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]
[5]
Colorectal cancer [ICD-11: 2B91]
[6]
Colorectal peritoneal carcinomatosis [ICD-11: 2D91]
[7]
Esophageal cancer [ICD-11: 2B70]
[8]
Gastric cancer [ICD-11: 2B72]
[9]
Kidney cancer [ICD-11: 2C90]
[10]
Liver cancer [ICD-11: 2C12]
[11]
Metastatic colorectal cancer [ICD-11: 2D85]
[13]
Neuroendocrine carcinoma [ICD-11: 2D4Y]
[14]
Pancreatic cancer [ICD-11: 2C10]
[15]
Disease(s) with Resistance Information Discovered by Cell Line Test for This Drug (13 diseases)
Melanoma [ICD-11: 2C30]
[12]
Breast cancer [ICD-11: 2C60]
[16]
Chronic myeloid leukemia [ICD-11: 2A20]
[17]
Colon cancer [ICD-11: 2B90]
[18]
Colorectal cancer [ICD-11: 2B91]
[1], [2]
Esophageal cancer [ICD-11: 2B70]
[19]
Gastric cancer [ICD-11: 2B72]
[20]
Liver cancer [ICD-11: 2C12]
[21]
Lung cancer [ICD-11: 2C25]
[22]
Oral squamous cell carcinoma [ICD-11: 2B6E]
[23]
Pancreatic cancer [ICD-11: 2C10]
[24]
Prostate cancer [ICD-11: 2C82]
[17]
Salivary gland carcinoma [ICD-11: 2E60]
[25]
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
  ADTT: Aberration of the Drug's Therapeutic Target
  EADR: Epigenetic Alteration of DNA, RNA or Protein
  IDUE: Irregularity in Drug Uptake and Drug Efflux
  MRAP: Metabolic Reprogramming via Altered Pathways
  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
Colorectal cancer [ICD-11: 2B91]
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
  Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Mast/stem cell growth factor receptor Kit (KIT) [26]
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colorectal cancer [ICD-11: 2B91]
The Specified Disease Colorectal cancer
The Studied Tissue Blood
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 9.75E-30
Fold-change: -3.66E-01
Z-score: -1.26E+01
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 blot 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: Neurogenic locus notch homolog protein 1 (NOTCH1) [27]
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colorectal cancer [ICD-11: 2B91]
The Specified Disease Colorectal cancer
The Studied Tissue Blood
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 3.67E-06
Fold-change: -9.64E-02
Z-score: -4.72E+00
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: Neurogenic locus notch homolog protein 1 (NOTCH1) [29]
Sensitive Disease Colorectal carcinoma [ICD-11: 2B91.3]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colorectal cancer [ICD-11: 2B91]
The Specified Disease Colorectal carcinoma
The Studied Tissue Blood
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 3.67E-06
Fold-change: -9.64E-02
Z-score: -4.72E+00
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: Cadherin-1 (CDH1) [30]
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colorectal cancer [ICD-11: 2B91]
The Specified Disease Colorectal cancer
The Studied Tissue Blood
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 8.45E-01
Fold-change: 6.54E-03
Z-score: 1.95E-01
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.
  Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: AT-rich interactive domain-containing protein 4B (ARID4B) [28]
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colorectal cancer [ICD-11: 2B91]
The Specified Disease Colorectal cancer
The Studied Tissue Blood
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 2.69E-05
Fold-change: -1.63E-01
Z-score: -4.24E+00
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.
Kidney cancer [ICD-11: 2C90]
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
  Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Metalloproteinase inhibitor 3 (TIMP3) [31]
Sensitive Disease Renal carcinoma [ICD-11: 2C90.2]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Kidney cancer [ICD-11: 2C90]
The Specified Disease Renal carcinoma
The Studied Tissue Blood
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 1.59E-02
Fold-change: 2.68E+00
Z-score: 3.16E+00
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) [36]
Sensitive Disease Kidney cancer [ICD-11: 2C90.1]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Kidney cancer [ICD-11: 2C90]
The Specified Disease Kidney cancer
The Studied Tissue Kidney
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 1.32E-08
Fold-change: -6.91E-01
Z-score: -6.01E+00
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 blot 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.
Key Molecule: Programmed cell death protein 4 (PDCD4) [31]
Sensitive Disease Renal carcinoma [ICD-11: 2C90.2]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Kidney cancer [ICD-11: 2C90]
The Specified Disease Renal carcinoma
The Studied Tissue Kidney
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 8.62E-01
Fold-change: 6.66E-03
Z-score: 1.74E-01
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.
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) [10]
Resistant Disease Kidney cancer [ICD-11: 2C90.1]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Kidney cancer [ICD-11: 2C90]
The Specified Disease Kidney cancer
The Studied Tissue Kidney
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 8.07E-35
Fold-change: 6.59E-01
Z-score: 1.61E+01
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.
Liver cancer [ICD-11: 2C12]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
  Metabolic Reprogramming via Altered Pathways (MRAP) Click to Show/Hide
Key Molecule: Proprotein convertase subtilisin/kexin type 9 (PCSK9) [32]
Metabolic Type Lipid metabolism
Resistant Disease Hepatocellular carcinoma [ICD-11: 2C12.02]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Liver cancer [ICD-11: 2C12]
The Specified Disease Hepatocellular carcinoma
The Studied Tissue Liver tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 5.96E-09
Fold-change: 4.92E-01
Z-score: 5.99E+00
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cholesterol metabolism Activation hsa04979
In Vivo Model Hepa1-6 hepatocellular carcinoma transplanted tumor model mice Mice
Experiment for
Molecule Alteration
Western blot analysis and immunohistochemical assays
Experiment for
Drug Resistance
Tumor volume assay
Mechanism Description ARBU significantly inhibited the proliferation of Hepa1-6 in vivo and in vitro, regulated cholesterol metabolism, and promoted the M1-type polarization of macrophages in the tumor microenvironment. ARBU inhibits cholesterol synthesis in the TME through the PCSK9/LDL-R signaling pathway, thereby blocking macrophage M2 polarization, promoting apoptosis of the tumor cells, and inhibiting their proliferation and migration.
