCK8 assay; Flow cytometric analysis
Mechanism Description miR19b-3p promotes colon cancer proliferation and oxaliplatin-based chemoresistance by targeting SMAD4.
Key Molecule: Cytoskeleton regulator RNA (CYTOR) [22]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AKT signaling pathway Activation hsa04151
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
SW620 cells Colon Homo sapiens (Human) CVCL_0547
CaCo2 cells Colon Homo sapiens (Human) CVCL_0025
NCM460 cells Colon Homo sapiens (Human) CVCL_0460
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
CCK8 assay; Annexin V-APC/PI Apoptosis assay
Mechanism Description Linc00152 modulates the expression of ERBB4 through competitively binding miR193a-3p. AkT activation mediated by ERBB4 contributes to Linc00152-conferred L-OHP resistance, Linc00152 contributed to L-OHP resistance at least partly through activating AkT pathway.
Key Molecule: hsa-mir-34 [10]
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 Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
WST-1 assay; Flow cytometry assay
Mechanism Description miR-34a positively regulates OAZ2 transcription by directly targeting its 3UTR and OAZ2 Overexpression Effectively Rescues the Chemosensitivity Impaired by miR-34a Deficiency.
Key Molecule: hsa-mir-126 [2]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
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
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
293T cells Breast Homo sapiens (Human) CVCL_0063
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Over-expression of miR-126 in colon cancer cell was able to inhibit cell proliferation, promote cell apoptosis and reduce the invasive ability. miR-126 significantly enhanced the sensitivity of the colon cancer cell to chemotherapeutic drug. It has been shown that IRS1, SLC75A and TOM1 were the potential target genes of miR-126 in colon cancer.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Solute carrier family 7 member 5 (SLC7A5) [2]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
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
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
293T cells Breast Homo sapiens (Human) CVCL_0063
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Over-expression of miR-126 in colon cancer cell was able to inhibit cell proliferation, promote cell apoptosis and reduce the invasive ability. miR-126 significantly enhanced the sensitivity of the colon cancer cell to chemotherapeutic drug. It has been shown that IRS1, SLC75A and TOM1 were the potential target genes of miR-126 in colon cancer.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Mothers against decapentaplegic homolog 4 (SMAD4) [21]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
In Vitro Model 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
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
NCM460 cells Colon Homo sapiens (Human) CVCL_0460
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Luciferase reporter assay; Western blot analysis
Experiment for
Drug Resistance
Annexin V-PE and 7-AAD double staining method to examine cell viabilityNA; CCK8 assay; Flow cytometric analysis
Mechanism Description miR19b-3p promotes colon cancer proliferation and oxaliplatin-based chemoresistance by targeting SMAD4.
Key Molecule: Receptor tyrosine-protein kinase erbB-4 (ERBB4) [22]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AKT signaling pathway Activation hsa04151
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
SW620 cells Colon Homo sapiens (Human) CVCL_0547
CaCo2 cells Colon Homo sapiens (Human) CVCL_0025
NCM460 cells Colon Homo sapiens (Human) CVCL_0460
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Annexin V-APC/PI Apoptosis assay
Mechanism Description Linc00152 modulates the expression of ERBB4 through competitively binding miR193a-3p. AkT activation mediated by ERBB4 contributes to Linc00152-conferred L-OHP resistance, Linc00152 contributed to L-OHP resistance at least partly through activating AkT pathway.
Key Molecule: Ornithine decarboxylase antizyme 2 (OAZ2) [10]
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 Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
WST-1 assay; Flow cytometry assay
Mechanism Description miR-34a positively regulates OAZ2 transcription by directly targeting its 3UTR and OAZ2 Overexpression Effectively Rescues the Chemosensitivity Impaired by miR-34a Deficiency.
Key Molecule: Insulin receptor substrate 1 (IRS1) [2]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Regulation hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
293T cells Breast Homo sapiens (Human) CVCL_0063
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Over-expression of miR-126 in colon cancer cell was able to inhibit cell proliferation, promote cell apoptosis and reduce the invasive ability. miR-126 significantly enhanced the sensitivity of the colon cancer cell to chemotherapeutic drug. It has been shown that IRS1, SLC75A and TOM1 were the potential target genes of miR-126 in colon cancer.
Key Molecule: Target of Myb protein 1 (TOM1) [2]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
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
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
293T cells Breast Homo sapiens (Human) CVCL_0063
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Over-expression of miR-126 in colon cancer cell was able to inhibit cell proliferation, promote cell apoptosis and reduce the invasive ability. miR-126 significantly enhanced the sensitivity of the colon cancer cell to chemotherapeutic drug. It has been shown that IRS1, SLC75A and TOM1 were the potential target genes of miR-126 in colon cancer.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-137 [23]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description microRNA-137 chemosensitizes colon cancer cells to the chemotherapeutic drug OXA by targeting YBX1, miR137 was involved in repression of YBX1 expression through targeting its 3'-untranslated region. Down-regulation of miR137 conferred OXA resistance in parental cells, while over-expression of miR137 sensitized resistant cells to OXA, which was partly rescued by YBX1 siRNA.
Key Molecule: hsa-miR-409-3p [24]
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 proliferation Inhibition hsa05200
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
FHC cells Colon Homo sapiens (Human) CVCL_3688
DLD1 cells Colon Homo sapiens (Human) CVCL_0248
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
RkO cells Colon Homo sapiens (Human) CVCL_0504
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
CCD-18Co cells Colon Homo sapiens (Human) CVCL_2379
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The overexpression of miR 409-3p inhibited Beclin-1 expression and autophagic activity by binding to the 3'-untranslated region of Beclin-1 mRNA. In addition, the overexpression of miR 409-3p (+) the chemosensitivity of the oxaliplatin-sensitive and oxaliplatin-resistant colon cancer cells. The restoration of Beclin-1 abrogated these effects of miR 409-3p. In a xenograft model using nude mice, we examined the effects of miR 409-3p on tumor growth during chemotherapy. miR 409-3p overexpression sensitized the tumor to chemotherapy, while inhibiting chemotherapy-induced autophagy in a manner dependent on Beclin-1.
Key Molecule: hsa-miR-219a-5p [25]
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
In Vitro Model 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
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
NCM460 cells Colon Homo sapiens (Human) CVCL_0460
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Luciferase assay; Wound healing assay; Transwell assay; Flow cytometry assay
Mechanism Description The aberrant expression of miR-219-5p and Sall4 in colon cancer specimens, and confirmed that Sall4 was the direct target of miR-219-5p. Additionally, by aid of gain and loss of function assays, miR-219-5p was observed to play an inhibitory effect on cell proliferation, invasion and drug resistance.
Key Molecule: hsa-mir-320 [26]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
Wnt/Beta-catenin signaling pathway Inhibition hsa04310
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-320 enhances the sensitivity of human colon cancer cells to chemoradiotherapy in vitro by targeting FOXM1.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Y-box-binding protein 1 (YBX1) [23]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
Experiment for
Molecule Alteration
Dual luciferase assay; Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description microRNA-137 chemosensitizes colon cancer cells to the chemotherapeutic drug OXA by targeting YBX1, miR137 was involved in repression of YBX1 expression through targeting its 3'-untranslated region. Down-regulation of miR137 conferred OXA resistance in parental cells, while over-expression of miR137 sensitized resistant cells to OXA, which was partly rescued by YBX1 siRNA.
Key Molecule: Beclin-1 (BECN1) [24]
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 proliferation Inhibition hsa05200
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
FHC cells Colon Homo sapiens (Human) CVCL_3688
DLD1 cells Colon Homo sapiens (Human) CVCL_0248
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
RkO cells Colon Homo sapiens (Human) CVCL_0504
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
CCD-18Co cells Colon Homo sapiens (Human) CVCL_2379
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The overexpression of miR 409-3p inhibited Beclin-1 expression and autophagic activity by binding to the 3'-untranslated region of Beclin-1 mRNA. In addition, the overexpression of miR 409-3p (+) the chemosensitivity of the oxaliplatin-sensitive and oxaliplatin-resistant colon cancer cells. The restoration of Beclin-1 abrogated these effects of miR 409-3p. In a xenograft model using nude mice, we examined the effects of miR 409-3p on tumor growth during chemotherapy. miR 409-3p overexpression sensitized the tumor to chemotherapy, while inhibiting chemotherapy-induced autophagy in a manner dependent on Beclin-1.
