Drug (ID: DG00049) and It's Reported Resistant Information
Name
Gemcitabine
Synonyms
Gemcitabine hydrochloride; DDFC; DFdC; DFdCyd; Folfugem; GEO; Gamcitabine; GemLip; Gemcel; Gemcin; Gemcitabina; Gemcitabinum; Gemtro; Gemzar; Zefei; Gemcitabine HCl; Gemcitabine stereoisomer; LY 188011; LY188011; Gemcitabina [INN-Spanish]; Gemcitabinum [INN-Latin]; Gemzar (TN); Gemzar (hydrochloride); Inno-D07001; LY-188011; Gemcitabine (USAN/INN)
    Click to Show/Hide
Indication
In total 1 Indication(s)
Solid tumour/cancer [ICD-11: 2A00-2F9Z]
Approved
[1]
Structure
Drug Resistance Disease(s)
Disease(s) with Clinically Reported Resistance for This Drug (6 diseases)
Bladder cancer [ICD-11: 2C94]
[2]
Breast cancer [ICD-11: 2C60]
[1]
Lung cancer [ICD-11: 2C25]
[3]
Mature T-cell lymphoma [ICD-11: 2A90]
[4]
Pancreatic cancer [ICD-11: 2C10]
[5]
Peritoneal cancer [ICD-11: 2C51]
[6]
Disease(s) with Resistance Information Discovered by Cell Line Test for This Drug (7 diseases)
Biliary tract cancer [ICD-11: 2C15]
[7]
Bladder cancer [ICD-11: 2C94]
[8]
Breast cancer [ICD-11: 2C60]
[9]
Liver cancer [ICD-11: 2C12]
[10]
Mature T-cell lymphoma [ICD-11: 2A90]
[11]
Osteosarcoma [ICD-11: 2B51]
[12]
Pancreatic cancer [ICD-11: 2C10]
[13]
Target Ribonucleoside-diphosphate reductase M2 (RRM2) RIR2_HUMAN [1]
Click to Show/Hide the Molecular Information and External Link(s) of This Drug
Formula
C9H11F2N3O4
IsoSMILES
C1=CN(C(=O)N=C1N)[C@H]2C([C@@H]([C@H](O2)CO)O)(F)F
InChI
1S/C9H11F2N3O4/c10-9(11)6(16)4(3-15)18-7(9)14-2-1-5(12)13-8(14)17/h1-2,4,6-7,15-16H,3H2,(H2,12,13,17)/t4-,6-,7-/m1/s1
InChIKey
SDUQYLNIPVEERB-QPPQHZFASA-N
PubChem CID
60750
ChEBI ID
CHEBI:175901
TTD Drug ID
D03UVS
VARIDT ID
DR00063
INTEDE ID
DR0765
DrugBank ID
DB00441
Type(s) of Resistant Mechanism of This Drug
  DISM: Drug Inactivation by Structure Modification
  EADR: Epigenetic Alteration of DNA, RNA or Protein
  IDUE: Irregularity in Drug Uptake and Drug Efflux
  RTDM: Regulation by the Disease Microenvironment
  UAPP: Unusual Activation of Pro-survival Pathway
Drug Resistance Data Categorized by Their Corresponding Diseases
ICD-02: Benign/in-situ/malignant neoplasm
Click to Show/Hide the Resistance Disease of This Class
Mature T-cell lymphoma [ICD-11: 2A90]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-187 [4]
Molecule Alteration Expression
Up-regulation
Resistant Disease Peripheral T-cell lymphoma [ICD-11: 2A90.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
In Vitro Model MOLT4 cells Bone marrow Homo sapiens (Human) CVCL_0013
HUT78 cells Lymph Homo sapiens (Human) CVCL_0337
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR187 downregulated tumor suppressor gene disabled homolog-2 (Dab2), decreased the interaction of Dab2 with adapter protein Grb2, resulting in Ras activation, phosphorylation/activation of extracellular signal-regulated kinase (ERk) and AkT, and subsequent stabilization of MYC oncoprotein. MiR187-overexpressing cells were resistant to chemotherapeutic agents like doxorubicin, cyclophosphamide, cisplatin and gemcitabine, but sensitive to the proteasome inhibitor bortezomib.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: ATP-binding cassette sub-family G2 (ABCG2) [14]
Molecule Alteration Expression
Up-regulation
Resistant Disease Natural killer/T-cell lymphoma [ICD-11: 2A90.2]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SNK-6 cells Oral Homo sapiens (Human) CVCL_A673
In Vivo Model Balb/c athymic nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description ABCG2 upregulated cell proliferation, increased clonogenicity, increased invasive ability and decreased apoptosis, in vivo and in vitro, when cells were treated with gemcitabine.