Key Molecule: Proprotein convertase subtilisin/kexin type 9 (PCSK9) [32]
Metabolic Type Lipid metabolism
Resistant Disease Hepatocellular carcinoma [ICD-11: 2C12.02]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Liver cancer [ICD-11: 2C12]
The Specified Disease Hepatocellular carcinoma
The Studied Tissue Liver tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 5.96E-09
Fold-change: 4.92E-01
Z-score: 5.99E+00
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cholesterol metabolism Activation hsa04979
In Vitro Model Hepa1-6 cells Liver Mus musculus (Mouse) CVCL_0327
Experiment for
Molecule Alteration
Western blot analysis and immunohistochemical assays
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description ARBU significantly inhibited the proliferation of Hepa1-6 in vivo and in vitro, regulated cholesterol metabolism, and promoted the M1-type polarization of macrophages in the tumor microenvironment. ARBU inhibits cholesterol synthesis in the TME through the PCSK9/LDL-R signaling pathway, thereby blocking macrophage M2 polarization, promoting apoptosis of the tumor cells, and inhibiting their proliferation and migration.
  Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Ubiquitin carboxyl-terminal hydrolase 22 (USP22) [41]
Resistant Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Liver cancer [ICD-11: 2C12]
The Specified Disease Hepatocellular carcinoma
The Studied Tissue Liver tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 1.48E-08
Fold-change: 3.19E-01
Z-score: 5.81E+00
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: High mobility group protein HMGI-C (HMGA2) [43]
Resistant Disease Liver cancer [ICD-11: 2C12.6]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Liver cancer [ICD-11: 2C12]
The Specified Disease Liver cancer
The Studied Tissue Liver tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 2.13E-05
Fold-change: 1.02E+00
Z-score: 4.33E+00
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: Nuclear receptor subfamily 2 group C2 (NR2C2) [44]
Resistant Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Liver cancer [ICD-11: 2C12]
The Specified Disease Liver cancer
The Studied Tissue Liver tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 2.03E-05
Fold-change: 7.29E-02
Z-score: 4.71E+00
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: Eukaryotic translation initiation factor 4E (EIF4E) [58]
Resistant Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Liver cancer [ICD-11: 2C12]
The Specified Disease Liver cancer
The Studied Tissue Liver tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 3.85E-01
Fold-change: -1.87E-02
Z-score: -8.76E-01
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: Dual specificity protein phosphatase 6 (DUSP6) [65]
Resistant Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Liver cancer [ICD-11: 2C12]
The Specified Disease Liver cancer
The Studied Tissue Liver tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 5.96E-02
Fold-change: -3.20E-02
Z-score: -1.92E+00
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: Phosphatase and tensin homolog (PTEN) [11]
Resistant Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Liver cancer [ICD-11: 2C12]
The Specified Disease Liver cancer
The Studied Tissue Liver tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 1.86E-04
Fold-change: -4.60E-02
Z-score: -3.95E+00
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 blot 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.
  Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: Small nucleolar RNA host gene 6 (SNHG6) [44]
Resistant Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Liver cancer [ICD-11: 2C12]
The Specified Disease Liver hepatocellular carcinoma
The Studied Tissue Liver
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 9.03E-20
Fold-change: 1.59E+00
Z-score: 9.61E+00
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.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
  Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Bcl-2-associated agonist of cell death (BAD) [39], [40]
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Liver cancer [ICD-11: 2C12]
The Specified Disease Hepatocellular carcinoma
The Studied Tissue Liver tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 3.19E-09
Fold-change: -2.62E-01
Z-score: -6.15E+00
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: Kelch-like ECH-associated protein 1 (KEAP1) [47]
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Liver cancer [ICD-11: 2C12]
The Specified Disease Liver cancer
The Studied Tissue Liver tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 1.51E-03
Fold-change: 3.81E-02
Z-score: 3.34E+00
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: High mobility group protein HMGI-C (HMGA2) [59]
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Liver cancer [ICD-11: 2C12]
The Specified Disease Liver cancer
The Studied Tissue Liver tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 3.56E-02
Fold-change: -2.08E-02
Z-score: -2.15E+00
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: Bcl-2-like protein 2 (BCL2L2) [64]
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Liver cancer [ICD-11: 2C12]
The Specified Disease Liver cancer
The Studied Tissue Liver tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 9.79E-03
Fold-change: -3.17E-02
Z-score: -2.69E+00
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 blot 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.
Key Molecule: Suppressor of cytokine signaling 6 (SOCS6) [67]
Sensitive Disease Hepatocellular cancer [ICD-11: 2C12.4]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Liver cancer [ICD-11: 2C12]
The Specified Disease Liver cancer
The Studied Tissue Liver tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 3.24E-04
Fold-change: -7.85E-02
Z-score: -3.85E+00
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 N.A.
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.
  Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) [52]
Sensitive Disease Liver cancer [ICD-11: 2C12.6]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Liver cancer [ICD-11: 2C12]
The Specified Disease Cholangiocarcinoma
The Studied Tissue Bile duct
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 1.43E-07
Fold-change: 2.16E+00
Z-score: 6.36E+00
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.
Lung cancer [ICD-11: 2C25]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
  Metabolic Reprogramming via Altered Pathways (MRAP) Click to Show/Hide
Key Molecule: Integrin beta-5 (ITGB5) [33]
Metabolic Type Redox metabolism
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Lung cancer [ICD-11: 2C25]
The Specified Disease Lung adenocarcinoma
The Studied Tissue Lung tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 3.87E-14
Fold-change: 4.20E-01
Z-score: 8.33E+00
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A5419 cells Lung Homo sapiens (Human) N.A.
LLC cells Lung Homo sapiens (Human) CVCL_A9AW
Experiment for
Molecule Alteration
qRT-PCR; Western blot analysis
Experiment for
Drug Resistance
IC50 assay
Mechanism Description Mechanistically, our proteomic analysis reveals a consistent up-regulation of sphingolipid metabolic enzyme ASAH2 and beta5-integrin expression in GemR pancreatic and lung cancer cells as well as stable beta5-integrin-expressing cells.