Key Molecule: Sal-like protein 4 (SALL4) [25]
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
In Vitro Model 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
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
NCM460 cells Colon Homo sapiens (Human) CVCL_0460
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Luciferase assay; Wound healing assay; Transwell assay; Flow cytometry assay
Mechanism Description The aberrant expression of miR-219-5p and Sall4 in colon cancer specimens, and confirmed that Sall4 was the direct target of miR-219-5p. Additionally, by aid of gain and loss of function assays, miR-219-5p was observed to play an inhibitory effect on cell proliferation, invasion and drug resistance.
Key Molecule: Forkhead box protein M1 (FOXM1) [26]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
Wnt/Beta-catenin signaling pathway Inhibition hsa04310
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-320 enhances the sensitivity of human colon cancer cells to chemoradiotherapy in vitro by targeting FOXM1.
Colorectal cancer [ICD-11: 2B91]
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: GIHCG inhibitor of miR-200b/200a/429 expression (GIHCG) [27]
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 Cell apoptosis Inhibition hsa04210
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
LOVO cells Colon Homo sapiens (Human) CVCL_0399
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Long noncoding RNA GIHCG induces cancer progression and chemoresistance and indicates poor prognosis in colorectal cancer.
Key Molecule: piR-hsa-54265 [28]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal adenocarcinoma [ICD-11: 2B91.2]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell proliferation Activation hsa05200
STAT3 signaling pathway Activation hsa04550
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
CCK8 assay; Colony formation assays
Mechanism Description piR-54265 binds PIWIL2 promotes CRC cell proliferation and invasiveness and 5-FU and oxaliplatin resistance via promoting oncogenic STAT3 signaling.
Key Molecule: hsa-let-7a [29]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
RkO cells Colon Homo sapiens (Human) CVCL_0504
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay; Transwell assays and wound healing assay; Flow cytometry assay
Mechanism Description ANRIL promotes chemoresistance via disturbing expression of ABCC1 by inhibiting the expression of Let-7a in colorectal cancer.
Key Molecule: hsa-mir-216b [30]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/PTEN/AKT signaling pathway Regulation hsa04151
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
SW620 cells Colon Homo sapiens (Human) CVCL_0547
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HCT8 cells Colon Homo sapiens (Human) CVCL_2478
COLO 205 cells Colon Homo sapiens (Human) CVCL_0218
CCD-18Co cells Colon Homo sapiens (Human) CVCL_2379
COLO-678 cells Colon Homo sapiens (Human) CVCL_1129
HT55 cells Colon Homo sapiens (Human) CVCL_1294
LS1034 cells Colon Homo sapiens (Human) CVCL_1382
SW1417 cells Colon Homo sapiens (Human) CVCL_1717
SW403 cells Colon Homo sapiens (Human) CVCL_0545
SW48 cells Colon Homo sapiens (Human) CVCL_1724
In Vivo Model BALB/c mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-216b promotes cell growth and enhances chemosensitivity of colorectal cancer by suppressing PDZ-binding kinase.
Key Molecule: hsa-mir-218 [31]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
HCT-116/L-OHP cells Kidney Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis of apoptosis
Mechanism Description Down-regulation of YEATS4 by miR218 sensitizes colorectal cancer cells to L-OHP-induced cell apoptosis by inhibiting cytoprotective autophagy.
Key Molecule: Maternally expressed 3 (MEG3) [32]
Molecule Alteration Expression
Down-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 viability Activation hsa05200
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Overexpression of MEG3 improved oxaliplatin sensitivity of HT29/OXA and HCT116/OXA cells via suppressing miR-141 expression and upregulating PDCD4.
Key Molecule: hsa-miR-625-3p [33]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal adenocarcinoma [ICD-11: 2B91.2]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
p38/MAPK signaling pathway Inhibition hsa04010
In Vitro Model HEK293 Flp pFRT/eGFP cells Kidney Homo sapiens (Human) CVCL_U427
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Inactivation of MAP2k6-p38 signalling as one likely mechanism of oxaliplatin resistance, and miR-625-3p induces oxaliplatin resistance by abrogating MAP2k6-p38-regulated apoptosis and cell cycle control networks.
Key Molecule: hsa-mir-520g [34]
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
p53/miR520g/p21 signaling pathway Regulation hsa05206
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
RkO cells Colon Homo sapiens (Human) CVCL_0504
FET cells Colon Homo sapiens (Human) CVCL_A604
GEO cells Colon Homo sapiens (Human) CVCL_0271
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay; ELISA assay
Mechanism Description p53 suppresses miR-520g expression and that deletion of p53 up-regulates miR-520g expression. Inhibition of miR-520g in p53 / cells increased their sensitivity to 5-FU treatment. miR-520g conferred resistance to 5-FU-induced apoptosis through the inhibition of p21 expression, which is a direct target of miR-520g. Rescued expression of p21 in miR-520g-expressing colon cancer cells sensitized them to 5-FU-induced apoptosis. Importantly, experiments in tumor xenograft mouse models demonstrate that miR-520g reduced the effectiveness of 5-FU in the inhibition of tumor growth in vivo. Moreover, studies of colorectal cancer specimens indicate a positive correlation between miR-520g expression and chemoresistance.
Key Molecule: hsa-miR-17-5p [35]
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 invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
PTEN/AKT/PI3K signaling pathway Activation hsa05235
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
COLO205 cells Colon Homo sapiens (Human) CVCL_F402
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 The expression level of miRNA-17-5p was found increased in chemoresistant patients. Significantly higher expression levels of miR-17-5p were found in CRC patients with distant metastases and higher clinical stages. kaplan-Meier analysis showed that CRC patients with higher levels of miR-17-5p had reduced survival, especially in patients who had previously received chemotherapy. Overexpression of miR-17-5p promoted COLO205 cell invasiveness. PTEN was a target of miR-17-5p in the colon cancer cells, and their context-specific interactions were responsible for multiple drug-resistance. Chemotherapy was found to increase the expression levels of miR-17-5p, which further repressed PTEN levels, contributing to the development of chemo-resistance.
Key Molecule: hsa-mir-19a [36]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Response evaluation criteria in solid tumors assay
Mechanism Description Aberrant expression of serum miR-19a in FOLFOX chemotherapy resistance patients, suggesting serum miR-19a could be a potential molecular biomarker for predicting and monitoring resistance to first-line FOLFOX chemotherapy regimens in advanced colorectal cancer patients.
Key Molecule: hsa-mir-203 [11]
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 Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
RkO cells Colon Homo sapiens (Human) CVCL_0504
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description We validated ATM as a bona fide target of miR-203 in CRC cells. Mutation of the putative miR-203 binding site in the 3' untranslated region (3'UTR) of the ATM mRNA abolished the inhibitory effect of miR-203 on ATM. Furthermore, stable knockdown of ATM induced resistance to oxaliplatin in chemo-na ve CRC cells.
Key Molecule: hsa-mir-153 [3]
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 invasion Activation hsa05200
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
SW48 cells Colon Homo sapiens (Human) CVCL_1724
COLO205 cells Colon Homo sapiens (Human) CVCL_F402
In Vivo Model SCID nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTS assay; Soft agar colony forming ability assay; Flow cytometry assay
Mechanism Description miR-153 promoted invasiveness indirectly by inducing MMP9 production, whereas drug resistance was mediated directly by inhibiting the Forkhead transcription factor FOXO3a.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: Long non-protein coding RNA (CCAL) [1]
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 Beta-catenin signaling pathway Activation hsa04520
Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description LncRNA CCAL can be transferred from CAFs to cancer cells via exosomes, and exosome-enriched CCAL promoted Oxa and 5-FU chemoresistance of CRC cells.
Key Molecule: hsa-miR-1229-5p [37]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PTEN/AKT signaling pathway Regulation hsa05235
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-1246, miR-21-5p, miR-96-5p and miR-1229-5p from serum exosomes involved in chemotherapy resistance may be new therapeutic targets, downregulating these miRNAs may promote CRC cell sensitivity to chemotherapeutic drugs.
Key Molecule: hsa-miR-1246 [37]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PTEN/AKT signaling pathway Regulation hsa05235
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-1246, miR-21-5p, miR-96-5p and miR-1229-5p from serum exosomes involved in chemotherapy resistance may be new therapeutic targets, downregulating these miRNAs may promote CRC cell sensitivity to chemotherapeutic drugs.
Key Molecule: hsa-miR-21-5p [37]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PTEN/AKT signaling pathway Regulation hsa05235
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-1246, miR-21-5p, miR-96-5p and miR-1229-5p from serum exosomes involved in chemotherapy resistance may be new therapeutic targets, downregulating these miRNAs may promote CRC cell sensitivity to chemotherapeutic drugs.