Key Molecule: ATP-binding cassette sub-family G2 (ABCG2) [14]
Molecule Alteration Expression
Up-regulation
Resistant Disease Natural killer/T-cell lymphoma [ICD-11: 2A90.2]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SNK-6 cells Oral Homo sapiens (Human) CVCL_A673
In Vivo Model Balb/c athymic nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description ABCG2 upregulated cell proliferation, increased clonogenicity, increased invasive ability and decreased apoptosis, in vivo and in vitro, when cells were treated with gemcitabine.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Protein zeta/delta 14-3-3 (YWHAZ) [11]
Molecule Alteration Expression
Up-regulation
Resistant Disease T-cell lymphoma [ICD-11: 2A60.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model YTS cells Pleural effusion Homo sapiens (Human) CVCL_D324
Experiment for
Molecule Alteration
Western blotting assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description Compared with YTS-gem cells, the level of Pro apoptotic protein Bax in YTS gem cells that down regulated 14-3-3-Zetawas significantly higher. In contrast, the levels of anti apoptotic proteins Bcl-2, Caspase-3, cleaved caspase-3 and cyclin D1 decreased significantly.
Key Molecule: Protein zeta/delta 14-3-3 (YWHAZ) [11]
Molecule Alteration Expression
Up-regulation
Resistant Disease Extranodal NK/T-cell lymphoma [ICD-11: 2A90.6]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell proliferation Activation hsa05200
In Vitro Model YTS cells Pleural effusion Homo sapiens (Human) CVCL_D324
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Mechanism Description 14-3-3-Zeta was up regulated in YTS gem cells, 14-3-3-Zeta promote cell proliferation and invasion, 14-3-3-Zeta protein induced enktl resistance to gemcitabine through anti apoptotic pathway.
Osteosarcoma [ICD-11: 2B51]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-152 [12]
Molecule Alteration Expression
Down-regulation
Resistant Disease Osteosarcoma [ICD-11: 2B51.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
c-Met/PI3K/AKT signaling pathway Activation hsa01521
In Vitro Model MG63 cells Bone marrow Homo sapiens (Human) CVCL_0426
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Soft agar assay
Mechanism Description LncRNAPVT1 targets miR-152 to enhance chemoresistance of osteosarcoma to gemcitabine through activating c-MET/PI3k/AkT pathway.
Key Molecule: Pvt1 oncogene (PVT1) [12]
Molecule Alteration Expression
Up-regulation
Resistant Disease Osteosarcoma [ICD-11: 2B51.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
c-Met/PI3K/AKT signaling pathway Activation hsa01521
In Vitro Model MG63 cells Bone marrow Homo sapiens (Human) CVCL_0426
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Soft agar assay
Mechanism Description LncRNAPVT1 targets miR-152 to enhance chemoresistance of osteosarcoma to gemcitabine through activating c-MET/PI3k/AkT pathway.
