Drug (ID: DG00069) and It's Reported Resistant Information
Name
Fluorouracil
Synonyms
5-Fluorouracil; 51-21-8; fluorouracil; 5-FU; Fluoroplex; Adrucil; Efudex; Carac; Fluracil; Fluoroblastin; 5-fluoropyrimidine-2,4(1H,3H)-dione; Kecimeton; Timazin; Carzonal; Efudix; Arumel; Fluril; Queroplex; Fluracilum; Ulup; 5-Fluoracil; Phthoruracil; Fluro Uracil; 5-Fluoro-2,4(1H,3H)-pyrimidinedione; Ftoruracil; Fluorouracilum; Efurix; Fluri; 5 Fluorouracil; Effluderm (free base); 5-fluoro-1H-pyrimidine-2,4-dione; Fluorouracilo; Fluroblastin; Phtoruracil; 2,4-Dihydroxy-5-fluoropyrimidine; 2,4(1H,3H)-Pyrimidinedione, 5-fluoro-; Adrucil; Effluderm; Fluorouracile; Fluoruracil; Fluracedyl; Flurodex; Neofluor; Onkofluor; Ribofluor; Tetratogen; URF; Allergan Brand of Fluorouracil; Biosyn Brand of Fluorouracil; CSP Brand of Fluorouracil; Cinco FU; Dakota Brand of Fluorouracil; Dermatech Brand of Fluorouracil; Dermik Brandof Fluorouracil; Ferrer Brand of Fluorouracil; Fluoro Uracile ICN; Fluorouracil GRY; Fluorouracil Mononitrate; Fluorouracil Monopotassium Salt; Fluorouracil Monosodium Salt; Fluorouracil Potassium Salt; Fluorouracil Teva Brand; Fluorouracile Dakota; Fluorouracile [DCIT]; Fluorouracilo Ferrer Far; Gry Brand of Fluorouracil; Haemato Brand of Fluorouracil; Haemato fu; Hexal Brand of Fluorouracil; ICN Brand of Fluorouracil; Inhibits thymilidate synthetase; Medac Brand of Fluorouracil; Neocorp Brand of Fluorouracil; Onkoworks Brand of Fluorouracil; Ribosepharm Brand of Fluorouracil; Riemser Brand of Fluorouracil; Roche Brand of Fluorouracil; Teva Brand of Fluorouracil; F 6627; F0151; IN1335; U 8953; Adrucil (TN); Carac (TN); Dakota, Fluorouracile; Efudex (TN); Fluoro-Uracile ICN; Fluoro-uracile; Fluoro-uracilo; Fluoroplex (TN); Fluorouracil-GRY; Fluorouracilo [INN-Spanish]; Fluorouracilum [INN-Latin]; Haemato-fu; Ro 2-9757; U-8953; Ro-2-9757; Fluorouracil (JP15/USP/INN); Fluorouracil [USAN:INN:BAN:JAN]; 1-fluoro-1h-pyrimidine-2,4-dione; 2,4-Dioxo-5-fluoropryimidine; 2,4-Dioxo-5-fluoropyrimidine; 5 FU Lederle; 5 FU medac; 5 Fluorouracil biosyn; 5 HU Hexal; 5-FU (TN); 5-FU Lederle; 5-FU medac; 5-Faracil; 5-Fluor-2,4(1H,3H)-pyrimidindion; 5-Fluor-2,4(1H,3H)-pyrimidindion [Czech]; 5-Fluor-2,4-dihydroxypyrimidin; 5-Fluor-2,4-dihydroxypyrimidin [Czech]; 5-Fluor-2,4-pyrimidindiol; 5-Fluor-2,4-pyrimidindiol [Czech]; 5-Fluoracil [German]; 5-Fluoracyl; 5-Fluoro-2,4-pyrimidinedione; 5-Fluoropyrimidin-2,4-diol; 5-Fluoropyrimidine-2,4-dione; 5-Fluorouracil-biosyn; 5-Fluoruracil; 5-Fluoruracil [German]; 5-Ftouracyl; 5-HU Hexal; 5-fluoro uracil; 5FU
    Click to Show/Hide
Indication
In total 2 Indication(s)
Solid tumour/cancer [ICD-11: 2A00-2F9Z]
Approved
[1], [2]
Colorectal cancer [ICD-11: 2B91]
Investigative
[1], [2]
Structure
Drug Resistance Disease(s)
Disease(s) with Clinically Reported Resistance for This Drug (11 diseases)
Breast cancer [ICD-11: 2C60]
[3]
Colon cancer [ICD-11: 2B90]
[4]
Colorectal cancer [ICD-11: 2B91]
[5]
Colorectal peritoneal carcinomatosis [ICD-11: 2D91]
[6]
Esophageal cancer [ICD-11: 2B70]
[7]
Gastric cancer [ICD-11: 2B72]
[8]
Kidney cancer [ICD-11: 2C90]
[9]
Liver cancer [ICD-11: 2C12]
[10]
Metastatic colorectal cancer [ICD-11: 2D85]
[11]
Neuroendocrine carcinoma [ICD-11: 2D4Y]
[12]
Pancreatic cancer [ICD-11: 2C10]
[13]
Disease(s) with Resistance Information Discovered by Cell Line Test for This Drug (10 diseases)
Breast cancer [ICD-11: 2C60]
[14]
Colon cancer [ICD-11: 2B90]
[15]
Colorectal cancer [ICD-11: 2B91]
[1], [2]
Esophageal cancer [ICD-11: 2B70]
[16]
Gastric cancer [ICD-11: 2B72]
[17]
Liver cancer [ICD-11: 2C12]
[18]
Lung cancer [ICD-11: 2C25]
[19]
Oral squamous cell carcinoma [ICD-11: 2B6E]
[20]
Pancreatic cancer [ICD-11: 2C10]
[21]
Salivary gland carcinoma [ICD-11: 2E60]
[22]
Target Candida Thymidylate synthase (Candi TMP1) TYSY_CANAL [1]
Dihydrothymine dehydrogenase (DPYD) DPYD_HUMAN [1]
TERT messenger RNA (TERT mRNA) TERT_HUMAN [1]
Thymidylate synthase messenger RNA (TYMS mRNA) TYSY_HUMAN [1]
Click to Show/Hide the Molecular Information and External Link(s) of This Drug
Formula
C4H3FN2O2
IsoSMILES
C1=C(C(=O)NC(=O)N1)F
InChI
1S/C4H3FN2O2/c5-2-1-6-4(9)7-3(2)8/h1H,(H2,6,7,8,9)
InChIKey
GHASVSINZRGABV-UHFFFAOYSA-N
PubChem CID
3385
ChEBI ID
CHEBI:46345
TTD Drug ID
D05LEO
VARIDT ID
DR00153
INTEDE ID
DR0020
DrugBank ID
DB00544
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
Acute lymphocytic leukemia [ICD-11: 2B33]
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-138 [23]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Leukemia [ICD-11: 2B33.6]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model HL60 cells Peripheral blood Homo sapiens (Human) CVCL_0002
Experiment for
Molecule Alteration
qRT-PCR; Northern blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-138 was found up-regulated in the vincristine-induced multidrug resistance (MDR) leukemia cell line HL-60/VCR as compared with HL-60 cells. Up-regulation of miR-138 could reverse resistance of both P-glycoprotein-related and P-glycoprotein-non-related drugs on HL-60/VCR cells, and promote adriamycin-induced apoptosis, accompanied by increased accumulation and decreased releasing amount of adriamycin.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [23]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Leukemia [ICD-11: 2B33.6]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model HL60 cells Peripheral blood Homo sapiens (Human) CVCL_0002
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-138 was found up-regulated in the vincristine-induced multidrug resistance (MDR) leukemia cell line HL-60/VCR as compared with HL-60 cells. Up-regulation of miR-138 could reverse resistance of both P-glycoprotein-related and P-glycoprotein-non-related drugs on HL-60/VCR cells, and promote adriamycin-induced apoptosis, accompanied by increased accumulation and decreased releasing amount of adriamycin.
Osteosarcoma [ICD-11: 2B51]
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-140 [24]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Osteosarcoma [ICD-11: 2B51.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model MG63 cells Bone marrow Homo sapiens (Human) CVCL_0426
U2OS cells Bone Homo sapiens (Human) CVCL_0042
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description miR-140 is involved in the chemoresistance by reduced cell proliferation via G1 and G2 phase arrest mediated in part.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Histone deacetylase 4 (HDAC4) [24]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Osteosarcoma [ICD-11: 2B51.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model MG63 cells Bone marrow Homo sapiens (Human) CVCL_0426
U2OS cells Bone Homo sapiens (Human) CVCL_0042
Experiment for
Molecule Alteration
Western blotting analysis; Immunofluorescence analysis
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description miR-140 is involved in the chemoresistance by reduced cell proliferation via G1 and G2 phase arrest mediated in part.
Nasopharyngeal cancer [ICD-11: 2B6B]
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-3188 [25]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Nasopharyngeal carcinoma [ICD-11: 2B6B.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
PI3K/AKT/mTOR signaling pathway Inhibition hsa04151
In Vitro Model HNE1 cells Nasopharynx Homo sapiens (Human) CVCL_0308
5-8F cells Nasopharynx Homo sapiens (Human) CVCL_C528
CNE2 cells Nasopharynx Homo sapiens (Human) CVCL_6889
C666-1 cells Throat Homo sapiens (Human) CVCL_7949
CNE1 cells Throat Homo sapiens (Human) CVCL_6888
HONE1 cells Throat Homo sapiens (Human) CVCL_8706
6-10B cells Nasopharynx Homo sapiens (Human) CVCL_C529
SUNE-1 cells Nasopharynx Homo sapiens (Human) CVCL_6946
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-3188 regulates nasopharyngeal carcinoma proliferation and chemosensitivity through a FOXO1-modulated positive feedback loop with mTOR-p-PI3k/AkT-c-JUN.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase mTOR (mTOR) [25]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Nasopharyngeal carcinoma [ICD-11: 2B6B.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
PI3K/AKT/mTOR signaling pathway Inhibition hsa04151
In Vitro Model HNE1 cells Nasopharynx Homo sapiens (Human) CVCL_0308
5-8F cells Nasopharynx Homo sapiens (Human) CVCL_C528
CNE2 cells Nasopharynx Homo sapiens (Human) CVCL_6889
C666-1 cells Throat Homo sapiens (Human) CVCL_7949
CNE1 cells Throat Homo sapiens (Human) CVCL_6888
HONE1 cells Throat Homo sapiens (Human) CVCL_8706
6-10B cells Nasopharynx Homo sapiens (Human) CVCL_C529
SUNE-1 cells Nasopharynx Homo sapiens (Human) CVCL_6946
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-3188 regulates nasopharyngeal carcinoma proliferation and chemosensitivity through a FOXO1-modulated positive feedback loop with mTOR-p-PI3k/AkT-c-JUN.
Oral squamous cell carcinoma [ICD-11: 2B6E]
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-654-5p [20]
Molecule Alteration Expression
Down-regulation
Resistant Disease Oral squamous cell carcinoma [ICD-11: 2B6E.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation MAPK/RAS signaling pathway Regulation hsa04010
In Vitro Model Tca8113 cells Tongue Homo sapiens (Human) CVCL_6851
CAL-27 cells Tongue Homo sapiens (Human) CVCL_1107
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR654-5p targets GRAP to promote proliferation, metastasis, and chemoresistance of oral squamous cell carcinoma through Ras/MAPk signaling.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: GRB2-related adapter protein (GRAP) [20]
Molecule Alteration Expression
Up-regulation
Resistant Disease Oral squamous cell carcinoma [ICD-11: 2B6E.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation MAPK/RAS signaling pathway Regulation hsa04010
In Vitro Model Tca8113 cells Tongue Homo sapiens (Human) CVCL_6851
CAL-27 cells Tongue Homo sapiens (Human) CVCL_1107
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR654-5p targets GRAP to promote proliferation, metastasis, and chemoresistance of oral squamous cell carcinoma through Ras/MAPk signaling.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-365a-3p [26]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Oral squamous cell carcinoma [ICD-11: 2B6E.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Beta5-integrin/c-Met signaling pathway Inhibition hsa01521
Cell viability Activation hsa05200
In Vitro Model C9-IV3 cells Oral Homo sapiens (Human) N.A.
CGHNC9 cells Oral Homo sapiens (Human) N.A.
OC-3 cells Oral Homo sapiens (Human) CVCL_WL09
In Vivo Model CB17-SCID mice xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-365-3p targets EHF to inhibit OSCC migration, invasion, and metastasis through kRT16.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: ETS homologous factor (EHF) [26]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Oral squamous cell carcinoma [ICD-11: 2B6E.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Beta5-integrin/c-Met signaling pathway Inhibition hsa01521
Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell viability Inhibition hsa05200
In Vitro Model C9-IV3 cells Oral Homo sapiens (Human) N.A.
CGHNC9 cells Oral Homo sapiens (Human) N.A.
OC-3 cells Oral Homo sapiens (Human) CVCL_WL09
In Vivo Model CB17-SCID mice xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-365-3p targets EHF to inhibit OSCC migration, invasion, and metastasis through kRT16.
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 [27]
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.
Key Molecule: hsa-mir-221 [7]
Molecule Alteration Expression
Up-regulation
Resistant Disease Esophageal adenocarcinoma [ICD-11: 2B70.2]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
Wnt/Beta-catenin/EMT signaling pathway Activation hsa04310
In Vitro Model OE19 cells Esophagus Homo sapiens (Human) CVCL_1622
OE33 cellss Esophagus Homo sapiens (Human) CVCL_0471
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Annexin V-fluorescein isothiocyanate (FITC) and propidium iodide (PI) staining assay
Mechanism Description miR-221 mediates chemoresistance of esophageal adenocarcinoma by direct targeting and reducing of Dkk2 expression.
Key Molecule: hsa-miR-193a-3p [16]
Molecule Alteration Expression
Up-regulation
Resistant Disease Esophageal cancer [ICD-11: 2B70.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model KYSE150 cells Esophagus Homo sapiens (Human) CVCL_1348
KYSE510 cells Esophagus Homo sapiens (Human) CVCL_1354
kYSE410 cells Esophagus Homo sapiens (Human) CVCL_1352
kYSE450 cells Esophagus Homo sapiens (Human) CVCL_1353
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Over-expression of miR-193a-3p increased the radioresistance and chemoresistance of oesophageal squamous cell carcinoma (ESCC) cells. In contrast, the down-regulation of miR-193a-3p decreased the radioresistance and chemoresistance of ESCC cells. In addition, miR-193a-3p inducing DNA damage has also been demonstrated through measuring the level of gamma-H2AX associated with miR-193a-3p. Moreover, a small interfering RNA(siRNA)-induced repression of the PSEN1 gene had an effect similar to that of miR-193a-3p up-regulation. The above processes also inhibited oesophageal cancer cells apoptosis. These findings suggest that miR-193a-3p contributes to the radiation and chemotherapy resistance of oesophageal carcinoma by down-regulating PSEN1.
Key Molecule: hsa-miR-193a-3p [16]
Molecule Alteration Expression
Up-regulation
Resistant Disease Esophageal cancer [ICD-11: 2B70.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model KYSE150 cells Esophagus Homo sapiens (Human) CVCL_1348
KYSE510 cells Esophagus Homo sapiens (Human) CVCL_1354
kYSE410 cells Esophagus Homo sapiens (Human) CVCL_1352
kYSE450 cells Esophagus Homo sapiens (Human) CVCL_1353
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Over-expression of miR-193a-3p increased the radioresistance and chemoresistance of oesophageal squamous cell carcinoma (ESCC) cells. The regulation role of miR-193a-3p on multi-chemoresistance and radioresistance were mediated by PSEN1.
Key Molecule: hsa-miR-193a-3p [16]
Molecule Alteration Expression
Up-regulation
Resistant Disease Esophageal cancer [ICD-11: 2B70.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model KYSE150 cells Esophagus Homo sapiens (Human) CVCL_1348
KYSE510 cells Esophagus Homo sapiens (Human) CVCL_1354
kYSE410 cells Esophagus Homo sapiens (Human) CVCL_1352
kYSE450 cells Esophagus Homo sapiens (Human) CVCL_1353
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Over-expression of miR-193a-3p increased the radioresistance and chemoresistance of oesophageal squamous cell carcinoma (ESCC) cells. The regulation role of miR-193a-3p on multi-chemoresistance and radioresistance were mediated by PSEN1.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Phosphatase and tensin homolog (PTEN) [27]
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: Dickkopf-related protein 2 (DKK2) [7]
Molecule Alteration Expression
Down-regulation
Resistant Disease Esophageal adenocarcinoma [ICD-11: 2B70.2]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell colony Activation hsa05200
Cell proliferation Activation hsa05200
Wnt/Beta-catenin/EMT signaling pathway Activation hsa04310
In Vitro Model OE19 cells Esophagus Homo sapiens (Human) CVCL_1622
OE33 cellss Esophagus Homo sapiens (Human) CVCL_0471
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Annexin V-fluorescein isothiocyanate (FITC) and propidium iodide (PI) staining assay
Mechanism Description miR-221 mediates chemoresistance of esophageal adenocarcinoma by direct targeting and reducing of Dkk2 expression.
Key Molecule: Presenilin-1 (PSEN1) [16]
Molecule Alteration Expression
Down-regulation
Resistant Disease Esophageal cancer [ICD-11: 2B70.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model KYSE150 cells Esophagus Homo sapiens (Human) CVCL_1348
KYSE510 cells Esophagus Homo sapiens (Human) CVCL_1354
kYSE410 cells Esophagus Homo sapiens (Human) CVCL_1352
kYSE450 cells Esophagus Homo sapiens (Human) CVCL_1353
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Over-expression of miR-193a-3p increased the radioresistance and chemoresistance of oesophageal squamous cell carcinoma (ESCC) cells. In contrast, the down-regulation of miR-193a-3p decreased the radioresistance and chemoresistance of ESCC cells. In addition, miR-193a-3p inducing DNA damage has also been demonstrated through measuring the level of gamma-H2AX associated with miR-193a-3p. Moreover, a small interfering RNA(siRNA)-induced repression of the PSEN1 gene had an effect similar to that of miR-193a-3p up-regulation. The above processes also inhibited oesophageal cancer cells apoptosis. These findings suggest that miR-193a-3p contributes to the radiation and chemotherapy resistance of oesophageal carcinoma by down-regulating PSEN1.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-130a-3p [28]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell migration Inhibition hsa04670
p53 signaling pathway Activation hsa04115
In Vitro Model KYSE-270 cells Esophagus Homo sapiens (Human) CVCL_1350
KYSE-410 cells Esophagus Homo sapiens (Human) CVCL_1352
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description The effect of miR-130a-3p downregulation on enhancement of protein levels was more pronounced for Bcl-2 compared to XIAP, whereas the increase of miR-130a-3p resulted in a more pronounced increase of protein levels of XIAP compared to Bcl-2. Both, up- and downregulation of miR-130a-3p and miR-148a-3p increased sensitivity towards chemotherapy in ESCC and complex role of miR-130a-3p and miR-148a-3p balance on drug resistance and tumor biology in esophageal squamous cell carcinoma.
Key Molecule: hsa-miR-130a-3p [28]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation p53 signaling pathway Activation hsa04115
In Vitro Model KYSE-270 cells Esophagus Homo sapiens (Human) CVCL_1350
KYSE-410 cells Esophagus Homo sapiens (Human) CVCL_1352
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description The effect of miR-130a-3p upregulation on suppression of protein levels was more pronounced for Bcl-2 compared to XIAP, whereas the inhibition of miR-130a-3p resulted in a more pronounced increase of protein levels of XIAP compared to Bcl-2. Both, up- and downregulation of miR-130a-3p and miR-148a-3p increased sensitivity towards chemotherapy in ESCC and complex role of miR-130a-3p and miR-148a-3p balance on drug resistance and tumor biology in esophageal squamous cell carcinoma.
Key Molecule: hsa-miR-148a-3p [28]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell migration Inhibition hsa04670
p53 signaling pathway Activation hsa04115
In Vitro Model KYSE-270 cells Esophagus Homo sapiens (Human) CVCL_1350
KYSE-410 cells Esophagus Homo sapiens (Human) CVCL_1352
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description The effect of miR-148a-3p downregulation on enhancement of protein levels was more pronounced for Bcl-2 compared to XIAP, whereas the increase of miR-130a-3p resulted in a more pronounced increase of protein levels of XIAP compared to Bcl-2. Both, up- and downregulation of miR-130a-3p and miR-148a-3p increased sensitivity towards chemotherapy in ESCC and complex role of miR-130a-3p and miR-148a-3p balance on drug resistance and tumor biology in esophageal squamous cell carcinoma.
Key Molecule: hsa-miR-148a-3p [28]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation p53 signaling pathway Activation hsa04115
In Vitro Model KYSE-270 cells Esophagus Homo sapiens (Human) CVCL_1350
KYSE-410 cells Esophagus Homo sapiens (Human) CVCL_1352
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description The effect of miR-148a-3p upregulation on suppression of protein levels was more pronounced for Bcl-2 compared to XIAP, whereas the inhibition of miR-130a-3p resulted in a more pronounced increase of protein levels of XIAP compared to Bcl-2. Both, up- and downregulation of miR-130a-3p and miR-148a-3p increased sensitivity towards chemotherapy in ESCC and complex role of miR-130a-3p and miR-148a-3p balance on drug resistance and tumor biology in esophageal squamous cell carcinoma.
Key Molecule: Long non-protein coding RNA 261 (LINC00261) [29]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Esophageal cancer [ICD-11: 2B70.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell colony Inhibition hsa05200
Cell viability Inhibition hsa05200
In Vitro Model ECA-109 cells Esophagus Homo sapiens (Human) CVCL_6898
TE-1 cells Esophagus Homo sapiens (Human) CVCL_1759
KYSE150 cells Esophagus Homo sapiens (Human) CVCL_1348
TE-5 cells Esophageal Homo sapiens (Human) CVCL_1764
In Vivo Model BALB/c nude mouse xenograft mode Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
WST-1 assay; Flow cytometry assay
Mechanism Description Long noncoding RNA LINC00261 induces chemosensitization to 5-fluorouracil by mediating methylation-dependent repression of DPYD in human esophageal cancer.
Key Molecule: hsa-mir-224 [30]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Inhibition hsa05200
DESC1/EGFR/AKT signaling pathway Regulation hsa04012
In Vitro Model KYSE30 cells Esophagus Homo sapiens (Human) CVCL_1351
EC9706 cells Esophagus Homo sapiens (Human) CVCL_E307
KYSE140 cells Esophagus Homo sapiens (Human) CVCL_1347
TE13 cells Esophageal Homo sapiens (Human) CVCL_4463
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description TUSC7 suppressed the proliferation and chemotherapy resistance of ESCC cells by increasing DESC1 expression via inhibiting miR-224.
Key Molecule: Tumor suppressor candidate 7 (TUSC7) [30]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Inhibition hsa05200
DESC1/EGFR/AKT signaling pathway Regulation hsa04012
In Vitro Model KYSE30 cells Esophagus Homo sapiens (Human) CVCL_1351
EC9706 cells Esophagus Homo sapiens (Human) CVCL_E307
KYSE140 cells Esophagus Homo sapiens (Human) CVCL_1347
TE13 cells Esophageal Homo sapiens (Human) CVCL_4463
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description TUSC7 suppressed the proliferation and chemotherapy resistance of ESCC cells by increasing DESC1 expression via inhibiting miR-224.
Key Molecule: hsa-mir-200c [31]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Esophageal cancer [ICD-11: 2B70.1]
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Endoscopy; Computed tomography assay; Positron emission tomography assay
Mechanism Description Serum miR-200c levels are useful for predicting the response to chemotherapy (cisplatin, 5-fluorouracil, and Adriamycin (ACF) or cisplatin, 5-fluorouracil, and docetaxel (DCF) ) in patients with esophageal cancer who underwent preoperative chemotherapy followed by surgery.
Key Molecule: hsa-mir-148a [32]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Esophageal adenocarcinoma [ICD-11: 2B70.2]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model kYSE410 cells Esophagus Homo sapiens (Human) CVCL_1352
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-148a sensitized chemotherapy-sensitive oesophageal cancer cell lines to cisplatin and, to a lesser extent, to 5-flurouracil and attenuated resistance in chemotherapy-resistant variants.
Key Molecule: hsa-mir-148a [32]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model kYSE410 cells Esophagus Homo sapiens (Human) CVCL_1352
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-148a sensitized chemotherapy-sensitive oesophageal cancer cell lines to cisplatin and, to a lesser extent, to 5-flurouracil and attenuated resistance in chemotherapy-resistant variants.
Key Molecule: hsa-mir-296 [33]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell growth Inhibition hsa05200
In Vitro Model ECA-109 cells Esophagus Homo sapiens (Human) CVCL_6898
Experiment for
Molecule Alteration
RT-PCR; Northern blotting analysis
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description Down-regulation of miR-296 could confer sensitivity of both P-glycoprotein-related and P-glycoprotein-nonrelated drugs on esophageal cancer cells, and might promote ADR-induced apoptosis, accompanied by increased accumulation and decreased releasing amount of ADR. Down-regulation of miR-296 could significantly decrease the expression of P-glycoprotein, Bcl-2, and the transcription of MDR1, but up-regulate the expression of Bax.
Key Molecule: hsa-mir-27a [34]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model ECA-109 cells Esophagus Homo sapiens (Human) CVCL_6898
TE13 cells Esophageal Homo sapiens (Human) CVCL_4463
Experiment for
Molecule Alteration
qRT-PCR; Northern blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Down-regulation of miR-27a significantly decreased expression of MDR1, but did not alter the expression of MRP, miR-27a could possibly mediate drug resistance, at least in part through regulation of MDR1 and apoptosis.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [33], [34]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell growth Inhibition hsa05200
In Vitro Model ECA-109 cells Esophagus Homo sapiens (Human) CVCL_6898
TE13 cells Esophageal Homo sapiens (Human) CVCL_4463
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description Down-regulation of miR-296 could confer sensitivity of both P-glycoprotein-related and P-glycoprotein-nonrelated drugs on esophageal cancer cells, and might promote ADR-induced apoptosis, accompanied by increased accumulation and decreased releasing amount of ADR. Down-regulation of miR-296 could significantly decrease the expression of P-glycoprotein, Bcl-2, and the transcription of MDR1, but up-regulate the expression of Bax. And down-regulation of miR-27a significantly decreased expression of MDR1, but did not alter the expression of MRP, miR-27a could possibly mediate drug resistance, at least in part through regulation of MDR1 and apoptosis.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Dihydropyrimidine dehydrogenase [NADP(+)] [29]
Molecule Alteration Methylation
Down-regulation
Sensitive Disease Esophageal cancer [ICD-11: 2B70.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell colony Inhibition hsa05200
Cell viability Inhibition hsa05200
In Vitro Model ECA-109 cells Esophagus Homo sapiens (Human) CVCL_6898
TE-1 cells Esophagus Homo sapiens (Human) CVCL_1759
KYSE150 cells Esophagus Homo sapiens (Human) CVCL_1348
TE-5 cells Esophageal Homo sapiens (Human) CVCL_1764
In Vivo Model BALB/c nude mouse xenograft mode Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
WST-1 assay; Flow cytometry assay
Mechanism Description Long noncoding RNA LINC00261 induces chemosensitization to 5-fluorouracil by mediating methylation-dependent repression of DPYD in human esophageal cancer.
Key Molecule: Transmembrane protease serine 11E (TM11E) [30]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Inhibition hsa05200
DESC1/EGFR/AKT signaling pathway Regulation hsa04012
In Vitro Model KYSE30 cells Esophagus Homo sapiens (Human) CVCL_1351
EC9706 cells Esophagus Homo sapiens (Human) CVCL_E307
KYSE140 cells Esophagus Homo sapiens (Human) CVCL_1347
TE13 cells Esophageal Homo sapiens (Human) CVCL_4463
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description TUSC7 suppressed the proliferation and chemotherapy resistance of ESCC cells by increasing DESC1 expression via inhibiting miR-224.
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: Pvt1 oncogene (PVT1) [35]
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 Cell apoptosis Inhibition hsa04210
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
In Vivo Model Nod/SCID mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description LncRNA PVT1 can inhibit apoptosis and enhance the 5-Fu resistance via Increasing Bcl2 expression in Gastric Cancer.
Key Molecule: Long non-protein coding RNA (XLOC_006753) [36]
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 Cell apoptosis Inhibition hsa04210
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
PI3K/AKT/mTOR signaling pathway Activation hsa04151
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Long non-coding RNA XLOC_006753 promotes the development of multidrug resistance in gastric cancer cells through the PI3k/Akt/mTOR signaling pathway.
Key Molecule: Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) [8]
Molecule Alteration Expression
Up-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
BGC823 cells Gastric Homo sapiens (Human) CVCL_3360
Experiment for
Molecule Alteration
RT-PCR; Luciferase reporter assay; Pull down assay
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description MALAT1 acts as a competing endogenous RNA for miR23b-3p and attenuates the inhibitory effect of miR23b-3p on ATG12, leading to chemo-induced autophagy and chemoresistance in GC cells.