Pancreatic cancer [ICD-11: 2C10]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
  Metabolic Reprogramming via Altered Pathways (MRAP) Click to Show/Hide
Key Molecule: Glucose-6-phosphate dehydrogenase (G6PD) [34]
Metabolic Type Glucose metabolism
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Pancreatic cancer [ICD-11: 2C10]
The Specified Disease Pancreatic ductal adenocarcinoma
The Studied Tissue Pancreas
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 3.01E-17
Fold-change: 4.09E-01
Z-score: 9.54E+00
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Adrenergic signaling in cardiomyocytes Activation hsa04261
In Vivo Model Female SCID mice of 6-week-old, with fresh tissue from patient Mice
Experiment for
Molecule Alteration
qRT-PCR; Western blot analysis
Experiment for
Drug Resistance
Tumor volume assay
Mechanism Description Glucomet-PDACs are more resistant to chemotherapy than lipomet-PDACs, and patients with glucomet-PDAC have a worse prognosis. Integrated analyses reveal that the GLUT1/aldolase B (ALDOB)/glucose-6-phosphate dehydrogenase (G6PD) axis induces chemotherapy resistance by remodeling glucose metabolism in glucomet-PDAC. Increased glycolytic flux, G6PD activity, and pyrimidine biosynthesis are identified in glucomet-PDAC with high GLUT1 and low ALDOB expression, and these phenotypes could be reversed by inhibiting GLUT1 expression or by increasing ALDOB expression.
Key Molecule: Solute carrier family 2 member 1 (SLC2A1) [34]
Metabolic Type Glucose metabolism
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Pancreatic cancer [ICD-11: 2C10]
The Specified Disease Pancreatic ductal adenocarcinoma
The Studied Tissue Pancreas
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 2.33E-08
Fold-change: 5.47E-01
Z-score: 5.83E+00
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Adrenergic signaling in cardiomyocytes Activation hsa04261
In Vivo Model Female SCID mice of 4-week-old, with fresh tissue from patient Mice
Experiment for
Molecule Alteration
qRT-PCR; Western blot analysis
Experiment for
Drug Resistance
Tumor volume assay
Mechanism Description Glucomet-PDACs are more resistant to chemotherapy than lipomet-PDACs, and patients with glucomet-PDAC have a worse prognosis. Integrated analyses reveal that the GLUT1/aldolase B (ALDOB)/glucose-6-phosphate dehydrogenase (G6PD) axis induces chemotherapy resistance by remodeling glucose metabolism in glucomet-PDAC. Increased glycolytic flux, G6PD activity, and pyrimidine biosynthesis are identified in glucomet-PDAC with high GLUT1 and low ALDOB expression, and these phenotypes could be reversed by inhibiting GLUT1 expression or by increasing ALDOB expression.
Key Molecule: N-acylsphingosine amidohydrolase 2 (ASAH2) [33]
Metabolic Type Redox metabolism
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Pancreatic cancer [ICD-11: 2C10]
The Specified Disease Pancreatic ductal adenocarcinoma
The Studied Tissue Pancreas
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 3.44E-01
Fold-change: 1.83E-01
Z-score: 9.68E-01
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model Panc1 cells Pancreas Homo sapiens (Human) CVCL_0480
TB32048 cells N.A. Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qRT-PCR; Western blot analysis
Experiment for
Drug Resistance
IC50 assay
Mechanism Description Mechanistically, our proteomic analysis reveals a consistent up-regulation of sphingolipid metabolic enzyme ASAH2 and beta5-integrin expression in GemR pancreatic and lung cancer cells as well as stable beta5-integrin-expressing cells.
Key Molecule: Integrin beta-5 (ITGB5) [33]
Metabolic Type Redox metabolism
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Pancreatic cancer [ICD-11: 2C10]
The Specified Disease Pancreatic ductal adenocarcinoma
The Studied Tissue Pancreas
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 1.91E-26
Fold-change: 7.66E-01
Z-score: 1.30E+01
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model Panc1 cells Pancreas Homo sapiens (Human) CVCL_0480
TB32048 cells N.A. Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qRT-PCR; Western blot analysis
Experiment for
Drug Resistance
IC50 assay
Mechanism Description Mechanistically, our proteomic analysis reveals a consistent up-regulation of sphingolipid metabolic enzyme ASAH2 and beta5-integrin expression in GemR pancreatic and lung cancer cells as well as stable beta5-integrin-expressing cells.
Key Molecule: Glucose-6-phosphate dehydrogenase (G6PD) [34]
Metabolic Type Glucose metabolism
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Pancreatic cancer [ICD-11: 2C10]
The Specified Disease Pancreatic ductal adenocarcinoma
The Studied Tissue Pancreas
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 3.01E-17
Fold-change: 4.09E-01
Z-score: 9.54E+00
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Adrenergic signaling in cardiomyocytes Activation hsa04261
In Vivo Model HCC patients Homo Sapiens
Experiment for
Molecule Alteration
qRT-PCR; Western blot analysis
Mechanism Description Glucomet-PDACs are more resistant to chemotherapy than lipomet-PDACs, and patients with glucomet-PDAC have a worse prognosis. Integrated analyses reveal that the GLUT1/aldolase B (ALDOB)/glucose-6-phosphate dehydrogenase (G6PD) axis induces chemotherapy resistance by remodeling glucose metabolism in glucomet-PDAC. Increased glycolytic flux, G6PD activity, and pyrimidine biosynthesis are identified in glucomet-PDAC with high GLUT1 and low ALDOB expression, and these phenotypes could be reversed by inhibiting GLUT1 expression or by increasing ALDOB expression.
Key Molecule: Solute carrier family 2 member 1 (SLC2A1) [34]
Metabolic Type Glucose metabolism
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Pancreatic cancer [ICD-11: 2C10]
The Specified Disease Pancreatic ductal adenocarcinoma
The Studied Tissue Pancreas
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 2.33E-08
Fold-change: 5.47E-01
Z-score: 5.83E+00
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Adrenergic signaling in cardiomyocytes Activation hsa04261
In Vivo Model HCC patients Homo Sapiens
Experiment for
Molecule Alteration
qRT-PCR; Western blot analysis
Mechanism Description Glucomet-PDACs are more resistant to chemotherapy than lipomet-PDACs, and patients with glucomet-PDAC have a worse prognosis. Integrated analyses reveal that the GLUT1/aldolase B (ALDOB)/glucose-6-phosphate dehydrogenase (G6PD) axis induces chemotherapy resistance by remodeling glucose metabolism in glucomet-PDAC. Increased glycolytic flux, G6PD activity, and pyrimidine biosynthesis are identified in glucomet-PDAC with high GLUT1 and low ALDOB expression, and these phenotypes could be reversed by inhibiting GLUT1 expression or by increasing ALDOB expression.