Key Molecule: hsa-miR-96-5p [37]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PTEN/AKT signaling pathway Regulation hsa05235
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-1246, miR-21-5p, miR-96-5p and miR-1229-5p from serum exosomes involved in chemotherapy resistance may be new therapeutic targets, downregulating these miRNAs may promote CRC cell sensitivity to chemotherapeutic drugs.
Key Molecule: Phosphatase and tensin homolog (PTEN) [37]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PTEN/AKT signaling pathway Regulation hsa05235
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-1246, miR-21-5p, miR-96-5p and miR-1229-5p from serum exosomes involved in chemotherapy resistance may be new therapeutic targets, downregulating these miRNAs may promote CRC cell sensitivity to chemotherapeutic drugs.
Key Molecule: Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) [38]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
FHC cells Colon Homo sapiens (Human) CVCL_3688
SW620 cells Colon Homo sapiens (Human) CVCL_0547
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Boyden chambers cell migration and invasion assays
Mechanism Description MALAT1 tethers EZH2 to CDH1 promoter and suppresses miR218 during oxaliplatin treatment, which finally promotes colorectal cancer cell EMT, metastasis, and chemoresistance.
Key Molecule: Cadherin-1 (CDH1) [38]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
FHC cells Colon Homo sapiens (Human) CVCL_3688
SW620 cells Colon Homo sapiens (Human) CVCL_0547
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Boyden chambers cell migration and invasion assays
Mechanism Description MALAT1 tethers EZH2 to CDH1 promoter and suppresses miR218 during oxaliplatin treatment, which finally promotes colorectal cancer cell EMT, metastasis, and chemoresistance.
Key Molecule: Histone-lysine N-methyltransferase EZH2 (EZH2) [38]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
FHC cells Colon Homo sapiens (Human) CVCL_3688
SW620 cells Colon Homo sapiens (Human) CVCL_0547
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Boyden chambers cell migration and invasion assays
Mechanism Description MALAT1 tethers EZH2 to CDH1 promoter and suppresses miR218 during oxaliplatin treatment, which finally promotes colorectal cancer cell EMT, metastasis, and chemoresistance. MALAT1 mediates oxaliplatin-induced EMT through EZH2 and interacts with miR218.
Key Molecule: hsa-mir-141 [39]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Beta-catenin signaling pathway Regulation hsa04520
Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
In Vivo Model NOD/SCID mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description CAF-derived exosomes transfer LncRNA H19 to colorectal cancer cells and H19 activated the beta-catenin pathway via acting as a competing endogenous RNA sponge for miR-141, while miR-141 inhibited the stemness of CRC cells.
Key Molecule: H19, imprinted maternally expressed transcript (H19) [39]
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 Beta-catenin signaling pathway Activation hsa04520
Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
In Vivo Model NOD/SCID mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description CAF-derived exosomes transfer LncRNA H19 to colorectal cancer cells and H19 activated the beta-catenin pathway via acting as a competing endogenous RNA sponge for miR-141, while miR-141 inhibited the stemness of CRC cells.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Piwi-like protein 2 (PIWIL2) [28]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal adenocarcinoma [ICD-11: 2B91.2]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell proliferation Activation hsa05200
STAT3 signaling pathway Activation hsa04550
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
CCK8 assay; Colony formation assays
Mechanism Description piR-54265 binds PIWIL2 promotes CRC cell proliferation and invasiveness and 5-FU and oxaliplatin resistance via promoting oncogenic STAT3 signaling.
Key Molecule: T-LAK cell-originated protein kinase(PBK) [30]
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 PI3K/PTEN/AKT signaling pathway Regulation hsa04151
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
SW620 cells Colon Homo sapiens (Human) CVCL_0547
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HCT8 cells Colon Homo sapiens (Human) CVCL_2478
COLO 205 cells Colon Homo sapiens (Human) CVCL_0218
CCD-18Co cells Colon Homo sapiens (Human) CVCL_2379
COLO-678 cells Colon Homo sapiens (Human) CVCL_1129
HT55 cells Colon Homo sapiens (Human) CVCL_1294
LS1034 cells Colon Homo sapiens (Human) CVCL_1382
SW1417 cells Colon Homo sapiens (Human) CVCL_1717
SW403 cells Colon Homo sapiens (Human) CVCL_0545
SW48 cells Colon Homo sapiens (Human) CVCL_1724
In Vivo Model BALB/c mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Luciferase activity assay; Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-216b promotes cell growth and enhances chemosensitivity of colorectal cancer by suppressing PDZ-binding kinase.
Key Molecule: YEATS domain-containing protein 4 (YEATS4) [31]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
HCT-116/L-OHP cells Kidney Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis of apoptosis
Mechanism Description Down-regulation of YEATS4 by miR218 sensitizes colorectal cancer cells to L-OHP-induced cell apoptosis by inhibiting cytoprotective autophagy.
Key Molecule: Programmed cell death protein 4 (PDCD4) [32]
Molecule Alteration Expression
Down-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 viability Activation hsa05200
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Western blot analysis; Luciferase reporter assay; RNA pull-down assay; RIP assay
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Overexpression of MEG3 improved oxaliplatin sensitivity of HT29/OXA and HCT116/OXA cells via suppressing miR-141 expression and upregulating PDCD4.
Key Molecule: MAPK/ERK kinase 6 (MEK6) [33]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colorectal adenocarcinoma [ICD-11: 2B91.2]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
p38/MAPK signaling pathway Inhibition hsa04010
In Vitro Model HEK293 Flp pFRT/eGFP cells Kidney Homo sapiens (Human) CVCL_U427
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Inactivation of MAP2k6-p38 signalling as one likely mechanism of oxaliplatin resistance, and miR-625-3p induces oxaliplatin resistance by abrogating MAP2k6-p38-regulated apoptosis and cell cycle control networks.
Key Molecule: Ribonuclease P protein subunit p21 (RPP21) [34]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
p53/miR520g/p21 signaling pathway Regulation hsa05206
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
RkO cells Colon Homo sapiens (Human) CVCL_0504
FET cells Colon Homo sapiens (Human) CVCL_A604
GEO cells Colon Homo sapiens (Human) CVCL_0271
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; ELISA assay
Mechanism Description p53 suppresses miR-520g expression and that deletion of p53 up-regulates miR-520g expression. Inhibition of miR-520g in p53 / cells increased their sensitivity to 5-FU treatment. miR-520g conferred resistance to 5-FU-induced apoptosis through the inhibition of p21 expression, which is a direct target of miR-520g. Rescued expression of p21 in miR-520g-expressing colon cancer cells sensitized them to 5-FU-induced apoptosis. Importantly, experiments in tumor xenograft mouse models demonstrate that miR-520g reduced the effectiveness of 5-FU in the inhibition of tumor growth in vivo. Moreover, studies of colorectal cancer specimens indicate a positive correlation between miR-520g expression and chemoresistance.
Key Molecule: Phosphatase and tensin homolog (PTEN) [35]
Molecule Alteration Expression
Down-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 invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
PTEN/AKT/PI3K signaling pathway Activation hsa05235
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
COLO205 cells Colon Homo sapiens (Human) CVCL_F402
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description The expression level of miRNA-17-5p was found increased in chemoresistant patients. Significantly higher expression levels of miR-17-5p were found in CRC patients with distant metastases and higher clinical stages. kaplan-Meier analysis showed that CRC patients with higher levels of miR-17-5p had reduced survival, especially in patients who had previously received chemotherapy. Overexpression of miR-17-5p promoted COLO205 cell invasiveness. PTEN was a target of miR-17-5p in the colon cancer cells, and their context-specific interactions were responsible for multiple drug-resistance. Chemotherapy was found to increase the expression levels of miR-17-5p, which further repressed PTEN levels, contributing to the development of chemo-resistance.
Key Molecule: Serine-protein kinase ATM (ATM) [11]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
RkO cells Colon Homo sapiens (Human) CVCL_0504
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description We validated ATM as a bona fide target of miR-203 in CRC cells. Mutation of the putative miR-203 binding site in the 3' untranslated region (3'UTR) of the ATM mRNA abolished the inhibitory effect of miR-203 on ATM. Furthermore, stable knockdown of ATM induced resistance to oxaliplatin in chemo-na ve CRC cells.