Key Molecule: hsa-miR-23b-3p [8]
Molecule Alteration Expression
Down-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
BGC823 cells Gastric Homo sapiens (Human) CVCL_3360
Experiment for
Molecule Alteration
RT-PCR; Luciferase reporter assay; Pull down assay
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description MALAT1 acts as a competing endogenous RNA for miR23b-3p and attenuates the inhibitory effect of miR23b-3p on ATG12, leading to chemo-induced autophagy and chemoresistance in GC cells. MALAT1 promotes autophagy-associated chemoresistance of GC cells via sequestration of miR23b-3p.
Key Molecule: hsa-mir-145 [37]
Molecule Alteration Expression
Down-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
Experiment for
Molecule Alteration
RT-PCR; qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description microRNA-145 exerts tumor-suppressive and chemo-resistance lowering effects by targeting CD44 in gastric cancer.
Key Molecule: hsa-mir-27b [38]
Molecule Alteration Expression
Down-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
SGC-7901/DDP cells Gastric Homo sapiens (Human) CVCL_0520
SGC-7901/FU cells Gastric Homo sapiens (Human) CVCL_0520
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Annexin V-FITC Apoptosis assay
Mechanism Description LncRNA urothelial carcinoma associated 1 (UCA1) increases multi-drug resistance of gastric cancer via downregulating miR27b.
Key Molecule: Urothelial cancer associated 1 (UCA1) [38]
Molecule Alteration Expression
Up-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
SGC-7901/DDP cells Gastric Homo sapiens (Human) CVCL_0520
SGC-7901/FU cells Gastric Homo sapiens (Human) CVCL_0520
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Annexin V-FITC Apoptosis assay
Mechanism Description LncRNA urothelial carcinoma associated 1 (UCA1) increases multi-drug resistance of gastric cancer via downregulating miR27b.
Key Molecule: hsa-mir-363 [39]
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 Cell proliferation 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
HGC27 cells Gastric Homo sapiens (Human) CVCL_1279
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-363 promotes gastric cancer cells proliferation by inhibiting FBW7 expression and was associated with chemo-resistance of gastric cancer cells. Silencing FBW7 largely phenocopied miR-363-induced resistance to chemotherapy agents and promoted proliferation in gastric cancer cells. In addition, an inverse correlation between miR-363 and FBW7 mRNA expression was observed in gastric cancer tissues.
Key Molecule: hsa-mir-19a [17]
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 Cell apoptosis Inhibition hsa04210
PTEN/AKT signaling pathway Inhibition hsa05235
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
SGC7901/VCR cells Gastric Homo sapiens (Human) CVCL_VU58
SGC7901/ADR cells Gastric Homo sapiens (Human) CVCL_VU57
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-19a/b are upregulated in multidrug-resistant gastric cancer cell line, miR-19a/b suppress the sensitivity of gastric cancer cells to anticancer drugs, miR-19a/b accelerate the efflux of ADR through P-gp upregulation.
Key Molecule: hsa-mir-19b [17]
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 Cell apoptosis Inhibition hsa04210
PTEN/AKT signaling pathway Inhibition hsa05235
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
SGC7901/VCR cells Gastric Homo sapiens (Human) CVCL_VU58
SGC7901/ADR cells Gastric Homo sapiens (Human) CVCL_VU57
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-19a/b are upregulated in multidrug-resistant gastric cancer cell line, miR-19a/b suppress the sensitivity of gastric cancer cells to anticancer drugs, miR-19a/b accelerate the efflux of ADR through P-gp upregulation.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: Transforming growth factor beta 1 (TGFB1) [40]
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 [40]
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) [40]
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: Apoptosis regulator Bcl-2 (BCL2) [35]
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 Cell apoptosis Inhibition hsa04210
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
In Vivo Model Nod/SCID mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description LncRNA PVT1 can inhibit apoptosis and enhance the 5-Fu resistance via Increasing Bcl2 expression in Gastric Cancer.
Key Molecule: Extracellular matrix receptor III (CD44) [37]
Molecule Alteration Expression
Up-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description microRNA-145 exerts tumor-suppressive and chemo-resistance lowering effects by targeting CD44 in gastric cancer.
Key Molecule: F-box/WD repeat-containing protein 7 (FBXW7) [39]
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 Cell proliferation 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
HGC27 cells Gastric Homo sapiens (Human) CVCL_1279
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-363 promotes gastric cancer cells proliferation by inhibiting FBW7 expression and was associated with chemo-resistance of gastric cancer cells. Silencing FBW7 largely phenocopied miR-363-induced resistance to chemotherapy agents and promoted proliferation in gastric cancer cells. In addition, an inverse correlation between miR-363 and FBW7 mRNA expression was observed in gastric cancer tissues.
Key Molecule: Phosphatase and tensin homolog (PTEN) [17]
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 Cell apoptosis Inhibition hsa04210
PTEN/AKT signaling pathway Inhibition hsa05235
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
SGC7901/VCR cells Gastric Homo sapiens (Human) CVCL_VU58
SGC7901/ADR cells Gastric Homo sapiens (Human) CVCL_VU57
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-19a/b are upregulated in multidrug-resistant gastric cancer cell line, miR-19a/b suppress the sensitivity of gastric cancer cells to anticancer drugs, miR-19a/b accelerate the efflux of ADR through P-gp upregulation.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: HOX transcript antisense RNA (HOTAIR) [41]
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 invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model BGC-823 cells Gastric Homo sapiens (Human) CVCL_3360
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Bromodeoxyuridine incorporation assay; Flow cytometry assay; Transwell assay
Mechanism Description Down-regulation of HOTAIR could promote chemosensitivity, induce apoptosis of GC cells, and significantly inhibit GC cell proliferation, invasion, and metastasis in vivo and in vitro.
Key Molecule: hsa-mir-147 [42]
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 Activation hsa04210
Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
PI3K/AKT signaling pathway Inhibition hsa04151
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
AGS cells Gastric Homo sapiens (Human) CVCL_0139
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description miR147 suppressed the proliferation and enhanced the chemosensitivity of gastric cancer cells to 5-FU by promoting cell apoptosis through directly targeting PTEN and regulating the PI3k/AkT signaling pathway. knockdown of pten reverses the effects of miR147 downregulation on gastric cancer cells.
Key Molecule: hsa-mir-31 [43]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description Induction of miR31 in MkN-45 followed by suppression of RhoA expression resulted in increased sensitivity to 5-fluorouracil, inhibition of cell proliferation, and invasion compared to the control groups.
Key Molecule: Hepatocellular carcinoma up-regulated long non-coding RNA (HULC) [44]
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 MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
GES-1 cells Gastric Homo sapiens (Human) CVCL_EQ22
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometric analysis
Mechanism Description Silencing LncRNA HULC could enhance chemotherapy induced apoptosis in GC cells.
Key Molecule: hsa-mir-939 [45]
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 Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
RAF/MEK/ERK signaling pathway Inhibition hsa04010
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
AGS cells Gastric Homo sapiens (Human) CVCL_0139
GES-1 cells Gastric Homo sapiens (Human) CVCL_EQ22
HGC27 cells Gastric Homo sapiens (Human) CVCL_1279
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Colony formation assay; Flow cytometric analysis; Wound-healing, migration and invasion assay
Mechanism Description Decreased expression of miR939 contributes to chemoresistance and metastasis of gastric cancer via dysregulation of SLC34A2 and Raf/MEk/ERk pathway. miR939 exerted its function mainly through inhibiting SLC34A2/Raf/MEk/ERk pathway, which is activated in GC.
Key Molecule: hsa-mir-31 [46]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
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
Cell viability Inhibition hsa05200
In Vitro Model AGS cells Gastric Homo sapiens (Human) CVCL_0139
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description MkN-45-miR-31 showed an increased sensitivity to 5-FU, decreased migration and cell invasion compared to the control groups and induction of miR-31 expression in MkN-45 caused a significant reduction of E2F6 and SMUG1 genes.
Key Molecule: hsa-mir-31 [47]
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 colony Inhibition hsa05200
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell viability Inhibition hsa05200
In Vitro Model AGS cells Gastric Homo sapiens (Human) CVCL_0139
SNU-1 cells Gastric Homo sapiens (Human) CVCL_0099
SNU-5 cells Gastric Homo sapiens (Human) CVCL_0078
SNU-16 cells Gastric Homo sapiens (Human) CVCL_0076
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description microRNA-31 triggers G 2/M cell cycle arrest, enhances the chemosensitivity and inhibits migration and invasion of human gastric cancer cells by downregulating the expression of zeste homolog 2 (ZH2).
Key Molecule: hsa-mir-495 [48]
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
mTOR signaling pathway Inhibition hsa04150
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description The miR-495 exerts promotive effects on GC chemosensitivity via inactivation of the mTOR signaling pathway by suppressing ERBB2.
Key Molecule: hsa-miR-195-5p [49]
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: hsa-miR-623 [50]
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 viability Inhibition 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
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description The restored miR-623 expression could inhibit the proliferation of GC cells and enhance their chemosensitivity to 5-FU via the cell apoptosis pathway and the recovered CCND1 expression counteracted the effects of miR-623 on GC cell proliferation, chemosensitivity, and 5-FU-induced apoptosis.
Key Molecule: Protein lin-28 homolog A (CSDD1) [51]
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) [51]
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 [51]
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 [52]
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-197 [53]
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 MAPK signaling pathway Inhibition hsa04010
In Vitro Model 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
Mechanism Description When miR-197 was overexpressed in SGC7901 cells, the protein levels of MAPk1 were downregulated. Furthermore, MAPk1 knockdown significantly increased the growth inhibition rate of the SGC7901/5-FU cells compared with those in the control group. These results indicated that miR-197 may influence the sensitivity of 5-FU treatment in a gastric cancer cell line by targeting MAPk1.
Key Molecule: hsa-miR-23b-3p [54]
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 miR23b-3p/ATG12/HMGB2/autophagy regulatory loop signaling pathway Regulation hsa05206
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
BGC823 cells Gastric Homo sapiens (Human) CVCL_3360
AGS cells Gastric Homo sapiens (Human) CVCL_0139
In Vivo Model SCID-SHO mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description ATG12 and HMGB2 were the direct targets of miR-23b-3p. Meanwhile, ATG12 and HMGB2 were positively associated with the occurrence of autophagy. Reducing the expression of these target genes by siRNA or inhibition of autophagy both sensitized GC cells to chemotherapy. These findings suggest that a miR-23b-3p/ATG12/HMGB2/autophagy-regulatory loop has a critical role in MDR in GC. In addition, miR-23b-3p could be used as a prognostic factor for overall survival in GC. miR-23b-3p inhibited autophagy mediated by ATG12 and HMGB2 and sensitized GC cells to chemotherapy, and suggested the potential application of miR-23b-3p in drug resistance prediction and treatment.
Key Molecule: hsa-miR-508-5p [55]
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 Inhibition hsa04210
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
SGC7901/VCR cells Gastric Homo sapiens (Human) CVCL_VU58
SGC7901/ADR cells Gastric Homo sapiens (Human) CVCL_VU57
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The overexpression of miR-508-5p was sufficient to reverse cancer cell resistance to multiple chemotherapeutics in vitro and sensitize tumours to chemotherapy in vivo. Further studies showed that miR-508-5p could directly target the 3'-untranslated regions of ABCB1 and Zinc ribbon domain-containing 1 (ZNRD1), and suppress their expression at the mRNA and protein levels. Meanwhile, the suppression of ZNRD1 led to a decrease in ABCB1.
Key Molecule: hsa-mir-204 [56]
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.
Key Molecule: hsa-mir-27a [57]
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 proliferation Inhibition hsa05200
Tumorigenesis Inhibition hsa05200
In Vitro Model MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Down-regulation of miR-27a could also confer sensitivity of drugs on gastric cancer cells, and might increase accumulation and decrease releasing amount of adriamycin in gastric cancer cells. Down-regulation of miR-27a could significantly decrease the expression of P-glycoprotein and the transcriptional activity of cyclin D1, and up-regulate the expression of p21.
Key Molecule: hsa-mir-181 [58]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
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
SGC7901/VCR cells Gastric Homo sapiens (Human) CVCL_VU58
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The antiapoptotic protein BCL2 is upregulated, whereas miR-181b is downregulated in both SGC7901/VCR and A549/CDDP cells, compared with SGC7901 and A549 cells, respectively. Enforced miR-181b expression reduced BCL2 protein level and sensitized SGC7901/VCR and A549/CDDP cells to VCR-induced and CDDP-induced apoptosis, respectively.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Solute carrier family 34 member 2 (SLC34A2) [45]
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 invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
RAF/MEK/ERK signaling pathway Inhibition hsa04010
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
AGS cells Gastric Homo sapiens (Human) CVCL_0139
GES-1 cells Gastric Homo sapiens (Human) CVCL_EQ22
HGC27 cells Gastric Homo sapiens (Human) CVCL_1279
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Colony formation assay; Flow cytometric analysis; Wound-healing, migration and invasion assay
Mechanism Description Decreased expression of miR939 contributes to chemoresistance and metastasis of gastric cancer via dysregulation of SLC34A2 and Raf/MEk/ERk pathway. miR939 exerted its function mainly through inhibiting SLC34A2/Raf/MEk/ERk pathway, which is activated in GC.
Key Molecule: ATP-binding cassette sub-family B5 (ABCB5) [52]
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.
Key Molecule: Multidrug resistance protein 1 (ABCB1) [55]
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 Inhibition hsa04210
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
SGC7901/VCR cells Gastric Homo sapiens (Human) CVCL_VU58
SGC7901/ADR cells Gastric Homo sapiens (Human) CVCL_VU57
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The overexpression of miR-508-5p was sufficient to reverse cancer cell resistance to multiple chemotherapeutics in vitro and sensitize tumours to chemotherapy in vivo. Further studies showed that miR-508-5p could directly target the 3'-untranslated regions of ABCB1 and Zinc ribbon domain-containing 1 (ZNRD1), and suppress their expression at the mRNA and protein levels. Meanwhile, the suppression of ZNRD1 led to a decrease in ABCB1.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: hsa-mir-30a [59]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
SGC-7901/DDP cells Gastric Homo sapiens (Human) CVCL_0520
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR30a can decrease multidrug resistance (MDR) of gastric cancer cells, miR30a overexpression decreased the expression of P-gp, a MDR-related protein. It is also an important miRNA modulating EMT of the cancer cells.
Key Molecule: Death effector domain-containing protein (DEDD) [60]
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 viability Inhibition hsa05200
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
BGC823 cells Gastric Homo sapiens (Human) CVCL_3360
AGS cells Gastric Homo sapiens (Human) CVCL_0139
HGC27 cells Gastric Homo sapiens (Human) CVCL_1279
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
Experiment for
Molecule Alteration
Western blot analysis; RIP assay; Luciferase reporter assay
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description The inhibition of miR-17 may have tumor suppressive effects on GC and enhance its chemosensitivity by promoting DEDD, impairing EMT in GC cells.
Key Molecule: hsa-mir-17 [60]
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 viability Inhibition hsa05200
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
BGC823 cells Gastric Homo sapiens (Human) CVCL_3360
AGS cells Gastric Homo sapiens (Human) CVCL_0139
HGC27 cells Gastric Homo sapiens (Human) CVCL_1279
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description The inhibition of miR-17 may have tumor suppressive effects on GC and enhance its chemosensitivity by promoting DEDD, impairing EMT in GC cells.
Key Molecule: TGF-beta receptor type II (TGFBR2) [61]
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 invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
Cell viability Inhibition hsa05200
TGF-beta signaling pathway Inhibition hsa04350
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
AGS cells Gastric Homo sapiens (Human) CVCL_0139
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Caspase 3 assay kit
Mechanism Description Sensitization of Gastric Cancer Cells to 5-FU by microRNA-204 Through Targeting the TGFBR2-Mediated Epithelial to Mesenchymal Transition.
Key Molecule: hsa-mir-204 [61]
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
Cell viability Inhibition hsa05200
TGF-beta signaling pathway Inhibition hsa04350
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
AGS cells Gastric Homo sapiens (Human) CVCL_0139
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Caspase 3 assay kit
Mechanism Description Sensitization of Gastric Cancer Cells to 5-FU by microRNA-204 Through Targeting the TGFBR2-Mediated Epithelial to Mesenchymal Transition.
Key Molecule: Long non-protein coding RNA (LEIGC) [62]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Gastric carcinoma [ICD-11: 2B72.Z]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
AGS cells Gastric Homo sapiens (Human) CVCL_0139
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description Overexpression of LEIGC suppressed tumor growth and cell proliferation, and (+) the sensitivity of gastric cancer cells to 5-fluorouracil (5-FU), whereas knockdown of LEIGC showed the opposite effect. We further demonstrated LEIGC functions by inhibiting the epithelial-to-mesenchymal transition (EMT) in gastric cancer.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Phosphatase and tensin homolog (PTEN) [42]
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 proliferation Inhibition hsa05200
PI3K/AKT signaling pathway Inhibition hsa04151
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
AGS cells Gastric Homo sapiens (Human) CVCL_0139
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
Experiment for
Molecule Alteration
Luciferase reporter assay; Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description miR147 suppressed the proliferation and enhanced the chemosensitivity of gastric cancer cells to 5-FU by promoting cell apoptosis through directly targeting PTEN and regulating the PI3k/AkT signaling pathway. knockdown of pten reverses the effects of miR147 downregulation on gastric cancer cells.
Key Molecule: Transforming protein RhoA (RHOA) [43]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
Experiment for
Molecule Alteration
qRT-PCR; Western blot analysis; Immunohistochemical assay
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description Induction of miR31 in MkN-45 followed by suppression of RhoA expression resulted in increased sensitivity to 5-fluorouracil, inhibition of cell proliferation, and invasion compared to the control groups.
Key Molecule: Transcription factor E2F6 (E2F6) [46]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
Cell viability Inhibition hsa05200
In Vitro Model AGS cells Gastric Homo sapiens (Human) CVCL_0139
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description MkN-45-miR-31 showed an increased sensitivity to 5-FU, decreased migration and cell invasion compared to the control groups and induction of miR-31 expression in MkN-45 caused a significant reduction of E2F6 and SMUG1 genes.
Key Molecule: Single-strand selective monofunctional uracil DNA glycosylase (SMUG1) [46]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
Cell viability Inhibition hsa05200
In Vitro Model AGS cells Gastric Homo sapiens (Human) CVCL_0139
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description MkN-45-miR-31 showed an increased sensitivity to 5-FU, decreased migration and cell invasion compared to the control groups and induction of miR-31 expression in MkN-45 caused a significant reduction of E2F6 and SMUG1 genes.
Key Molecule: Zeste homolog 2 (ZH2) [47]
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 colony Inhibition hsa05200
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell viability Inhibition hsa05200
In Vitro Model AGS cells Gastric Homo sapiens (Human) CVCL_0139
SNU-1 cells Gastric Homo sapiens (Human) CVCL_0099
SNU-5 cells Gastric Homo sapiens (Human) CVCL_0078
SNU-16 cells Gastric Homo sapiens (Human) CVCL_0076
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description microRNA-31 triggers G 2/M cell cycle arrest, enhances the chemosensitivity and inhibits migration and invasion of human gastric cancer cells by downregulating the expression of zeste homolog 2 (ZH2).
Key Molecule: Receptor tyrosine-protein kinase erbB-2 (ERBB2) [48]
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
mTOR signaling pathway Inhibition hsa04150
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description The miR-495 exerts promotive effects on GC chemosensitivity via inactivation of the mTOR signaling pathway by suppressing ERBB2.
Key Molecule: Zinc finger protein with KRAB and SCAN domains 1 (ZKSCAN1) [49]
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: G1/S-specific cyclin-D1 (CCND1) [50]
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 viability Inhibition 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
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description The restored miR-623 expression could inhibit the proliferation of GC cells and enhance their chemosensitivity to 5-FU via the cell apoptosis pathway and the recovered CCND1 expression counteracted the effects of miR-623 on GC cell proliferation, chemosensitivity, and 5-FU-induced apoptosis.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [52]
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) [52]
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: Mitogen-activated protein kinase 1 (MAPK1) [53]
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 MAPK signaling pathway Inhibition hsa04010
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description When miR-197 was overexpressed in SGC7901 cells, the protein levels of MAPk1 were downregulated. Furthermore, MAPk1 knockdown significantly increased the growth inhibition rate of the SGC7901/5-FU cells compared with those in the control group. These results indicated that miR-197 may influence the sensitivity of 5-FU treatment in a gastric cancer cell line by targeting MAPk1.
Key Molecule: Ubiquitin-like protein ATG12 (ATG12) [54]
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 miR23b-3p/ATG12/HMGB2/autophagy regulatory loop signaling pathway Regulation hsa05206
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
BGC823 cells Gastric Homo sapiens (Human) CVCL_3360
AGS cells Gastric Homo sapiens (Human) CVCL_0139
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description ATG12 and HMGB2 were the direct targets of miR-23b-3p. Meanwhile, ATG12 and HMGB2 were positively associated with the occurrence of autophagy. Reducing the expression of these target genes by siRNA or inhibition of autophagy both sensitized GC cells to chemotherapy. These findings suggest that a miR-23b-3p/ATG12/HMGB2/autophagy-regulatory loop has a critical role in MDR in GC. In addition, miR-23b-3p could be used as a prognostic factor for overall survival in GC. miR-23b-3p inhibited autophagy mediated by ATG12 and HMGB2 and sensitized GC cells to chemotherapy, and suggested the potential application of miR-23b-3p in drug resistance prediction and treatment.
Key Molecule: High mobility group protein B2 (HMGB2) [54]
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 miR23b-3p/ATG12/HMGB2/autophagy regulatory loop signaling pathway Regulation hsa05206
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
BGC823 cells Gastric Homo sapiens (Human) CVCL_3360
AGS cells Gastric Homo sapiens (Human) CVCL_0139
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description ATG12 and HMGB2 were the direct targets of miR-23b-3p. Meanwhile, ATG12 and HMGB2 were positively associated with the occurrence of autophagy. Reducing the expression of these target genes by siRNA or inhibition of autophagy both sensitized GC cells to chemotherapy. These findings suggest that a miR-23b-3p/ATG12/HMGB2/autophagy-regulatory loop has a critical role in MDR in GC. In addition, miR-23b-3p could be used as a prognostic factor for overall survival in GC. miR-23b-3p inhibited autophagy mediated by ATG12 and HMGB2 and sensitized GC cells to chemotherapy, and suggested the potential application of miR-23b-3p in drug resistance prediction and treatment.
Key Molecule: DNA-directed RNA polymerase I subunit RPA12 (RPA12) [55]
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 Inhibition hsa04210
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
SGC7901/VCR cells Gastric Homo sapiens (Human) CVCL_VU58
SGC7901/ADR cells Gastric Homo sapiens (Human) CVCL_VU57
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The overexpression of miR-508-5p was sufficient to reverse cancer cell resistance to multiple chemotherapeutics in vitro and sensitize tumours to chemotherapy in vivo. Further studies showed that miR-508-5p could directly target the 3'-untranslated regions of ABCB1 and Zinc ribbon domain-containing 1 (ZNRD1), and suppress their expression at the mRNA and protein levels. Meanwhile, the suppression of ZNRD1 led to a decrease in ABCB1.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [56]
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.
Key Molecule: G1/S-specific cyclin-D1 (CCND1) [57]
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 proliferation Inhibition hsa05200
Tumorigenesis Inhibition hsa05200
In Vitro Model MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Down-regulation of miR-27a could also confer sensitivity of drugs on gastric cancer cells, and might increase accumulation and decrease releasing amount of adriamycin in gastric cancer cells. Down-regulation of miR-27a could significantly decrease the expression of P-glycoprotein and the transcriptional activity of cyclin D1, and up-regulate the expression of p21.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [58]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
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
SGC7901/VCR cells Gastric Homo sapiens (Human) CVCL_VU58
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The antiapoptotic protein BCL2 is upregulated, whereas miR-181b is downregulated in both SGC7901/VCR and A549/CDDP cells, compared with SGC7901 and A549 cells, respectively. Enforced miR-181b expression reduced BCL2 protein level and sensitized SGC7901/VCR and A549/CDDP cells to VCR-induced and CDDP-induced apoptosis, respectively.
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-195 [63]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description Inhibition of miR195 sensitized resistant cells to 5-FU by downregulating WEE1 and CHk1.
Key Molecule: hsa-miR-15b-5p [64]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
NF-kappaB signaling pathway Inhibition hsa04064
In Vitro Model DLD1 cells Colon Homo sapiens (Human) CVCL_0248
SW620 cells Colon Homo sapiens (Human) CVCL_0547
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
NCM460 cells Colon Homo sapiens (Human) CVCL_0460
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CellTiter-Glo Luminescent Cell Viability Assay; CCK8 assay; Flow cytometric analysis
Mechanism Description miR15b-5p resensitizes colon cancer cells to 5-fluorouracil by promoting apoptosis via the NF-kB/XIAP axis. miR15b-5p results in significant reductions in the levels of NF-kB1 and Ikk-alpha, two key modulators in inflammation and cell apoptosis.
Key Molecule: hsa-miR-206 [65]
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 proliferation Activation hsa05200
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
RkO cells Colon Homo sapiens (Human) CVCL_0504
In Vivo Model SCID mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTS assay; Flow cytometry assay
Mechanism Description miR-206 downregulation modulates 5-FU resistance in HCT116 cells by upregulating Bcl-2.
Key Molecule: hsa-mir-21 [66]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
PI3K/AKT signaling pathway Regulation hsa04151
In Vitro Model RkO cells Colon Homo sapiens (Human) CVCL_0504
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-21 can mediate the drug resistance to 5-FU by inhibiting its target PDCD4, which can regulate the expression of ABCC5 and CD44 genes.
Key Molecule: Long non-protein coding RNA (snaR) [67]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SNU-C4R cells Colon Homo sapiens (Human) CVCL_5111
SNU-C5R cells Colon Homo sapiens (Human) CVCL_5112
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Down-regulation of snaR decreased cell death after 5-FU treatment, which indicates that snaR loss decreases in vitro sensitivity to 5-FU in human colon cancer cells.
Key Molecule: hsa-mir-21 [68], [69]
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 migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model HT-29 cells Colon Homo sapiens (Human) CVCL_0320
HT-29/5-FU cells Colon Homo sapiens (Human) CVCL_0I27
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-21 targeted the human mutS homolog2 (hMSH2), and indirectly regulated the expression of thymidine phosphorylase (TP) and dihydropyrimidine dehydrogenase (DPD). These results demonstrate that miR-21 may play an important role in the 5-FU resistance of colon cancer cells. And high miR-21 expression was significantly associated with poor therapeutic outcome (P = 0.0001) and adjuvant therapy was associated with improved survival in patients with low miR-21.
Key Molecule: hsa-mir-19b [15]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model DLD1 cells Colon Homo sapiens (Human) CVCL_0248
KM12C cells Colon Homo sapiens (Human) CVCL_9547
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-19b and miR-21 were over-expressed in 5-FU-resistant cells. Ingenuity pathway analysis of mRNA targets significantly (P < 0.05) indicated the category "Cell Cycle" as a probable area of the molecular and cellular function related with 5-FU resistance. Among candidate mRNA targets, SFPQ and MYBL2 have been linked to cell cycle functions.
Key Molecule: hsa-mir-34 [4]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PI3K/AKT signaling pathway Activation hsa04151
In Vitro Model DLD1 cells Colon Homo sapiens (Human) CVCL_0248
DLD-1/5FU cells Colon Homo sapiens (Human) CVCL_0248
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Trypan blue dye exclusion assay
Mechanism Description The ectopic expression of miR-34a in the 5-FU-resistant cells inhibited growth, as in the parental cells, and attenuated the resistance to 5-FU through the down-regulation of Sirt1 and E2F3.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase Chk1 (CHK1) [63]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description Inhibition of miR195 sensitized resistant cells to 5-FU by downregulating WEE1 and CHk1.
Key Molecule: Wee1-like protein kinase (WEE1) [63]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description Inhibition of miR195 sensitized resistant cells to 5-FU by downregulating WEE1 and CHk1.
Key Molecule: Nuclear factor kappa-B kinase subunit alpha inhibitor (IKKalpha) [64]
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 Activation hsa04210
NF-kappaB signaling pathway Inhibition hsa04064
In Vitro Model DLD1 cells Colon Homo sapiens (Human) CVCL_0248
SW620 cells Colon Homo sapiens (Human) CVCL_0547
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
NCM460 cells Colon Homo sapiens (Human) CVCL_0460
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Dual-Luciferase Reporter Assay
Experiment for
Drug Resistance
CellTiter-Glo Luminescent Cell Viability Assay; CCK8 assay; Flow cytometric analysis
Mechanism Description miR15b-5p resensitizes colon cancer cells to 5-fluorouracil by promoting apoptosis via the NF-kB/XIAP axis. miR15b-5p results in significant reductions in the levels of NF-kB1 and Ikk-alpha, two key modulators in inflammation and cell apoptosis.