Gastric cancer [ICD-11: 2B72]
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-34 [15]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation p53 signaling pathway Inhibition hsa04115
In Vitro Model AGS cells Gastric Homo sapiens (Human) CVCL_0139
NCI-N87 cells Gastric Homo sapiens (Human) CVCL_1603
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
KATO-3 cells Gastric Homo sapiens (Human) CVCL_0371
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Human gastric cancer kato III cells with miR-34 restoration reduced the expression of target genes Bcl-2, Notch, and HMGA2. MicroRNA miR-34 was recently found to be a direct target of p53, functioning downstream of the p53 pathway as a tumor suppressor, miR-34 impaired cell growth, accumulated the cells in G1 phase, increased caspase-3 activation, and, more significantly, inhibited tumorsphere formation and growth.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [15]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation p53 signaling pathway Inhibition hsa04115
In Vitro Model AGS cells Gastric Homo sapiens (Human) CVCL_0139
NCI-N87 cells Gastric Homo sapiens (Human) CVCL_1603
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
KATO-3 cells Gastric Homo sapiens (Human) CVCL_0371
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Human gastric cancer kato III cells with miR-34 restoration reduced the expression of target genes Bcl-2, Notch, and HMGA2. MicroRNA miR-34 was recently found to be a direct target of p53, functioning downstream of the p53 pathway as a tumor suppressor, miR-34 impaired cell growth, accumulated the cells in G1 phase, increased caspase-3 activation, and, more significantly, inhibited tumorsphere formation and growth.
Key Molecule: High mobility group protein HMGI-C (HMGA2) [15]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation p53 signaling pathway Inhibition hsa04115
In Vitro Model AGS cells Gastric Homo sapiens (Human) CVCL_0139
NCI-N87 cells Gastric Homo sapiens (Human) CVCL_1603
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
KATO-3 cells Gastric Homo sapiens (Human) CVCL_0371
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Human gastric cancer kato III cells with miR-34 restoration reduced the expression of target genes Bcl-2, Notch, and HMGA2. MicroRNA miR-34 was recently found to be a direct target of p53, functioning downstream of the p53 pathway as a tumor suppressor, miR-34 impaired cell growth, accumulated the cells in G1 phase, increased caspase-3 activation, and, more significantly, inhibited tumorsphere formation and growth.
Key Molecule: Neurogenic locus notch homolog protein (NOTCH) [15]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation p53 signaling pathway Inhibition hsa04115
In Vitro Model AGS cells Gastric Homo sapiens (Human) CVCL_0139
NCI-N87 cells Gastric Homo sapiens (Human) CVCL_1603
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
KATO-3 cells Gastric Homo sapiens (Human) CVCL_0371
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Human gastric cancer kato III cells with miR-34 restoration reduced the expression of target genes Bcl-2, Notch, and HMGA2. MicroRNA miR-34 was recently found to be a direct target of p53, functioning downstream of the p53 pathway as a tumor suppressor, miR-34 impaired cell growth, accumulated the cells in G1 phase, increased caspase-3 activation, and, more significantly, inhibited tumorsphere formation and growth.
Colon cancer [ICD-11: 2B90]
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-330 [16]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HT-29 cells Colon Homo sapiens (Human) CVCL_0320
Colo320 cells Colon Homo sapiens (Human) CVCL_1989
WiDR cells Colon Homo sapiens (Human) CVCL_2760
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Sulforhodamide B (SRB) test assay
Mechanism Description Deoxycytidine kinase (dCk) is essential for phosphorylation of natural deoxynucleosides andanalogs, such as gemcitabine and cytarabine, two widely used anticancer compounds. miR-330 expression negatively correlated withdCk mRNA expression, suggesting a role of miR-330 in post-transcriptional regulationof dCk. Expression of miR-330 in various colon and lung cancer cell lines,as measured by QRT-PCR, varied five-fold between samples and correlated with in-vitro gemcitabineresistance.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Deoxycytidine kinase (DCK) [16]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HT-29 cells Colon Homo sapiens (Human) CVCL_0320
Colo320 cells Colon Homo sapiens (Human) CVCL_1989
WiDR cells Colon Homo sapiens (Human) CVCL_2760
Experiment for
Molecule Alteration
qRT -PCR
Experiment for
Drug Resistance
Sulforhodamide B (SRB) test assay
Mechanism Description Deoxycytidine kinase (dCk) is essential for phosphorylation of natural deoxynucleosides andanalogs, such as gemcitabine and cytarabine, two widely used anticancer compounds. miR-330 expression negatively correlated withdCk mRNA expression, suggesting a role of miR-330 in post-transcriptional regulationof dCk. Expression of miR-330 in various colon and lung cancer cell lines,as measured by QRT-PCR, varied five-fold between samples and correlated with in-vitro gemcitabineresistance.