Key Molecule: Forkhead box protein O3 (FOXO3) [3]
Molecule Alteration Expression
Down-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 invasion Activation hsa05200
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
SW48 cells Colon Homo sapiens (Human) CVCL_1724
COLO205 cells Colon Homo sapiens (Human) CVCL_F402
In Vivo Model SCID nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay; Soft agar colony forming ability assay; Flow cytometry assay
Mechanism Description miR-153 promoted invasiveness indirectly by inducing MMP9 production, whereas drug resistance was mediated directly by inhibiting the Forkhead transcription factor FOXO3a.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-340 [40]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
NCM460 cells Colon Homo sapiens (Human) CVCL_0460
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 The ectopic overexpression of miR340 in CRC cell lines resulted in growth inhibition, apoptosis and enhanced chemosensitivity in vitro and in vivo, which was mediated by directly targeting RLIP76.
Key Molecule: hsa-mir-145 [41]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HCT116/L-OHP cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR145 inhibits drug resistance to L-OHP of HCT116 cells through suppressing the expression of target gene GPR98.
Key Molecule: hsa-miR-503-5p [42]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal carcinoma [ICD-11: 2B91.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation P53/PUMA signaling pathway Activation hsa04115
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
TUNEL and ki67 staining; Caspase-3 activity assay; MTT assay
Mechanism Description miR503-5p induces oxaliplatin resistance through the inhibition of apoptosis by reducing PUMA expression, which could direct target by miR503-5p. P53 suppresses miR503-5p expression.
Key Molecule: hsa-mir-506 [43]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Wnt/Beta-catenin signaling pathway Inhibition hsa04310
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT116-OxR cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description miR-506 overexpression in HCT116-OxR cells enhances oxaliplatin sensitivity by inhibiting MDR1/P-gp expression via down-regulation of the Wnt/beta-catenin pathway.
Key Molecule: hsa-mir-141 [44]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW620 cells Colon Homo sapiens (Human) CVCL_0547
HEY cells Ovary Homo sapiens (Human) CVCL_0297
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
Flow cytometry assay; MTS assay
Mechanism Description miR141 inhibited CRC cell proliferation via targeting cyclin D2, which is involved in cell cycle regulation, and inhibited the mainte.nce of CSC stemness, thereby enhancing drug susceptibility.
Key Molecule: hsa-miR-139-5p [45]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal carcinoma [ICD-11: 2B91.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT8 cells Colon Homo sapiens (Human) CVCL_2478
In Vivo Model Mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay
Mechanism Description miR139-5p reverses CD44+/CD133+-associated multidrug resistance by downregulating NOTCH1 in colorectal carcinoma cells.
Key Molecule: hsa-mir-122 [46]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description The recovery of miR-122 expression can sensitize SW480/OR and HT29/OR cells to oxaliplatin-mediated apoptosis through the inhibition of XIAP expression.
Key Molecule: hsa-mir-141 [32]
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 apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Overexpression of MEG3 improved oxaliplatin sensitivity of HT29/OXA and HCT116/OXA cells via suppressing miR-141 expression and upregulating PDCD4.
Key Molecule: Maternally expressed 3 (MEG3) [32]
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 apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Overexpression of MEG3 improved oxaliplatin sensitivity of HT29/OXA and HCT116/OXA cells via suppressing miR-141 expression and upregulating PDCD4.
Key Molecule: hsa-mir-135b [47]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
FOXO1 signaling pathway Activation hsa04068
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
SW620 cells Colon Homo sapiens (Human) CVCL_0547
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Knockdown of Mir-135b Sensitizes Colorectal Cancer Cells to Oxaliplatin-Induced Apoptosis Through Increase of FOXO1.
Key Molecule: hsa-miR-425-5p [48]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CellTiter-Glo assay
Mechanism Description miR-425-5p is up-regulated in HCT116-R cells with acquired resistance to 5-fluouracil and OX compared with the parental HCT116 cells. Inhibition of miR-425-5p increases sensitivity to anti-cancer drugs by regulating apoptosis-related protein PDCD10 both in vitro and in vivo.
Key Molecule: hsa-miR-204-5p [49]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
miR204-5p/RAB22A signaling pathway Regulation hsa05206
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
HCT8 cells Colon Homo sapiens (Human) CVCL_2478
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-204-5p is frequently downregulated in colorectal cancer tissues, and survival analysis showed that the downregulation of miR-204-5p in colorectal cancer was associated with poor prognoses. Ectopic miR-204-5p expression repressed colorectal cancer cell growth both in vitro and in vivo. Moreover, restoring miR-204-5p expression inhibited colorectal cancer migration and invasion and promoted tumor sensitivity to chemotherapy. Mechanistic investigations revealed that RAB22A, a member of the RAS oncogene family, is a direct functional target of miR-204-5p in colorectal cancer. Furthermore, RAB22A protein levels in colorectal cancer tissues were frequently increased and negatively associated with miR-204-5p levels and survival time.
Key Molecule: hsa-mir-143 [50]
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 apoptosis Activation hsa04210
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
PI3K/AKT/HIF-1/VEGF signaling pathway Activation hsa04151
In Vitro Model SW1116 cells Colon Homo sapiens (Human) CVCL_0544
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Overexpression of miR-143 inhibited cell proliferation, migration, tumor growth and angiogenesis and increased chemosensitivity to oxaliplatin treatment in an IGF-IR-dependent manner.
Key Molecule: hsa-mir-222 [51]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal carcinoma [ICD-11: 2B91.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description ADAM-17 (a desintegrin and metalloproteases 17) is a novel multidrug resistance (MDR) mechanism in multidrug-resistant colorectal carcinoma (CRC). The presence of miR-222 was consistently inversely proportionate to the expression levels of ADAM-17. The loss of miR-222 in the HCT116/L-OHP and HCT-8/VCR MDR cell lines contributed to the overexpression of ADAM-17 and sensitized the HCT116/L-OHP and HCT-8/VCR MDR cells to some anticancer drugs.
Key Molecule: hsa-miR-297 [52]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal carcinoma [ICD-11: 2B91.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
RT-PCR; qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description MRP-2 (MDR-associated protein 2) is an important MDR protein in platinum-drug-resistance cells, miR-297 in MDR colorectal carcinoma cells reduced MRP-2 protein level and sensitized these cells to anti-cancer drugs in vitro and in vivo.
Key Molecule: hsa-mir-1915 [53]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal carcinoma [ICD-11: 2B91.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT-116/L-OHP cells Kidney Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Elevated levels of miR-1915 in the mimics-transfected HCT116/L-OHP cells reduced Bcl-2 protein level and the luciferase activity of a Bcl-2 3'-untranslated region-based reporter, and also sensitized these cells to some anticancer drugs. miR-1915 could play a role in the development of MDR in colorectal carcinoma cells at least in part by modulation of apoptosis via targeting Bcl-2.
Key Molecule: hsa-mir-20a [54]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal adenocarcinoma [ICD-11: 2B91.2]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
SW620 cells Colon Homo sapiens (Human) CVCL_0547
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-20a overexpression resulted in resistance to these chemotherapy agents, while miR-20a knockdown led to sensitization, miR-20a down-regulated both BNIP2 mRNA and BNIP2 protein levels. miR-20a down-regulated the expression of the proapoptotic factor BNIP2, leading to an imbalance of anti-apoptosis and pro-apoptosis factors, resulting in the blockage of events leading to apoptosis.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [43]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Wnt/Beta-catenin signaling pathway Inhibition hsa04310
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT116-OxR cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Western blot analysis; Immunofluorescence staining assay
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description miR-506 overexpression in HCT116-OxR cells enhances oxaliplatin sensitivity by inhibiting MDR1/P-gp expression via down-regulation of the Wnt/beta-catenin pathway.
Key Molecule: ATP-binding cassette sub-family C2 (ABCC2) [52]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal carcinoma [ICD-11: 2B91.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description MRP-2 (MDR-associated protein 2) is an important MDR protein in platinum-drug-resistance cells, miR-297 in MDR colorectal carcinoma cells reduced MRP-2 protein level and sensitized these cells to anti-cancer drugs in vitro and in vivo.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: hsa-mir-218 [38]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
FHC cells Colon Homo sapiens (Human) CVCL_3688
SW620 cells Colon Homo sapiens (Human) CVCL_0547
Experiment for
Molecule Alteration
RT-PCR; qRT-PCR
Experiment for
Drug Resistance
Boyden chambers cell migration and invasion assays
Mechanism Description miR218 is a tumor-suppressor gene and could significantly suppress the EMT process, miR218 promoted cell apoptosis and enhanced 5-FU-based chemosensitivity in colorectal cancer cells by targeting BIRC5.