Key Molecule: DNA-binding factor KBF1 (p105) (NFKB1) [64]
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 Activation hsa04210
NF-kappaB signaling pathway Inhibition hsa04064
In Vitro Model DLD1 cells Colon Homo sapiens (Human) CVCL_0248
SW620 cells Colon Homo sapiens (Human) CVCL_0547
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
NCM460 cells Colon Homo sapiens (Human) CVCL_0460
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Dual-Luciferase Reporter Assay
Experiment for
Drug Resistance
CellTiter-Glo Luminescent Cell Viability Assay; CCK8 assay; Flow cytometric analysis
Mechanism Description miR15b-5p resensitizes colon cancer cells to 5-fluorouracil by promoting apoptosis via the NF-kB/XIAP axis. miR15b-5p results in significant reductions in the levels of NF-kB1 and Ikk-alpha, two key modulators in inflammation and cell apoptosis.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [65]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
RkO cells Colon Homo sapiens (Human) CVCL_0504
In Vivo Model SCID mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay; Flow cytometry assay
Mechanism Description miR-206 downregulation modulates 5-FU resistance in HCT116 cells by upregulating Bcl-2.
Key Molecule: Programmed cell death protein 4 (PDCD4) [66]
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
PI3K/AKT signaling pathway Regulation hsa04151
In Vitro Model RkO cells Colon Homo sapiens (Human) CVCL_0504
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-21 can mediate the drug resistance to 5-FU by inhibiting its target PDCD4, which can regulate the expression of ABCC5 and CD44 genes.
Key Molecule: DNA mismatch repair protein Msh2 (MSH2) [69]
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 invasion Activation hsa05200
Cell proliferation Activation hsa05200
In Vitro Model HT-29 cells Colon Homo sapiens (Human) CVCL_0320
HT-29/5-FU cells Colon Homo sapiens (Human) CVCL_0I27
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-21 targeted the human mutS homolog2 (hMSH2), and indirectly regulated the expression of thymidine phosphorylase (TP) and dihydropyrimidine dehydrogenase (DPD). These results demonstrate that miR-21 may play an important role in the 5-FU resistance of colon cancer cells.
Key Molecule: Myb-related protein B (MYBL2) [15]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model DLD1 cells Colon Homo sapiens (Human) CVCL_0248
KM12C cells Colon Homo sapiens (Human) CVCL_9547
Experiment for
Molecule Alteration
mRNA immunoprecipitation assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-19b and miR-21 were over-expressed in 5-FU-resistant cells. Ingenuity pathway analysis of mRNA targets significantly (P < 0.05) indicated the category "Cell Cycle" as a probable area of the molecular and cellular function related with 5-FU resistance. Among candidate mRNA targets, SFPQ and MYBL2 have been linked to cell cycle functions.
Key Molecule: Splicing factor/proline/glutamine-rich (SFPQ) [15]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model DLD1 cells Colon Homo sapiens (Human) CVCL_0248
KM12C cells Colon Homo sapiens (Human) CVCL_9547
Experiment for
Molecule Alteration
mRNA immunoprecipitation assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-19b and miR-21 were over-expressed in 5-FU-resistant cells. Ingenuity pathway analysis of mRNA targets significantly (P < 0.05) indicated the category "Cell Cycle" as a probable area of the molecular and cellular function related with 5-FU resistance. Among candidate mRNA targets, SFPQ and MYBL2 have been linked to cell cycle functions.
Key Molecule: Transcription factor E2F3 (E2F3) [4]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PI3K/AKT signaling pathway Activation hsa04151
In Vitro Model DLD1 cells Colon Homo sapiens (Human) CVCL_0248
DLD-1/5FU cells Colon Homo sapiens (Human) CVCL_0248
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Trypan blue dye exclusion assay
Mechanism Description The ectopic expression of miR-34a in the 5-FU-resistant cells inhibited growth, as in the parental cells, and attenuated the resistance to 5-FU through the down-regulation of Sirt1 and E2F3.
Key Molecule: NAD-dependent protein deacetylase sirtuin-1 (SIRT1) [4]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PI3K/AKT signaling pathway Activation hsa04151
In Vitro Model DLD1 cells Colon Homo sapiens (Human) CVCL_0248
DLD-1/5FU cells Colon Homo sapiens (Human) CVCL_0248
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Trypan blue dye exclusion assay
Mechanism Description The ectopic expression of miR-34a in the 5-FU-resistant cells inhibited growth, as in the parental cells, and attenuated the resistance to 5-FU through the down-regulation of Sirt1 and E2F3.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-338-3p [70]
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 miR338-3p/mTOR signaling pathway Activation hsa05206
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description miR338-3p increased 5-FU resistance by reducing the expression of its target gene, mTOR; and miR338-3p inhibitor sensitized HT29 (mutant p53) and HCT116 p53-/- (deficient p53) cells by activating mTOR; and miR338-3p-mTOR-autophagy was in the competition with 5-FU-induced apoptosis and contributed to the subsequent 5-FU resistance. (Inhibition of mTOR induces autophagy and depresses apoptosis to confer resistance to 5-FU).
Key Molecule: E3 ubiquitin-protein ligase XIAP (XIAP) [64]
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 migration Inhibition hsa04670
In Vitro Model DLD1 cells Colon Homo sapiens (Human) CVCL_0248
SW620 cells Colon Homo sapiens (Human) CVCL_0547
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
NCM460 cells Colon Homo sapiens (Human) CVCL_0460
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CellTiter-Glo Luminescent Cell Viability Assay; CCK8 assay; Flow cytometric analysis
Mechanism Description Overexpression of XIAP decreases the inhibitory effects of miR15b-5p on drug resistance in colon cancer cells. miR15b-5p mediates NF- B regulation by targeting the anti-apoptosis protein XIAP in vitro.
Key Molecule: hsa-mir-101 [71]
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 proliferation Inhibition hsa05200
In Vitro Model RkO cells Colon Homo sapiens (Human) CVCL_0504
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Colony formation assay; AO/EB double staining; Transwell invasion assay
Mechanism Description Upregulation of miR101 enhances the cytotoxic effect of anticancer drugs through inhibition of colon cancer cell proliferation. The upregulated expression of miR101 inhibited proliferation and migration, and increased the sensitivity of colon cancer cells to chemotherapy.
Key Molecule: hsa-mir-20b [72]
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 ADAM9/EGFR signaling pathway Inhibition hsa01521
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT116-R cells Colon Homo sapiens (Human) CVCL_AU09
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Annexin V apoptosis assay
Mechanism Description miR20b suppresses cell proliferation and apoptosis and regulates cell cycle progression by targeting ADAM9 in HCT116-R cells, miR20b reduces 5-FU resistance by suppressing the ADAM9/EGFR signaling pathway in colon cancer.
Key Molecule: hsa-mir-302a [73]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Trypan blue dye-exclusion assay; Annexin V-FITC apoptosis assay; Flow cytometer
Mechanism Description Both miR 302a and si IGF 1R inhibited Akt signaling. MiR 302a targeted IGF 1R and enhanced 5 FU induced cell death and viability inhibition in human colon cancer cells.
Key Molecule: hsa-mir-214 [74]
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 viability Inhibition hsa05200
In Vitro Model LOVO cells Colon Homo sapiens (Human) CVCL_0399
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; TUNEL assay
Mechanism Description miR-214 targeted heat shock protein 27 and could sensitize non-resistant colon cancer cells and 5-FU-resistant colon cancer cellsto 5-FU while overexpression of Hsp27 could block miR-214 with an effect on the sensitivity of colon cancer cells to 5-FU.
Key Molecule: hsa-miR-219a-5p [75]
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-494 [76]
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 5-Fu catabolic signaling pathway Regulation hsa00983
Cell apoptosis Activation hsa04210
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HCT8 cells Colon Homo sapiens (Human) CVCL_2478
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-494 also negatively regulated endogenous DPYD expression in SW480 cells. Overexpression or knockdown of DPYD could attenuate miR-494 mediated 5-Fu sensitivity regulation, suggesting the dependence of DPYD regulation in miR-494 activity. miR-494 inhibited SW480/5-Fu derived xenograft tumors growth in vivo at present of 5-Fu.
Key Molecule: hsa-mir-320 [77]
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.
Key Molecule: hsa-mir-34 [78]
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 DLD1 cells Colon Homo sapiens (Human) CVCL_0248
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description LDHA was shown to be a direct target of miR 34a. Overexpression of miR 34a reduced the expression of LDHA, probably through binding to the 3' untranslated region, leading to the re sensitization of 5 FU resistant cancer cells to 5 FU. Additionally, overexpression of LDHA rendered colon cancer cells resistant to 5 FU, suggesting that the miR 34a induced sensitization to 5 FU is mediated through the inhibition of LDHA. The current study showed that miR 34a is involved in sensitivity to 5 FU in part through its effects on LDHA expression.
Key Molecule: hsa-miR-142-3p [79]
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 CaCo2 cells Colon Homo sapiens (Human) CVCL_0025
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
In Vivo Model HT-29 xenograft mouse model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Annexin V assay
Mechanism Description The miR-142-3p was markedly decreased in coloncancer specimens, in which it was negatively correlated withthe expression of CD133, Lgr5, and ABCG2. Transfection of miR-142-3p mimics in colon cancer cells downregulated cyclin D1expression, induced G1phase cell cycle arrest, and elevatedthe sensitivity of the cells to 5-fluorouracil. Furthermore,OCT4 suppressed miR-142-3p, and hypomethylation of theOCT4promoter was associated with a reduction in miR-142-3p.
Key Molecule: hsa-mir-140 [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 proliferation Inhibition hsa05200
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description miR-140 is involved in the chemoresistance by reduced cell proliferation via G1 and G2 phase arrest mediated in part.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: ATP-binding cassette sub-family G2 (ABCG2) [79]
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 CaCo2 cells Colon Homo sapiens (Human) CVCL_0025
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
In Vivo Model HT-29 xenograft mouse model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Annexin V assay
Mechanism Description The miR-142-3p was markedly decreased in coloncancer specimens, in which it was negatively correlated withthe expression of CD133, Lgr5, and ABCG2. Transfection of miR-142-3p mimics in colon cancer cells downregulated cyclin D1expression, induced G1phase cell cycle arrest, and elevatedthe sensitivity of the cells to 5-fluorouracil. Furthermore,OCT4 suppressed miR-142-3p, and hypomethylation of theOCT4promoter was associated with a reduction in miR-142-3p.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase mTOR (mTOR) [70]
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 miR338-3p/mTOR signaling pathway Activation hsa05206
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Western blot analysis; Dual luciferase reporter assay
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description miR338-3p increased 5-FU resistance by reducing the expression of its target gene, mTOR; and miR338-3p inhibitor sensitized HT29 (mutant p53) and HCT116 p53-/- (deficient p53) cells by activating mTOR; and miR338-3p-mTOR-autophagy was in the competition with 5-FU-induced apoptosis and contributed to the subsequent 5-FU resistance. (Inhibition of mTOR induces autophagy and depresses apoptosis to confer resistance to 5-FU).
Key Molecule: Myeloma cell metalloproteinase (ADAM9) [72]
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 ADAM9/EGFR signaling pathway Inhibition hsa01521
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT116-R cells Colon Homo sapiens (Human) CVCL_AU09
Experiment for
Molecule Alteration
Western blot analysis; Luciferase reporter assay
Experiment for
Drug Resistance
MTT assay; Annexin V apoptosis assay
Mechanism Description miR20b suppresses cell proliferation and apoptosis and regulates cell cycle progression by targeting ADAM9 in HCT116-R cells, miR20b reduces 5-FU resistance by suppressing the ADAM9/EGFR signaling pathway in colon cancer.
Key Molecule: Insulin-like growth factor 1 receptor (IGF1R) [73]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Trypan blue dye-exclusion assay; Annexin V-FITC apoptosis assay; Flow cytometer
Mechanism Description Both miR 302a and si IGF 1R inhibited Akt signaling. MiR 302a targeted IGF 1R and enhanced 5 FU induced cell death and viability inhibition in human colon cancer cells.
Key Molecule: Heat shock protein beta-1 (HSPB1) [74]
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 viability Inhibition hsa05200
In Vitro Model LOVO cells Colon Homo sapiens (Human) CVCL_0399
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; TUNEL assay
Mechanism Description miR-214 targeted heat shock protein 27 and could sensitize non-resistant colon cancer cells and 5-FU-resistant colon cancer cellsto 5-FU while overexpression of Hsp27 could block miR-214 with an effect on the sensitivity of colon cancer cells to 5-FU.
Key Molecule: Sal-like protein 4 (SALL4) [75]
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: Dihydropyrimidine dehydrogenase [NADP(+)] [76]
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 5-Fu catabolic signaling pathway Regulation hsa00983
Cell apoptosis Activation hsa04210
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HCT8 cells Colon Homo sapiens (Human) CVCL_2478
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-494 also negatively regulated endogenous DPYD expression in SW480 cells. Overexpression or knockdown of DPYD could attenuate miR-494 mediated 5-Fu sensitivity regulation, suggesting the dependence of DPYD regulation in miR-494 activity. miR-494 inhibited SW480/5-Fu derived xenograft tumors growth in vivo at present of 5-Fu.
Key Molecule: Forkhead box protein M1 (FOXM1) [77]
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 apoptosis Activation hsa04210
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.
Key Molecule: Lactate dehydrogenase A (LDHA) [78]
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 DLD1 cells Colon Homo sapiens (Human) CVCL_0248
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description LDHA was shown to be a direct target of miR 34a. Overexpression of miR 34a reduced the expression of LDHA, probably through binding to the 3' untranslated region, leading to the re sensitization of 5 FU resistant cancer cells to 5 FU. Additionally, overexpression of LDHA rendered colon cancer cells resistant to 5 FU, suggesting that the miR 34a induced sensitization to 5 FU is mediated through the inhibition of LDHA. The current study showed that miR 34a is involved in sensitivity to 5 FU in part through its effects on LDHA expression.
Key Molecule: Prominin-1 (PROM1) [79]
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 CaCo2 cells Colon Homo sapiens (Human) CVCL_0025
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
In Vivo Model HT-29 xenograft mouse model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Annexin V assay
Mechanism Description The miR-142-3p was markedly decreased in coloncancer specimens, in which it was negatively correlated withthe expression of CD133, Lgr5, and ABCG2. Transfection of miR-142-3p mimics in colon cancer cells downregulated cyclin D1expression, induced G1phase cell cycle arrest, and elevatedthe sensitivity of the cells to 5-fluorouracil. Furthermore,OCT4 suppressed miR-142-3p, and hypomethylation of theOCT4promoter was associated with a reduction in miR-142-3p.
Key Molecule: Leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5) [79]
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 CaCo2 cells Colon Homo sapiens (Human) CVCL_0025
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
In Vivo Model HT-29 xenograft mouse model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Annexin V assay
Mechanism Description The miR-142-3p was markedly decreased in coloncancer specimens, in which it was negatively correlated withthe expression of CD133, Lgr5, and ABCG2. Transfection of miR-142-3p mimics in colon cancer cells downregulated cyclin D1expression, induced G1phase cell cycle arrest, and elevatedthe sensitivity of the cells to 5-fluorouracil. Furthermore,OCT4 suppressed miR-142-3p, and hypomethylation of theOCT4promoter was associated with a reduction in miR-142-3p.
Key Molecule: Histone deacetylase 4 (HDAC4) [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 proliferation Inhibition hsa05200
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Western blotting analysis; Immunofluorescence analysis
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description miR-140 is involved in the chemoresistance by reduced cell proliferation via G1 and G2 phase arrest mediated in part.
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: HOX transcript antisense RNA (HOTAIR) [1], [2]
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 migration Inhibition hsa04670
NF-kB signaling pathway Activation hsa04218
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description HOTAIR was associated with EZH2, which subsequently suppressed miR-218 expression, and HOTAIR contributes to 5FU resistance through suppressing miR-218 and activating NF-kB signaling in CRC. Thus, HOTAIR may serve as a promising therapeutic target for CRC patients.
Key Molecule: Long non-protein coding RNA 958 (LINC00958) [80]
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 invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model DLD1 cells Colon Homo sapiens (Human) CVCL_0248
SW620 cells Colon Homo sapiens (Human) CVCL_0547
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Colony formation assay
Mechanism Description BLACAT2 contributes to the cell proliferation, its levels were significantly increased in 5-fluorouracil-resistant cells, and overexpression of BLACAT2 was markedly associated with a low cell inhibition rate.
Key Molecule: hsa-mir-31 [81]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
DLD1 cells Colon Homo sapiens (Human) CVCL_0248
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Trypan blue dye-exclusion assay
Mechanism Description The increased expression level of miR-31 caused 5-FU resistance in colorectal cancer through silencing FIH-1, which is associated with cancer-specific energy metabolism.
Key Molecule: Small nucleolar RNA host gene 15 (SNHG15) [82]
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
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
In Vivo Model BALB/c-Rag2/-IL2cc/immunodeficient mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometry assay; Colony formation assay; MTS kit assay
Mechanism Description The levels of SNHG15 are related with the capacity of CRC cells to cope with the cytotoxic stress caused by 5-FU, which could be mediated by its interaction with AIF.
Key Molecule: GIHCG inhibitor of miR-200b/200a/429 expression (GIHCG) [83]
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 invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
Cell viability 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 [84]
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 metastasis Activation hsa05205
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 [85]
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: CDKN2B antisense RNA 1 (CDKN2B-AS1) [85]
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 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 regulating the expression of Let-7a in colorectal cancer.
Key Molecule: hsa-mir-218 [2]
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 metastasis Activation hsa05205
NF-kB signaling pathway Activation hsa04218
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
Experiment for
Molecule Alteration
qRT-PCR; luciferase reporter assay;ChIP
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description HOTAIR contributes to 5FU resistance through suppressing miR-218 and activating NF-kB signaling in CRC.
Key Molecule: Cytoskeleton regulator RNA (CYTOR) [86]
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 metastasis Activation hsa05205
Cell proliferation Activation hsa05200
Chemoresistance Activation hsa05207
miR139-5p/Notch1 signaling pathway Regulation hsa05206
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
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
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
RT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Long non-coding RNA LINC00152 promotes cell proliferation, metastasis, and confers 5-FU resistance in colorectal cancer by inhibiting miR139-5p. LINC00152 could regulate the expression of NOTCH1 through sponging miR139-5p and inhibiting its activity from promoting CRC progression and development.
Key Molecule: hsa-miR-139-5p [86]
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 metastasis Activation hsa05205
Cell proliferation Activation hsa05200
miR139-5p/Notch1 signaling pathway Regulation hsa05206
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
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
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 Long non-coding RNA LINC00152 promotes cell proliferation, metastasis, and confers 5-FU resistance in colorectal cancer by inhibiting miR139-5p. LINC00152 could regulate the expression of NOTCH1 through sponging miR139-5p and inhibiting its activity from promoting CRC progression and development.
Key Molecule: Novel transcript, antisense to MYRFL (ENST00000547547) [87]
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
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HCT116/5-FU cells Colon Homo sapiens (Human) CVCL_AU09
LOVO/5-FU cells Colon Homo sapiens (Human) CVCL_0399
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description Knockdown of miR31 increased the 5-FU sensitivity of CRC cells at least partly by upregulation of apoptosis. Overexpression of ENST00000547547 suppressed the anti-apoptotic effect of miR31 via competitive binding to it. ENST00000547547 reduces the 5-FU resistance via competitive binding to miR31 in CRC cells.
Key Molecule: hsa-mir-31 [87]
Molecule Alteration Function
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
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HCT116/5-FU cells Colon Homo sapiens (Human) CVCL_AU09
LOVO/5-FU cells Colon Homo sapiens (Human) CVCL_0399
Experiment for
Molecule Alteration
RNA immunoprecipitation (RIP) assay; Dual-luciferase reporter assay
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description Knockdown of miR31 increased the 5-FU sensitivity of CRC cells at least partly by upregulation of apoptosis. Overexpression of ENST00000547547 suppressed the anti-apoptotic effect of miR31 via competitive binding to it. ENST00000547547 reduces the 5-FU resistance via competitive binding to miR31 in CRC cells.
Key Molecule: HOX transcript antisense RNA (HOTAIR) [2]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation NF-kB signaling pathway Activation hsa04218
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
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
CCK8 assay; Colony formation assays
Mechanism Description LncRNA HOTAIR contributes to 5fu resistance through suppressing miR-218 and activating NF-kB/TS signaling in colorectal cancer.
Key Molecule: hsa-mir-218 [2]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation NF-kB signaling pathway Activation hsa04218
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
Experiment for
Molecule Alteration
qPCR; Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Colony formation assays
Mechanism Description LncRNA HOTAIR contributes to 5fu resistance through suppressing miR-218 and activating NF-kB/TS signaling in colorectal cancer.
Key Molecule: hsa-mir-135b [88]
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 proliferation Activation hsa05200
PI3K/AKT signaling pathway Activation hsa04151
In Vitro Model HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HCT-8/5-FU cells Colon Homo sapiens (Human) CVCL_2478
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Upregulation of microRNA-135b and microRNA-182 promotes chemoresistance of colorectal cancer by targeting ST6GALNAC2 via PI3k/AkT pathway. Inhibition of the PI3k/AkT pathway enhanced the chemosensitivity to 5-FU in HCT-8/5-FU and LoVo/5-FU.
Key Molecule: hsa-mir-182 [88]
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 proliferation Activation hsa05200
PI3K/AKT signaling pathway Activation hsa04151
In Vitro Model HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HCT-8/5-FU cells Colon Homo sapiens (Human) CVCL_2478
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Upregulation of microRNA-135b and microRNA-182 promotes chemoresistance of colorectal cancer by targeting ST6GALNAC2 via PI3k/AkT pathway. Inhibition of the PI3k/AkT pathway enhanced the chemosensitivity to 5-FU in HCT-8/5-FU and LoVo/5-FU.
Key Molecule: Sialyltransferase 7B (SIAT7B) [88]
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 proliferation Activation hsa05200
PI3K/AKT signaling pathway Activation hsa04151
In Vitro Model HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HCT-8/5-FU cells Colon Homo sapiens (Human) CVCL_2478
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Reporter gene assay; RT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Upregulation of microRNA-135b and microRNA-182 promotes chemoresistance of colorectal cancer by targeting ST6GALNAC2 via PI3k/AkT pathway. Inhibition of the PI3k/AkT pathway enhanced the chemosensitivity to 5-FU in HCT-8/5-FU and LoVo/5-FU.
Key Molecule: Long non-protein coding RNA (RP11-708H21.4) [89]
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 AKT/mTOR signaling pathway Activation hsa04150
Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
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
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR; Sequencing assay
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay
Mechanism Description Overexpressed RP11-708H21.4 suppresses CRC cell proliferation through inducing G1 arrest. Moreover, up-regulation of RP11-708H21.4 inhibits cell migration and invasion, causes cell apoptosis, and enhances 5-FU sensitivity of CRC cells.
Key Molecule: hsa-mir-106a [90]
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 viability Activation hsa05200
In Vitro Model SW620 cells Colon Homo sapiens (Human) CVCL_0547
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-106a Reduces 5-Fluorouracil (5-FU) Sensitivity of Colorectal Cancer by downregulating Dual-Specificity Phosphatases 2 (DUSP2).
Key Molecule: Pvt1 oncogene (PVT1) [91]
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 viability Activation hsa05200
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description The overexpression of PVT1 increased the mRNA and protein expression levels of multidrug resistance associated protein 1, P glycoprotein, serine/threonine protein kinase mTOR and apoptosis regulator Bcl2.
Key Molecule: Pvt1 oncogene (PVT1) [91]
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 viability Activation hsa05200
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-qPCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description The overexpression of PVT1 increased the mRNA and protein expression levels of multidrug resistance associated protein 1, P glycoprotein, serine/threonine protein kinase mTOR and apoptosis regulator Bcl2.
Key Molecule: hsa-miR-204-5p [92]
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
UCA1/miR204-5p ceRNA signaling pathway Regulation hsa05206
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
HCT8 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
CCK8 assay
Mechanism Description LncRNA-UCA1 enhances cell proliferation and 5-fluorouracil resistance in colorectal cancer by inhibiting miR-204-5p.We found that UCA1 was up-regulated in CRCs and negatively correlated with survival time in two CRC cohorts. Further mechanistic studies revealed that UCA1 could sponge endogenous miR-204-5p and inhibit its activity. We also identified CREB1 as a new target of miR-204-5p. The protein levels of CREB1 were significantly up-regulated in CRCs, negatively associated with survival time and positively correlated with the UCA1 expression. The present work provides the first evidence of a UCA1-miR-204-5p-CREB1/BCL2/RAB22A regulatory network in CRC and reveals that UCA1 and CREB1 are potential new oncogenes and prognostic factors for CRC.
Key Molecule: Urothelial cancer associated 1 (UCA1) [92]
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
UCA1/miR204-5p ceRNA signaling pathway Regulation hsa05206
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
HCT8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
qRT-PCR; Northern blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description LncRNA-UCA1 enhances cell proliferation and 5-fluorouracil resistance in colorectal cancer by inhibiting miR-204-5p.We found that UCA1 was up-regulated in CRCs and negatively correlated with survival time in two CRC cohorts. Further mechanistic studies revealed that UCA1 could sponge endogenous miR-204-5p and inhibit its activity. We also identified CREB1 as a new target of miR-204-5p. The protein levels of CREB1 were significantly up-regulated in CRCs, negatively associated with survival time and positively correlated with the UCA1 expression. The present work provides the first evidence of a UCA1-miR-204-5p-CREB1/BCL2/RAB22A regulatory network in CRC and reveals that UCA1 and CREB1 are potential new oncogenes and prognostic factors for CRC.
Key Molecule: Urothelial cancer associated 1 (UCA1) [92]
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
UCA1/miR204-5p ceRNA signaling pathway Regulation hsa05206
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
HCT8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
qRT-PCR; Northern blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description LncRNA-UCA1 enhances cell proliferation and 5-fluorouracil resistance in colorectal cancer by inhibiting miR-204-5p.We found that UCA1 was up-regulated in CRCs and negatively correlated with survival time in two CRC cohorts. Further mechanistic studies revealed that UCA1 could sponge endogenous miR-204-5p and inhibit its activity. We also identified CREB1 as a new target of miR-204-5p. The protein levels of CREB1 were significantly up-regulated in CRCs, negatively associated with survival time and positively correlated with the UCA1 expression. The present work provides the first evidence of a UCA1-miR-204-5p-CREB1/BCL2/RAB22A regulatory network in CRC and reveals that UCA1 and CREB1 are potential new oncogenes and prognostic factors for CRC.
Key Molecule: hsa-miR-450b-5p [93]
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 proliferation Activation hsa05200
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
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-450b-5p inhibited stemness and development of chemoresistance to 5-FU by targeting SOX2 in CRC cells.
Key Molecule: RAC serine/threonine-protein kinase (AKT) [94]
Molecule Alteration Phosphorylation
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
miR587/PPP2R1B/pAKT/XIAP signaling pathway Inhibition 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
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description AkT activation mediated by PPP2R1B contributes to miR-587-conferred 5-FU resistance in colon cancer cells.
Key Molecule: hsa-miR-587 [94]
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 Activation hsa04210
miR587/PPP2R1B/pAKT/XIAP signaling pathway Inhibition 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
RT-PCR; RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description microRNA-587 antagonizes 5-FU-induced apoptosis and confers drug resistance by inhibiting PPP2R1B expression in colorectal cancer.
Key Molecule: hsa-mir-520g [95]
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 [96]
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 [97]
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: Bcl-2-like protein 11 (BCL2L11) [98]
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 miR10b/BIM signaling pathway Activation hsa05206
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Luciferase assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-10b directly inhibits pro-apoptotic BIM, and the overexpression of miR-10b confers chemoresistance in colorectal cancer cells to 5-FU.
Key Molecule: hsa-mir-10b [98]
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 miR10b/BIM signaling pathway Activation hsa05206
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-10b directly inhibits pro-apoptotic BIM, and the overexpression of miR-10b confers chemoresistance in colorectal cancer cells to 5-FU.
Key Molecule: hsa-mir-21 [5]
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 proliferation Inhibition hsa05200
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
COLO 320DM cells Colon Homo sapiens (Human) CVCL_0219
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
FACS analysis
Mechanism Description The mismatch repair (MMR) system is involved in DNA damage recognition and repair. Human mutS homolog 2 (hMSH2) and human mutL homolog 1 (hMLH1) function as core MMR proteins and form heterodimers with protein homologs hMSH3 or hMSH6 and hMLH3 or hPMS2, respectively. Colorectal tumors that express a high level of miR-21 display reduced hMSH2 protein expression. Cells that overproduce miR-21 exhibit significantly reduced 5-fluorouracil (5-FU) -induced G2/M damage arrest and apoptosis that is characteristic of defects in the core MMR component.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance-associated protein 1 (MRP1) [85]
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 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: ABC-type oligopeptide transporter ABCB9 (ABCB9) [87]
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 invasion Activation hsa05200
Cell migration Activation hsa04670
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HCT116/5-FU cells Colon Homo sapiens (Human) CVCL_AU09
LOVO/5-FU cells Colon Homo sapiens (Human) CVCL_0399
Experiment for
Molecule Alteration
Western blot analysis; qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description ENST00000547547 promotes ABCB9 expression by acting as a sponge of miR31 and reduces the 5-FU resistance of CRC cells.