Pancreatic cancer [ICD-11: 2C10]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Drug Inactivation by Structure Modification (DISM) Click to Show/Hide
Key Molecule: Activation induced cytidine deaminase (AICDA) [17]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Identified from the Human Clinical Data
Mechanism Description The main mechanism for gemcitabine inactivation is through deamination by cytidine deaminase (CDA) to difluorodeoxyuridine (dFdU). Since dFdU is not a substrate for pyrimidine nucleoside phosphorylases, the drug is degraded and excreted out of the cell.
Key Molecule: Deoxycytidine kinase (DCK) [17]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Identified from the Human Clinical Data
Mechanism Description Once taken up into the cell, gemcitabine is phosphorylated by deoxycytidine kinase (dCK) to produce dFdCMP. In turn, dFdCMP is converted by other pyrimidine kinases to its active diphosphate and triphosphate derivatives, dFdCDP and dFdCTP. Due to the central role of dCK in gemcitabine metabolism, its deficiency is a major contributor to gemcitabine resistance.
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-188-3p [18]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model 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
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Soft agar assay
Mechanism Description Long non-coding RNA LINC00346 promotes pancreatic cancer growth and gemcitabine resistance by sponging miR-188-3p to derepress BRD4 expression.
Key Molecule: P53 regulated carcinoma associated Stat3 activating long intergenic non-protein coding transcript (PRECSIT) [18]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model 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
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Soft agar assay
Mechanism Description Long non-coding RNA LINC00346 promotes pancreatic cancer growth and gemcitabine resistance by sponging miR-188-3p to derepress BRD4 expression.
Key Molecule: hsa-mir-301 [19]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic carcinoma [ICD-11: 2C10.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Capan-2 cells Pancreas Homo sapiens (Human) CVCL_0026
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Colorimetric methylene blue assay; Flow cytometry assay
Mechanism Description Gemcitabine-resistant Capan-2 and Panc-1 cells exhibited increased miR-301 expression, and miR-301 overepression can enhance apoptosis and inhibit cell invasiveness and exhibit strong gemcitabine resistance.
Key Molecule: Maternally expressed 3 (MEG3) [20]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
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
COLO357 cells Pancreas Homo sapiens (Human) CVCL_0221
T3M4 cells Pancreas Homo sapiens (Human) CVCL_4056
HTERT-HPNE cells Pancreas Homo sapiens (Human) CVCL_C466
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
CCK8 assay; Boyden chamber assay; Sphere formation assay; Flow cytometric analysis
Mechanism Description Decreased expression of MEG3 could promote PC cell migration and invasion, as well as chemoresistance by regulating the EMT process and CSC properties.
Key Molecule: HOX transcript antisense RNA (HOTAIR) [21]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Gemcitabine treatment causes resistance and malignancy of pancreatic cancer stem-like cells via induction of LncRNA HOTAIR.
Key Molecule: Taurine up-regulated 1 (TUG1) [22]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell colony Activation hsa05200
Cell viability Activation hsa05200
ERK signaling pathway Activation hsa04210
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
PANC-28 cells Pancreatic Homo sapiens (Human) CVCL_3917
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description TUG1 promoted the viability of PDAC cells and enhanced its resistance of gemcitabine and overexpression of TUG1 increased ERk phosphorylation.
Key Molecule: hsa-mir-301 [23]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Activation hsa04151
Cell viability Activation hsa05200
In Vitro Model HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
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
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
HPAF-II cells Pancreatic Homo sapiens (Human) CVCL_0313
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR-301a upregulation promoted resistance to gemcitabine under hypoxia through downregulation of TAp63.
Key Molecule: hsa-miR-455-3p [24]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
In Vitro Model HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
HPDE6-C7 cells Pancreas Homo sapiens (Human) CVCL_0P38
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Down-regulation of microRNA-455-3p Links to Proliferation and Drug Resistance of Pancreatic Cancer Cells via Targeting TAZ.
Key Molecule: hsa-mir-125a [25]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
In Vitro Model HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-125a may promote chemo-resistance to gemcitabine in pancreatic cell lines through targeting A20, which may provide novel therapeutic targets or molecular biomarkers for cancer therapy and improve tumor diagnosis or predictions of therapeutic responses.