Key Molecule: hsa-miR-139-5p [55]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation BCL2 signaling pathway Regulation hsa04210
Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
SW620 cells Colon Homo sapiens (Human) CVCL_0547
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
RkO cells Colon Homo sapiens (Human) CVCL_0504
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
LS174T cells Colon Homo sapiens (Human) CVCL_1384
COLO205 cells Colon Homo sapiens (Human) CVCL_F402
In Vivo Model Mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Transwell assay
Mechanism Description BCL2 is a direct target of miR-139-5p in colorectal cancer cells and showed that the tumour suppressor activity of miR-139-5p is mediated by the modulation of BCL2 expression. BCL2 family proteins regulate and contribute to programmed cell death or apoptosis. The cell apoptosis results showed the induction of apoptotic cells contributed greatly to 5-Fu and OXA drug sensitivity, which was consistent with the multidrug resistance mechanisms. miR-139-5p is downregulated in colorectal cancer cells and tissues, and its inhibitory effects on cell migration, invasion, and drug sensitivity are mediated by the downregulation of its target BCL2.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: RalA-binding protein 1 (RALBP1) [40]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
NCM460 cells Colon Homo sapiens (Human) CVCL_0460
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Luciferase reporter assay
Experiment for
Drug Resistance
CCK8 assay; Flow cytometric analysis
Mechanism Description The ectopic overexpression of miR340 in CRC cell lines resulted in growth inhibition, apoptosis and enhanced chemosensitivity in vitro and in vivo, which was mediated by directly targeting RLIP76.
Key Molecule: Adhesion G-protein coupled receptor V1 (ADGRV1) [41]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HCT116/L-OHP cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR145 inhibits drug resistance to L-OHP of HCT116 cells through suppressing the expression of target gene GPR98.
Key Molecule: Bcl-2-binding component 3 (BBC3) [42]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal carcinoma [ICD-11: 2B91.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation P53/PUMA signaling pathway Activation hsa04115
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Luciferase reporter assay
Experiment for
Drug Resistance
TUNEL and ki67 staining; Caspase-3 activity assay; MTT assay
Mechanism Description miR503-5p induces oxaliplatin resistance through the inhibition of apoptosis by reducing PUMA expression, which could direct target by miR503-5p. P53 suppresses miR503-5p expression.
Key Molecule: G1/S-specific cyclin-D2 (CCND2) [44]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW620 cells Colon Homo sapiens (Human) CVCL_0547
HEY cells Ovary Homo sapiens (Human) CVCL_0297
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay; MTS assay
Mechanism Description miR141 inhibited CRC cell proliferation via targeting cyclin D2, which is involved in cell cycle regulation, and inhibited the mainte.nce of CSC stemness, thereby enhancing drug susceptibility.
Key Molecule: Neurogenic locus notch homolog protein 1 (NOTCH1) [45]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal carcinoma [ICD-11: 2B91.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT8 cells Colon Homo sapiens (Human) CVCL_2478
In Vivo Model Mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay
Mechanism Description miR139-5p reverses CD44+/CD133+-associated multidrug resistance by downregulating NOTCH1 in colorectal carcinoma cells.
Key Molecule: E3 ubiquitin-protein ligase XIAP (XIAP) [46]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description The recovery of miR-122 expression can sensitize SW480/OR and HT29/OR cells to oxaliplatin-mediated apoptosis through the inhibition of XIAP expression.
Key Molecule: Programmed cell death protein 4 (PDCD4) [32]
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 apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
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
MTT assay; Flow cytometry assay
Mechanism Description Overexpression of MEG3 improved oxaliplatin sensitivity of HT29/OXA and HCT116/OXA cells via suppressing miR-141 expression and upregulating PDCD4.
Key Molecule: Forkhead box protein O1 (FOXO1) [47]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
FOXO1 signaling pathway Activation hsa04068
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
SW620 cells Colon Homo sapiens (Human) CVCL_0547
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Knockdown of Mir-135b Sensitizes Colorectal Cancer Cells to Oxaliplatin-Induced Apoptosis Through Increase of FOXO1.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [55]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation BCL2 signaling pathway Regulation hsa04210
Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
SW620 cells Colon Homo sapiens (Human) CVCL_0547
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
RkO cells Colon Homo sapiens (Human) CVCL_0504
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
LS174T cells Colon Homo sapiens (Human) CVCL_1384
COLO205 cells Colon Homo sapiens (Human) CVCL_F402
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Transwell assay
Mechanism Description BCL2 is a direct target of miR-139-5p in colorectal cancer cells and showed that the tumour suppressor activity of miR-139-5p is mediated by the modulation of BCL2 expression. BCL2 family proteins regulate and contribute to programmed cell death or apoptosis. The cell apoptosis results showed the induction of apoptotic cells contributed greatly to 5-Fu and OXA drug sensitivity, which was consistent with the multidrug resistance mechanisms. miR-139-5p is downregulated in colorectal cancer cells and tissues, and its inhibitory effects on cell migration, invasion, and drug sensitivity are mediated by the downregulation of its target BCL2.
Key Molecule: Programmed cell death protein 10 (PDCD10) [48]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model 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
CellTiter-Glo assay
Mechanism Description miR-425-5p is up-regulated in HCT116-R cells with acquired resistance to 5-fluouracil and OX compared with the parental HCT116 cells. Inhibition of miR-425-5p increases sensitivity to anti-cancer drugs by regulating apoptosis-related protein PDCD10 both in vitro and in vivo.
Key Molecule: Ras-related protein Rab-22A (RAP22A) [49]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
miR204-5p/RAB22A signaling pathway Regulation hsa05206
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
HCT8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-204-5p is frequently downregulated in colorectal cancer tissues, and survival analysis showed that the downregulation of miR-204-5p in colorectal cancer was associated with poor prognoses. Ectopic miR-204-5p expression repressed colorectal cancer cell growth both in vitro and in vivo. Moreover, restoring miR-204-5p expression inhibited colorectal cancer migration and invasion and promoted tumor sensitivity to chemotherapy. Mechanistic investigations revealed that RAB22A, a member of the RAS oncogene family, is a direct functional target of miR-204-5p in colorectal cancer. Furthermore, RAB22A protein levels in colorectal cancer tissues were frequently increased and negatively associated with miR-204-5p levels and survival time.
Key Molecule: Insulin-like growth factor 1 receptor (IGF1R) [50]
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 migration Activation hsa04670
Cell proliferation Inhibition hsa05200
PI3K/AKT/HIF-1/VEGF signaling pathway Activation hsa04151
In Vitro Model SW1116 cells Colon Homo sapiens (Human) CVCL_0544
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Overexpression of miR-143 inhibited cell proliferation, migration, tumor growth and angiogenesis and increased chemosensitivity to oxaliplatin treatment in an IGF-IR-dependent manner.
Key Molecule: TNF alpha converting enzyme (ADAM17) [51]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal carcinoma [ICD-11: 2B91.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description ADAM-17 (a desintegrin and metalloproteases 17) is a novel multidrug resistance (MDR) mechanism in multidrug-resistant colorectal carcinoma (CRC). The presence of miR-222 was consistently inversely proportionate to the expression levels of ADAM-17. The loss of miR-222 in the HCT116/L-OHP and HCT-8/VCR MDR cell lines contributed to the overexpression of ADAM-17 and sensitized the HCT116/L-OHP and HCT-8/VCR MDR cells to some anticancer drugs.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [53]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal carcinoma [ICD-11: 2B91.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT-116/L-OHP cells Kidney Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Elevated levels of miR-1915 in the mimics-transfected HCT116/L-OHP cells reduced Bcl-2 protein level and the luciferase activity of a Bcl-2 3'-untranslated region-based reporter, and also sensitized these cells to some anticancer drugs. miR-1915 could play a role in the development of MDR in colorectal carcinoma cells at least in part by modulation of apoptosis via targeting Bcl-2.
Key Molecule: Bcl-2/adenovirus E1B 19 kDa protein-interacting protein 2 (BNIP2) [54]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal adenocarcinoma [ICD-11: 2B91.2]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
SW620 cells Colon Homo sapiens (Human) CVCL_0547
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-20a overexpression resulted in resistance to these chemotherapy agents, while miR-20a knockdown led to sensitization, miR-20a down-regulated both BNIP2 mRNA and BNIP2 protein levels. miR-20a down-regulated the expression of the proapoptotic factor BNIP2, leading to an imbalance of anti-apoptosis and pro-apoptosis factors, resulting in the blockage of events leading to apoptosis.