Key Molecule: Glucose-6-phosphate dehydrogenase (G6PD) [34]
Metabolic Type Glucose metabolism
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Pancreatic cancer [ICD-11: 2C10]
The Specified Disease Pancreatic ductal adenocarcinoma
The Studied Tissue Pancreas
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 3.01E-17
Fold-change: 4.09E-01
Z-score: 9.54E+00
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Adrenergic signaling in cardiomyocytes Activation hsa04261
In Vivo Model HCC patients Homo Sapiens
Experiment for
Molecule Alteration
qRT-PCR; Western blot analysis
Mechanism Description Glucomet-PDACs are more resistant to chemotherapy than lipomet-PDACs, and patients with glucomet-PDAC have a worse prognosis. Integrated analyses reveal that the GLUT1/aldolase B (ALDOB)/glucose-6-phosphate dehydrogenase (G6PD) axis induces chemotherapy resistance by remodeling glucose metabolism in glucomet-PDAC. Increased glycolytic flux, G6PD activity, and pyrimidine biosynthesis are identified in glucomet-PDAC with high GLUT1 and low ALDOB expression, and these phenotypes could be reversed by inhibiting GLUT1 expression or by increasing ALDOB expression.
Key Molecule: Solute carrier family 2 member 1 (SLC2A1) [34]
Metabolic Type Glucose metabolism
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Pancreatic cancer [ICD-11: 2C10]
The Specified Disease Pancreatic ductal adenocarcinoma
The Studied Tissue Pancreas
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 2.33E-08
Fold-change: 5.47E-01
Z-score: 5.83E+00
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Adrenergic signaling in cardiomyocytes Activation hsa04261
In Vivo Model HCC patients Homo Sapiens
Experiment for
Molecule Alteration
qRT-PCR; Western blot analysis
Mechanism Description Glucomet-PDACs are more resistant to chemotherapy than lipomet-PDACs, and patients with glucomet-PDAC have a worse prognosis. Integrated analyses reveal that the GLUT1/aldolase B (ALDOB)/glucose-6-phosphate dehydrogenase (G6PD) axis induces chemotherapy resistance by remodeling glucose metabolism in glucomet-PDAC. Increased glycolytic flux, G6PD activity, and pyrimidine biosynthesis are identified in glucomet-PDAC with high GLUT1 and low ALDOB expression, and these phenotypes could be reversed by inhibiting GLUT1 expression or by increasing ALDOB expression.
  Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Protein salvador homolog 1 (SAV1) [38]
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Pancreatic cancer [ICD-11: 2C10]
The Specified Disease Pancreatic cancer
The Studied Tissue Pancreas
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 7.75E-14
Fold-change: -5.70E-01
Z-score: -8.32E+00
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Hippo signaling pathway Regulation N.A.
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: Programmed cell death protein 4 (PDCD4) [70], [71]
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Pancreatic cancer [ICD-11: 2C10]
The Specified Disease Pancreatic cancer
The Studied Tissue Pancreas
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 1.56E-02
Fold-change: -2.17E-01
Z-score: -2.68E+00
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 N.A.
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.
  Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: Urothelial cancer associated 1 (UCA1) [45]
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Pancreatic cancer [ICD-11: 2C10]
The Specified Disease Pancreatic adenocarcinoma
The Studied Tissue Pancreas
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 2.90E-08
Fold-change: 5.54E+00
Z-score: 5.81E+00
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: Growth arrest specific 5 (GAS5) [72]
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Pancreatic cancer [ICD-11: 2C10]
The Specified Disease Pancreatic adenocarcinoma
The Studied Tissue Pancreas
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 6.05E-15
Fold-change: -8.27E-01
Z-score: -8.25E+00
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: DiGeorge syndrome critical region gene 5 (DGCR5) [15]
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Pancreatic cancer [ICD-11: 2C10]
The Specified Disease Pancreatic adenocarcinoma
The Studied Tissue Pancreas
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 7.83E-10
Fold-change: -1.00E+00
Z-score: -6.33E+00
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.
Colon cancer [ICD-11: 2B90]
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
  Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Histone deacetylase 4 (HDAC4) [35]
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colon cancer [ICD-11: 2B90]
The Specified Disease Colon cancer
The Studied Tissue Colon tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 9.80E-14
Fold-change: -3.79E-01
Z-score: -8.17E+00
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 blot 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.
Key Molecule: Dihydropyrimidine dehydrogenase [NADP(+)] [61]
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colon cancer [ICD-11: 2B90]
The Specified Disease Colon cancer
The Studied Tissue Colon tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 1.29E-04
Fold-change: -2.76E-02
Z-score: -3.90E+00
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation 5-Fu catabolic signaling pathway Regulation N.A.
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: Heat shock protein beta-1 (HSPB1) [62]
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colon cancer [ICD-11: 2B90]
The Specified Disease Colon cancer
The Studied Tissue Colon tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 1.98E-05
Fold-change: -2.80E-02
Z-score: -4.32E+00
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: Insulin-like growth factor 1 receptor (IGF1R) [63]
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colon cancer [ICD-11: 2B90]
The Specified Disease Colon cancer
The Studied Tissue Colon tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 5.32E-03
Fold-change: -3.07E-02
Z-score: -2.81E+00
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: Prominin-1 (PROM1) [68]
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colon cancer [ICD-11: 2B90]
The Specified Disease Colon cancer
The Studied Tissue Colon tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 4.04E-12
Fold-change: -9.18E-02
Z-score: -7.18E+00
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 blot 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.
  Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: E3 ubiquitin-protein ligase XIAP (XIAP) [42]
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colon cancer [ICD-11: 2B90]
The Specified Disease Colon cancer
The Studied Tissue Colon tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 3.85E-04
Fold-change: 1.37E-01
Z-score: 3.61E+00
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.
  Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: ATP-binding cassette sub-family G2 (ABCG2) [68]
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colon cancer [ICD-11: 2B90]
The Specified Disease Colon cancer
The Studied Tissue Colon tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 1.83E-95
Fold-change: -9.70E-01
Z-score: -4.32E+01
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 blot 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.