Key Molecule: Multidrug resistance-associated protein 1 (MRP1) [91]
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 viability Activation hsa05200
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 blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description The overexpression of PVT1 increased the mRNA and protein expression levels of multidrug resistance associated protein 1, P glycoprotein, serine/threonine protein kinase mTOR and apoptosis regulator Bcl2.
Key Molecule: ATP-binding cassette sub-family B5 (ABCB5) [91]
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 viability Activation hsa05200
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 blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description The overexpression of PVT1 increased the mRNA and protein expression levels of multidrug resistance associated protein 1, P glycoprotein, serine/threonine protein kinase mTOR and apoptosis regulator Bcl2.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: Long non-protein coding RNA (CCAL) [99]
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 [100]
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 [100]
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 [100]
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 [100]
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) [100]
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: SLC25A25 antisense RNA 1 (SLC25A25-AS1) [101]
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 migration Activation hsa04670
Cell proliferation Activation hsa05200
ERK/p38 signaling pathway Inhibition hsa04210
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description SLC25A25-AS1 overexpression significantly inhibited proliferation and colony formation in colorectal cancer cell lines, and downregulation of SLC25A25-AS1 obviously (+) chemoresistance and promoted EMT process in vitro associated with Erk and p38 signaling pathway activation. Therefore, SLC25A25-AS1 was determined to play a tumor suppressive role in CRC.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Histone-lysine N-methyltransferase EZH2 (EZH2) [1], [2]
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 metastasis Activation hsa05205
NF-kB signaling pathway Activation hsa04218
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
Experiment for
Molecule Alteration
RIP experiments
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description HOTAIR directly recruits EZH2 and subsequently suppresses miR-218 expression by binding to its promoter and contributes to 5FU resistance through activating NF-kB/TS Signaling in colorectal cancer.
Key Molecule: Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) [81]
Molecule Alteration Expression
Down-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
DLD1 cells Colon Homo sapiens (Human) CVCL_0248
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
Trypan blue dye-exclusion assay
Mechanism Description The increased expression level of miR-31 caused 5-FU resistance in colorectal cancer through silencing FIH-1, which is associated with cancer-specific energy metabolism.
Key Molecule: Apoptosis-inducing factor 1 (AIFM1) [82]
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
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
In Vivo Model BALB/c-Rag2/-IL2cc/immunodeficient mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RIP experiments
Experiment for
Drug Resistance
Flow cytometry assay; Colony formation assay; MTS kit assay
Mechanism Description The levels of SNHG15 are related with the capacity of CRC cells to cope with the cytotoxic stress caused by 5-FU, which could be mediated by its interaction with AIF.
Key Molecule: Piwi-like protein 2 (PIWIL2) [84]
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: DNA-binding factor KBF1 (p105) (NFKB1) [2]
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 metastasis Activation hsa05205
NF-kB signaling pathway Activation hsa04218
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
Experiment for
Molecule Alteration
Western blot analysis; luciferase reporter assay;ChIP
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description HOTAIR contributes to 5FU resistance through suppressing miR-218 and activating NF-kB signaling in CRC.
Key Molecule: EGFR-coamplified and overexpressed protein (ECOP) [2]
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 metastasis Activation hsa05205
NF-kB signaling pathway Activation hsa04218
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
SW480 cells Colon Homo sapiens (Human) CVCL_0546
Experiment for
Molecule Alteration
Western blot analysis; luciferase reporter assay;ChIP
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description VOPP1 overexpression partially reversed the miR-218-induced enhanced susceptibility to 5FU in the HT29 5FU-R subline and HOTAIR knockdown partially reversed 5FU resistance through promoting miR-218 and inactivating NF-kB signaling.
Key Molecule: DNA-binding factor KBF1 (p105) (NFKB1) [2]
Molecule Alteration Phosphorylation
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation NF-kB signaling pathway Activation hsa04218
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
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Colony formation assays
Mechanism Description LncRNA HOTAIR contributes to 5fu resistance through suppressing miR-218 and activating NF-kB/TS signaling in colorectal cancer.
Key Molecule: EGFR-coamplified and overexpressed protein (ECOP) [2]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation NF-kB signaling pathway Activation hsa04218
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
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
CCK8 assay; Colony formation assays
Mechanism Description LncRNA HOTAIR contributes to 5fu resistance through suppressing miR-218 and the activation of VOPP1 expression and activating NF-kB/TS signaling in colorectal cancer.
Key Molecule: RAC-alpha serine/threonine-protein kinase (AKT1) [89]
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 AKT/mTOR signaling pathway Activation hsa04150
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
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay
Mechanism Description Overexpressed RP11-708H21.4 suppresses CRC cell proliferation through inducing G1 arrest. Moreover, up-regulation of RP11-708H21.4 inhibits cell migration and invasion, causes cell apoptosis, and enhances 5-FU sensitivity of CRC cells.
Key Molecule: Serine/threonine-protein kinase mTOR (mTOR) [89]
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 AKT/mTOR signaling pathway Activation hsa04150
Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
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
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay
Mechanism Description Overexpressed RP11-708H21.4 suppresses CRC cell proliferation through inducing G1 arrest. Moreover, up-regulation of RP11-708H21.4 inhibits cell migration and invasion, causes cell apoptosis, and enhances 5-FU sensitivity of CRC cells.
Key Molecule: Ribosomal protein S6 kinase beta-1 (RPS6KB1) [89]
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 AKT/mTOR signaling pathway Activation hsa04150
Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
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
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay
Mechanism Description Overexpressed RP11-708H21.4 suppresses CRC cell proliferation through inducing G1 arrest. Moreover, up-regulation of RP11-708H21.4 inhibits cell migration and invasion, causes cell apoptosis, and enhances 5-FU sensitivity of CRC cells.
Key Molecule: Dual specificity protein phosphatase 2 (DUSP2) [90]
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 viability Activation hsa05200
In Vitro Model SW620 cells Colon Homo sapiens (Human) CVCL_0547
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-106a Reduces 5-Fluorouracil (5-FU) Sensitivity of Colorectal Cancer by downregulating Dual-Specificity Phosphatases 2 (DUSP2).
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [91]
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 viability Activation hsa05200
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 blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description The overexpression of PVT1 increased the mRNA and protein expression levels of multidrug resistance associated protein 1, P glycoprotein, serine/threonine protein kinase mTOR and apoptosis regulator Bcl2.
Key Molecule: Serine/threonine-protein kinase mTOR (mTOR) [91]
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 viability Activation hsa05200
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 blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description The overexpression of PVT1 increased the mRNA and protein expression levels of multidrug resistance associated protein 1, P glycoprotein, serine/threonine protein kinase mTOR and apoptosis regulator Bcl2.
Key Molecule: Cyclic AMP-responsive element-binding protein 1 (CREB1) [92]
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
UCA1/miR204-5p ceRNA signaling pathway Regulation hsa05206
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
HCT8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description LncRNA-UCA1 enhances cell proliferation and 5-fluorouracil resistance in colorectal cancer by inhibiting miR-204-5p.We found that UCA1 was up-regulated in CRCs and negatively correlated with survival time in two CRC cohorts. Further mechanistic studies revealed that UCA1 could sponge endogenous miR-204-5p and inhibit its activity. We also identified CREB1 as a new target of miR-204-5p. The protein levels of CREB1 were significantly up-regulated in CRCs, negatively associated with survival time and positively correlated with the UCA1 expression. The present work provides the first evidence of a UCA1-miR-204-5p-CREB1/BCL2/RAB22A regulatory network in CRC and reveals that UCA1 and CREB1 are potential new oncogenes and prognostic factors for CRC.
Key Molecule: Transcription factor SOX-2 (SOX2) [93]
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 proliferation Activation hsa05200
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
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-450b-5p inhibited stemness and development of chemoresistance to 5-FU by targeting SOX2 in CRC cells.
Key Molecule: PP2A subunit A isoform R1-beta (PPP2R1B) [94]
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 Activation hsa04210
miR587/PPP2R1B/pAKT/XIAP signaling pathway Inhibition 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
Mechanism Description microRNA-587 antagonizes 5-FU-induced apoptosis and confers drug resistance by inhibiting PPP2R1B expression in colorectal cancer.
Key Molecule: Ribonuclease P protein subunit p21 (RPP21) [95]
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) [96]
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: DNA mismatch repair protein Msh2 (MSH2) [5]
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 proliferation Inhibition hsa05200
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
COLO 320DM cells Colon Homo sapiens (Human) CVCL_0219
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
FACS analysis
Mechanism Description The mismatch repair (MMR) system is involved in DNA damage recognition and repair. Human mutS homolog 2 (hMSH2) and human mutL homolog 1 (hMLH1) function as core MMR proteins and form heterodimers with protein homologs hMSH3 or hMSH6 and hMLH3 or hPMS2, respectively. Colorectal tumors that express a high level of miR-21 display reduced hMSH2 protein expression. Cells that overproduce miR-21 exhibit significantly reduced 5-fluorouracil (5-FU) -induced G2/M damage arrest and apoptosis that is characteristic of defects in the core MMR component.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-31 [87]
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
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HCT116/5-FU cells Colon Homo sapiens (Human) CVCL_AU09
LOVO/5-FU cells Colon Homo sapiens (Human) CVCL_0399
Experiment for
Molecule Alteration
RNA immunoprecipitation (RIP) assay; Dual-luciferase reporter assay
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description ABCB9 is a transporter which was reported to be targeted by miR31, involved in cisplatin-induced apoptosis, thus knockdown of miR31 increases the 5-FU sensitivity of CRC cell lines. ENST00000547547 reduces the 5-FU resistance via competitive binding to miR31.
Key Molecule: Novel transcript, antisense to MYRFL (ENST00000547547) [87]
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
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HCT116/5-FU cells Colon Homo sapiens (Human) CVCL_AU09
LOVO/5-FU cells Colon Homo sapiens (Human) CVCL_0399
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description ABCB9 is a transporter which was reported to be targeted by miR31, involved in cisplatin-induced apoptosis, thus knockdown of miR31 increases the 5-FU sensitivity of CRC cell lines. ENST00000547547 reduces the 5-FU resistance via competitive binding to miR31.
Key Molecule: hsa-miR-874-3p [102]
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 Hippo signaling pathway Activation hsa04391
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Caspase-9 or 3 activity assays; Flow cytometric analysis
Mechanism Description Down-regulation of miR874-3p promotes chemotherapeutic resistance in colorectal cancer via inactivation of the Hippo signaling pathway. miR874-3p directly inhibited the expression of transcriptional co-activators YAP and TAZ of the Hippo signaling pathway, resulting in the inactivation of the TEAD transcription.
Key Molecule: hsa-miR-3190-5p [103]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
CHO cells Ovary Homo sapiens (Human) CVCL_0213
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The expression of ABCC4 was inhibited by miR3190-5p through binding to the 3'-UTR of the ABCC4 gene, this regulatory role of miR3190-5p was disrupted by rs3742106. The rs3742106 T allele offers a binding-site for miR3190-5p, which results in low-expression of ABCC4, increased intracellular concentration of 5-FU, and enhanced sensitivity to 5-FU treatment.
Key Molecule: hsa-miR-196b-5p [104]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation JAKT2/STAT3 signaling pathway Inhibition hsa04030
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
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
COLO 205 cells Colon Homo sapiens (Human) CVCL_0218
COLO 320DM cells Colon Homo sapiens (Human) CVCL_0219
CW-2 cells Colon Homo sapiens (Human) CVCL_1151
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
LS174T cells Colon Homo sapiens (Human) CVCL_1384
NCI-H716 cells Colon Homo sapiens (Human) CVCL_1581
SW948 cells Colon Homo sapiens (Human) CVCL_0632
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Caspase-9 or -3 activity assays; Spheroid formation assay; Flow cytometric analysis; MTT assay
Mechanism Description miR196b-5p promotes stemness and chemoresistance of CRC cells to 5-fluorouracil via targeting negative regulators SOCS1 and SOCS3 of STAT3 signaling pathway, giving rise to activation of STAT3 signaling.
Key Molecule: Long non-protein coding RNA (RP11-708H21.4) [89]
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 AKT/mTOR signaling pathway Inhibition hsa04150
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
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay
Mechanism Description Overexpressed RP11-708H21.4 suppresses CRC cell proliferation through inducing G1 arrest. Moreover, up-regulation of RP11-708H21.4 inhibits cell migration and invasion, causes cell apoptosis, and enhances 5-FU sensitivity of CRC cells.
Key Molecule: hsa-mir-200c [105]
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 AKT signaling pathway Inhibition hsa04151
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Annexin V/ PI staining; Caspase-3 activity assay
Mechanism Description Levels of PTEN and E-cadherin were reduced by knockdown of miR200c in HCT-116 cells, PTEN inactivate the AkT signaling pathway, and E-cadherin is one of the major downstream regulators of miRNA-200c contributing to EMT, which is also important to inhibit tumor invasion and proliferation as well as to induce cell apoptosis.
Key Molecule: hsa-mir-125b [106]
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 autophagy Activation hsa04140
Wnt/Beta-catenin signaling pathway Activation hsa04310
In Vitro Model SW620 cells Colon Homo sapiens (Human) CVCL_0547
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Annexin V-FITC/PI staining assay
Mechanism Description CXCL12/CXCR4 axis induced miR125b promotes invasion and confers 5-fluorouracil resistance through enhancing autophagy in colorectal cancer There was a negative correlation of the expression of miR125b with APC mRNA in paired human colorectal tissue specimens. The upregulation of miR125b activated the Wnt/beta-catenin signaling by targeting APC gene and contributed to 5-FU resistance through enhancing cell autophagy.
Key Molecule: hsa-miR-577 [107]
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
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
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; BrdU incorporation assay; Flow cytometric analysis
Mechanism Description Ectopic expression of miR577 enhanced 5-FU sensitivity in SW480/5-FU cells by down-regulating HSP27. Enforced expression of HSP27 reversed the effects of miR577 on CRC cell growth and 5-FU sensitivity.
Key Molecule: hsa-mir-141 [108]
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 [109]
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-543 [110]
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
PTEN/PI3K/AKT signaling pathway Activation hsa05235
In Vitro Model HCT8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Transwell assay; Flow cytometry assay
Mechanism Description miR-543 enhanced drug resistance by down-regulating the expression of phosphatase and tensin homolog (PTEN), which negatively regulates protein kinase B (AkT) activation while an elevated expression of PTEN reversed the chemoresistance of miR-543-overexpressing HCT8 cells to 5-FU.
Key Molecule: hsa-miR-195-5p [111]
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
Notch signaling pathway Inhibition hsa04330
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
SW620 cells Colon Homo sapiens (Human) CVCL_0547
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
HCT-160 cells Colorectal Homo sapiens (Human) N.A.
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Caspase-3 activity
Mechanism Description miR-195-5p regulates CRC cell stemness and 5-FU resistance through Notch2 and RBPJ.
Key Molecule: AT-rich interactive domain-containing protein 4B (ARID4B) [112]
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 proliferation Inhibition hsa05200
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 Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR-519b-3p mimics promoted HCT116 and SW480 cells more sensitive to chemoradiation treatment while ectopic expression of ARID4B in the meantime decreased the sensitivity.
Key Molecule: hsa-miR-519b-3p [112]
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 proliferation Inhibition hsa05200
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 Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR-519b-3p mimics promoted HCT116 and SW480 cells more sensitive to chemoradiation treatment while ectopic expression of ARID4B in the meantime decreased the sensitivity.
Key Molecule: hsa-miR-761 [113]
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 viability Inhibition 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
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-761 suppressed colorectal cancer cell proliferation and invasion by downregulating FOXM1.
Key Molecule: hsa-mir-497 [114], [115]
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
Cell proliferation Inhibition hsa05200
MAPK/ERK signaling pathway Inhibition hsa04010
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miRNA-497 targeted Smurf1 in CRC cells and the Smurf1 expression level was dramatically increased in neoadjuvant therapy-resistant patients compared with treatment-sensitive patients. These results indicate that down-regulation of miRNA-497 in colorectal cancer may contribute to the resistance of CRC cells to 5-FU treatment. Thus, miRNA-497 has the potential to be a novel biomarker for predicting the neoadjuvant chemotherapy sensitivity in CRC patients.
Key Molecule: hsa-mir-149 [116]
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 AKT/mTOR signaling pathway Regulation hsa04150
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
LOVO cells Colon Homo sapiens (Human) CVCL_0399
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Reduced miR-149 is a critical factor in the mechanisms by which CRC cells resist the cytotoxicity of 5-FU. Also, re-expression of miR-149 could increase the 5-FU sensitivity of CRC cells via enhancing 5-FU-inducing apoptosis by targeting FOXM1.
Key Molecule: hsa-mir-874 [117]
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 proliferation Inhibition hsa05200
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
LOVO cells Colon Homo sapiens (Human) CVCL_0399
NCM460 cells Colon Homo sapiens (Human) CVCL_0460
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
HCT-116 cells Colon Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-874 inhibits growth, increases apoptosis and enhances chemosensitivity in CRC cells by targeting XIAP.
Key Molecule: hsa-miR-139-5p [118]
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 proliferation Inhibition hsa05200
IGF-1R/AKT/S6 signaling pathway Inhibition hsa05225
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
LOVO cells Colon Homo sapiens (Human) CVCL_0399
In Vivo Model Mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Transwell assay
Mechanism Description Ectopic expression of miR-139-5p sensitized CRC cells to 5-FU by increasing 5-FU-induced apoptosis. In addition, miR-139-5p inhibited the expression of the miR-139-5p target gene NOTCH-1 and its downstream molecules MRP-1 and BCL-2, two key MDR-associated genes. Furthermore, silencing NOTCH-1 expression promoted the chemotherapeutic effects of 5-FU, and up-regulation of NOTCH-1 abrogated miR-139-5p-mediated sensitization to 5-FU in LoVo and HCT-116 cells.
Key Molecule: hsa-miR-204-5p [92]
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
UCA1/miR204-5p ceRNA signaling pathway Regulation hsa05206
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
HCT8 cells Colon Homo sapiens (Human) CVCL_2478
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR; Northern blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description LncRNA-UCA1 enhances cell proliferation and 5-fluorouracil resistance in colorectal cancer by inhibiting miR-204-5p.We found that UCA1 was up-regulated in CRCs and negatively correlated with survival time in two CRC cohorts. Further mechanistic studies revealed that UCA1 could sponge endogenous miR-204-5p and inhibit its activity. We also identified CREB1 as a new target of miR-204-5p. The protein levels of CREB1 were significantly up-regulated in CRCs, negatively associated with survival time and positively correlated with the UCA1 expression. The present work provides the first evidence of a UCA1-miR-204-5p-CREB1/BCL2/RAB22A regulatory network in CRC and reveals that UCA1 and CREB1 are potential new oncogenes and prognostic factors for CRC.
Key Molecule: hsa-miR-425-5p [119]
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-22 [120]
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 invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model SW620 cells Colon Homo sapiens (Human) CVCL_0547
RkO cells Colon Homo sapiens (Human) CVCL_0504
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR; RT-PCR
Experiment for
Drug Resistance
Trypan blue exclusion assay
Mechanism Description Tumor cells undergoing autophagy may affect the sensitivity of 5-FU by repressing miR-22 expression. miR-22 will facilitate 5-FU to kill tumor cells when it was exotically introduced into the tumor cells, and tumor cells with higher levels of miR-22 were more sensitive to 5-FU. starvation induced up-regulation of BTG1 in CRC cells was inversely correlated with miR-22, which further demonstrated that miR-22 may influence cells under stress. More importantly, BTG1 can reverse the inhibition of autophagy induced by overexpression of miR-22, and the knockdown of BTG1 can reduce the level of autophagy resulting from the down-regulation of miR-22 in CRC cells with 5-FU treatment. Similarly, the data from clinical samples indicated that the miR-22 level was inversely correlated with the expression of BTG1, and the tumors with higher miR-22 level were more sensitive to 5-FU.
Key Molecule: hsa-mir-23a [121]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation APAF-1/caspase-9 apoptotic signaling pathway Activation hsa04210
Cell apoptosis Inhibition hsa04210
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
RT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-23a may inhibit 5-FU-induced apoptosis through the APAF-1/caspase-9 pathway and provide new insight into CRC treatment.
Key Molecule: hsa-mir-34 [122]
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 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
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
A real-time cell analyzer assay
Mechanism Description c-KIT was shown to mediate chemo-resistance (kike 5-FU) in ovarian tumor initiating cells, miR-34a inhibits Erk signaling and colony formation by down-regulation of c-kit, miR-34a can inhibit this effect via down-regulation of c-kit and therefore sensitize cells to chemotherapeutic treatment.
Key Molecule: hsa-mir-129 [123]
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 proliferation Inhibition hsa05200
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
In Vivo Model CRC nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
WST assay
Mechanism Description microRNA-129 (miR-129) trigger apoptosis by suppressing key anti-apoptotic protein, B-cell lymphoma 2 (BCL2), enhanced the cytotoxic effect of 5-fluorouracil both in vitro and in vivo.
Key Molecule: hsa-mir-497 [124]
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
MEK/ERK signaling pathway Inhibition hsa04011
PI3K/AKT signaling pathway Inhibition hsa04151
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
COLO 205 cells Colon Homo sapiens (Human) CVCL_0218
HCT28 cells Colon Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description IGF1-R has an important role in mediating activation of the PI3k/Akt pathway, miR-497 inhibits PI3k/Akt signalling. Down-regulation of miR-497 is an important mechanism of upregulation of IGF1-R in CRC cells that contributes to malignancy of CRC.
Key Molecule: hsa-mir-222 [125]
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-20a [126]
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.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: hsa-mir-218 [127]
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 [128]
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.
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [129]
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 proliferation Inhibition hsa05200
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT asssay; Innocyte invasion assay
Mechanism Description microRNA-224 was differentially expressed in dysplastic colorectal disease and in isogeneic kRAS WT and mutant HCT116 cells. Antagomir-mediated miR-224 silencing in HCT116 kRAS WT cells phenocopied kRAS mutation, increased kRAS activity and ERk and AkT phosphorylation. 5-FU chemosensitivity was significantly increased in miR-224 knockdown cells, and in NIH3T3 cells expressing kRAS and BRAF mutant proteins. Bioinformatics analysis of predicted miR-224 target genes predicted altered cell proliferation, invasion and epithelial-mesenchymal transition (EMT) phenotypes that were experimentally confirmed in miR-224 knockdown cells.
Key Molecule: GTPase KRas (KRAS) [129]
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 proliferation Inhibition hsa05200
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT asssay; Innocyte invasion assay
Mechanism Description microRNA-224 was differentially expressed in dysplastic colorectal disease and in isogeneic kRAS WT and mutant HCT116 cells. Antagomir-mediated miR-224 silencing in HCT116 kRAS WT cells phenocopied kRAS mutation, increased kRAS activity and ERk and AkT phosphorylation. 5-FU chemosensitivity was significantly increased in miR-224 knockdown cells, and in NIH3T3 cells expressing kRAS and BRAF mutant proteins. Bioinformatics analysis of predicted miR-224 target genes predicted altered cell proliferation, invasion and epithelial-mesenchymal transition (EMT) phenotypes that were experimentally confirmed in miR-224 knockdown cells.
Key Molecule: hsa-mir-224 [129]
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 proliferation Inhibition hsa05200
In Vitro Model 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
MTT asssay; Innocyte invasion assay
Mechanism Description microRNA-224 was differentially expressed in dysplastic colorectal disease and in isogeneic kRAS WT and mutant HCT116 cells. Antagomir-mediated miR-224 silencing in HCT116 kRAS WT cells phenocopied kRAS mutation, increased kRAS activity and ERk and AkT phosphorylation. 5-FU chemosensitivity was significantly increased in miR-224 knockdown cells, and in NIH3T3 cells expressing kRAS and BRAF mutant proteins. Bioinformatics analysis of predicted miR-224 target genes predicted altered cell proliferation, invasion and epithelial-mesenchymal transition (EMT) phenotypes that were experimentally confirmed in miR-224 knockdown cells.
Key Molecule: hsa-mir-145 [130]
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 Activation hsa05200
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
DLD1 cells Colon Homo sapiens (Human) CVCL_0248
SW620 cells Colon Homo sapiens (Human) CVCL_0547
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LS174T cells Colon Homo sapiens (Human) CVCL_1384
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Trypan blue assay; Sulforhodamine B assay
Mechanism Description Inhibition of SNAI2 directly with short hairpin sequence for SNAI2 and miR145 replacement therapy both decreased vimentin expression and increased in vitro 5FU sensitivity.
Key Molecule: Zinc finger protein SNAI2 (SNAI2) [130]
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 Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
DLD1 cells Colon Homo sapiens (Human) CVCL_0248
SW620 cells Colon Homo sapiens (Human) CVCL_0547
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LS174T cells Colon Homo sapiens (Human) CVCL_1384
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Trypan blue assay; Sulforhodamine B assay
Mechanism Description Inhibition of SNAI2 directly with short hairpin sequence for SNAI2 and miR145 replacement therapy both decreased vimentin expression and increased in vitro 5FU sensitivity.
Key Molecule: hsa-mir-143 [131]
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 proliferation Inhibition hsa05200
NF-kappaB signaling pathway Regulation hsa04064
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-143 increases the sensitivity of colon cancer cells to 5-fluorouracil, probably acting through extracellular-regulated protein kinase 5/nuclear factor-kB regulated pathways.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Tafazzin (TAZ) [102]
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 Hippo signaling pathway Activation hsa04391
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Luciferase assay; miRNA immunoprecipitation assay
Experiment for
Drug Resistance
Caspase-9 or 3 activity assays; Flow cytometric analysis
Mechanism Description Down-regulation of miR874-3p promotes chemotherapeutic resistance in colorectal cancer via inactivation of the Hippo signaling pathway. miR874-3p directly inhibited the expression of transcriptional co-activators YAP and TAZ of the Hippo signaling pathway, resulting in the inactivation of the TEAD transcription.
Key Molecule: Transcriptional coactivator YAP1 (YAP1) [102]
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 Hippo signaling pathway Activation hsa04391
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Luciferase assay; miRNA immunoprecipitation assay
Experiment for
Drug Resistance
Caspase-9 or 3 activity assays; Flow cytometric analysis
Mechanism Description Down-regulation of miR874-3p promotes chemotherapeutic resistance in colorectal cancer via inactivation of the Hippo signaling pathway. miR874-3p directly inhibited the expression of transcriptional co-activators YAP and TAZ of the Hippo signaling pathway, resulting in the inactivation of the TEAD transcription.
Key Molecule: Suppressor of cytokine signaling 1 (SOCS1) [104]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation JAKT2/STAT3 signaling pathway Inhibition hsa04030
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
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
COLO 205 cells Colon Homo sapiens (Human) CVCL_0218
COLO 320DM cells Colon Homo sapiens (Human) CVCL_0219
CW-2 cells Colon Homo sapiens (Human) CVCL_1151
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
LS174T cells Colon Homo sapiens (Human) CVCL_1384
NCI-H716 cells Colon Homo sapiens (Human) CVCL_1581
SW948 cells Colon Homo sapiens (Human) CVCL_0632
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Luciferase reporter assay
Experiment for
Drug Resistance
Caspase-9 or -3 activity assays; Spheroid formation assay; Flow cytometric analysis; MTT assay
Mechanism Description miR196b-5p promotes stemness and chemoresistance of CRC cells to 5-fluorouracil via targeting negative regulators SOCS1 and SOCS3 of STAT3 signaling pathway, giving rise to activation of STAT3 signaling.
Key Molecule: Suppressor of cytokine signaling 3 (SOCS3) [104]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation JAKT2/STAT3 signaling pathway Inhibition hsa04030
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
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
COLO 205 cells Colon Homo sapiens (Human) CVCL_0218
COLO 320DM cells Colon Homo sapiens (Human) CVCL_0219
CW-2 cells Colon Homo sapiens (Human) CVCL_1151
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
LS174T cells Colon Homo sapiens (Human) CVCL_1384
NCI-H716 cells Colon Homo sapiens (Human) CVCL_1581
SW948 cells Colon Homo sapiens (Human) CVCL_0632
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Luciferase reporter assay
Experiment for
Drug Resistance
Caspase-9 or -3 activity assays; Spheroid formation assay; Flow cytometric analysis; MTT assay
Mechanism Description miR196b-5p promotes stemness and chemoresistance of CRC cells to 5-fluorouracil via targeting negative regulators SOCS1 and SOCS3 of STAT3 signaling pathway, giving rise to activation of STAT3 signaling.