Liver cancer [ICD-11: 2C12]
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: Hepatocellular carcinoma up-regulated long non-coding RNA (HULC) [13]
Molecule Alteration Expression
Up-regulation
Resistant 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
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
PLC cells Liver Homo sapiens (Human) CVCL_0485
L02 cells Liver Homo sapiens (Human) CVCL_6926
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description LncRNA HULC triggers autophagy via stabilizing Sirt1 and attenuates the chemosensitivity of HCC cells. HULC inhibits the expression and activity of miR6825-5p, miR6845-5p and miR6886-3p.
Key Molecule: hsa-miR-6825-5p [13]
Molecule Alteration Expression
Down-regulation
Resistant 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
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
PLC cells Liver Homo sapiens (Human) CVCL_0485
L02 cells Liver Homo sapiens (Human) CVCL_6926
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR6825-5p, miR6845-5p and miR6886-3p could decrease the level of USP22 protein by binding to the 3'-untranlated region of USP22 mRNA. All the three microRNAs (miRNAs) were downregulated by HULC, which resulted in the elevation of USP22. The pathway 'HULC/USP22/Sirt1/ protective autophagy' attenuates the sensitivity of HCC cells to chemotherapeutic agents.
Key Molecule: hsa-miR-6845-5p [13]
Molecule Alteration Expression
Down-regulation
Resistant 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
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
PLC cells Liver Homo sapiens (Human) CVCL_0485
L02 cells Liver Homo sapiens (Human) CVCL_6926
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR6825-5p, miR6845-5p and miR6886-3p could decrease the level of USP22 protein by binding to the 3'-untranlated region of USP22 mRNA. All the three microRNAs (miRNAs) were downregulated by HULC, which resulted in the elevation of USP22. The pathway 'HULC/USP22/Sirt1/ protective autophagy' attenuates the sensitivity of HCC cells to chemotherapeutic agents.
Key Molecule: hsa-miR-6886-3p [13]
Molecule Alteration Expression
Down-regulation
Resistant 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
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
PLC cells Liver Homo sapiens (Human) CVCL_0485
L02 cells Liver Homo sapiens (Human) CVCL_6926
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR6825-5p, miR6845-5p and miR6886-3p could decrease the level of USP22 protein by binding to the 3'-untranlated region of USP22 mRNA. All the three microRNAs (miRNAs) were downregulated by HULC, which resulted in the elevation of USP22. The pathway 'HULC/USP22/Sirt1/ protective autophagy' attenuates the sensitivity of HCC cells to chemotherapeutic agents.
Key Molecule: hsa-miR-7-5p [6]
Molecule Alteration Expression
Down-regulation
Resistant Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
Cell viability Activation hsa05200
miR7-5p/ABCC1 signaling pathway Regulation hsa05206
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
Skhep1 cells Liver Homo sapiens (Human) CVCL_0525
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Overexpression of kCNQ1OT1 enhances OXA resistance through downregulating miR-7-5p and upregulating ABCC1 in HCC cells.
Key Molecule: KCNQ1 opposite strand/antisense transcript 1 (KCNQ1OT1) [6]
Molecule Alteration Expression
Up-regulation
Resistant Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
Cell viability Activation hsa05200
miR7-5p/ABCC1 signaling pathway Regulation hsa05206
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
Skhep1 cells Liver Homo sapiens (Human) CVCL_0525
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Overexpression of kCNQ1OT1 enhances OXA resistance through downregulating miR-7-5p and upregulating ABCC1 in HCC cells.
Key Molecule: NR2F1 antisense RNA 1 (NR2F1-AS1) [56]
Molecule Alteration Expression
Up-regulation
Resistant Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell viability Activation hsa05200
Epithelial mesenchymal transition signaling pathway Activation hsa01521
NR2F1/AS1/miR363/ABCC1 signaling pathway Regulation hsa05206
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Transwell assay
Mechanism Description Both NR2F1-AS1 and ABCC1 were up-regulated in oxaliplatin-resistant HCC cells,and miR-363 expression was increased in Huh7/OXA and HepG2/OXA cells transfected with NR2F1-AS1 siRNA compared to empty vector-transfected cells.
Key Molecule: hsa-mir-363 [56]
Molecule Alteration Expression
Down-regulation
Resistant Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell viability Activation hsa05200
NR2F1/AS1/miR363/ABCC1 signaling pathway Regulation hsa05206
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Transwell assay
Mechanism Description Both NR2F1-AS1 and ABCC1 were up-regulated in oxaliplatin-resistant HCC cells,and miR-363 expression was increased in Huh7/OXA and HepG2/OXA cells transfected with NR2F1-AS1 siRNA compared to empty vector-transfected cells.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance-associated protein 1 (MRP1) [6]
Molecule Alteration Expression
Up-regulation
Resistant Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell viability Activation hsa05200
miR7-5p/ABCC1 signaling pathway Regulation hsa05206
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
Skhep1 cells Liver Homo sapiens (Human) CVCL_0525
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Overexpression of kCNQ1OT1 enhances OXA resistance through downregulating miR-7-5p and upregulating ABCC1 in HCC cells.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Ubiquitin carboxyl-terminal hydrolase 22 (USP22) [13]
Molecule Alteration Expression
Up-regulation
Resistant 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
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
PLC cells Liver Homo sapiens (Human) CVCL_0485
L02 cells Liver Homo sapiens (Human) CVCL_6926
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR6825-5p, miR6845-5p and miR6886-3p could decrease the level of USP22 protein by binding to the 3'-untranlated region of USP22 mRNA. All the three microRNAs (miRNAs) were downregulated by HULC, which resulted in the elevation of USP22. The pathway 'HULC/USP22/Sirt1/ protective autophagy' attenuates the sensitivity of HCC cells to chemotherapeutic agents.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-122 [57]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
Wnt/Beta-catenin signaling pathway Inhibition hsa04310
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-122 inhibits MDR1 expression via suppression of Wnt/beta-catenin pathway, thereby enhancing HCC sensitivity to OXA.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [57]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
Wnt/Beta-catenin signaling pathway Inhibition hsa04310
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-122 inhibits MDR1 expression via suppression of Wnt/beta-catenin pathway, thereby enhancing HCC sensitivity to OXA.
Ovarian cancer [ICD-11: 2C73]
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: Cancer susceptibility 11 (CASC11) [9]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian squamous cell carcinoma [ICD-11: 2C73.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model UWB1.289 cells Ovary Homo sapiens (Human) CVCL_B079
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Overexpression of CASC11 in ovarian squamous cell carcinoma mediates the development of cancer cell resistance to chemotherapy (oxaliplatin, tetraplatin, cisplatin, and carboplatin).
Kidney cancer [ICD-11: 2C90]
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-21 [58]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Renal carcinoma [ICD-11: 2C90.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model 786-O cells Kidney Homo sapiens (Human) CVCL_1051
ACHN cells Pleural effusion Homo sapiens (Human) CVCL_1067
HK-2 cells Kidney Homo sapiens (Human) CVCL_0302
RCC10 cells Kidney Homo sapiens (Human) CVCL_6265
RCC4 cells Kidney Homo sapiens (Human) CVCL_0498
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
Celltiter96 Aqueous Non Radioactive Cell Proliferation Assay
Mechanism Description Tumor suppressor genes like PTEN, PDCD4 and TIMP3, are target genes of miR21. PTEN is a potent inhibitor of PI3k/Akt pathway, as well as a direct target of miR21.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Programmed cell death protein 4 (PDCD4) [58]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Renal carcinoma [ICD-11: 2C90.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model 786-O cells Kidney Homo sapiens (Human) CVCL_1051
ACHN cells Pleural effusion Homo sapiens (Human) CVCL_1067
HK-2 cells Kidney Homo sapiens (Human) CVCL_0302
RCC10 cells Kidney Homo sapiens (Human) CVCL_6265
RCC4 cells Kidney Homo sapiens (Human) CVCL_0498
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Celltiter96 Aqueous Non Radioactive Cell Proliferation Assay
Mechanism Description Tumor suppressor genes like PTEN, PDCD4 and TIMP3, are target genes of miR21. PTEN is a potent inhibitor of PI3k/Akt pathway, as well as a direct target of miR21.
Key Molecule: Phosphatase and tensin homolog (PTEN) [58]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Renal carcinoma [ICD-11: 2C90.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model 786-O cells Kidney Homo sapiens (Human) CVCL_1051
ACHN cells Pleural effusion Homo sapiens (Human) CVCL_1067
HK-2 cells Kidney Homo sapiens (Human) CVCL_0302
RCC10 cells Kidney Homo sapiens (Human) CVCL_6265
RCC4 cells Kidney Homo sapiens (Human) CVCL_0498
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Celltiter96 Aqueous Non Radioactive Cell Proliferation Assay
Mechanism Description Tumor suppressor genes like PTEN, PDCD4 and TIMP3, are target genes of miR21. PTEN is a potent inhibitor of PI3k/Akt pathway, as well as a direct target of miR21.