Drug Resistance Data Categorized by Their Corresponding Mechanisms
  Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Programmed cell death protein 4 (PDCD4) [37]
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colon cancer [ICD-11: 2B90]
The Specified Disease Colon cancer
The Studied Tissue Colon tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 1.69E-35
Fold-change: -7.24E-01
Z-score: -1.54E+01
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
PI3K/AKT signaling pathway Regulation N.A.
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: Serine/threonine-protein kinase Chk1 (CHK1) [49]
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colon cancer [ICD-11: 2B90]
The Specified Disease Colon cancer
The Studied Tissue Colon tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 6.28E-72
Fold-change: 2.53E-01
Z-score: 2.53E+01
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: Transcription factor E2F3 (E2F3) [5]
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colon cancer [ICD-11: 2B90]
The Specified Disease Colon cancer
The Studied Tissue Colon tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 1.02E-82
Fold-change: 1.51E-01
Z-score: 2.75E+01
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 blot 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) [5]
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colon cancer [ICD-11: 2B90]
The Specified Disease Colon cancer
The Studied Tissue Colon tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 2.34E-02
Fold-change: 1.46E-02
Z-score: 2.28E+00
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 blot 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: Wee1-like protein kinase (WEE1) [49]
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colon cancer [ICD-11: 2B90]
The Specified Disease Colon cancer
The Studied Tissue Colon tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 4.92E-41
Fold-change: 1.05E-01
Z-score: 1.63E+01
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: DNA-binding factor KBF1 (p105) (NFKB1) [42]
Resistant Disease Colon cancer [ICD-11: 2B90.1]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Colon cancer [ICD-11: 2B90]
The Specified Disease Colon cancer
The Studied Tissue Colon tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 3.70E-46
Fold-change: -7.40E-02
Z-score: -1.70E+01
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.
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: Urothelial cancer associated 1 (UCA1) [46]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Gastric cancer [ICD-11: 2B72]
The Specified Disease Stomach adenocarcinoma
The Studied Tissue Stomach
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 7.51E-08
Fold-change: 5.46E+00
Z-score: 5.48E+00
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: Pvt1 oncogene (PVT1) [82]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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) [83]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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) [9]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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 [9]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
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 [53]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
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 [46]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
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 [84]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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 [20]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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 [20]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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) [50]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Gastric cancer [ICD-11: 2B72]
The Specified Disease Gastric cancer
The Studied Tissue Gastric tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 5.13E-01
Fold-change: 2.49E-02
Z-score: 7.76E-01
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 [50]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
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) [50]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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: Extracellular matrix receptor III (CD44) [53]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Gastric cancer [ICD-11: 2B72]
The Specified Disease Gastric cancer
The Studied Tissue Gastric tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 2.62E-01
Fold-change: 1.66E-01
Z-score: 1.54E+00
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: Apoptosis regulator Bcl-2 (BCL2) [82]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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: F-box/WD repeat-containing protein 7 (FBXW7) [84]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
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) [20]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
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 blot 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.
Key Molecule: Serine/threonine-protein kinase mTOR (mTOR) [86]
Resistant Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Molecule Alteration Expression
Down-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT/mTOR signaling pathway Inhibition hsa04151
In Vitro Model MKN-45/R cells Gastric Homo sapiens (Human) N.A.
MKN-74/R cells Gastric Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot assay; qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The PI3K/Akt/mTOR signaling pathway was activated in drug-resistant GC cells and tumor tissues of patients refractory to 5-FU chemotherapy, as evidenced by high PI3K, Akt, and mTOR levels in MKN-45/R, MKN-74/R, and GC tissues resistant to 5-FU. Silencing of the PI3K/Akt/mTOR signaling pathway suppressed the 5-FU resistance of GC cells.
Key Molecule: Serine/threonine-protein kinase mTOR (mTOR) [86]
Resistant Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Molecule Alteration Phosphorylation
Down-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT/mTOR signaling pathway Inhibition hsa04151
In Vitro Model MKN-45/R cells Gastric Homo sapiens (Human) N.A.
MKN-74/R cells Gastric Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot assay; qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The PI3K/Akt/mTOR signaling pathway was activated in drug-resistant GC cells and tumor tissues of patients refractory to 5-FU chemotherapy, as evidenced by high PI3K, Akt, and mTOR levels in MKN-45/R, MKN-74/R, and GC tissues resistant to 5-FU. Silencing of the PI3K/Akt/mTOR signaling pathway suppressed the 5-FU resistance of GC cells.
Key Molecule: Phosphatidylinositol 3-kinase (PI3K) [86]
Resistant Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Molecule Alteration Expression
Down-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT/mTOR signaling pathway Inhibition hsa04151
In Vitro Model MKN-45/R cells Gastric Homo sapiens (Human) N.A.
MKN-74/R cells Gastric Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot assay; qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The PI3K/Akt/mTOR signaling pathway was activated in drug-resistant GC cells and tumor tissues of patients refractory to 5-FU chemotherapy, as evidenced by high PI3K, Akt, and mTOR levels in MKN-45/R, MKN-74/R, and GC tissues resistant to 5-FU. Silencing of the PI3K/Akt/mTOR signaling pathway suppressed the 5-FU resistance of GC cells.
Key Molecule: Phosphatidylinositol 3-kinase (PI3K) [86]
Resistant Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Molecule Alteration Phosphorylation
Down-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT/mTOR signaling pathway Inhibition hsa04151
In Vitro Model MKN-45/R cells Gastric Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot assay; qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The PI3K/Akt/mTOR signaling pathway was activated in drug-resistant GC cells and tumor tissues of patients refractory to 5-FU chemotherapy, as evidenced by high PI3K, Akt, and mTOR levels in MKN-45/R, MKN-74/R, and GC tissues resistant to 5-FU. Silencing of the PI3K/Akt/mTOR signaling pathway suppressed the 5-FU resistance of GC cells.
Key Molecule: Phosphatidylinositol 3-kinase (PI3K) [86]
Resistant Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Molecule Alteration Phosphorylation
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT/mTOR signaling pathway Inhibition hsa04151
In Vitro Model MKN-74/R cells Gastric Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot assay; qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The PI3K/Akt/mTOR signaling pathway was activated in drug-resistant GC cells and tumor tissues of patients refractory to 5-FU chemotherapy, as evidenced by high PI3K, Akt, and mTOR levels in MKN-45/R, MKN-74/R, and GC tissues resistant to 5-FU. Silencing of the PI3K/Akt/mTOR signaling pathway suppressed the 5-FU resistance of GC cells.