Key Molecule: Cadherin-1 (CDH1) [105]
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 AKT signaling pathway Inhibition hsa04151
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Annexin V/ PI staining; Caspase-3 activity assay
Mechanism Description Levels of PTEN and E-cadherin were reduced by knockdown of miR200c in HCT-116 cells, PTEN inactivate the AkT signaling pathway, and E-cadherin is one of the major downstream regulators of miRNA-200c contributing to EMT, which is also important to inhibit tumor invasion and proliferation as well as to induce cell apoptosis.
Key Molecule: Phosphatase and tensin homolog (PTEN) [105]
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 AKT signaling pathway Inhibition hsa04151
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Annexin V/ PI staining; Caspase-3 activity assay
Mechanism Description Levels of PTEN and E-cadherin were reduced by knockdown of miR200c in HCT-116 cells, PTEN inactivate the AkT signaling pathway, and E-cadherin is one of the major downstream regulators of miRNA-200c contributing to EMT, which is also important to inhibit tumor invasion and proliferation as well as to induce cell apoptosis.
Key Molecule: Adenomatous polyposis coli protein (APC) [106]
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 colony Inhibition hsa05200
Cell proliferation Inhibition hsa05200
Wnt/Beta-catenin signaling pathway Activation hsa04310
In Vitro Model SW620 cells Colon Homo sapiens (Human) CVCL_0547
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
MTT assay; Annexin V-FITC/PI staining assay
Mechanism Description CXCL12/CXCR4 axis induced miR125b promotes invasion and confers 5-fluorouracil resistance through enhancing autophagy in colorectal cancer There was a negative correlation of the expression of miR125b with APC mRNA in paired human colorectal tissue specimens. The upregulation of miR125b activated the Wnt/beta-catenin signaling by targeting APC gene and contributed to 5-FU resistance through enhancing cell autophagy.
Key Molecule: Heat shock protein beta-1 (HSPB1) [107]
Molecule Alteration Expression
Down-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
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
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; BrdU incorporation assay; Flow cytometric analysis
Mechanism Description Ectopic expression of miR577 enhanced 5-FU sensitivity in SW480/5-FU cells by down-regulating HSP27. Enforced expression of HSP27 reversed the effects of miR577 on CRC cell growth and 5-FU sensitivity.
Key Molecule: G1/S-specific cyclin-D2 (CCND2) [108]
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) [109]
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: Phosphatase and tensin homolog (PTEN) [110]
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
PTEN/PI3K/AKT signaling pathway Activation hsa05235
In Vitro Model HCT8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Transwell assay; Flow cytometry assay
Mechanism Description miR-543 enhanced drug resistance by down-regulating the expression of phosphatase and tensin homolog (PTEN), which negatively regulates protein kinase B (AkT) activation while an elevated expression of PTEN reversed the chemoresistance of miR-543-overexpressing HCT8 cells to 5-FU.
Key Molecule: Neurogenic locus notch homolog protein 2 (NOTCH2) [111]
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
Notch signaling pathway Inhibition hsa04330
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
SW620 cells Colon Homo sapiens (Human) CVCL_0547
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
HCT-160 cells Colorectal Homo sapiens (Human) N.A.
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; RIP assay; Luciferase reporter assay
Experiment for
Drug Resistance
MTT assay; Caspase-3 activity
Mechanism Description miR-195-5p regulates CRC cell stemness and 5-FU resistance through Notch2 and RBPJ.
Key Molecule: Recombining binding protein suppressor of hairless (RBPJ) [111]
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
Notch signaling pathway Inhibition hsa04330
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
SW620 cells Colon Homo sapiens (Human) CVCL_0547
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
HCT-160 cells Colorectal Homo sapiens (Human) N.A.
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; RIP assay; Luciferase reporter assay
Experiment for
Drug Resistance
MTT assay; Caspase-3 activity
Mechanism Description miR-195-5p regulates CRC cell stemness and 5-FU resistance through Notch2 and RBPJ.
Key Molecule: Forkhead box protein M1 (FOXM1) [113]
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 viability Inhibition 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
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-761 suppressed colorectal cancer cell proliferation and invasion by downregulating FOXM1.
Key Molecule: Forkhead box protein M1 (FOXM1) [116]
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 AKT/mTOR signaling pathway Regulation hsa04150
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
LOVO cells Colon Homo sapiens (Human) CVCL_0399
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Reduced miR-149 is a critical factor in the mechanisms by which CRC cells resist the cytotoxicity of 5-FU. Also, re-expression of miR-149 could increase the 5-FU sensitivity of CRC cells via enhancing 5-FU-inducing apoptosis by targeting FOXM1.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [128]
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: E3 ubiquitin-protein ligase XIAP (XIAP) [117]
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 proliferation Inhibition hsa05200
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
LOVO cells Colon Homo sapiens (Human) CVCL_0399
NCM460 cells Colon Homo sapiens (Human) CVCL_0460
SW1116 cells Colon Homo sapiens (Human) CVCL_0544
HCT-116 cells Colon Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
RT-qPCR; Western blot analysiss
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-874 inhibits growth, increases apoptosis and enhances chemosensitivity in CRC cells by targeting XIAP.
Key Molecule: Neurogenic locus notch homolog protein 1 (NOTCH1) [118]
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 proliferation Inhibition hsa05200
IGF-1R/AKT/S6 signaling pathway Inhibition hsa05225
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
LOVO cells Colon Homo sapiens (Human) CVCL_0399
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Ectopic expression of miR-139-5p sensitized CRC cells to 5-FU by increasing 5-FU-induced apoptosis. In addition, miR-139-5p inhibited the expression of the miR-139-5p target gene NOTCH-1 and its downstream molecules MRP-1 and BCL-2, two key MDR-associated genes. Furthermore, silencing NOTCH-1 expression promoted the chemotherapeutic effects of 5-FU, and up-regulation of NOTCH-1 abrogated miR-139-5p-mediated sensitization to 5-FU in LoVo and HCT-116 cells.
Key Molecule: Cyclic AMP-responsive element-binding protein 1 (CREB1) [92]
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 proliferation Inhibition hsa05200
UCA1/miR204-5p ceRNA signaling pathway Regulation hsa05206
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
HCT8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description LncRNA-UCA1 enhances cell proliferation and 5-fluorouracil resistance in colorectal cancer by inhibiting miR-204-5p.We found that UCA1 was up-regulated in CRCs and negatively correlated with survival time in two CRC cohorts. Further mechanistic studies revealed that UCA1 could sponge endogenous miR-204-5p and inhibit its activity. We also identified CREB1 as a new target of miR-204-5p. The protein levels of CREB1 were significantly up-regulated in CRCs, negatively associated with survival time and positively correlated with the UCA1 expression. The present work provides the first evidence of a UCA1-miR-204-5p-CREB1/BCL2/RAB22A regulatory network in CRC and reveals that UCA1 and CREB1 are potential new oncogenes and prognostic factors for CRC.
Key Molecule: Kinase suppressor of Ras 1 (KSR1) [115]
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
Cell proliferation Inhibition hsa05200
MAPK/ERK signaling pathway Inhibition hsa04010
In Vitro Model SW1116 cells Colon Homo sapiens (Human) CVCL_0544
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-497 expression levels were downregulated in human CRC specimens compared to the adjacent normal tissues. miR-497 expression levels were strongly correlated with clinical stages and lymph node metastases. Furthermore, kinase suppressor of ras 1 (kSR1), a known oncogene, was a direct target of miR-497, and kSR1 expression levels were inversely correlated with miR-497 expression levels in human CRC specimens. Overexpression of miR-497 inhibited cell proliferation, migration, invasion and increased chemosensitivity to 5-fluorouracil treatment, whereas forced expression of kSR1 had the opposite effect.
Key Molecule: Programmed cell death protein 10 (PDCD10) [119]
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: E3 ubiquitin-protein ligase SMURF1 (SMURF1) [114]
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 SW480 cells Colon Homo sapiens (Human) CVCL_0546
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miRNA-497 targeted Smurf1 in CRC cells and the Smurf1 expression level was dramatically increased in neoadjuvant therapy-resistant patients compared with treatment-sensitive patients. These results indicate that down-regulation of miRNA-497 in colorectal cancer may contribute to the resistance of CRC cells to 5-FU treatment. Thus, miRNA-497 has the potential to be a novel biomarker for predicting the neoadjuvant chemotherapy sensitivity in CRC patients.
Key Molecule: B-cell translocation gene 1 (BTG1) [120]
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 invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model SW620 cells Colon Homo sapiens (Human) CVCL_0547
RkO cells Colon Homo sapiens (Human) CVCL_0504
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Trypan blue exclusion assay
Mechanism Description Tumor cells undergoing autophagy may affect the sensitivity of 5-FU by repressing miR-22 expression. miR-22 will facilitate 5-FU to kill tumor cells when it was exotically introduced into the tumor cells, and tumor cells with higher levels of miR-22 were more sensitive to 5-FU. starvation induced up-regulation of BTG1 in CRC cells was inversely correlated with miR-22, which further demonstrated that miR-22 may influence cells under stress. More importantly, BTG1 can reverse the inhibition of autophagy induced by overexpression of miR-22, and the knockdown of BTG1 can reduce the level of autophagy resulting from the down-regulation of miR-22 in CRC cells with 5-FU treatment. Similarly, the data from clinical samples indicated that the miR-22 level was inversely correlated with the expression of BTG1, and the tumors with higher miR-22 level were more sensitive to 5-FU.
Key Molecule: Apoptotic protease-activating factor 1 (APAF1) [121]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation APAF-1/caspase-9 apoptotic signaling pathway Activation hsa04210
Cell apoptosis Inhibition hsa04210
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 blotting analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-23a may inhibit 5-FU-induced apoptosis through the APAF-1/caspase-9 pathway and provide new insight into CRC treatment.
Key Molecule: Mast/stem cell growth factor receptor Kit (KIT) [122]
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 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
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
A real-time cell analyzer assay
Mechanism Description c-KIT was shown to mediate chemo-resistance (kike 5-FU) in ovarian tumor initiating cells, miR-34a inhibits Erk signaling and colony formation by down-regulation of c-kit, miR-34a can inhibit this effect via down-regulation of c-kit and therefore sensitize cells to chemotherapeutic treatment.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [123]
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 proliferation Inhibition hsa05200
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
In Vivo Model CRC nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis; Immunofluorescence analysis
Experiment for
Drug Resistance
WST assay
Mechanism Description microRNA-129 (miR-129) trigger apoptosis by suppressing key anti-apoptotic protein, B-cell lymphoma 2 (BCL2), enhanced the cytotoxic effect of 5-fluorouracil both in vitro and in vivo.
Key Molecule: Insulin-like growth factor 1 receptor (IGF1R) [124]
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 migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
MEK/ERK signaling pathway Inhibition hsa04011
PI3K/AKT signaling pathway Inhibition hsa04151
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
COLO 205 cells Colon Homo sapiens (Human) CVCL_0218
HCT28 cells Colon Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description IGF1-R has an important role in mediating activation of the PI3k/Akt pathway, miR-497 inhibits PI3k/Akt signalling. Down-regulation of miR-497 is an important mechanism of upregulation of IGF1-R in CRC cells that contributes to malignancy of CRC.
Key Molecule: TNF alpha converting enzyme (ADAM17) [125]
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: Bcl-2/adenovirus E1B 19 kDa protein-interacting protein 2 (BNIP2) [126]
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.
Key Molecule: Mitogen-activated protein kinase 7 (MAPK7) [131]
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 proliferation Inhibition hsa05200
NF-kappaB signaling pathway Regulation hsa04064
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Immunoblotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-143 increases the sensitivity of colon cancer cells to 5-fluorouracil, probably acting through extracellular-regulated protein kinase 5/nuclear factor-kB regulated pathways.
Pancreatic cancer [ICD-11: 2C10]
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: DiGeorge syndrome critical region gene 5 (DGCR5) [13]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
HAPC cells Pancreas Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description DGCR5 and miR320a regulate each other in a reciprocal manner and that DGCR5 reverses the inhibition of PDCD4 by miR320a, which is involved in the regulation of the PDAC cell phenotype and response to 5-FU. miR320a is involved in 5-FU resistance modulated by DGCR5.
Key Molecule: Growth arrest specific 5 (GAS5) [132]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Hippo signaling pathway Inhibition hsa04390
In Vitro Model SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
5-FU cells Colon Homo sapiens (Human) CVCL_1846
PATU8988 Pancreas Homo sapiens (Human) CVCL_1847
PATU8988 cells Pancreas Homo sapiens (Human) CVCL_1846
SW1990/GEM cells Pancreas Homo sapiens (Human) CVCL_ZW98
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description GAS5 regualtes Hippo signaling pathway via miR181c-5p to antagonize the development of multidrug resistance in pancreatic cancer cells. GAS5 regulated chemoresistance and Hippo pathway of pancreatic cancer cells via miR181c-5p/Hippo.
Key Molecule: hsa-miR-221-3p [133]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
PATU8988 cells Pancreas Homo sapiens (Human) CVCL_1846
293TN cells Pancreas Homo sapiens (Human) CVCL_UL49
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
5-FU and gemcitabine assay; CCK8 assay; Wound healing assay; Transwell chamber invasion assay
Mechanism Description miRNA-221-3p desensitizes pancreatic cancer cells to 5-fluorouracil by targeting RB1. miR221-3p down-regulated RB1 expression by directly binding to its 3'-UTR and therefore caused increased several aspects of pancreatic cancer pathogenesis, including proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT).
Key Molecule: Urothelial cancer associated 1 (UCA1) [134]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Activation hsa04151
Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
ERK signaling pathway Activation hsa04210
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Capan-1 cells Pancreas Homo sapiens (Human) CVCL_0237
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
CFPAC1 cells Pancreas Homo sapiens (Human) CVCL_1119
HPAC cells Pancreas Homo sapiens (Human) CVCL_3517
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Wound-healing assay
Mechanism Description CUDR overexpression inhibits cell apoptosis and promotes drug resistance in PDAC and CUDR overexpression in Panc-1 cells significantly increased phosphorylated (p-) focal adhesion kinase (FAk) and p-AkT levels, whereas the total FAk and AkT were not altered compared with in Panc-1 cells transfected with an empty vector.
Key Molecule: hsa-mir-21 [135]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
PI3K/AKT/mTOR signaling pathway Regulation hsa04151
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
PATU8988 cells Pancreas Homo sapiens (Human) CVCL_1846
293TN cells Pancreas Homo sapiens (Human) CVCL_UL49
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-21 regulates 5-FU drug resistance in pancreatic cancer by reducing the expression of its targets, PTEN and PDCD4. And PTEN and PDCD4, as tumor suppressors, not only can inhibit tumor growth and invasion, but also can downregulate the 5-FU resistance induced by miR-21 in pancreatic cancer cells.
Key Molecule: hsa-mir-181c [136]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Hippo signaling pathway Regulation hsa04392
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-181c directly repressed MST1, LATS2, MOB1 and SAV1 expression in human pancreatic cancer cells. Overexpression of miR-181c induced hyperactivation of the YAP/TAZ and (+) expression of the Hippo signaling downstream genes CTGF, BIRC5 and BLC2L1, leading to pancreatic cancer cell survival and chemoresistance in vitro and in vivo. Importantly, high miR-181c levels were significantly correlated with Hippo signaling inactivation in pancreatic cancer samples, and predicted a poor patient overall survival.
Key Molecule: hsa-miR-1246 [21]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model Panc1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The in vitro drug sensitivity of pancreatic cancer cells was altered according to the miR-1246 expression via CCNG2. In vivo, we found that miR-1246 could increase tumour-initiating potential and induced drug resistance. A high expression level of miR-1246 was correlated with a worse prognosis and CCNG2 expression was significantly lower in those patients. miR-1246 expression was associated with chemoresistance and CSC-like properties via CCNG2, and could predict worse prognosis in pancreatic cancer patients.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: hsa-mir-320 [137]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
PATU8988 cells Pancreas Homo sapiens (Human) CVCL_1846
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay; Wound Healing assay; Matrigel transmembrane invasion assay
Mechanism Description miR-320a was up-regulated in 5-FU resistant pancreatic cancer cells and that miR-320a could promote pancreatic cancer cell proliferation, migration and invasion then contributed to the increased 5-FU resistance. Researchers think miR-320a could suppress cell apoptosis by inhibiting PDCD4 and further contribute to drug-resistance, which will be studied in future.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Retinoblastoma-associated protein (RB1) [133]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
PATU8988 cells Pancreas Homo sapiens (Human) CVCL_1846
293TN cells Pancreas Homo sapiens (Human) CVCL_UL49
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
5-FU and gemcitabine assay; CCK8 assay; Wound healing assay; Transwell chamber invasion assay
Mechanism Description miRNA-221-3p desensitizes pancreatic cancer cells to 5-fluorouracil by targeting RB1. miR221-3p down-regulated RB1 expression by directly binding to its 3'-UTR and therefore caused increased several aspects of pancreatic cancer pathogenesis, including proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT).
Key Molecule: RAC serine/threonine-protein kinase (AKT) [134]
Molecule Alteration Phosphorylation
Up-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Activation hsa04151
Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
ERK signaling pathway Activation hsa04210
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Capan-1 cells Pancreas Homo sapiens (Human) CVCL_0237
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
CFPAC1 cells Pancreas Homo sapiens (Human) CVCL_1119
HPAC cells Pancreas Homo sapiens (Human) CVCL_3517
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; Wound-healing assay
Mechanism Description CUDR overexpression inhibits cell apoptosis and promotes drug resistance in PDAC and CUDR overexpression in Panc-1 cells significantly increased phosphorylated (p-) focal adhesion kinase (FAk) and p-AkT levels, whereas the total FAk and AkT were not altered compared with in Panc-1 cells transfected with an empty vector.
Key Molecule: Mitogen-activated protein kinase (MAPK) [134]
Molecule Alteration Phosphorylation
Up-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Activation hsa04151
Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
ERK signaling pathway Activation hsa04210
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Capan-1 cells Pancreas Homo sapiens (Human) CVCL_0237
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
CFPAC1 cells Pancreas Homo sapiens (Human) CVCL_1119
HPAC cells Pancreas Homo sapiens (Human) CVCL_3517
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; Wound-healing assay
Mechanism Description CUDR overexpression inhibits cell apoptosis and promotes drug resistance in PDAC and CUDR overexpression in Panc-1 cells significantly increased phosphorylated (p-) focal adhesion kinase (FAk) and p-AkT levels, whereas the total FAk and AkT were not altered compared with in Panc-1 cells transfected with an empty vector.
Key Molecule: Programmed cell death protein 4 (PDCD4) [135], [137]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
PI3K/AKT/mTOR signaling pathway Regulation hsa04151
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
PATU8988 cells Pancreas Homo sapiens (Human) CVCL_1846
293TN cells Pancreas Homo sapiens (Human) CVCL_UL49
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Wound Healing assay; Matrigel transmembrane invasion assay
Mechanism Description miR-21 regulates 5-FU drug resistance in pancreatic cancer by reducing the expression of its targets, PTEN and PDCD4. And PTEN and PDCD4, as tumor suppressors, not only can inhibit tumor growth and invasion, but also can downregulate the 5-FU resistance induced by miR-21 in pancreatic cancer cells.
Key Molecule: Phosphatase and tensin homolog (PTEN) [135]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
PI3K/AKT/mTOR signaling pathway Regulation hsa04151
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
PATU8988 cells Pancreas Homo sapiens (Human) CVCL_1846
293TN cells Pancreas Homo sapiens (Human) CVCL_UL49
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-21 regulates 5-FU drug resistance in pancreatic cancer by reducing the expression of its targets, PTEN and PDCD4. And PTEN and PDCD4, as tumor suppressors, not only can inhibit tumor growth and invasion, but also can downregulate the 5-FU resistance induced by miR-21 in pancreatic cancer cells.
Key Molecule: Serine/threonine-protein kinase LATS2 (LATS2) [136]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Hippo signaling pathway Regulation hsa04392
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-181c directly repressed MST1, LATS2, MOB1 and SAV1 expression in human pancreatic cancer cells. Overexpression of miR-181c induced hyperactivation of the YAP/TAZ and (+) expression of the Hippo signaling downstream genes CTGF, BIRC5 and BLC2L1, leading to pancreatic cancer cell survival and chemoresistance in vitro and in vivo. Importantly, high miR-181c levels were significantly correlated with Hippo signaling inactivation in pancreatic cancer samples, and predicted a poor patient overall survival.
Key Molecule: MOB kinase activator 1A (MOB1A) [136]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Hippo signaling pathway Regulation hsa04392
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-181c directly repressed MST1, LATS2, MOB1 and SAV1 expression in human pancreatic cancer cells. Overexpression of miR-181c induced hyperactivation of the YAP/TAZ and (+) expression of the Hippo signaling downstream genes CTGF, BIRC5 and BLC2L1, leading to pancreatic cancer cell survival and chemoresistance in vitro and in vivo. Importantly, high miR-181c levels were significantly correlated with Hippo signaling inactivation in pancreatic cancer samples, and predicted a poor patient overall survival.
Key Molecule: Serine/threonine-protein kinase 4 (MST1) [136]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Hippo signaling pathway Regulation hsa04392
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-181c directly repressed MST1, LATS2, MOB1 and SAV1 expression in human pancreatic cancer cells. Overexpression of miR-181c induced hyperactivation of the YAP/TAZ and (+) expression of the Hippo signaling downstream genes CTGF, BIRC5 and BLC2L1, leading to pancreatic cancer cell survival and chemoresistance in vitro and in vivo. Importantly, high miR-181c levels were significantly correlated with Hippo signaling inactivation in pancreatic cancer samples, and predicted a poor patient overall survival.
Key Molecule: Protein salvador homolog 1 (SAV1) [136]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Hippo signaling pathway Regulation hsa04392
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-181c directly repressed MST1, LATS2, MOB1 and SAV1 expression in human pancreatic cancer cells. Overexpression of miR-181c induced hyperactivation of the YAP/TAZ and (+) expression of the Hippo signaling downstream genes CTGF, BIRC5 and BLC2L1, leading to pancreatic cancer cell survival and chemoresistance in vitro and in vivo. Importantly, high miR-181c levels were significantly correlated with Hippo signaling inactivation in pancreatic cancer samples, and predicted a poor patient overall survival.
Key Molecule: Cyclin-G2 (CCNG2) [21]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model Panc1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The in vitro drug sensitivity of pancreatic cancer cells was altered according to the miR-1246 expression via CCNG2. In vivo, we found that miR-1246 could increase tumour-initiating potential and induced drug resistance. A high expression level of miR-1246 was correlated with a worse prognosis and CCNG2 expression was significantly lower in those patients. miR-1246 expression was associated with chemoresistance and CSC-like properties via CCNG2, and could predict worse prognosis in pancreatic cancer patients.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-320 [13]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model SUDHL-4 cells Peritoneal effusion Homo sapiens (Human) CVCL_0539
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description DGCR5 and miR320a regulate each other in a reciprocal manner and that DGCR5 reverses the inhibition of PDCD4 by miR320a, which is involved in the regulation of the PDAC cell phenotype and response to 5-FU. miR320a is involved in 5-FU resistance modulated by DGCR5. DGCR5 reversed the inhibition of the miR320a target gene PDCD4, which in turn inhibited the proliferation, migration and 5-FU resistance of PDAC cells.
Key Molecule: hsa-miR-181c-5p [132]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Hippo signaling pathway Inhibition hsa04390
In Vitro Model SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
5-FU cells Colon Homo sapiens (Human) CVCL_1846
PATU8988 Pancreas Homo sapiens (Human) CVCL_1847
PATU8988 cells Pancreas Homo sapiens (Human) CVCL_1846
SW1990/GEM cells Pancreas Homo sapiens (Human) CVCL_ZW98
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Long non-coding RNA GAS5 antagonizes the chemoresistance of pancreatic cancer cells through down-regulation of miR181c-5p. GAS5 negatively regulated miR181c-5p, and miR181c-5p dramatically promoted pancreatic cancer cell chemoresistance through inactivating the Hippo signaling.
Key Molecule: hsa-mir-21 [138]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
HPAC cells Pancreas Homo sapiens (Human) CVCL_3517
HPAF-II cells Pancreatic Homo sapiens (Human) CVCL_0313
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
SRB (sulforhodamine-B) assay
Mechanism Description Low miR-21 expression was associated with benefit from adjuvant treatment in two independent cohorts of PDAC cases, and anti-miR-21 increased anticancer drug activity in vitro.
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: hsa-mir-33a [139]
Molecule Alteration Expression
Down-regulation
Resistant Disease Liver cancer [ICD-11: 2C12.6]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Downregulated LncRNA CRNDE could up-regulate miR-33a expression and inhibit HMGA2 expression, thus it could significantly promote apoptosis of liver cancer drug-resistant cells on different chemotherapeutic drugs (ADM, DDP, 5-FU)and inhibit its proliferation, migration, invasion and drug resistance.
Key Molecule: Colorectal neoplasia differentially expressed (CRNDE) [139]
Molecule Alteration Expression
Up-regulation
Resistant Disease Liver cancer [ICD-11: 2C12.6]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Downregulated LncRNA CRNDE could up-regulate miR-33a expression and inhibit HMGA2 expression, thus it could significantly promote apoptosis of liver cancer drug-resistant cells on different chemotherapeutic drugs (ADM, DDP, 5-FU)and inhibit its proliferation, migration, invasion and drug resistance.
Key Molecule: hsa-miR-200a-3p [140]
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 Hep3B cells Liver Homo sapiens (Human) CVCL_0326
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTS assay; Flow cytometric analysis; Colony forming assay
Mechanism Description miR200a-3p enhances anti-cancer drug resistance by decreasing DUSP6 expression.
Key Molecule: Hepatocellular carcinoma up-regulated long non-coding RNA (HULC) [141]
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 [141]
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 [141]
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 [141]
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-503 [142]
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 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
L02 cells Liver Homo sapiens (Human) CVCL_6926
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR503 inhibits proliferation making human hepatocellular carcinoma cells susceptible to 5 fluorouracil by targeting EIF4E.
Key Molecule: hsa-mir-122 [18]
Molecule Alteration Expression
Up-regulation
Resistant Disease Hepatic carcinoma [ICD-11: 2C12.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation miR122/PCDH20/AKT/mTOR signaling pathway Activation hsa05206
In Vitro Model HepG2 cells Liver Homo sapiens (Human) CVCL_0027
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
SNU449 cells Liver Homo sapiens (Human) CVCL_0454
MHCC97 cells Liver Homo sapiens (Human) CVCL_4971
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR; RT-qPCR
Experiment for
Drug Resistance
MTT assay; CellTiter 96 Aqueous One Solution cell proliferation assay
Mechanism Description Targeting PCDH20 gene by microRNA-122 confers 5-FU resistance in hepatic carcinoma. Rescue of PCDH20 expression in miR122-expressing cells decreases Akt and mTOR phosphorylation, re-sensitizing hepatocellular carcinoma cell to 5-fluorouracil induced apoptosis.
Key Molecule: hsa-mir-21 [10]
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
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
PLC/PRF/5 cells Liver Homo sapiens (Human) CVCL_0485
HLE cells Liver Homo sapiens (Human) CVCL_1281
HLF cells Liver Homo sapiens (Human) CVCL_2947
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Hepatocellular carcinoma cells transfected with pre-miR-21 were significantly resistant to IFN-alpha/5-FU. Transfection of anti-miR-21 rendered HCC cells sensitive to IFN-alpha/5-FU, and such sensitivity was weakened by transfection of siRNAs of target molecules, PETN and PDCD4, miR-21 induces chemoresistance to IFN-alpha and 5-FU, mediated through PETN and PDCD4.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: Phosphatase and tensin homolog (PTEN) [143]
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 proliferation Activation hsa05200
Epithelial mesenchymal transition signaling pathway Activation hsa01521
PI3K/AKT signaling pathway Activation hsa04151
In Vitro Model BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
MHCC97-H cells Liver Homo sapiens (Human) CVCL_4972
Bel/5-FU cells Liver Homo sapiens (Human) CVCL_5493
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Wound healing assay; Transwell assay
Mechanism Description Over-expression of miR-32-5p activated the PI3k/Akt pathway by suppressing PTEN and induced multidrug resistance via exosomes through promoting angiogenesis and epithelial-mesenchymal transition.
Key Molecule: hsa-miR-32-5p [143]
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 invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
Cell viability Activation hsa05200
PI3K/AKT signaling pathway Activation hsa04151
In Vitro Model BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
MHCC97-H cells Liver Homo sapiens (Human) CVCL_4972
Bel/5-FU cells Liver Homo sapiens (Human) CVCL_5493
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Wound healing assay; Transwell assay
Mechanism Description Over-expression of miR-32-5p activated the PI3k/Akt pathway by suppressing PTEN and induced multidrug resistance via exosomes through promoting angiogenesis and epithelial-mesenchymal transition.