Key Molecule: Metalloproteinase inhibitor 3 (TIMP3) [58]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Renal carcinoma [ICD-11: 2C90.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model 786-O cells Kidney Homo sapiens (Human) CVCL_1051
ACHN cells Pleural effusion Homo sapiens (Human) CVCL_1067
HK-2 cells Kidney Homo sapiens (Human) CVCL_0302
RCC10 cells Kidney Homo sapiens (Human) CVCL_6265
RCC4 cells Kidney Homo sapiens (Human) CVCL_0498
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Celltiter96 Aqueous Non Radioactive Cell Proliferation Assay
Mechanism Description Tumor suppressor genes like PTEN, PDCD4 and TIMP3, are target genes of miR21. PTEN is a potent inhibitor of PI3k/Akt pathway, as well as a direct target of miR21.
References
Ref 1 Long noncoding RNA CCAL transferred from fibroblasts by exosomes promotes chemoresistance of colorectal cancer cells. Int J Cancer. 2020 Mar 15;146(6):1700-1716. doi: 10.1002/ijc.32608. Epub 2019 Aug 27.
Ref 2 [miR-126 inhibits colon cancer proliferation and invasion through targeting IRS1, SLC7A5 and TOM1 gene]. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2013 Aug;38(8):809-17. doi: 10.3969/j.issn.1672-7347.2013.08.009.
Ref 3 miR-153 supports colorectal cancer progression via pleiotropic effects that enhance invasion and chemotherapeutic resistance. Cancer Res. 2013 Nov 1;73(21):6435-47. doi: 10.1158/0008-5472.CAN-12-3308. Epub 2013 Aug 15.
Ref 4 Involvement of microRNA-141-3p in 5-fluorouracil and oxaliplatin chemo-resistance in esophageal cancer cells via regulation of PTEN. Mol Cell Biochem. 2016 Nov;422(1-2):161-170. doi: 10.1007/s11010-016-2816-9. Epub 2016 Sep 19.
Ref 5 miR-135a promotes gastric cancer progression and resistance to oxaliplatin. Oncotarget. 2016 Oct 25;7(43):70699-70714. doi: 10.18632/oncotarget.12208.
Ref 6 Long non-coding RNA KCNQ1OT1 modulates oxaliplatin resistance in hepatocellular carcinoma through miR-7-5p/ ABCC1 axis. Biochem Biophys Res Commun. 2018 Sep 18;503(4):2400-2406. doi: 10.1016/j.bbrc.2018.06.168. Epub 2018 Jul 4.
Ref 7 Benefit of rebiopsy for deciding treatment strategy in rectal cancer: A case reportOncol Lett. 2017 Sep;14(3):3697-3700. doi: 10.3892/ol.2017.6601. Epub 2017 Jul 18.
Ref 8 Regulation of cellular sphingosine-1-phosphate by sphingosine kinase 1 and sphingosine-1-phopshate lyase determines chemotherapy resistance in gastroesophageal cancer .BMC Cancer. 2015 Oct 22;15:762. doi: 10.1186/s12885-015-1718-7. 10.1186/s12885-015-1718-7
Ref 9 Overexpression of CASC11 in ovarian squamous cell carcinoma mediates the development of cancer cell resistance to chemotherapy. Gene. 2019 Aug 20;710:363-366. doi: 10.1016/j.gene.2019.06.011. Epub 2019 Jun 7.
Ref 10 miR-34a Regulates Multidrug Resistance via Positively Modulating OAZ2 Signaling in Colon Cancer Cells. J Immunol Res. 2018 Aug 2;2018:7498514. doi: 10.1155/2018/7498514. eCollection 2018.
Ref 11 miR-203 induces oxaliplatin resistance in colorectal cancer cells by negatively regulating ATM kinase. Mol Oncol. 2014 Feb;8(1):83-92. doi: 10.1016/j.molonc.2013.09.004. Epub 2013 Oct 8.
Ref 12 MSC-regulated lncRNA MACC1-AS1 promotes stemness and chemoresistance through fatty acid oxidation in gastric cancer. Oncogene. 2019 Jun;38(23):4637-4654. doi: 10.1038/s41388-019-0747-0. Epub 2019 Feb 11.
Ref 13 LncRNA HULC triggers autophagy via stabilizing Sirt1 and attenuates the chemosensitivity of HCC cells. Oncogene. 2017 Jun 22;36(25):3528-3540. doi: 10.1038/onc.2016.521. Epub 2017 Feb 6.
Ref 14 Epigenetic inactivation of the p53-induced long noncoding RNA TP53 target 1 in human cancer. Proc Natl Acad Sci U S A. 2016 Nov 22;113(47):E7535-E7544. doi: 10.1073/pnas.1608585113. Epub 2016 Nov 7.
Ref 15 Gene-expression signature regulated by the KEAP1-NRF2-CUL3 axis is associated with a poor prognosis in head and neck squamous cell cancer .BMC Cancer. 2018 Jan 6;18(1):46. doi: 10.1186/s12885-017-3907-z. 10.1186/s12885-017-3907-z
Ref 16 Long noncoding RNA BLACAT1 modulates ABCB1 to promote oxaliplatin resistance of gastric cancer via sponging miR-361. Biomed Pharmacother. 2018 Mar;99:832-838. doi: 10.1016/j.biopha.2018.01.130. Epub 2018 Feb 20.
Ref 17 miR 195 5p regulates multi drug resistance of gastric cancer cells via targeting ZNF139. Oncol Rep. 2018 Sep;40(3):1370-1378. doi: 10.3892/or.2018.6524. Epub 2018 Jun 25.
Ref 18 Overexpression of Lin28 Decreases the Chemosensitivity of Gastric Cancer Cells to Oxaliplatin, Paclitaxel, Doxorubicin, and Fluorouracil in Part via microRNA-107. PLoS One. 2015 Dec 4;10(12):e0143716. doi: 10.1371/journal.pone.0143716. eCollection 2015.
Ref 19 MiR-218 inhibits multidrug resistance (MDR) of gastric cancer cells by targeting Hedgehog/smoothened. Int J Clin Exp Pathol. 2015 Jun 1;8(6):6397-406. eCollection 2015.
Ref 20 miR-204 targets Bcl-2 expression and enhances responsiveness of gastric cancer. Cell Death Dis. 2012 Nov 15;3(11):e423. doi: 10.1038/cddis.2012.160.
Ref 21 miR-19b-3p promotes colon cancer proliferation and oxaliplatin-based chemoresistance by targeting SMAD4: validation by bioinformatics and experimental analyses. J Exp Clin Cancer Res. 2017 Sep 22;36(1):131. doi: 10.1186/s13046-017-0602-5.
Ref 22 Linc00152 Functions as a Competing Endogenous RNA to Confer Oxaliplatin Resistance and Holds Prognostic Values in Colon Cancer. Mol Ther. 2016 Dec;24(12):2064-2077. doi: 10.1038/mt.2016.180. Epub 2016 Sep 16.
Ref 23 MicroRNA-137 chemosensitizes colon cancer cells to the chemotherapeutic drug oxaliplatin (OXA) by targeting YBX1. Cancer Biomark. 2017;18(1):1-9. doi: 10.3233/CBM-160650.
Ref 24 miR-409-3p sensitizes colon cancer cells to oxaliplatin by inhibiting Beclin-1-mediated autophagy. Int J Mol Med. 2016 Apr;37(4):1030-8. doi: 10.3892/ijmm.2016.2492. Epub 2016 Feb 18.
Ref 25 miR-219-5p plays a tumor suppressive role in colon cancer by targeting oncogene Sall4. Oncol Rep. 2015 Oct;34(4):1923-32. doi: 10.3892/or.2015.4168. Epub 2015 Aug 4.
Ref 26 miR-320 enhances the sensitivity of human colon cancer cells to chemoradiotherapy in vitro by targeting FOXM1. Biochem Biophys Res Commun. 2015 Feb 6;457(2):125-32. doi: 10.1016/j.bbrc.2014.11.039. Epub 2014 Nov 21.
Ref 27 Long noncoding RNA GIHCG induces cancer progression and chemoresistance and indicates poor prognosis in colorectal cancer. Onco Targets Ther. 2019 Feb 7;12:1059-1070. doi: 10.2147/OTT.S192290. eCollection 2019.