Key Molecule: AKT serine/threonine kinase (AKT) [86]
Resistant Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT/mTOR signaling pathway Inhibition hsa04151
In Vitro Model MKN-45/R cells Gastric Homo sapiens (Human) N.A.
MKN-74/R cells Gastric Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot assay; qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The PI3K/Akt/mTOR signaling pathway was activated in drug-resistant GC cells and tumor tissues of patients refractory to 5-FU chemotherapy, as evidenced by high PI3K, Akt, and mTOR levels in MKN-45/R, MKN-74/R, and GC tissues resistant to 5-FU. Silencing of the PI3K/Akt/mTOR signaling pathway suppressed the 5-FU resistance of GC cells.
Key Molecule: AKT serine/threonine kinase (AKT) [86]
Resistant Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Molecule Alteration Phosphorylation
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT/mTOR signaling pathway Inhibition hsa04151
In Vitro Model MKN-45/R cells Gastric Homo sapiens (Human) N.A.
MKN-74/R cells Gastric Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot assay; qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The PI3K/Akt/mTOR signaling pathway was activated in drug-resistant GC cells and tumor tissues of patients refractory to 5-FU chemotherapy, as evidenced by high PI3K, Akt, and mTOR levels in MKN-45/R, MKN-74/R, and GC tissues resistant to 5-FU. Silencing of the PI3K/Akt/mTOR signaling pathway suppressed the 5-FU resistance of GC cells.
  Metabolic Reprogramming via Altered Pathways (MRAP) Click to Show/Hide
Key Molecule: Dihydroorotate dehydrogenase (DHODH) [85]
Metabolic Type Nucleic acid metabolism
Resistant Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model AGS cells Gastric Homo sapiens (Human) CVCL_0139
HGC27 cells Gastric Homo sapiens (Human) CVCL_1279
Experiment for
Molecule Alteration
qRT-PCR; Western blot analysis
Experiment for
Drug Resistance
IC50 assay
Mechanism Description Mechanistically, pyrimidine biosynthesis augmented Notch signaling and transcriptionally increased c-Myc expression, leading to up-regulation of critical glycolytic enzymes. Further studies revealed that pyrimidine synthesis could stabilize gamma-secretase subunit Nicastrin at post-translational N-linked glycosylation level, thereby inducing the cleavage and activation of Notch. Besides, we found that up-regulation of the key enzymes for de novo pyrimidine synthesis CAD and DHODH conferred the chemotherapeutic resistance of gastric cancer via accelerating glycolysis, and pharmacologic inhibition of pyrimidine biosynthetic pathway sensitized cancer cells to chemotherapy in vitro and in vivo.
Key Molecule: Carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase (CAD) [85]
Metabolic Type Nucleic acid metabolism
Resistant Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model AGS cells Gastric Homo sapiens (Human) CVCL_0139
HGC27 cells Gastric Homo sapiens (Human) CVCL_1279
Experiment for
Molecule Alteration
qRT-PCR; Western blot analysis
Experiment for
Drug Resistance
IC50 assay
Mechanism Description Mechanistically, pyrimidine biosynthesis augmented Notch signaling and transcriptionally increased c-Myc expression, leading to up-regulation of critical glycolytic enzymes. Further studies revealed that pyrimidine synthesis could stabilize gamma-secretase subunit Nicastrin at post-translational N-linked glycosylation level, thereby inducing the cleavage and activation of Notch. Besides, we found that up-regulation of the key enzymes for de novo pyrimidine synthesis CAD and DHODH conferred the chemotherapeutic resistance of gastric cancer via accelerating glycolysis, and pharmacologic inhibition of pyrimidine biosynthetic pathway sensitized cancer cells to chemotherapy in vitro and in vivo.
Key Molecule: Pyruvate kinase muscle isozyme 1 (PKM1) [17]
Metabolic Type Mitochondrial metabolism
Resistant Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MKN45 cells Liver Homo sapiens (Human) CVCL_0434
MKN-45/F2R cells Stomach Homo sapiens (Human) CVCL_0434
NUGC3 cells Gastric Homo sapiens (Human) CVCL_1612
NUGC-3/5-FUR cells Stomach Homo sapiens (Human) CVCL_1612
Experiment for
Molecule Alteration
Expression profiles
Experiment for
Drug Resistance
Cell viability assay
Mechanism Description The overexpression of PKM1 resulted in resistance of the parental cells to 5-FU and oxaliplatin.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
  Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: Protein lin-28 homolog A (CSDD1) [54]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Gastric cancer [ICD-11: 2B72]
The Specified Disease Gastric cancer
The Studied Tissue Gastric tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 2.28E-02
Fold-change: 1.60E-01
Z-score: 4.12E+00
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
Lin28/miR107 pathway Regulation N.A.
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: HOX transcript antisense RNA (HOTAIR) [87]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
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 [88]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
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 [66]
Sensitive Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Molecule Alteration Expression
Up-regulation
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) [89]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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 [90]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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 [91]
Sensitive Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Molecule Alteration Expression
Up-regulation
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 [92]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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 [93]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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 [94]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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 [95]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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 B (CSDD2) [54]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
Lin28/miR107 pathway Regulation N.A.
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 [54]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
Lin28/miR107 pathway Regulation N.A.
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 [55]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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 [96]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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 [97]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation miR23b-3p/ATG12/HMGB2/autophagy regulatory loop signaling pathway Regulation N.A.