Key Molecule: Small nucleolar RNA host gene 6 (SNHG6) [144]
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 apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
Skhep1 cells Liver Homo sapiens (Human) CVCL_0525
MHCC97-H cells Liver Homo sapiens (Human) CVCL_4972
HCC-LM3 cells Liver Homo sapiens (Human) CVCL_6832
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8; Flow cytometry assay; EdU assay
Mechanism Description Ectopic expression of SNHG6-003 in HCC cells promoted cell proliferation and induced drug resistance, whereas SNHG6-003 knockdown promoted apoptosis. Moreover, SNHG6-003 functioned as a competitive endogenous RNA (ceRNA), effectively becoming sponge for miR-26a/b and thereby modulating the expression of transforming growth factor-beta-activated kinase 1 (TAk1). Importantly, expression analysis revealed that both SNHG6-003 and TAk1 were upregulated in human cancers, exhibiting a co-expression pattern. In HCC patients, high expression of SNHG6-003 closely correlated with tumor progression and shorter survival.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: High mobility group protein HMGI-C (HMGA2) [139]
Molecule Alteration Expression
Up-regulation
Resistant Disease Liver cancer [ICD-11: 2C12.6]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR; Luciferase activity assay
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Downregulated LncRNA CRNDE could up-regulate miR-33a expression and inhibit HMGA2 expression, thus it could significantly promote apoptosis of liver cancer drug-resistant cells on different chemotherapeutic drugs (ADM, DDP, 5-FU)and inhibit its proliferation, migration, invasion and drug resistance.
Key Molecule: Dual specificity protein phosphatase 6 (DUSP6) [140]
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 Hep3B cells Liver Homo sapiens (Human) CVCL_0326
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay; Flow cytometric analysis; Colony forming assay
Mechanism Description miR200a-3p enhances anti-cancer drug resistance by decreasing DUSP6 expression.
Key Molecule: Ubiquitin carboxyl-terminal hydrolase 22 (USP22) [141]
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.
Key Molecule: Eukaryotic translation initiation factor 4E (EIF4E) [142]
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 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
L02 cells Liver Homo sapiens (Human) CVCL_6926
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR503 inhibits proliferation making human hepatocellular carcinoma cells susceptible to 5 fluorouracil by targeting EIF4E.
Key Molecule: Protocadherin-20 (PCDH20) [18]
Molecule Alteration Expression
Down-regulation
Resistant Disease Hepatic carcinoma [ICD-11: 2C12.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation miR122/PCDH20/AKT/mTOR signaling pathway Activation hsa05206
In Vitro Model HepG2 cells Liver Homo sapiens (Human) CVCL_0027
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
SNU449 cells Liver Homo sapiens (Human) CVCL_0454
MHCC97 cells Liver Homo sapiens (Human) CVCL_4971
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; CellTiter 96 Aqueous One Solution cell proliferation assay
Mechanism Description Targeting PCDH20 gene by microRNA-122 confers 5-FU resistance in hepatic carcinoma. Rescue of PCDH20 expression in miR122-expressing cells decreases Akt and mTOR phosphorylation, re-sensitizing hepatocellular carcinoma cell to 5-fluorouracil induced apoptosis.
Key Molecule: Nuclear receptor subfamily 2 group C2 (NR2C2) [144]
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 apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
Skhep1 cells Liver Homo sapiens (Human) CVCL_0525
MHCC97-H cells Liver Homo sapiens (Human) CVCL_4972
HCC-LM3 cells Liver Homo sapiens (Human) CVCL_6832
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8; Flow cytometry assay; EdU assay
Mechanism Description Ectopic expression of SNHG6-003 in HCC cells promoted cell proliferation and induced drug resistance, whereas SNHG6-003 knockdown promoted apoptosis. Moreover, SNHG6-003 functioned as a competitive endogenous RNA (ceRNA), effectively becoming sponge for miR-26a/b and thereby modulating the expression of transforming growth factor-beta-activated kinase 1 (TAk1). Importantly, expression analysis revealed that both SNHG6-003 and TAk1 were upregulated in human cancers, exhibiting a co-expression pattern. In HCC patients, high expression of SNHG6-003 closely correlated with tumor progression and shorter survival.
Key Molecule: Programmed cell death protein 4 (PDCD4) [10]
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 viability Activation hsa05200
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
PLC/PRF/5 cells Liver Homo sapiens (Human) CVCL_0485
HLE cells Liver Homo sapiens (Human) CVCL_1281
HLF cells Liver Homo sapiens (Human) CVCL_2947
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Hepatocellular carcinoma cells transfected with pre-miR-21 were significantly resistant to IFN-alpha/5-FU. Transfection of anti-miR-21 rendered HCC cells sensitive to IFN-alpha/5-FU, and such sensitivity was weakened by transfection of siRNAs of target molecules, PETN and PDCD4, miR-21 induces chemoresistance to IFN-alpha and 5-FU, mediated through PETN and PDCD4.
Key Molecule: Phosphatase and tensin homolog (PTEN) [10]
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 viability Activation hsa05200
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
PLC/PRF/5 cells Liver Homo sapiens (Human) CVCL_0485
HLE cells Liver Homo sapiens (Human) CVCL_1281
HLF cells Liver Homo sapiens (Human) CVCL_2947
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Hepatocellular carcinoma cells transfected with pre-miR-21 were significantly resistant to IFN-alpha/5-FU. Transfection of anti-miR-21 rendered HCC cells sensitive to IFN-alpha/5-FU, and such sensitivity was weakened by transfection of siRNAs of target molecules, PETN and PDCD4, miR-21 induces chemoresistance to IFN-alpha and 5-FU, mediated through PETN and PDCD4.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-379 [145]
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 IGF1/IGF1R signaling pathway Inhibition hsa05200
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Propidium Iodide (PI) Staining
Mechanism Description IGF1 is a hub gene in HCC and is involved in the p53 signaling pathway regulation. miR379 can sensitize HCC cells to chemotherapeutic reagents via targeting IGF1R and suppressing its expression, and suppressing the IGF1/IGF1R signaling pathway.
Key Molecule: Long non-protein coding RNA 607 (LINC00607) [146]
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 proliferation Inhibition hsa05200
NF-kappaB p65/p53 signaling pathway Regulation hsa04064
In Vitro Model HepG2 cells Liver Homo sapiens (Human) CVCL_0027
HCCLM3 cells Liver Homo sapiens (Human) CVCL_6832
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
PLC cells Liver Homo sapiens (Human) CVCL_0485
MHCC97-H cells Liver Homo sapiens (Human) CVCL_4972
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Real-time RT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description LncRNA 00607 overexpression leads to decreased HCC cell proliferation in vitro and in vivo, enhanced apoptosis and chemotherapeutic drug sensitivity, inhibiting the p65 transcription by binding to the p65 promoter region, therefore contributing to increased p53 levels in HCC.
Key Molecule: Long non-protein coding RNA (KRAL) [147]
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
Nrf2 signaling pathway Inhibition hsa05208
In Vitro Model 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
CCK8 assay; Flow cytometry assay
Mechanism Description Cells with kRAL overexpression exhibited a reversal in the resistance against 5-FU, with a significant decrease in the IC50 and a dramatic increase in cellular apoptosis, while silencing keap1 or ectopically expressing miR-141 partially rescued this effect.
Key Molecule: hsa-mir-141 [147]
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
Nrf2 signaling pathway Inhibition hsa05208
In Vitro Model 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
CCK8 assay; Flow cytometry assay
Mechanism Description Cells with kRAL overexpression exhibited a reversal in the resistance against 5-FU, with a significant decrease in the IC50 and a dramatic increase in cellular apoptosis, while silencing keap1 or ectopically expressing miR-141 partially rescued this effect.
Key Molecule: Long non-protein coding RNA (KRAL) [147]
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
Nrf2 signaling pathway Inhibition hsa05208
In Vitro Model 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
CCK8 assay; Flow cytometry assay
Mechanism Description Cells with kRAL overexpression exhibited a reversal in the resistance against 5-FU, with a significant decrease in the IC50 and a dramatic increase in cellular apoptosis, while silencing keap1 or ectopically expressing miR-141 partially rescued this effect.
Key Molecule: hsa-mir-183 [148]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Hepatocellular cancer [ICD-11: 2C12.4]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
miR183/IDH2/SOCS6/HIF1alpha feedback loop signaling pathway Regulation hsa05206
In Vitro Model BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description IDH2 knockdown resulted in significantly increased HIF-1alpha expression in both BEL-7402 and BEL-7402/5-FU cells. knockdown of SOCS6 had similar but stronger effect as miR-183 in promoting MRP2, P-gp, p-STAT3 and HIF-1alpha expression in BEL-7402 cells, while SOCS6 overexpression also showed similar but stronger effect as miR-183 inhibition in reducing MRP2, P-gp, p-STAT3 and HIF-1alpha levels in BEL-7402/5-FU cells. Both SOCS6 overexpression and miR-183 knockdown significantly increased the sensitivity of BEL-7402/5-FU cells to 5-FU. miR-183 overexpression partly abrogated the effect of SOCS6 in enhancing 5-FU sensitivity.
Key Molecule: hsa-mir-133a [149]
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 proliferation Inhibition hsa05200
Mitochondrial signaling pathway Activation hsa04217
In Vitro Model HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-133a and miR-326 share a common target gene, Bcl-xl. Expression levels of miR-133a and miR-326 are significantly upregulated subsequent to transfection. miR-133a and miR-326 downregulate the mRNA expression of Bcl-xl. miR-133a and miR-326 sensitize HepG2 cells to 5-FU and DDP.
Key Molecule: hsa-miR-326 [149]
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 proliferation Inhibition hsa05200
Mitochondrial signaling pathway Activation hsa04217
In Vitro Model HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-133a and miR-326 share a common target gene, Bcl-xl. Expression levels of miR-133a and miR-326 are significantly upregulated subsequent to transfection. miR-133a and miR-326 downregulate the mRNA expression of Bcl-xl. miR-133a and miR-326 sensitize HepG2 cells to 5-FU and DDP.
Key Molecule: hsa-let-7b [150]
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
In Vitro Model BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Let-7b increased 5 FU sensitivity by repressing Bcl xl expression in HCC cells.
Key Molecule: hsa-mir-125b [151]
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 proliferation Inhibition hsa05200
In Vitro Model HepG2 cells Liver Homo sapiens (Human) CVCL_0027
THLE-2 cells Liver Homo sapiens (Human) CVCL_3803
THLE-3 cells Liver Homo sapiens (Human) CVCL_3804
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Compared with 5-FU-sensitive cells, 5-FU-resistant cells exhibited reduced expression levels of miR-125b, and transfection of pre-miR-125b into liver cancer cells resulted in an increased sensitivity of 5-FU-resistant cells to 5-FU. Since drug resistance is a phenotype of malignant cancer cells, the finding that miR-125b expression levels are negatively correlated with 5-FU resistance in HCC cells is consistent with the reported functions of miR-125b. In addition, 5-FU-resistant cells exhibited higher glucose metabolic activity than 5-FU-sensitive cells, and miR-125 was identified to downregulate glucose metabolism by directly targeting Hk II. These results identified miR-125b as a tumor suppressor-like microRNA, which has great potential as a diagnostic and prognostic biomarker.
Key Molecule: hsa-let-7g [152]
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 cycle Inhibition hsa04110
In Vitro Model Bel-7402/5-Fu cells Liver Homo sapiens (Human) CVCL_5493
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Let-7g microRNA contributed to an increase of 5-Fu-induced cell cycle inhibit in human hepatoma cell and sensitized cells to 5-Fu, leading to increased the effectiveness of the drug in treating hepatoma cancer.
Key Molecule: hsa-mir-101 [153]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
SNU182 cells Liver Homo sapiens (Human) CVCL_0090
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description miR-101-mediated EZH2 silencing sensitized hepatoblastoma cells to 5-FU- and doxorubicin-induced apoptosis, whereas antagomiR-mediated downregulation of endogenous miR-101 reversed the pro-apoptotic effect.
Key Molecule: hsa-mir-200b [154]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
In Vitro Model QBC939 cells Bile duct Homo sapiens (Human) CVCL_6942
TFk-1 cells Bile duct Homo sapiens (Human) CVCL_2214
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
WST cell counting kit-8
Mechanism Description miR-200b/c influenced the tumourigenesis of cholangiocarcinoma cells including their tumour-initiating capacity, sphere formation, and drug resistance (like fluorouracil). We further found that miR-200b/c regulated migration and invasion capacities by directly targeting rho-kinase 2 and regulated tumorigenic properties by directly targeting SUZ12 (a subunit of a polycomb repressor complex).
Key Molecule: hsa-mir-200c [154]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
In Vitro Model QBC939 cells Bile duct Homo sapiens (Human) CVCL_6942
TFk-1 cells Bile duct Homo sapiens (Human) CVCL_2214
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
WST cell counting kit-8
Mechanism Description miR-200b/c influenced the tumourigenesis of cholangiocarcinoma cells including their tumour-initiating capacity, sphere formation, and drug resistance (like fluorouracil). We further found that miR-200b/c regulated migration and invasion capacities by directly targeting rho-kinase 2 and regulated tumorigenic properties by directly targeting SUZ12 (a subunit of a polycomb repressor complex).
Key Molecule: hsa-miR-193a-3p [155]
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
In Vitro Model BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
QGY-7703 cells Liver Homo sapiens (Human) CVCL_6715
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
PLC cells Liver Homo sapiens (Human) CVCL_0485
FOCUS cells Liver Homo sapiens (Human) CVCL_7955
YY-8103 cells Liver Homo sapiens (Human) CVCL_WY40
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description SRSF2 preferentially up-regulates the proapoptotic splicing form of caspase 2 (CASP2L) and sensitizes HCC cells to 5-FU. miR-193a-3p Dictates Resistance of Hepatocellular Carcinoma to 5-Fluorouracil via Repression of SRSF2 Expression.
Key Molecule: hsa-mir-195 [156]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
Bel-7402/5-Fu cells Liver Homo sapiens (Human) CVCL_5493
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-195 antisense oligonucleotide induced drug resistance in BEL-7402/5-FU cells. miR-195 overexpression repressed Bcl-w protein level. miR-195, one of the down-regulated miRNAs in BEL-7402/5-FU cells, was demonstrated to play a role in the development of drug resistance in hepatocellular carcinoma cells by targeting the antiapoptotic gene, Bcl-w.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) [157]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Liver cancer [ICD-11: 2C12.6]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell colony Activation hsa05200
Cell viability Activation hsa05200
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
HCCLM3 cells Liver Homo sapiens (Human) CVCL_6832
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
MHCC97-L cells Liver Homo sapiens (Human) CVCL_4973
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
CCK8 analysis; EdU analysis; Boyden chamber assay; Transwell assay; Flow cytometry assay
Mechanism Description MALAT1 deficiency related increase in sensitivity of liver cancer cells was associated with regulation of NF-kB.
Key Molecule: DNA-binding factor KBF1 (p105) (NFKB1) [157]
Molecule Alteration Phosphorylation
Down-regulation
Sensitive Disease Liver cancer [ICD-11: 2C12.6]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell colony Activation hsa05200
Cell viability Activation hsa05200
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
HCCLM3 cells Liver Homo sapiens (Human) CVCL_6832
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
MHCC97-L cells Liver Homo sapiens (Human) CVCL_4973
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 analysis; EdU analysis; Boyden chamber assay; Transwell assay; Flow cytometry assay
Mechanism Description MALAT1 deficiency related increase in sensitivity of liver cancer cells was associated with regulation of NF-kB.
Key Molecule: Poly[ADP-ribose] synthase 1 (PARP1) [157]
Molecule Alteration Phosphorylation
Down-regulation
Sensitive Disease Liver cancer [ICD-11: 2C12.6]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell colony Activation hsa05200
Cell viability Activation hsa05200
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
HCCLM3 cells Liver Homo sapiens (Human) CVCL_6832
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
MHCC97-L cells Liver Homo sapiens (Human) CVCL_4973
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 analysis; EdU analysis; Transwell assay; Flow cytometry assay
Mechanism Description MALAT1 deficiency related increase in sensitivity of liver cancer cells was associated with regulation of NF-kB.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Insulin-like growth factor 1 receptor (IGF1R) [145]
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 IGF1/IGF1R signaling pathway Inhibition hsa05200
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Experiment for
Molecule Alteration
Dual luciferase assay; Western blot analysis
Experiment for
Drug Resistance
Propidium Iodide (PI) Staining
Mechanism Description IGF1 is a hub gene in HCC and is involved in the p53 signaling pathway regulation. miR379 can sensitize HCC cells to chemotherapeutic reagents via targeting IGF1R and suppressing its expression, and suppressing the IGF1/IGF1R signaling pathway.
Key Molecule: Cellular tumor antigen p53 (TP53) [146]
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 proliferation Inhibition hsa05200
NF-kappaB p65/p53 signaling pathway Regulation hsa04064
In Vitro Model HepG2 cells Liver Homo sapiens (Human) CVCL_0027
HCCLM3 cells Liver Homo sapiens (Human) CVCL_6832
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
PLC cells Liver Homo sapiens (Human) CVCL_0485
MHCC97-H cells Liver Homo sapiens (Human) CVCL_4972
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 LncRNA 00607 overexpression leads to decreased HCC cell proliferation in vitro and in vivo, enhanced apoptosis and chemotherapeutic drug sensitivity, inhibiting the p65 transcription by binding to the p65 promoter region, therefore contributing to increased p53 levels in HCC.
Key Molecule: Kelch-like ECH-associated protein 1 (KEAP1) [147]
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
Nrf2 signaling pathway Inhibition hsa05208
In Vitro Model 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
CCK8 assay; Flow cytometry assay
Mechanism Description Cells with kRAL overexpression exhibited a reversal in the resistance against 5-FU, with a significant decrease in the IC50 and a dramatic increase in cellular apoptosis, while silencing keap1 or ectopically expressing miR-141 partially rescued this effect.
Key Molecule: Bcl-2-associated agonist of cell death (BAD) [149], [150]
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 proliferation Inhibition hsa05200
Mitochondrial signaling pathway Activation hsa04217
In Vitro Model BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; MTT assay
Mechanism Description Let-7b increased 5 FU sensitivity by repressing Bcl xl expression in HCC cells. And miR-133a and miR-326 share a common target gene, Bcl-xl. Expression levels of miR-133a and miR-326 are significantly upregulated subsequent to transfection. miR-133a and miR-326 downregulate the mRNA expression of Bcl-xl. miR-133a and miR-326 sensitize HepG2 cells to 5-FU and DDP.
Key Molecule: Suppressor of cytokine signaling 6 (SOCS6) [148]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Hepatocellular cancer [ICD-11: 2C12.4]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
miR183/IDH2/SOCS6/HIF1alpha feedback loop signaling pathway Regulation hsa05206
In Vitro Model BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description IDH2 knockdown resulted in significantly increased HIF-1alpha expression in both BEL-7402 and BEL-7402/5-FU cells. knockdown of SOCS6 had similar but stronger effect as miR-183 in promoting MRP2, P-gp, p-STAT3 and HIF-1alpha expression in BEL-7402 cells, while SOCS6 overexpression also showed similar but stronger effect as miR-183 inhibition in reducing MRP2, P-gp, p-STAT3 and HIF-1alpha levels in BEL-7402/5-FU cells. Both SOCS6 overexpression and miR-183 knockdown significantly increased the sensitivity of BEL-7402/5-FU cells to 5-FU. miR-183 overexpression partly abrogated the effect of SOCS6 in enhancing 5-FU sensitivity.
Key Molecule: Hexokinase-2 (HK2) [151]
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 proliferation Inhibition hsa05200
In Vitro Model HepG2 cells Liver Homo sapiens (Human) CVCL_0027
THLE-2 cells Liver Homo sapiens (Human) CVCL_3803
THLE-3 cells Liver Homo sapiens (Human) CVCL_3804
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Compared with 5-FU-sensitive cells, 5-FU-resistant cells exhibited reduced expression levels of miR-125b, and transfection of pre-miR-125b into liver cancer cells resulted in an increased sensitivity of 5-FU-resistant cells to 5-FU. Since drug resistance is a phenotype of malignant cancer cells, the finding that miR-125b expression levels are negatively correlated with 5-FU resistance in HCC cells is consistent with the reported functions of miR-125b. In addition, 5-FU-resistant cells exhibited higher glucose metabolic activity than 5-FU-sensitive cells, and miR-125 was identified to downregulate glucose metabolism by directly targeting Hk II. These results identified miR-125b as a tumor suppressor-like microRNA, which has great potential as a diagnostic and prognostic biomarker.
Key Molecule: High mobility group protein HMGI-C (HMGA2) [152]
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 Inhibition hsa04210
Cell colony Activation hsa05200
Cell cycle Activation hsa04110
In Vitro Model Bel-7402/5-Fu cells Liver Homo sapiens (Human) CVCL_5493
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Let-7g microRNA contributed to an increase of 5-Fu-induced cell cycle inhibit in human hepatoma cell and sensitized cells to 5-Fu, leading to increased the effectiveness of the drug in treating hepatoma cancer.
Key Molecule: Histone-lysine N-methyltransferase EZH2 (EZH2) [153]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
SNU182 cells Liver Homo sapiens (Human) CVCL_0090
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description miR-101-mediated EZH2 silencing sensitized hepatoblastoma cells to 5-FU- and doxorubicin-induced apoptosis, whereas antagomiR-mediated downregulation of endogenous miR-101 reversed the pro-apoptotic effect.
Key Molecule: Rho-associated protein kinase 2 (ROCK2) [154]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
In Vitro Model QBC939 cells Bile duct Homo sapiens (Human) CVCL_6942
TFk-1 cells Bile duct Homo sapiens (Human) CVCL_2214
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Northern blotting analysis
Experiment for
Drug Resistance
WST cell counting kit-8
Mechanism Description miR-200b/c influenced the tumourigenesis of cholangiocarcinoma cells including their tumour-initiating capacity, sphere formation, and drug resistance (like fluorouracil). We further found that miR-200b/c regulated migration and invasion capacities by directly targeting rho-kinase 2 and regulated tumorigenic properties by directly targeting SUZ12 (a subunit of a polycomb repressor complex).
Key Molecule: Polycomb protein SUZ12 (SUZ12) [154]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
In Vitro Model QBC939 cells Bile duct Homo sapiens (Human) CVCL_6942
TFk-1 cells Bile duct Homo sapiens (Human) CVCL_2214
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Northern blotting analysis
Experiment for
Drug Resistance
WST cell counting kit-8
Mechanism Description miR-200b/c influenced the tumourigenesis of cholangiocarcinoma cells including their tumour-initiating capacity, sphere formation, and drug resistance (like fluorouracil). We further found that miR-200b/c regulated migration and invasion capacities by directly targeting rho-kinase 2 and regulated tumorigenic properties by directly targeting SUZ12 (a subunit of a polycomb repressor complex).
Key Molecule: Transcription factor E2F1 (E2F1) [155]
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
In Vitro Model BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
QGY-7703 cells Liver Homo sapiens (Human) CVCL_6715
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
PLC cells Liver Homo sapiens (Human) CVCL_0485
FOCUS cells Liver Homo sapiens (Human) CVCL_7955
YY-8103 cells Liver Homo sapiens (Human) CVCL_WY40
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
ELISA assay
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description SRSF2 preferentially up-regulates the proapoptotic splicing form of caspase 2 (CASP2L) and sensitizes HCC cells to 5-FU. miR-193a-3p Dictates Resistance of Hepatocellular Carcinoma to 5-Fluorouracil via Repression of SRSF2 Expression.
Key Molecule: Serine/arginine-rich splicing factor 2 (SRSF2) [155]
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
In Vitro Model BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
QGY-7703 cells Liver Homo sapiens (Human) CVCL_6715
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
PLC cells Liver Homo sapiens (Human) CVCL_0485
FOCUS cells Liver Homo sapiens (Human) CVCL_7955
YY-8103 cells Liver Homo sapiens (Human) CVCL_WY40
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
ELISA assay
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description SRSF2 preferentially up-regulates the proapoptotic splicing form of caspase 2 (CASP2L) and sensitizes HCC cells to 5-FU. miR-193a-3p Dictates Resistance of Hepatocellular Carcinoma to 5-Fluorouracil via Repression of SRSF2 Expression.
Key Molecule: Bcl-2-like protein 2 (BCL2L2) [156]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
Bel-7402/5-Fu cells Liver Homo sapiens (Human) CVCL_5493
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-195 antisense oligonucleotide induced drug resistance in BEL-7402/5-FU cells. miR-195 overexpression repressed Bcl-w protein level. miR-195, one of the down-regulated miRNAs in BEL-7402/5-FU cells, was demonstrated to play a role in the development of drug resistance in hepatocellular carcinoma cells by targeting the antiapoptotic gene, Bcl-w.
Lung cancer [ICD-11: 2C25]
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-128 [19]
Molecule Alteration Expression
Up-regulation
Resistant Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT signaling pathway Activation hsa04151
In Vitro Model H1299 cells Lung Homo sapiens (Human) CVCL_0060
A459 cells Lung Homo sapiens (Human) CVCL_0023
H1299 clone 23 cells Lung Homo sapiens (Human) N.A.
H1299 clone 41 cells Lung Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description The expression of the transcriptional repressor E2F5, a target of miR-128-2, strongly decreases after miR-128-2 exogenous expression. This leads to the abrogation of E2F5 repressive activity on p21waf1 promoter and, consequently, to the transcriptional induction of p21waf1. The newly synthesized p21waf1 protein is mainly localized into the cytoplasmic compartment, where it exerts an anti-apoptotic function in response to anticancer drug treatments.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Protein LYRIC (MTDH) [19]
Molecule Alteration Expression
Down-regulation
Resistant Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT signaling pathway Activation hsa04151
In Vitro Model H1299 cells Lung Homo sapiens (Human) CVCL_0060
A459 cells Lung Homo sapiens (Human) CVCL_0023
H1299 clone 23 cells Lung Homo sapiens (Human) N.A.
H1299 clone 41 cells Lung Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Luciferase assay
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description The expression of the transcriptional repressor E2F5, a target of miR-128-2, strongly decreases after miR-128-2 exogenous expression. This leads to the abrogation of E2F5 repressive activity on p21waf1 promoter and, consequently, to the transcriptional induction of p21waf1. The newly synthesized p21waf1 protein is mainly localized into the cytoplasmic compartment, where it exerts an anti-apoptotic function in response to anticancer drug treatments.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-181 [58]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
A549/CDDP cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The antiapoptotic protein BCL2 is upregulated, whereas miR-181b is downregulated in both SGC7901/VCR and A549/CDDP cells, compared with SGC7901 and A549 cells, respectively. Enforced miR-181b expression reduced BCL2 protein level and sensitized SGC7901/VCR and A549/CDDP cells to VCR-induced and CDDP-induced apoptosis, respectively.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: CX3C chemokine receptor 1 (CX3CR1) [158]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
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 Calu3 cells Lung Homo sapiens (Human) CVCL_0609
A549 cells Lung Homo sapiens (Human) CVCL_0023
SPC-A1 cells Lung Homo sapiens (Human) CVCL_6955
HCC827 cells Lung Homo sapiens (Human) CVCL_2063
NCI-H358 cells Lung Homo sapiens (Human) CVCL_1559
H157 cells Lung Homo sapiens (Human) CVCL_2458
D6 cells Lung Homo sapiens (Human) N.A.
LAX cells Lung Homo sapiens (Human) N.A.
LTEP-2 cells Lung Homo sapiens (Human) CVCL_6929
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR296-3p inhibited NSCLC cell proliferation, enhance the drug resistance, and apoptosis. Data of luciferase reporter assays demonstrated that the CX3CR1 gene was a direct regulator of tumorsuppressive miR296-3p. Moreover, overexpressed CX3CR1 was confirmed in NSCLC clinical specimens. Inhibition of CX3CR1 could inhibit cancer cellular survival and increase chemotherapy sensitivity. There was a negative relationship between miR296-3p and CX3CR1 expression in NSCLC tissues.
Key Molecule: hsa-miR-296-3p [158]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
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 Calu3 cells Lung Homo sapiens (Human) CVCL_0609
A549 cells Lung Homo sapiens (Human) CVCL_0023
SPC-A1 cells Lung Homo sapiens (Human) CVCL_6955
HCC827 cells Lung Homo sapiens (Human) CVCL_2063
NCI-H358 cells Lung Homo sapiens (Human) CVCL_1559
H157 cells Lung Homo sapiens (Human) CVCL_2458
D6 cells Lung Homo sapiens (Human) N.A.
LAX cells Lung Homo sapiens (Human) N.A.
LTEP-2 cells Lung Homo sapiens (Human) CVCL_6929
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR296-3p inhibited NSCLC cell proliferation, enhance the drug resistance, and apoptosis. Data of luciferase reporter assays demonstrated that the CX3CR1 gene was a direct regulator of tumorsuppressive miR296-3p. Moreover, overexpressed CX3CR1 was confirmed in NSCLC clinical specimens. Inhibition of CX3CR1 could inhibit cancer cellular survival and increase chemotherapy sensitivity. There was a negative relationship between miR296-3p and CX3CR1 expression in NSCLC tissues.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [58]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
A549/CDDP cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The antiapoptotic protein BCL2 is upregulated, whereas miR-181b is downregulated in both SGC7901/VCR and A549/CDDP cells, compared with SGC7901 and A549 cells, respectively. Enforced miR-181b expression reduced BCL2 protein level and sensitized SGC7901/VCR and A549/CDDP cells to VCR-induced and CDDP-induced apoptosis, respectively.