Ref 28 PIWI-interacting RNA-54265 is oncogenic and a potential therapeutic target in colorectal adenocarcinoma. Theranostics. 2018 Oct 6;8(19):5213-5230. doi: 10.7150/thno.28001. eCollection 2018.
Ref 29 ANRIL promotes chemoresistance via disturbing expression of ABCC1 by regulating the expression of Let-7a in colorectal cancer. Biosci Rep. 2018 Nov 20;38(6):BSR20180620. doi: 10.1042/BSR20180620. Print 2018 Dec 21.
Ref 30 miR-216b promotes cell growth and enhances chemosensitivity of colorectal cancer by suppressing PDZ-binding kinase. Biochem Biophys Res Commun. 2017 Jun 24;488(2):247-252. doi: 10.1016/j.bbrc.2017.03.162. Epub 2017 Mar 31.
Ref 31 Downregulation of YEATS4 by miR-218 sensitizes colorectal cancer cells to L-OHP-induced cell apoptosis by inhibiting cytoprotective autophagy. Oncol Rep. 2016 Dec;36(6):3682-3690. doi: 10.3892/or.2016.5195. Epub 2016 Oct 21.
Ref 32 Overexpression of MEG3 sensitizes colorectal cancer cells to oxaliplatin through regulation of miR-141/PDCD4 axis. Biomed Pharmacother. 2018 Oct;106:1607-1615. doi: 10.1016/j.biopha.2018.07.131. Epub 2018 Jul 29.
Ref 33 miR-625-3p regulates oxaliplatin resistance by targeting MAP2K6-p38 signalling in human colorectal adenocarcinoma cells. Nat Commun. 2016 Aug 16;7:12436. doi: 10.1038/ncomms12436.
Ref 34 MicroRNA-520g confers drug resistance by regulating p21 expression in colorectal cancer. J Biol Chem. 2015 Mar 6;290(10):6215-25. doi: 10.1074/jbc.M114.620252. Epub 2015 Jan 23.
Ref 35 MicroRNA-17-5p promotes chemotherapeutic drug resistance and tumour metastasis of colorectal cancer by repressing PTEN expression. Oncotarget. 2014 May 30;5(10):2974-87. doi: 10.18632/oncotarget.1614.
Ref 36 Serum miR-19a predicts resistance to FOLFOX chemotherapy in advanced colorectal cancer cases. Asian Pac J Cancer Prev. 2013;14(12):7421-6. doi: 10.7314/apjcp.2013.14.12.7421.
Ref 37 A panel of serum exosomal microRNAs as predictive markers for chemoresistance in advanced colorectal cancer. Cancer Chemother Pharmacol. 2019 Aug;84(2):315-325. doi: 10.1007/s00280-019-03867-6. Epub 2019 May 14.
Ref 38 MALAT1 Is Associated with Poor Response to Oxaliplatin-Based Chemotherapy in Colorectal Cancer Patients and Promotes Chemoresistance through EZH2. Mol Cancer Ther. 2017 Apr;16(4):739-751. doi: 10.1158/1535-7163.MCT-16-0591. Epub 2017 Jan 9.
Ref 39 Carcinoma-associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics. 2018 Jun 24;8(14):3932-3948. doi: 10.7150/thno.25541. eCollection 2018.
Ref 40 miR-340 suppresses tumor growth and enhances chemosensitivity of colorectal cancer by targeting RLIP76. Eur Rev Med Pharmacol Sci. 2017 Jun;21(12):2875-2886.
Ref 41 [MiR-145 inhibits drug resistance to Oxaliplatin in colorectal cancer cells through regulating G protein coupled receptor 98]. Zhonghua Wei Chang Wai Ke Za Zhi. 2017 May 25;20(5):566-570.
Ref 42 miR-503-5p confers drug resistance by targeting PUMA in colorectal carcinoma. Oncotarget. 2017 Mar 28;8(13):21719-21732. doi: 10.18632/oncotarget.15559.
Ref 43 miR-506 enhances the sensitivity of human colorectal cancer cells to oxaliplatin by suppressing MDR1/P-gp expression. Cell Prolif. 2017 Jun;50(3):e12341. doi: 10.1111/cpr.12341. Epub 2017 Feb 19.
Ref 44 MicroRNA-141 inhibits tumor growth and minimizes therapy resistance in colorectal cancer. Mol Med Rep. 2017 Mar;15(3):1037-1042. doi: 10.3892/mmr.2017.6135. Epub 2017 Jan 23.
Ref 45 MiR-139-5p reverses CD44+/CD133+-associated multidrug resistance by downregulating NOTCH1 in colorectal carcinoma cells. Oncotarget. 2016 Nov 15;7(46):75118-75129. doi: 10.18632/oncotarget.12611.
Ref 46 miR-122 Targets X-Linked Inhibitor of Apoptosis Protein to Sensitize Oxaliplatin-Resistant Colorectal Cancer Cells to Oxaliplatin-Mediated Cytotoxicity. Cell Physiol Biochem. 2018;51(5):2148-2159. doi: 10.1159/000495832. Epub 2018 Dec 6.
Ref 47 Knockdown of Mir-135b Sensitizes Colorectal Cancer Cells to Oxaliplatin-Induced Apoptosis Through Increase of FOXO1. Cell Physiol Biochem. 2018;48(4):1628-1637. doi: 10.1159/000492284. Epub 2018 Aug 2.
Ref 48 MicroRNA-425-5p regulates chemoresistance in colorectal cancer cells via regulation of Programmed Cell Death 10. J Cell Mol Med. 2016 Feb;20(2):360-9. doi: 10.1111/jcmm.12742. Epub 2015 Dec 9.
Ref 49 miR-204-5p inhibits proliferation and invasion and enhances chemotherapeutic sensitivity of colorectal cancer cells by downregulating RAB22A. Clin Cancer Res. 2014 Dec 1;20(23):6187-99. doi: 10.1158/1078-0432.CCR-14-1030. Epub 2014 Oct 7.
Ref 50 MicroRNA-143 inhibits tumor growth and angiogenesis and sensitizes chemosensitivity to oxaliplatin in colorectal cancers. Cell Cycle. 2013 May 1;12(9):1385-94. doi: 10.4161/cc.24477. Epub 2013 Apr 8.
Ref 51 MiR-222 modulates multidrug resistance in human colorectal carcinoma by down-regulating ADAM-17. Exp Cell Res. 2012 Oct 15;318(17):2168-77. doi: 10.1016/j.yexcr.2012.04.014. Epub 2012 Jun 4.
Ref 52 miR-297 modulates multidrug resistance in human colorectal carcinoma by down-regulating MRP-2. Biochem J. 2012 Sep 1;446(2):291-300. doi: 10.1042/BJ20120386.
Ref 53 miR-1915 inhibits Bcl-2 to modulate multidrug resistance by increasing drug-sensitivity in human colorectal carcinoma cells. Mol Carcinog. 2013 Jan;52(1):70-8. doi: 10.1002/mc.21832. Epub 2011 Nov 28.
Ref 54 miR-20a targets BNIP2 and contributes chemotherapeutic resistance in colorectal adenocarcinoma SW480 and SW620 cell lines. Acta Biochim Biophys Sin (Shanghai). 2011 Mar;43(3):217-25. doi: 10.1093/abbs/gmq125. Epub 2011 Jan 17.
Ref 55 miR-139-5p Inhibits the Epithelial-Mesenchymal Transition and Enhances the Chemotherapeutic Sensitivity of Colorectal Cancer Cells by Downregulating BCL2. Sci Rep. 2016 May 31;6:27157. doi: 10.1038/srep27157.
Ref 56 LncRNA NR2F1-AS1 regulates hepatocellular carcinoma oxaliplatin resistance by targeting ABCC1 via miR-363. J Cell Mol Med. 2018 Jun;22(6):3238-3245. doi: 10.1111/jcmm.13605. Epub 2018 Mar 30.
Ref 57 miR-122 enhances sensitivity of hepatocellular carcinoma to oxaliplatin via inhibiting MDR1 by targeting Wnt/Beta-catenin pathway. Exp Mol Pathol. 2019 Feb;106:34-43. doi: 10.1016/j.yexmp.2018.10.009. Epub 2018 Oct 26.
Ref 58 Targeting miR-21 decreases expression of multi-drug resistant genes and promotes chemosensitivity of renal carcinoma. Tumour Biol. 2017 Jul;39(7):1010428317707372. doi: 10.1177/1010428317707372.

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