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 [98]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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 [99]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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 [100]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
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 [101]
Sensitive Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Molecule Alteration Expression
Up-regulation
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: ATP-binding cassette sub-family B5 (ABCB5) [55]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Gastric cancer [ICD-11: 2B72]
The Specified Disease Gastric cancer
The Studied Tissue Gastric tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 9.75E-01
Fold-change: -5.59E-04
Z-score: -3.48E-02
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: Solute carrier family 34 member 2 (SLC34A2) [90]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
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: Multidrug resistance protein 1 (ABCB1) [98]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
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 blot 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: TGF-beta receptor type II (TGFBR2) [56]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Gastric cancer [ICD-11: 2B72]
The Specified Disease Gastric cancer
The Studied Tissue Gastric tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 9.24E-01
Fold-change: -4.81E-03
Z-score: -1.07E-01
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-30a [102]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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) [103]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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 [103]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model 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: hsa-mir-204 [56]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Up-regulation
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) [104]
Sensitive Disease Gastric carcinoma [ICD-11: 2B72.Z]
Molecule Alteration Expression
Up-regulation
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: Transforming protein RhoA (RHOA) [66]
Sensitive Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Gastric cancer [ICD-11: 2B72]
The Specified Disease Gastric cancer
The Studied Tissue Gastric tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 2.78E-02
Fold-change: -4.29E-02
Z-score: -4.37E+00
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: Smoothened homolog (SMO) [55]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Gastric cancer [ICD-11: 2B72]
The Specified Disease Gastric cancer
The Studied Tissue Gastric tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 3.86E-01
Fold-change: -1.09E-01
Z-score: -1.10E+00
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: Phosphatase and tensin homolog (PTEN) [88]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
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: Transcription factor E2F6 (E2F6) [91]
Sensitive Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Molecule Alteration Expression
Down-regulation
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) [91]
Sensitive Disease Gastric adenocarcinoma [ICD-11: 2B72.0]
Molecule Alteration Expression
Down-regulation
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) [92]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Molecule Alteration Expression
Down-regulation
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).
Esophageal cancer [ICD-11: 2B70]
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: Long non-protein coding RNA 261 (LINC00261) [48]
Sensitive Disease Esophageal cancer [ICD-11: 2B70.1]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Esophageal cancer [ICD-11: 2B70]
The Specified Disease Esophageal carcinoma
The Studied Tissue Esophagus
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 9.38E-04
Fold-change: 2.73E+00
Z-score: 3.38E+00
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: Tumor suppressor candidate 7 (TUSC7) [51]
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Molecule Alteration Expression
Up-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Esophageal cancer [ICD-11: 2B70]
The Specified Disease Esophageal carcinoma
The Studied Tissue Esophagus
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 2.61E-01
Fold-change: 2.29E+00
Z-score: 1.16E+00
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Inhibition hsa05200
DESC1/EGFR/AKT signaling pathway Regulation N.A.
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-130a-3p [77]
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Molecule Alteration Expression
Down-regulation
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 [77]
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Molecule Alteration Expression
Up-regulation
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 [77]
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Molecule Alteration Expression
Down-regulation
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 [77]
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Molecule Alteration Expression
Up-regulation
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: hsa-mir-224 [51]
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Molecule Alteration Expression
Down-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Inhibition hsa05200
DESC1/EGFR/AKT signaling pathway Regulation N.A.
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 [78]
Sensitive Disease Esophageal cancer [ICD-11: 2B70.1]
Molecule Alteration Expression
Down-regulation
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 [79]
Sensitive Disease Esophageal adenocarcinoma [ICD-11: 2B70.2]
Molecule Alteration Expression
Up-regulation
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 [79]
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Molecule Alteration Expression
Up-regulation
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 [80]
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Molecule Alteration Expression
Down-regulation
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 [81]
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Molecule Alteration Expression
Down-regulation
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) [80], [81]
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Molecule Alteration Expression
Down-regulation
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 blot 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(+)] [48]
Sensitive Disease Esophageal cancer [ICD-11: 2B70.1]
Molecule Alteration Methylation
Down-regulation
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) [51]
Sensitive Disease Esophageal squamous cell carcinoma [ICD-11: 2B70.3]
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Inhibition hsa05200
DESC1/EGFR/AKT signaling pathway Regulation N.A.
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.
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 [76]
Resistant Disease Esophageal cancer [ICD-11: 2B70.1]
Molecule Alteration Expression
Up-regulation
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 [8]
Resistant Disease Esophageal adenocarcinoma [ICD-11: 2B70.2]
Molecule Alteration Expression
Up-regulation
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 [19]
Resistant Disease Esophageal cancer [ICD-11: 2B70.1]
Molecule Alteration Expression
Up-regulation
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 [19]
Resistant Disease Esophageal cancer [ICD-11: 2B70.1]
Molecule Alteration Expression
Up-regulation
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 [19]
Resistant Disease Esophageal cancer [ICD-11: 2B70.1]
Molecule Alteration Expression
Up-regulation
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) [76]
Resistant Disease Esophageal cancer [ICD-11: 2B70.1]
Molecule Alteration Expression
Down-regulation
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) [8]
Resistant Disease Esophageal adenocarcinoma [ICD-11: 2B70.2]
Molecule Alteration Expression
Down-regulation
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) [19]
Resistant Disease Esophageal cancer [ICD-11: 2B70.1]
Molecule Alteration Expression
Down-regulation
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.
Breast cancer [ICD-11: 2C60]
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: Caspase-9 (CASP9) [57]
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Breast cancer [ICD-11: 2C60]
The Specified Disease Breast cancer
The Studied Tissue Breast tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 1.78E-02
Fold-change: -1.19E-02
Z-score: -2.38E+00
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.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
  Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Nuclear factor of activated T-cells 3 (NFATC3) [60]
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Breast cancer [ICD-11: 2C60]
The Specified Disease Breast cancer
The Studied Tissue Breast tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 1.18E-08
Fold-change: -2.53E-02
Z-score: -5.81E+00
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
TRPC5 signaling pathway Regulation N.A.
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.
Oral squamous cell carcinoma [ICD-11: 2B6E]
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
  Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: ETS homologous factor (EHF) [69]
Sensitive Disease Oral squamous cell carcinoma [ICD-11: 2B6E.0]
Molecule Alteration Expression
Down-regulation
Differential expression of the molecule in resistant disease
Classification of Disease Oral squamous cell carcinoma [ICD-11: 2B6E]
The Specified Disease Oral cancer
The Studied Tissue Oral tissue
The Expression Level of Disease Section Compare with the Healthy Individual Tissue
p-value: 1.86E-05
Fold-change: -1.81E-01
Z-score: -4.92E+00
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.
  Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-365a-3p [69]
Sensitive Disease Oral squamous cell carcinoma [ICD-11: 2B6E.0]
Molecule Alteration Expression
Up-regulation
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.