Breast cancer [ICD-11: 2C60]
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: LncRNA in non-homologous end joining pathway 1 (LINP1) [159]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Epithelial mesenchymal transition signaling pathway Activation hsa01521
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Long noncoding RNA LINP1 acts as an oncogene and promotes chemoresistance against 5-fluoroutacil and doxorubicin by inhibiting chemotherapeutics-induced apoptosis in breast cancer.
Key Molecule: hsa-mir-149 [160]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
HS signaling pathway Inhibition hsa00534
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR; RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-149 modulated chemoresistance through targeting the expression of GlcNAc N-deacetylase/N-sulfotransferase-1 (NDST1). With downregulated miR-149, NDST1 expression was increased in chemoresistant MCF-7/ADM cells versus control MCF-7 wild-type cells. The increased NDST1 then activated a heparan sulfate-related pathway involving activation of heparanase. Finally, expression of miR-149 and NDST1 was confirmed in clinical chemoresistant samples of breast cancers receiving anthracycline/taxane-based chemotherapies. The high expression of NDST1 was also an unfavorable predictor for distant relapse-free survival in Her2 and basal breast cancers.
Key Molecule: hsa-mir-204 [161]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Activation hsa05200
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Low miR-024 expression was enhancing chemotherapeutic resistance of breast cancer patients.
Key Molecule: hsa-mir-125b [3], [162]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation p53 signaling pathway Inhibition hsa04115
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
T47D cells Breast Homo sapiens (Human) CVCL_0553
BT20 cells Breast Homo sapiens (Human) CVCL_0178
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Sphere formation assay
Mechanism Description E2F3, and in some settings E2F1, induce apoptosis through p53-dependent or -independent pathways, Overexpression of miR-125b in MCF-7 cells significantly down-regulated E2F3 protein level, overexpression of miR-125b caused a marked inhibition of anticancer drug activity and increased resistance in breast cancer cells in vitro. And elevated miR-125b expression in chemoresistant cancer cells were due to high percentage of SP cells.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: High mobility group protein HMGI-C (HMGA2) [14]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
T47D cells Breast Homo sapiens (Human) CVCL_0553
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
MCF10A cells Breast Homo sapiens (Human) CVCL_0598
In Vivo Model Mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Luciferase reporter assay
Experiment for
Drug Resistance
Transwell migration assay
Mechanism Description NEAT1 down-regulation decreased the endogenous HMGA2 expression, and HMGA2 down-regulation mimicked LncRNA NEAT1 down-regulation's effect on cell migration, invasion, EMT phenotype and chemo-sensitivity.
Key Molecule: NAD-dependent protein deacetylase sirtuin-1 (SIRT1) [163]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
miR4766-5p/SIRT1 signaling pathway Regulation hsa05206
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; PE Annexin V assay; TUNEL assay; Invasion assay
Mechanism Description The promotion of cell proliferation and metastasis, the acquisition of chemoresistance, and the increased expression of SIRT1 induced by LncRNA-PRLB overexpression could be partly abrogated by ectopic expression of miR-4766-5p.
Key Molecule: SIRT1 regulating LncRNA tumor promoter (SIRLNT) [163]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell metastasis Activation hsa05205
Cell proliferation Activation hsa05200
Chemoresistance Activation hsa05207
miR4766-5p/SIRT1 signaling pathway Regulation hsa05206
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qPCR; Microarray
Experiment for
Drug Resistance
MTT assay; PE Annexin V assay; TUNEL assay; Invasion assay
Mechanism Description The promotion of cell proliferation and metastasis, the acquisition of chemoresistance, and the increased expression of SIRT1 induced by LncRNA-PRLB overexpression could be partly abrogated by ectopic expression of miR-4766-5p.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Apoptosis regulator BAX (BAX) [159]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Epithelial mesenchymal transition signaling pathway Activation hsa01521
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
Experiment for
Molecule Alteration
Western blot analysis; TUNEL assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description Long noncoding RNA LINP1 acts as an oncogene and promotes chemoresistance against 5-fluoroutacil and doxorubicin by inhibiting chemotherapeutics-induced apoptosis (apoptosis-related proteins such as caspase-8, caspase-9 and Bax proteins) in breast cancer.
Key Molecule: Caspase-8 (CASP8) [159]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Epithelial mesenchymal transition signaling pathway Activation hsa01521
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
Experiment for
Molecule Alteration
Western blot analysis; TUNEL assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description Long noncoding RNA LINP1 acts as an oncogene and promotes chemoresistance against 5-fluoroutacil and doxorubicin by inhibiting chemotherapeutics-induced apoptosis (apoptosis-related proteins such as caspase-8, caspase-9 and Bax proteins) in breast cancer.
Key Molecule: Caspase-9 (CASP9) [159]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Epithelial mesenchymal transition signaling pathway Activation hsa01521
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
Experiment for
Molecule Alteration
Western blot analysis; TUNEL assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description Long noncoding RNA LINP1 acts as an oncogene and promotes chemoresistance against 5-fluoroutacil and doxorubicin by inhibiting chemotherapeutics-induced apoptosis (apoptosis-related proteins such as caspase-8, caspase-9 and Bax proteins) in breast cancer.
Key Molecule: Bifunctional heparan sulfate N-deacetylase/sulfotransferase 1 (NDST1) [160]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
HS signaling pathway Inhibition hsa00534
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-149 modulated chemoresistance through targeting the expression of GlcNAc N-deacetylase/N-sulfotransferase-1 (NDST1). With downregulated miR-149, NDST1 expression was increased in chemoresistant MCF-7/ADM cells versus control MCF-7 wild-type cells. The increased NDST1 then activated a heparan sulfate-related pathway involving activation of heparanase. Finally, expression of miR-149 and NDST1 was confirmed in clinical chemoresistant samples of breast cancers receiving anthracycline/taxane-based chemotherapies. The high expression of NDST1 was also an unfavorable predictor for distant relapse-free survival in Her2 and basal breast cancers.
Key Molecule: Transcription factor E2F3 (E2F3) [3]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation p53 signaling pathway Inhibition hsa04115
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
T47D cells Breast Homo sapiens (Human) CVCL_0553
BT20 cells Breast Homo sapiens (Human) CVCL_0178
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Trypan blue dye exclusion assay
Mechanism Description E2F3, and in some settings E2F1, induce apoptosis through p53-dependent or -independent pathways, Overexpression of miR-125b in MCF-7 cells significantly down-regulated E2F3 protein level, overexpression of miR-125b caused a marked inhibition of anticancer drug activity and increased resistance in breast cancer cells in vitro.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-320 [164]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
TRPC5 signaling pathway Regulation hsa05206
In Vitro Model MCF-7/ADM cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The overexpression of miR-320a, which downregulated TRPC5 and NFATC3, colud inreduce chemoresistance.
Key Molecule: Polycomb complex protein BMI-1 (BMI1) [165]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-200c and miR-203 overexpression in breast cancer cells downregulated Bmi1 expression accompanied with reversion of resistance to 5-Fu mediated by Bmi1.
Key Molecule: hsa-mir-200c [165]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-200c and miR-203 overexpression in breast cancer cells downregulated Bmi1 expression accompanied with reversion of resistance to 5-Fu mediated by Bmi1.
Key Molecule: hsa-mir-203 [165]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-200c and miR-203 overexpression in breast cancer cells downregulated Bmi1 expression accompanied with reversion of resistance to 5-Fu mediated by Bmi1.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: Nuclear paraspeckle assembly transcript 1 (NEAT1) [14]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Epithelial mesenchymal transition signaling pathway Inhibition hsa01521
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
T47D cells Breast Homo sapiens (Human) CVCL_0553
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
MCF10A cells Breast Homo sapiens (Human) CVCL_0598
In Vivo Model Mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Transwell migration assay
Mechanism Description NEAT1 promoted invasion through inducing Epithelial-mesenchymal transition (EMT), NEAT1 down-regulation inhibited cell motility and invasion by reversing the EMT phenotype and increased breast cancer cells chemo-sensitivity. There may be a reciprocal repression between miR211 and NEAT1.
Key Molecule: hsa-mir-211 [14]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Epithelial mesenchymal transition signaling pathway Inhibition hsa01521
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
T47D cells Breast Homo sapiens (Human) CVCL_0553
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
MCF10A cells Breast Homo sapiens (Human) CVCL_0598
In Vivo Model Mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Transwell migration assay
Mechanism Description NEAT1 promoted invasion through inducing Epithelial-mesenchymal transition (EMT), NEAT1 down-regulation inhibited cell motility and invasion by reversing the EMT phenotype and increased breast cancer cells chemo-sensitivity. There may be a reciprocal repression between miR211 and NEAT1.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Nuclear factor of activated T-cells 3 (NFATC3) [164]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
TRPC5 signaling pathway Regulation hsa05206
In Vitro Model MCF-7/ADM cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The overexpression of miR-320a, which downregulated TRPC5 and NFATC3, colud inreduce chemoresistance.
Ovarian cancer [ICD-11: 2C73]
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-206 [166]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description As a potential tumor suppressor, miR206 directly targets CDk4 to suppress the cell growth and enhance the chemotherapy sensitivity to 5-Fu in ovarian cancer cells in vitro.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Cyclin-dependent kinase 4 (CDK4) [166]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
Experiment for
Molecule Alteration
Luciferase reporter assay; Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description As a potential tumor suppressor, miR206 directly targets CDk4 to suppress the cell growth and enhance the chemotherapy sensitivity to 5-Fu in ovarian cancer cells in vitro.
Cervical cancer [ICD-11: 2C77]
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-135a [167]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Cervical cancer [ICD-11: 2C77.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model Hela cells Cervix uteri Homo sapiens (Human) CVCL_0030
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Promega viability kit assay
Mechanism Description Down-regulated of FAk increased sensitivity of HeLa cancer cells to Fluorouracil chemotherapy, targeting and down-regulation of FAk expression by miR-135 and miR-138, overexpressed miR-138 and miR-135 had increased sensitivity to chemotherapy.
Key Molecule: hsa-mir-138 [167]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Cervical cancer [ICD-11: 2C77.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model Hela cells Cervix uteri Homo sapiens (Human) CVCL_0030
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Promega viability kit assay
Mechanism Description Down-regulated of FAk increased sensitivity of HeLa cancer cells to Fluorouracil chemotherapy, targeting and down-regulation of FAk expression by miR-135 and miR-138, overexpressed miR-138 and miR-135 had increased sensitivity to chemotherapy.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Focal adhesion kinase 1 (FAK1) [167]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Cervical cancer [ICD-11: 2C77.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model Hela cells Cervix uteri Homo sapiens (Human) CVCL_0030
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Promega viability kit assay
Mechanism Description Down-regulated of FAk increased sensitivity of HeLa cancer cells to Fluorouracil chemotherapy, targeting and down-regulation of FAk expression by miR-135 and miR-138, overexpressed miR-138 and miR-135 had increased sensitivity to chemotherapy.
Kidney cancer [ICD-11: 2C90]
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: SET and MYND domain containing 2 (SMYD2) [9]
Molecule Alteration Expression
Up-regulation
Resistant Disease Kidney cancer [ICD-11: 2C90.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
HK-2 cells Kidney Homo sapiens (Human) CVCL_0302
In Vivo Model Balb/c athymic nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting assay
Experiment for
Drug Resistance
MTS assay
Mechanism Description SMYD2 is a histone methyltransferase.The estimated IC50 values of cisplatin, doxorubicin, or 5-FU (but not docetaxel) for AZ505-treated RCC cells were significantly lower than those for the control cells, indicating that the SMYD2 inhibition enhanced the drug sensitivity in renal cancer cells.
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 [168]
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.
Key Molecule: hsa-mir-381 [169]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Kidney cancer [ICD-11: 2C90.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell colony Inhibition hsa05200
Cell proliferation Inhibition hsa05200
WEE1/Cdc2 signaling pathway Activation hsa04110
In Vitro Model 786-O cells Kidney Homo sapiens (Human) CVCL_1051
HK-2 cells Kidney Homo sapiens (Human) CVCL_0302
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-381 increases sensitivity of 786-O cells to 5-FU by inhibitory WEE1 and increase of Cdc2activity.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Programmed cell death protein 4 (PDCD4) [168]
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) [168]
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) [168]
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: Wee1-like protein kinase (WEE1) [169]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Kidney cancer [ICD-11: 2C90.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
WEE1/Cdc2 signaling pathway Activation hsa04110
In Vitro Model 786-O cells Kidney Homo sapiens (Human) CVCL_1051
HK-2 cells Kidney Homo sapiens (Human) CVCL_0302
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-381 increases sensitivity of 786-O cells to 5-FU by inhibitory WEE1 and increase of Cdc2activity.
Head and neck cancer [ICD-11: 2D42]
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-let-7d [170]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Head and neck squamous cell carcinoma [ICD-11: 2D42.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
FaDu cells Pharynx Homo sapiens (Human) CVCL_1218
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Clonogenic assay; MTT assay
Mechanism Description The level of let-7d expression is an important factor for cell response to irradiation and chemotherapeutics. Overexpressed let-7d inhibited chemoresistance to cisplatin and paclitaxel in OSCC-ALDH1+ cells.
Key Molecule: hsa-miR-24-3p [171]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Head and neck squamous cell carcinoma [ICD-11: 2D42.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model SCC15 cells Tongue Homo sapiens (Human) CVCL_1681
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-24-3p is involved in regulating cell proliferation, clonogenecity and chemosensitivity in HNSCC cells. CHD5 is the critical downstream mediator implicated in this regulation. Importantly, miR-24-3p is upregulated in HNSCC patient samples. Inhibition of miR-24-3p conferred sensitivity to chemo drugs which was reversed with CHD5 knockdown.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Chromodomain-helicase-DNA-binding protein 5 (CHD5) [171]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Head and neck squamous cell carcinoma [ICD-11: 2D42.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model SCC15 cells Tongue Homo sapiens (Human) CVCL_1681
Experiment for
Molecule Alteration
Northern blotting
Experiment for
Drug Resistance
CellTiter-Glo assay
Mechanism Description miR-24-3p is involved in regulating cell proliferation, clonogenecity and chemosensitivity in HNSCC cells. CHD5 is the critical downstream mediator implicated in this regulation. Importantly, miR-24-3p is upregulated in HNSCC patient samples. Inhibition of miR-24-3p conferred sensitivity to chemo drugs which was reversed with CHD5 knockdown.
Metastatic colorectal cancer [ICD-11: 2D85]
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: Mediator of RNA polymerase II transcription subunit 12 (MED12) [11]
Molecule Alteration Expression
Down-regulation
Resistant Disease Middle eastern colorectal cancer [ICD-11: 2D85.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
TGF-beta signaling pathway Inhibition hsa04350
In Vitro Model DLD1 cells Colon Homo sapiens (Human) CVCL_0248
LOVO cells Colon Homo sapiens (Human) CVCL_0399
Experiment for
Molecule Alteration
Whole genome sequencing assay; Next-generation sequencing assay
Experiment for
Drug Resistance
Clinical manifestations assay
Mechanism Description However, it is worth noting that 50% of MED12-mutated patients with CRC who underwent chemotherapy (4/8) showed poor response to standard chemotherapy.
Key Molecule: Mediator of RNA polymerase II transcription subunit 12 (MED12) [11]
Molecule Alteration Expression
Down-regulation
Resistant Disease Middle eastern colorectal cancer [ICD-11: 2D85.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
TGF-beta signaling pathway Inhibition hsa04350
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
DLD1 cells Colon Homo sapiens (Human) CVCL_0248
CaCo2 cells Colon Homo sapiens (Human) CVCL_0025
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
CL34 cells Colon Homo sapiens (Human) CVCL_1980
Colo-320 cells Colon Homo sapiens (Human) CVCL_1989
Experiment for
Molecule Alteration
Immunoblotted assay; Fluorescence immunostaining assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description We also show that MED12 deficiency confers resistance to chemotherapy at the tissue culture level. MED12 is a likely tumour suppressor that modulates TGF-beta-induced epithelial-mesenchymal transition signalling in CRC cell lines.
Key Molecule: Mediator of RNA polymerase II transcription subunit 12 (MED12) [11]
Molecule Alteration Mutation
.
Resistant Disease Middle eastern colorectal cancer [ICD-11: 2D85.1]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
TGF-beta signaling pathway Inhibition hsa04350
Experiment for
Molecule Alteration
Whole genome sequencing assay; Next-generation sequencing assay
Experiment for
Drug Resistance
Clinical manifestations assay
Mechanism Description However, it is worth noting that 50% of MED12-mutated patients with CRC who underwent chemotherapy (4/8) showed poor response to standard chemotherapy.
Colorectal peritoneal carcinomatosis [ICD-11: 2D91]
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: Adipocytic Glutamine Synthetase (GS) [6]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal peritoneal carcinomatosis [CD-11: 2D91.Y]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation mTOR signaling pathway Activation hsa04150
In Vitro Model B16 cells Skin Homo sapiens (Human) CVCL_F936
HCT-116 cells Colon Homo sapiens (Human) N.A.
CT26 cells Colon Mus musculus (Mouse) CVCL_7254
MC38 cells Colon Mus musculus (Mouse) CVCL_B288
SW-480 cells Colon Homo sapiens (Human) CVCL_0546
3T3-L1 cells Nasopharynx Mus musculus (Mouse) CVCL_0123
In Vivo Model BALB/c mice xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR; Western blotting assay
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Adipocytic Glutamine Synthetase Upregulation via Altered Histone Methylation Promotes 5FU Chemoresistance in Peritoneal Carcinomatosis of Colorectal Cancer
Salivary gland carcinoma [ICD-11: 2E60]
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: Prominin-1 (PROM1) [22]
Molecule Alteration Expression
Up-regulation
Resistant Disease Salivary gland adenoid cystic carcinoma [ICD-11: 2E60.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation MET/PI3K/AKT/mTOR signaling pathway Activation hsa04150
Cell migration Activation hsa04670
Cell invasion Activation hsa05200
In Vitro Model KOA-1 cells Skin Homo sapiens (Human) CVCL_L997
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Mechanism Description CD133 activates the PI3K/AKT, AKT/Wnt and other signaling pathways and affects the behavior of CD133+ cells, thereby playing a major role in cancer therapy. In addition, CD133 is also involved in the regulation of tumor resistance. Long-term chemotherapy leads to a significant increase in CD133 expression. Targeting CD133 can reverse drug resistance in colorectal cancer via the AKT/NF-kappa-B/multidrug resistance protein (MDR)1 pathway.
References
Ref 1 Long noncoding RNA HOTAIR regulates polycomb-dependent chromatin modification and is associated with poor prognosis in colorectal cancers. Cancer Res. 2011 Oct 15;71(20):6320-6. doi: 10.1158/0008-5472.CAN-11-1021. Epub 2011 Aug 23.
Ref 2 lncRNA HOTAIR Contributes to 5FU Resistance through Suppressing miR-218 and Activating NF-kB/TS Signaling in Colorectal Cancer. Mol Ther Nucleic Acids. 2017 Sep 15;8:356-369. doi: 10.1016/j.omtn.2017.07.007. Epub 2017 Jul 8.
Ref 3 Circulating MiR-125b as a marker predicting chemoresistance in breast cancer. PLoS One. 2012;7(4):e34210. doi: 10.1371/journal.pone.0034210. Epub 2012 Apr 16.
Ref 4 Dysregulation of microRNA-34a expression causes drug-resistance to 5-FU in human colon cancer DLD-1 cells. Cancer Lett. 2011 Jan 28;300(2):197-204. doi: 10.1016/j.canlet.2010.10.006. Epub 2010 Nov 9.
Ref 5 MicroRNA-21 induces resistance to 5-fluorouracil by down-regulating human DNA MutS homolog 2 (hMSH2). Proc Natl Acad Sci U S A. 2010 Dec 7;107(49):21098-103. doi: 10.1073/pnas.1015541107. Epub 2010 Nov 15.
Ref 6 Adipocytic Glutamine Synthetase Upregulation via Altered Histone Methylation Promotes 5FU Chemoresistance in Peritoneal Carcinomatosis of Colorectal Cancer .Front Oncol. 2021 Oct 12;11:748730. doi: 10.3389/fonc.2021.748730. eCollection 2021. 10.3389/fonc.2021.748730
Ref 7 miR-221 Mediates Chemoresistance of Esophageal Adenocarcinoma by Direct Targeting of DKK2 Expression. Ann Surg. 2016 Nov;264(5):804-814. doi: 10.1097/SLA.0000000000001928.
Ref 8 Long noncoding RNA MALAT1 regulates autophagy associated chemoresistance via miR-23b-3p sequestration in gastric cancer. Mol Cancer. 2017 Nov 21;16(1):174. doi: 10.1186/s12943-017-0743-3.
Ref 9 Inhibition of SMYD2 suppresses tumor progression by down-regulating microRNA-125b and attenuates multi-drug resistance in renal cell carcinoma .Theranostics. 2019 Oct 22;9(26):8377-8391. doi: 10.7150/thno.37628. eCollection 2019. 10.7150/thno.37628
Ref 10 MicroRNA-21 induces resistance to the anti-tumour effect of interferon-Alpha/5-fluorouracil in hepatocellular carcinoma cells. Br J Cancer. 2010 Nov 9;103(10):1617-26. doi: 10.1038/sj.bjc.6605958. Epub 2010 Oct 26.
Ref 11 MED12 is recurrently mutated in Middle Eastern colorectal cancer. Gut. 2018 Apr;67(4):663-671. doi: 10.1136/gutjnl-2016-313334. Epub 2017 Feb 9.
Ref 12 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 13 Reciprocal regulation of DGCR5 and miR-320a affects the cellular malignant phenotype and 5-FU response in pancreatic ductal adenocarcinoma. Oncotarget. 2017 Jun 6;8(53):90868-90878. doi: 10.18632/oncotarget.18377. eCollection 2017 Oct 31.
Ref 14 The lncRNA NEAT1 facilitates cell growth and invasion via the miR-211/HMGA2 axis in breast cancer. Int J Biol Macromol. 2017 Dec;105(Pt 1):346-353. doi: 10.1016/j.ijbiomac.2017.07.053. Epub 2017 Jul 16.
Ref 15 Role of miR-19b and its target mRNAs in 5-fluorouracil resistance in colon cancer cells. J Gastroenterol. 2012 Aug;47(8):883-95. doi: 10.1007/s00535-012-0547-6. Epub 2012 Mar 1.
Ref 16 miR-193a-3p regulation of chemoradiation resistance in oesophageal cancer cells via the PSEN1 gene. Gene. 2016 Apr 1;579(2):139-45. doi: 10.1016/j.gene.2015.12.060. Epub 2015 Dec 29.
Ref 17 MicroRNA-19a/b regulates multidrug resistance in human gastric cancer cells by targeting PTEN. Biochem Biophys Res Commun. 2013 May 10;434(3):688-94. doi: 10.1016/j.bbrc.2013.04.010. Epub 2013 Apr 16.
Ref 18 Targeting PCDH20 gene by microRNA-122 confers 5-FU resistance in hepatic carcinoma. Am J Cancer Res. 2016 Aug 1;6(8):1681-94. eCollection 2016.
Ref 19 MicroRNA-128-2 targets the transcriptional repressor E2F5 enhancing mutant p53 gain of function. Cell Death Differ. 2012 Jun;19(6):1038-48. doi: 10.1038/cdd.2011.190. Epub 2011 Dec 23.
Ref 20 miR-654-5p Targets GRAP to Promote Proliferation, Metastasis, and Chemoresistance of Oral Squamous Cell Carcinoma Through Ras/MAPK Signaling. DNA Cell Biol. 2018 Apr;37(4):381-388. doi: 10.1089/dna.2017.4095. Epub 2018 Jan 24.
Ref 21 MicroRNA-1246 expression associated with CCNG2-mediated chemoresistance and stemness in pancreatic cancer. Br J Cancer. 2014 Oct 14;111(8):1572-80. doi: 10.1038/bjc.2014.454. Epub 2014 Aug 12.
Ref 22 Effects of CD133 expression on chemotherapy and drug sensitivity of adenoid cystic carcinoma .Mol Med Rep. 2022 Jan;25(1):18. doi: 10.3892/mmr.2021.12534. Epub 2021 Nov 18. 10.3892/mmr.2021.12534
Ref 23 miR-138 might reverse multidrug resistance of leukemia cells. Leuk Res. 2010 Aug;34(8):1078-82. doi: 10.1016/j.leukres.2009.10.002. Epub 2009 Nov 6.
Ref 24 Mechanism of chemoresistance mediated by miR-140 in human osteosarcoma and colon cancer cells. Oncogene. 2009 Nov 19;28(46):4065-74. doi: 10.1038/onc.2009.274. Epub 2009 Sep 7.
Ref 25 miR-3188 regulates nasopharyngeal carcinoma proliferation and chemosensitivity through a FOXO1-modulated positive feedback loop with mTOR-p-PI3K/AKT-c-JUN. Nat Commun. 2016 Apr 20;7:11309. doi: 10.1038/ncomms11309.
Ref 26 A novel miR-365-3p/EHF/keratin 16 axis promotes oral squamous cell carcinoma metastasis, cancer stemness and drug resistance via enhancing Beta5-integrin/c-met signaling pathway. J Exp Clin Cancer Res. 2019 Feb 19;38(1):89. doi: 10.1186/s13046-019-1091-5.
Ref 27 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 28 Complex role of miR-130a-3p and miR-148a-3p balance on drug resistance and tumor biology in esophageal squamous cell carcinoma. Sci Rep. 2018 Dec 3;8(1):17553. doi: 10.1038/s41598-018-35799-1.
Ref 29 Long noncoding RNA LINC00261 induces chemosensitization to 5-fluorouracil by mediating methylation-dependent repression of DPYD in human esophageal cancer. FASEB J. 2019 Feb;33(2):1972-1988. doi: 10.1096/fj.201800759R. Epub 2018 Sep 18.
Ref 30 LncRNA-TUSC7/miR-224 affected chemotherapy resistance of esophageal squamous cell carcinoma by competitively regulating DESC1. J Exp Clin Cancer Res. 2018 Mar 12;37(1):56. doi: 10.1186/s13046-018-0724-4.
Ref 31 Circulating miR-200c levels significantly predict response to chemotherapy and prognosis of patients undergoing neoadjuvant chemotherapy for esophageal cancer. Ann Surg Oncol. 2013 Dec;20 Suppl 3:S607-15. doi: 10.1245/s10434-013-3093-4. Epub 2013 Jul 10.
Ref 32 Mir-148a improves response to chemotherapy in sensitive and resistant oesophageal adenocarcinoma and squamous cell carcinoma cells. J Gastrointest Surg. 2011 Mar;15(3):429-38. doi: 10.1007/s11605-011-1418-9. Epub 2011 Jan 19.
Ref 33 The prognostic and chemotherapeutic value of miR-296 in esophageal squamous cell carcinoma. Ann Surg. 2010 Jun;251(6):1056-63. doi: 10.1097/SLA.0b013e3181dd4ea9.
Ref 34 Down-regulation of miR-27a might reverse multidrug resistance of esophageal squamous cell carcinoma. Dig Dis Sci. 2010 Sep;55(9):2545-51. doi: 10.1007/s10620-009-1051-6. Epub 2009 Dec 4.
Ref 35 LncRNA PVT1 Mediates Antiapoptosis and 5-Fluorouracil Resistance via Increasing Bcl2 Expression in Gastric Cancer. J Oncol. 2019 May 8;2019:9325407. doi: 10.1155/2019/9325407. eCollection 2019.
Ref 36 Long Non-Coding RNA XLOC_006753 Promotes the Development of Multidrug Resistance in Gastric Cancer Cells Through the PI3K/AKT/mTOR Signaling Pathway. Cell Physiol Biochem. 2018;51(3):1221-1236. doi: 10.1159/000495499. Epub 2018 Nov 27.
Ref 37 MicroRNA-145 exerts tumor-suppressive and chemo-resistance lowering effects by targeting CD44 in gastric cancer. World J Gastroenterol. 2017 Apr 7;23(13):2337-2345. doi: 10.3748/wjg.v23.i13.2337.
Ref 38 Long Non-Coding RNA (LncRNA) Urothelial Carcinoma Associated 1 (UCA1) Increases Multi-Drug Resistance of Gastric Cancer via Downregulating miR-27b. Med Sci Monit. 2016 Oct 1;22:3506-3513. doi: 10.12659/msm.900688.
Ref 39 miR-363 promotes proliferation and chemo-resistance of human gastric cancer via targeting of FBW7 ubiquitin ligase expression. Oncotarget. 2016 Jun 7;7(23):35284-92. doi: 10.18632/oncotarget.9169.
Ref 40 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 41 Effect and mechanism of long noncoding RNAs HOTAIR on occurrence and development of gastric cancer. J Cell Biochem. 2017 Dec 13. doi: 10.1002/jcb.26594. Online ahead of print.
Ref 42 Downregulation of MicroRNA-147 Inhibits Cell Proliferation and Increases the Chemosensitivity of Gastric Cancer Cells to 5-Fluorouracil by Directly Targeting PTEN. Oncol Res. 2018 Jul 5;26(6):901-911. doi: 10.3727/096504017X15061902533715. Epub 2017 Sep 26.
Ref 43 MicroRNA-31 inhibits RhoA-mediated tumor invasion and chemotherapy resistance in MKN-45 gastric adenocarcinoma cells. Exp Biol Med (Maywood). 2017 Dec;242(18):1842-1847. doi: 10.1177/1535370217728460. Epub 2017 Aug 24.
Ref 44 Silencing of LncRNA HULC Enhances Chemotherapy Induced Apoptosis in Human Gastric Cancer. J Med Biochem. 2016 Apr;35(2):137-143. doi: 10.1515/jomb-2015-0016. Epub 2016 May 9.
Ref 45 Decreased expression of miR-939 contributes to chemoresistance and metastasis of gastric cancer via dysregulation of SLC34A2 and Raf/MEK/ERK pathway. Mol Cancer. 2017 Jan 23;16(1):18. doi: 10.1186/s12943-017-0586-y.
Ref 46 Induction of miR-31 causes increased sensitivity to 5-FU and decreased migration and cell invasion in gastric adenocarcinoma. Bratisl Lek Listy. 2019;120(1):35-39. doi: 10.4149/BLL_2019_005.
Ref 47 MicroRNA-31 triggers G(2)/M cell cycle arrest, enhances the chemosensitivity and inhibits migration and invasion of human gastric cancer cells by downregulating the expression of zeste homolog 2 (ZH2). Arch Biochem Biophys. 2019 Mar 15;663:269-275. doi: 10.1016/j.abb.2019.01.023. Epub 2019 Jan 21.
Ref 48 MicroRNA-495 Confers Increased Sensitivity to Chemotherapeutic Agents in Gastric Cancer via the Mammalian Target of Rapamycin (mTOR) Signaling Pathway by Interacting with Human Epidermal Growth Factor Receptor 2 (ERBB2). Med Sci Monit. 2018 Aug 27;24:5960-5972. doi: 10.12659/MSM.909458.
Ref 49 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 50 MicroRNA-623 Targets Cyclin D1 to Inhibit Cell Proliferation and Enhance the Chemosensitivity of Cells to 5-Fluorouracil in Gastric Cancer. Oncol Res. 2018 Dec 27;27(1):19-27. doi: 10.3727/096504018X15193469240508. Epub 2018 Mar 1.
Ref 51 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 52 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 53 MicroRNA 197 reverses the drug resistance of fluorouracil induced SGC7901 cells by targeting mitogen activated protein kinase 1. Mol Med Rep. 2015 Oct;12(4):5019-25. doi: 10.3892/mmr.2015.4052. Epub 2015 Jul 7.
Ref 54 miR-23b-3p regulates the chemoresistance of gastric cancer cells by targeting ATG12 and HMGB2. Cell Death Dis. 2015 May 21;6(5):e1766. doi: 10.1038/cddis.2015.123.
Ref 55 miR-508-5p regulates multidrug resistance of gastric cancer by targeting ABCB1 and ZNRD1. Oncogene. 2014 Jun 19;33(25):3267-76. doi: 10.1038/onc.2013.297. Epub 2013 Jul 29.
Ref 56 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 57 Down-regulation of miR-27a might inhibit proliferation and drug resistance of gastric cancer cells. J Exp Clin Cancer Res. 2011 May 13;30(1):55. doi: 10.1186/1756-9966-30-55.
Ref 58 miR-181b modulates multidrug resistance by targeting BCL2 in human cancer cell lines. Int J Cancer. 2010 Dec 1;127(11):2520-9. doi: 10.1002/ijc.25260.
Ref 59 MiR-30a Decreases Multidrug Resistance (MDR) of Gastric Cancer Cells. Med Sci Monit. 2016 Nov 23;0:0.
Ref 60 MicroRNA-17 inhibition overcomes chemoresistance and suppresses epithelial-mesenchymal transition through a DEDD-dependent mechanism in gastric cancer. Int J Biochem Cell Biol. 2018 Sep;102:59-70. doi: 10.1016/j.biocel.2018.06.007. Epub 2018 Jun 25.
Ref 61 Sensitization of Gastric Cancer Cells to 5-FU by MicroRNA-204 Through Targeting the TGFBR2-Mediated Epithelial to Mesenchymal Transition. Cell Physiol Biochem. 2018;47(4):1533-1545. doi: 10.1159/000490871. Epub 2018 Jun 21.
Ref 62 LEIGC long non-coding RNA acts as a tumor suppressor in gastric carcinoma by inhibiting the epithelial-to-mesenchymal transition. BMC Cancer. 2014 Dec 11;14:932. doi: 10.1186/1471-2407-14-932.
Ref 63 MicroRNA-195 desensitizes HCT116 human colon cancer cells to 5-fluorouracil. Cancer Lett. 2018 Jan 1;412:264-271. doi: 10.1016/j.canlet.2017.10.022. Epub 2017 Nov 5.
Ref 64 miR-15b-5p resensitizes colon cancer cells to 5-fluorouracil by promoting apoptosis via the NF-kB/XIAP axis. Sci Rep. 2017 Jun 23;7(1):4194. doi: 10.1038/s41598-017-04172-z.
Ref 65 miR-206 regulates 5-FU resistance by targeting Bcl-2 in colon cancer cells. Onco Targets Ther. 2018 Mar 29;11:1757-1765. doi: 10.2147/OTT.S159093. eCollection 2018.
Ref 66 [Drug resistance of colon cancer cells to 5-fluorouracil mediated by microRNA-21]. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 2015 Oct;32(5):620-4. doi: 10.3760/cma.j.issn.1003-9406.2015.05.003.
Ref 67 A long non-coding RNA snaR contributes to 5-fluorouracil resistance in human colon cancer cells. Mol Cells. 2014 Jul;37(7):540-6. doi: 10.14348/molcells.2014.0151. Epub 2014 Jul 31.
Ref 68 High miR-21 expression from FFPE tissues is associated with poor survival and response to adjuvant chemotherapy in colon cancer. Int J Cancer. 2014 Apr 15;134(8):1926-34. doi: 10.1002/ijc.28522. Epub 2013 Nov 8.
Ref 69 Targeting miR-21 enhances the sensitivity of human colon cancer HT-29 cells to chemoradiotherapy in vitro. Biochem Biophys Res Commun. 2014 Jan 17;443(3):789-95. doi: 10.1016/j.bbrc.2013.11.064. Epub 2013 Nov 23.
Ref 70 miR-338-3p confers 5-fluorouracil resistance in p53 mutant colon cancer cells by targeting the mammalian target of rapamycin. Exp Cell Res. 2017 Nov 15;360(2):328-336. doi: 10.1016/j.yexcr.2017.09.023. Epub 2017 Sep 18.
Ref 71 Upregulation of miR-101 enhances the cytotoxic effect of anticancer drugs through inhibition of colon cancer cell proliferation. Oncol Rep. 2017 Jul;38(1):100-108. doi: 10.3892/or.2017.5666. Epub 2017 May 24.
Ref 72 miR-20b reduces 5-FU resistance by suppressing the ADAM9/EGFR signaling pathway in colon cancer. Oncol Rep. 2017 Jan;37(1):123-130. doi: 10.3892/or.2016.5259. Epub 2016 Nov 18.
Ref 73 MicroRNA-302a enhances 5-fluorouracil-induced cell death in human colon cancer cells. Oncol Rep. 2017 Jan;37(1):631-639. doi: 10.3892/or.2016.5237. Epub 2016 Nov 8.
Ref 74 MiR-214 sensitizes human colon cancer cells to 5-FU by targeting Hsp27. Cell Mol Biol Lett. 2019 Mar 14;24:22. doi: 10.1186/s11658-019-0143-3. eCollection 2019.
Ref 75 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 76 MicroRNA-494 sensitizes colon cancer cells to fluorouracil through regulation of DPYD. IUBMB Life. 2015 Mar;67(3):191-201. doi: 10.1002/iub.1361. Epub 2015 Apr 15.
Ref 77 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 78 Inhibition of lactate dehydrogenase A by microRNA-34a resensitizes colon cancer cells to 5-fluorouracil. Mol Med Rep. 2015 Jan;11(1):577-82. doi: 10.3892/mmr.2014.2726. Epub 2014 Oct 21.
Ref 79 MiR-142-3p functions as a tumor suppressor by targeting CD133, ABCG2, and Lgr5 in colon cancer cells. J Mol Med (Berl). 2013 Aug;91(8):989-1000. doi: 10.1007/s00109-013-1037-x. Epub 2013 Apr 26.
Ref 80 Long non-coding RNA bladder cancer-associated transcript 2 contributes to disease progression, chemoresistance and poor survival of patients with colorectal cancer. Oncol Lett. 2019 Aug;18(2):2050-2058. doi: 10.3892/ol.2019.10487. Epub 2019 Jun 18.
Ref 81 Induced miR-31 by 5-fluorouracil exposure contributes to the resistance in colorectal tumors. Cancer Sci. 2019 Aug;110(8):2540-2548. doi: 10.1111/cas.14090. Epub 2019 Jul 23.
Ref 82 SNHG15 is a bifunctional MYC-regulated noncoding locus encoding a lncRNA that promotes cell proliferation, invasion and drug resistance in colorectal cancer by interacting with AIF. J Exp Clin Cancer Res. 2019 Apr 24;38(1):172. doi: 10.1186/s13046-019-1169-0.
Ref 83 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 84 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 85 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 86 Long non-coding RNA LINC00152 promotes cell proliferation, metastasis, and confers 5-FU resistance in colorectal cancer by inhibiting miR-139-5p. Oncogenesis. 2017 Nov 28;6(11):395. doi: 10.1038/s41389-017-0008-4.
Ref 87 The long non-coding RNA ENST00000547547 reduces 5-fluorouracil resistance of colorectal cancer cells via competitive binding to microRNA-31. Oncol Rep. 2018 Jan;39(1):217-226. doi: 10.3892/or.2017.6082. Epub 2017 Nov 7.
Ref 88 Upregulation of microRNA-135b and microRNA-182 promotes chemoresistance of colorectal cancer by targeting ST6GALNAC2 via PI3K/AKT pathway. Mol Carcinog. 2017 Dec;56(12):2669-2680. doi: 10.1002/mc.22710. Epub 2017 Aug 21.
Ref 89 Down-regulation of long non-coding RNA RP11-708H21.4 is associated with poor prognosis for colorectal cancer and promotes tumorigenesis through regulating AKT/mTOR pathway. Oncotarget. 2017 Apr 25;8(17):27929-27942. doi: 10.18632/oncotarget.15846.
Ref 90 miR-106a Reduces 5-Fluorouracil (5-FU) Sensitivity of Colorectal Cancer by Targeting Dual-Specificity Phosphatases 2 (DUSP2). Med Sci Monit. 2018 Jul 16;24:4944-4951. doi: 10.12659/MSM.910016.
Ref 91 Knockdown of long non coding RNA PVT1 reverses multidrug resistance in colorectal cancer cells. Mol Med Rep. 2018 Jun;17(6):8309-8315. doi: 10.3892/mmr.2018.8907. Epub 2018 Apr 20.
Ref 92 LncRNA-UCA1 enhances cell proliferation and 5-fluorouracil resistance in colorectal cancer by inhibiting miR-204-5p. Sci Rep. 2016 Apr 5;6:23892. doi: 10.1038/srep23892.
Ref 93 miR-450b-5p Suppresses Stemness and the Development of Chemoresistance by Targeting SOX2 in Colorectal Cancer. DNA Cell Biol. 2016 May;35(5):249-56. doi: 10.1089/dna.2015.3120. Epub 2016 Feb 4.
Ref 94 MicroRNA-587 antagonizes 5-FU-induced apoptosis and confers drug resistance by regulating PPP2R1B expression in colorectal cancer. Cell Death Dis. 2015 Aug 6;6(8):e1845. doi: 10.1038/cddis.2015.200.
Ref 95 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 96 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 97 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 98 MicroRNA-10b is a prognostic indicator in colorectal cancer and confers resistance to the chemotherapeutic agent 5-fluorouracil in colorectal cancer cells. Ann Surg Oncol. 2012 Sep;19(9):3065-71. doi: 10.1245/s10434-012-2246-1. Epub 2012 Feb 10.
Ref 99 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 100 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 101 Decreased expression of LncRNA SLC25A25-AS1 promotes proliferation, chemoresistance, and EMT in colorectal cancer cells. Tumour Biol. 2016 Oct;37(10):14205-14215. doi: 10.1007/s13277-016-5254-0. Epub 2016 Aug 23.
Ref 102 Downregulation of miR-874-3p promotes chemotherapeutic resistance in colorectal cancer via inactivation of the Hippo signaling pathway. Oncol Rep. 2017 Dec;38(6):3376-3386. doi: 10.3892/or.2017.6041. Epub 2017 Oct 17.
Ref 103 A polymorphism in ABCC4 is related to efficacy of 5-FU/capecitabine-based chemotherapy in colorectal cancer patients. Sci Rep. 2017 Aug 1;7(1):7059. doi: 10.1038/s41598-017-07491-3.
Ref 104 Maintenance of cancer stemness by miR-196b-5p contributes to chemoresistance of colorectal cancer cells via activating STAT3 signaling pathway. Oncotarget. 2017 Jul 25;8(30):49807-49823. doi: 10.18632/oncotarget.17971.
Ref 105 The Effect of miR-200c Inhibition on Chemosensitivity (5- FluoroUracil) in Colorectal Cancer. Pathol Oncol Res. 2018 Jan;24(1):145-151. doi: 10.1007/s12253-017-0222-6. Epub 2017 Apr 14.
Ref 106 CXCL12/CXCR4 axis induced miR-125b promotes invasion and confers 5-fluorouracil resistance through enhancing autophagy in colorectal cancer. Sci Rep. 2017 Feb 8;7:42226. doi: 10.1038/srep42226.
Ref 107 microRNA-577 suppresses tumor growth and enhances chemosensitivity in colorectal cancer. J Biochem Mol Toxicol. 2017 Jun;31(6). doi: 10.1002/jbt.21888. Epub 2017 Feb 2.
Ref 108 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 109 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 110 Down-regulation of miR-543 expression increases the sensitivity of colorectal cancer cells to 5-Fluorouracil through the PTEN/PI3K/AKT pathway. Biosci Rep. 2019 Mar 22;39(3):BSR20190249. doi: 10.1042/BSR20190249. Print 2019 Mar 29.
Ref 111 Overcoming stemness and chemoresistance in colorectal cancer through miR-195-5p-modulated inhibition of notch signaling. Int J Biol Macromol. 2018 Oct 1;117:445-453. doi: 10.1016/j.ijbiomac.2018.05.151. Epub 2018 May 28.
Ref 112 miR-519b-3p promotes responsiveness to preoperative chemoradiotherapy in rectal cancer patients by targeting ARID4B. Gene. 2018 May 20;655:84-90. doi: 10.1016/j.gene.2018.02.056. Epub 2018 Mar 22.
Ref 113 MicroRNA-761 promotes the sensitivity of colorectal cancer cells to 5-Fluorouracil through targeting FOXM1. Oncotarget. 2017 Aug 10;9(1):321-331. doi: 10.18632/oncotarget.20109. eCollection 2018 Jan 2.
Ref 114 miRNA-497 Enhances the Sensitivity of Colorectal Cancer Cells to Neoadjuvant Chemotherapeutic Drug. Curr Protein Pept Sci. 2015;16(4):310-5. doi: 10.2174/138920371604150429154142.
Ref 115 MicroRNA-497 inhibits tumor growth and increases chemosensitivity to 5-fluorouracil treatment by targeting KSR1. Oncotarget. 2016 Jan 19;7(3):2660-71. doi: 10.18632/oncotarget.6545.
Ref 116 MicroRNA-149 Increases the Sensitivity of Colorectal Cancer Cells to 5-Fluorouracil by Targeting Forkhead Box Transcription Factor FOXM1. Cell Physiol Biochem. 2016;39(2):617-29. doi: 10.1159/000445653. Epub 2016 Jul 15.
Ref 117 MicroRNA-874 inhibits growth, induces apoptosis and reverses chemoresistance in colorectal cancer by targeting X-linked inhibitor of apoptosis protein. Oncol Rep. 2016 Jul;36(1):542-50. doi: 10.3892/or.2016.4810. Epub 2016 May 16.
Ref 118 miR-139-5p sensitizes colorectal cancer cells to 5-fluorouracil by targeting NOTCH-1. Pathol Res Pract. 2016 Jul;212(7):643-9. doi: 10.1016/j.prp.2016.04.011. Epub 2016 May 3.
Ref 119 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 120 MiR-22 regulates 5-FU sensitivity by inhibiting autophagy and promoting apoptosis in colorectal cancer cells. Cancer Lett. 2015 Jan 28;356(2 Pt B):781-90. doi: 10.1016/j.canlet.2014.10.029. Epub 2014 Oct 29.
Ref 121 MicroRNA-23a antisense enhances 5-fluorouracil chemosensitivity through APAF-1/caspase-9 apoptotic pathway in colorectal cancer cells. J Cell Biochem. 2014 Apr;115(4):772-84. doi: 10.1002/jcb.24721.
Ref 122 Repression of c-Kit by p53 is mediated by miR-34 and is associated with reduced chemoresistance, migration and stemness. Oncotarget. 2013 Sep;4(9):1399-415. doi: 10.18632/oncotarget.1202.
Ref 123 miR-129 promotes apoptosis and enhances chemosensitivity to 5-fluorouracil in colorectal cancer. Cell Death Dis. 2013 Jun 6;4(6):e659. doi: 10.1038/cddis.2013.193.
Ref 124 MicroRNA-497 targets insulin-like growth factor 1 receptor and has a tumour suppressive role in human colorectal cancer. Oncogene. 2013 Apr 11;32(15):1910-20. doi: 10.1038/onc.2012.214. Epub 2012 Jun 18.
Ref 125 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 126 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 127 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 128 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 129 MicroRNA-224 is associated with colorectal cancer progression and response to 5-fluorouracil-based chemotherapy by KRAS-dependent and -independent mechanisms. Br J Cancer. 2015 Apr 28;112(9):1480-90. doi: 10.1038/bjc.2015.125.
Ref 130 SNAI2 modulates colorectal cancer 5-fluorouracil sensitivity through miR145 repression. Mol Cancer Ther. 2014 Nov;13(11):2713-26. doi: 10.1158/1535-7163.MCT-14-0207. Epub 2014 Sep 23.
Ref 131 MicroRNA-143 reduces viability and increases sensitivity to 5-fluorouracil in HCT116 human colorectal cancer cells. FEBS J. 2009 Nov;276(22):6689-700. doi: 10.1111/j.1742-4658.2009.07383.x. Epub 2009 Oct 16.
Ref 132 Long non-coding RNA GAS5 antagonizes the chemoresistance of pancreatic cancer cells through down-regulation of miR-181c-5p. Biomed Pharmacother. 2018 Jan;97:809-817. doi: 10.1016/j.biopha.2017.10.157. Epub 2017 Nov 6.
Ref 133 MiRNA-221-3p desensitizes pancreatic cancer cells to 5-fluorouracil by targeting RB1. Tumour Biol. 2016 Oct 10. doi: 10.1007/s13277-016-5445-8. Online ahead of print.
Ref 134 Long non-coding RNA CUDR promotes malignant phenotypes in pancreatic ductal adenocarcinoma via activating AKT and ERK signaling pathways. Int J Oncol. 2018 Dec;53(6):2671-2682. doi: 10.3892/ijo.2018.4574. Epub 2018 Sep 27.
Ref 135 MicroRNA-21 induces 5-fluorouracil resistance in human pancreatic cancer cells by regulating PTEN and PDCD4. Cancer Med. 2016 Apr;5(4):693-702. doi: 10.1002/cam4.626. Epub 2016 Feb 10.
Ref 136 Upregulation of miR-181c contributes to chemoresistance in pancreatic cancer by inactivating the Hippo signaling pathway. Oncotarget. 2015 Dec 29;6(42):44466-79. doi: 10.18632/oncotarget.6298.
Ref 137 MicroRNA-320a promotes 5-FU resistance in human pancreatic cancer cells. Sci Rep. 2016 Jun 9;6:27641. doi: 10.1038/srep27641.
Ref 138 Identification of microRNA-21 as a biomarker for chemoresistance and clinical outcome following adjuvant therapy in resectable pancreatic cancer. PLoS One. 2010 May 14;5(5):e10630. doi: 10.1371/journal.pone.0010630.
Ref 139 Downregulation of long noncoding RNA CRNDE suppresses drug resistance of liver cancer cells by increasing microRNA-33a expression and decreasing HMGA2 expression. Cell Cycle. 2019 Oct;18(19):2524-2537. doi: 10.1080/15384101.2019.1652035. Epub 2019 Aug 15.
Ref 140 microRNA-200a-3p increases 5-fluorouracil resistance by regulating dual specificity phosphatase 6 expression. Exp Mol Med. 2017 May 12;49(5):e327. doi: 10.1038/emm.2017.33.
Ref 141 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 142 miR-503 inhibits proliferation making human hepatocellular carcinoma cells susceptible to 5 fluorouracil by targeting EIF4E. Oncol Rep. 2017 Jan;37(1):563-570. doi: 10.3892/or.2016.5220. Epub 2016 Nov 7.
Ref 143 Exosomal microRNA-32-5p induces multidrug resistance in hepatocellular carcinoma via the PI3K/Akt pathway. J Exp Clin Cancer Res. 2018 Mar 12;37(1):52. doi: 10.1186/s13046-018-0677-7.
Ref 144 The long non-coding RNA, SNHG6-003, functions as a competing endogenous RNA to promote the progression of hepatocellular carcinoma. Oncogene. 2017 Feb 23;36(8):1112-1122. doi: 10.1038/onc.2016.278. Epub 2016 Aug 15.
Ref 145 Bioinformatic identification of IGF1 as a hub gene in hepatocellular carcinoma (HCC) and in-vitro analysis of the chemosensitizing effect of miR-379 via suppressing the IGF1/IGF1R signaling pathway. Eur Rev Med Pharmacol Sci. 2016 Dec;20(24):5098-5106.
Ref 146 Long non-coding RNA 00607 as a tumor suppressor by modulating NF-kB p65/p53 signaling axis in hepatocellular carcinoma. Carcinogenesis. 2018 Dec 31;39(12):1438-1446. doi: 10.1093/carcin/bgy113.
Ref 147 lncRNA KRAL reverses 5-fluorouracil resistance in hepatocellular carcinoma cells by acting as a ceRNA against miR-141. Cell Commun Signal. 2018 Aug 17;16(1):47. doi: 10.1186/s12964-018-0260-z.
Ref 148 MiR-183 modulates multi-drug resistance in hepatocellular cancer (HCC) cells via miR-183-IDH2/SOCS6-HIF-1Alpha feedback loop. Eur Rev Med Pharmacol Sci. 2016 May;20(10):2020-7.
Ref 149 MicroRNA 133a and microRNA 326 co contribute to hepatocellular carcinoma 5 fluorouracil and cisplatin sensitivity by directly targeting B cell lymphoma extra large. Mol Med Rep. 2015 Oct;12(4):6235-40. doi: 10.3892/mmr.2015.4134. Epub 2015 Jul 29.
Ref 150 Let-7b binding site polymorphism in the B-cell lymphoma-extra large 3'UTR is associated with fluorouracil resistance of hepatocellular carcinoma. Mol Med Rep. 2015 Jan;11(1):677-81. doi: 10.3892/mmr.2014.2692. Epub 2014 Oct 17.
Ref 151 Overexpression of microRNA-125b sensitizes human hepatocellular carcinoma cells to 5-fluorouracil through inhibition of glycolysis by targeting hexokinase II. Mol Med Rep. 2014 Aug;10(2):995-1002. doi: 10.3892/mmr.2014.2271. Epub 2014 May 26.
Ref 152 Let-7 g microRNA sensitizes fluorouracil-resistant human hepatoma cells. Pharmazie. 2014 Apr;69(4):287-92.
Ref 153 MicroRNA-101 inhibits human hepatocellular carcinoma progression through EZH2 downregulation and increased cytostatic drug sensitivity. J Hepatol. 2014 Mar;60(3):590-8. doi: 10.1016/j.jhep.2013.10.028. Epub 2013 Nov 6.
Ref 154 Direct targeting of SUZ12/ROCK2 by miR-200b/c inhibits cholangiocarcinoma tumourigenesis and metastasis. Br J Cancer. 2013 Dec 10;109(12):3092-104. doi: 10.1038/bjc.2013.655. Epub 2013 Oct 29.
Ref 155 DNA methylation-regulated miR-193a-3p dictates resistance of hepatocellular carcinoma to 5-fluorouracil via repression of SRSF2 expression. J Biol Chem. 2012 Feb 17;287(8):5639-49. doi: 10.1074/jbc.M111.291229. Epub 2011 Nov 23.
Ref 156 miRNA-195 sensitizes human hepatocellular carcinoma cells to 5-FU by targeting BCL-w. Oncol Rep. 2012 Jan;27(1):250-7. doi: 10.3892/or.2011.1472. Epub 2011 Sep 22.
Ref 157 Inhibition of MALAT1 sensitizes liver cancer cells to 5-flurouracil by regulating apoptosis through IKKAlpha/NF-kB pathway. Biochem Biophys Res Commun. 2018 Jun 18;501(1):33-40. doi: 10.1016/j.bbrc.2018.04.116. Epub 2018 May 7.
Ref 158 miRNA-296-3p modulates chemosensitivity of lung cancer cells by targeting CX3CR1. Am J Transl Res. 2016 Apr 15;8(4):1848-56. eCollection 2016.
Ref 159 Long noncoding RNA LINP1 acts as an oncogene and promotes chemoresistance in breast cancer. Cancer Biol Ther. 2018 Feb 1;19(2):120-131. doi: 10.1080/15384047.2017.1394543. Epub 2018 Jan 2.
Ref 160 Methylation-regulated miR-149 modulates chemoresistance by targeting GlcNAc N-deacetylase/N-sulfotransferase-1 in human breast cancer. FEBS J. 2014 Oct;281(20):4718-30. doi: 10.1111/febs.13012. Epub 2014 Sep 30.
Ref 161 Decreased expression of miR-204 is associated with poor prognosis in patients with breast cancer. Int J Clin Exp Pathol. 2014 May 15;7(6):3287-92. eCollection 2014.
Ref 162 miR-125b regulates side population in breast cancer and confers a chemoresistant phenotype. J Cell Biochem. 2013 Oct;114(10):2248-57. doi: 10.1002/jcb.24574.
Ref 163 A novel long non-coding RNA-PRLB acts as a tumor promoter through regulating miR-4766-5p/SIRT1 axis in breast cancer. Cell Death Dis. 2018 May 1;9(5):563. doi: 10.1038/s41419-018-0582-1.
Ref 164 A methylation-based regulatory network for microRNA 320a in chemoresistant breast cancer. Mol Pharmacol. 2014 Nov;86(5):536-47. doi: 10.1124/mol.114.092759. Epub 2014 Aug 26.
Ref 165 A Bmi1-miRNAs cross-talk modulates chemotherapy response to 5-fluorouracil in breast cancer cells. PLoS One. 2013 Sep 9;8(9):e73268. doi: 10.1371/journal.pone.0073268. eCollection 2013.
Ref 166 [Role of miR-206/CDK4 in modulating the growth and chemotlerapy sensitivity of ovarian cancer cells]. Nan Fang Yi Ke Da Xue Xue Bao. 2017 Mar 20;37(3):393-397. doi: 10.3969/j.issn.1673-4254.2017.03.20.
Ref 167 MiR-138 and MiR-135 directly target focal adhesion kinase, inhibit cell invasion, and increase sensitivity to chemotherapy in cancer cells. Anticancer Agents Med Chem. 2014 Jan;14(1):18-28. doi: 10.2174/187152061401140108113435.
Ref 168 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.
Ref 169 miR-381, a novel intrinsic WEE1 inhibitor, sensitizes renal cancer cells to 5-FU by up-regulation of Cdc2 activities in 786-O. J Chemother. 2013 Aug;25(4):229-38. doi: 10.1179/1973947813Y.0000000092. Epub 2013 May 7.
Ref 170 Different levels of let-7d expression modulate response of FaDu cells to irradiation and chemotherapeutics. PLoS One. 2017 Jun 30;12(6):e0180265. doi: 10.1371/journal.pone.0180265. eCollection 2017.
Ref 171 miRNA-24-3p promotes cell proliferation and regulates chemosensitivity in head and neck squamous cell carcinoma by targeting CHD5. Future Oncol. 2016 Dec;12(23):2701-2712. doi: 10.2217/fon-2016-0179. Epub 2016 Aug 11.

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