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 [23]
Resistant Disease Oral squamous cell carcinoma [ICD-11: 2B6E.0]
Molecule Alteration Expression
Down-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation MAPK/RAS signaling pathway Regulation N.A.
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) [23]
Resistant Disease Oral squamous cell carcinoma [ICD-11: 2B6E.0]
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation MAPK/RAS signaling pathway Regulation N.A.
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.
Chronic myeloid leukemia [ICD-11: 2A20]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
  Metabolic Reprogramming via Altered Pathways (MRAP) Click to Show/Hide
Key Molecule: Pyruvate kinase muscle isozyme 1 (PKM1) [17]
Metabolic Type Mitochondrial metabolism
Resistant Disease Chronic myeloid leukemia [ICD-11: 2A20.0]
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model DLD-1 cells Colon Homo sapiens (Human) CVCL_0248
K562 cells Blood Homo sapiens (Human) CVCL_0004
Experiment for
Molecule Alteration
Expression profiles
Experiment for
Drug Resistance
Cell viability assay
Mechanism Description The overexpression of PKM1 resulted in resistance of the parental cells to 5-FU and oxaliplatin.
Acute myeloid leukemia [ICD-11: 2A60]
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
  Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Cysteine and glycine-rich protein 1 (CSRP1) [73]
Sensitive Disease Acute myeloid leukemia [ICD-11: 2A60.0]
Molecule Alteration Expression
Up-regulation
Experimental Note Discovered Using In-vivo Testing Model
Cell Pathway Regulation Rap1 signaling pathway Activation hsa04015
HIF-1 signaling pathway Activation hsa04066
JAK-STAT signaling pathway Activation hsa04630
In Vivo Model Patient-derived advanced AML model Homo sapiens
Experiment for
Drug Resistance
OncoPredict assay
Mechanism Description Based on the findings, the high?CSRP1?groups of patients in the TCGA datasets showed higher sensitivity to 5-fluorouracil, gemcitabine, rapamycin, and cisplatin and lower sensitivity to fludarabine. CSRP1 may serve as a potential prognostic marker and a therapeutic target for AML in the future.
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 [74]
Sensitive Disease Leukemia [ICD-11: 2B33.6]
Molecule Alteration Expression
Up-regulation
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) [74]
Sensitive Disease Leukemia [ICD-11: 2B33.6]
Molecule Alteration Expression
Down-regulation
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 blot 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 [35]
Sensitive Disease Osteosarcoma [ICD-11: 2B51.0]
Molecule Alteration Expression
Up-regulation
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) [35]
Sensitive Disease Osteosarcoma [ICD-11: 2B51.0]
Molecule Alteration Expression
Down-regulation
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 blot 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 [75]
Sensitive Disease Nasopharyngeal carcinoma [ICD-11: 2B6B.0]
Molecule Alteration Expression
Up-regulation
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) [75]
Sensitive Disease Nasopharyngeal carcinoma [ICD-11: 2B6B.0]
Molecule Alteration Expression
Down-regulation
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.
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 LncRNA-FGD5-AS1 promotes 5-Fu resistance of cervical cancer cells through modulating the miR-130a-3p-YTHDF2 axis. J Chemother. 2024 Dec 6:1-13.
Ref 5 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 6 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 7 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 8 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 9 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 10 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 11 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 12 3-deazaneplanocin A, a histone methyltransferase inhibitor, improved the chemoresistance induced under hypoxia in melanoma cells. Biochem Biophys Res Commun. 2023 Oct 15;677:26-30.
Ref 13 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 14 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 15 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 16 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 17 PKM1 is involved in resistance to anti-cancer drugs. Biochem Biophys Res Commun. 2016 Apr 22;473(1):174-180.
Ref 18 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 19 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 20 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 21 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 22 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 23 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 24 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 25 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 26 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 27 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 28 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 29 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 30 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 31 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 32 Arenobufagin modulation of PCSK9-mediated cholesterol metabolism induces tumor-associated macrophages polarisation to inhibit hepatocellular carcinoma progression. Phytomedicine. 2024 Jun;128:155532.
Ref 33 Targeting Src reactivates pyroptosis to reverse chemoresistance in lung and pancreatic cancer models. Sci Transl Med. 2023 Jan 11;15(678):eabl7895.
Ref 34 Metabolic classification suggests the GLUT1/ALDOB/G6PD axis as a therapeutic target in chemotherapy-resistant pancreatic cancer. Cell Rep Med. 2023 Sep 19;4(9):101162.
Ref 35 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 36 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 37 [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 38 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 39 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 40 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 41 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 42 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 43 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 44 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 45 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 46 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 47 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 48 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 49 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 50 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 51 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 52 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 53 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 54 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 55 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 56 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 57 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 58 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 59 Let-7 g microRNA sensitizes fluorouracil-resistant human hepatoma cells. Pharmazie. 2014 Apr;69(4):287-92.
Ref 60 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 61 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 62 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 63 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 64 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 65 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 66 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 67 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 68 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 69 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 70 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 71 MicroRNA-320a promotes 5-FU resistance in human pancreatic cancer cells. Sci Rep. 2016 Jun 9;6:27641. doi: 10.1038/srep27641.
Ref 72 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 73 Cysteine- and glycine-rich protein 1 predicts prognosis and therapy response in patients with acute myeloid leukemia. Clin Exp Med. 2024 Mar 28;24(1):57.
Ref 74 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 75 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 76 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 77 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 78 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 79 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 80 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 81 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 82 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 83 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 84 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 85 De novo pyrimidine synthesis fuels glycolysis and confers chemoresistance in gastric cancer. Cancer Lett. 2022 Nov 28;549:215837.
Ref 86 Inhibition of PI3K/Akt/mTOR Signaling Pathway Suppresses 5-Fluorouracil Resistance in Gastric Cancer. Mol Biotechnol. 2024 Dec;66(12):3640-3654.
Ref 87 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 88 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 89 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 90 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 91 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 92 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 93 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 94 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 95 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 96 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 97 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 98 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 99 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 100 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 101 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 102 MiR-30a Decreases Multidrug Resistance (MDR) of Gastric Cancer Cells. Med Sci Monit. 2016 Nov 23;0:0.
Ref 103 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 104 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.

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