Key Molecule: hsa-mir-181c [26]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Hippo signaling pathway Regulation hsa04392
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-181c directly repressed MST1, LATS2, MOB1 and SAV1 expression in human pancreatic cancer cells. Overexpression of miR-181c induced hyperactivation of the YAP/TAZ and (+) expression of the Hippo signaling downstream genes CTGF, BIRC5 and BLC2L1, leading to pancreatic cancer cell survival and chemoresistance in vitro and in vivo. Importantly, high miR-181c levels were significantly correlated with Hippo signaling inactivation in pancreatic cancer samples, and predicted a poor patient overall survival.
Key Molecule: HOXA distal transcript antisense RNA (HOTTIP) [27]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
HOTTIP/HOXA13 signaling pathway Activation hsa05202
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-2 cells Pancreas Homo sapiens (Human) CVCL_0026
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description The long non-coding RNA HOTTIP promotes progression and gemcitabine resistance by regulating HOXA13 in pancreatic cancer.Microarray analyses revealed that HOTTIP was one of the most significantly upregulated LncRNAs in PDAC tissues compared with pancreatic tissues.Furthermore, knockdown of HOXA13 by RNA interference (siHOXA13) revealed that HOTTIP promoted PDAC cell proliferation, invasion, and chemoresistance, at least partly through regulating HOXA13. As a crucial tumor promoter, HOTTIP promotes cell proliferation, invasion, and chemoresistance by modulating HOXA13. Therefore, the HOTTIP/HOXA13 axis is a potential therapeutic target and molecular biomarker for PDAC.
Key Molecule: hsa-miR-1246 [28]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model Panc1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The in vitro drug sensitivity of pancreatic cancer cells was altered according to the miR-1246 expression via CCNG2. In vivo, we found that miR-1246 could increase tumour-initiating potential and induced drug resistance. A high expression level of miR-1246 was correlated with a worse prognosis and CCNG2 expression was significantly lower in those patients. miR-1246 expression was associated with chemoresistance and CSC-like properties via CCNG2, and could predict worse prognosis in pancreatic cancer patients.
Key Molecule: hsa-mir-21 [5], [13], [29]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AKT signaling pathway Activation hsa04151
Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
FasL/Fas signaling pathway Inhibition 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
LPc006 cells Pancreas Homo sapiens (Human) N.A.
LPc028 cells Pancreas Homo sapiens (Human) N.A.
LPc033 cells Pancreas Homo sapiens (Human) N.A.
LPc067 cells Pancreas Homo sapiens (Human) N.A.
LPc111 cells Pancreas Homo sapiens (Human) N.A.
LPc167 cells Pancreas Homo sapiens (Human) N.A.
PP437 cells Pancreas Homo sapiens (Human) N.A.
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
WST-8 assay; Fluorescence microscopy
Mechanism Description miR-21 regulates expression of PTEN and phosphorylation of its downstream kinase Akt and (b) the reduction of phospho-Akt (pAkt) correlated with the enhancement of gemcitabine-induced apoptosis and antitumor activity in vitro and in vivo, suggesting that Akt pathway plays a significant role in mediating drug resistance in PDAC cells.
Key Molecule: hsa-mir-21 [30]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell migration Activation hsa04670
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
HPAC cells Pancreas Homo sapiens (Human) CVCL_3517
BxPc3 cells Pancreas Homo sapiens (Human) CVCL_0186
Capan cells Pancreas Homo sapiens (Human) CVCL_0237
HPAF cells Pancreas Homo sapiens (Human) CVCL_B284
PL-45 cells Pancreas Homo sapiens (Human) CVCL_3567
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Histone acetylation levels at miR-21 promoter were increased in PDAC cells after treatment with gemcitabine. Enhanced invasion and metastasis, increased miR-21 expression, decreased PTEN, elevated pAkT level were demonstrated in gemcitabine-resistant HPAC and PANC-1 cells. Pre-miR-21 transfection or TSA treatment further increased invasion and metastasis ability, decreased PTEN, and elevated pAkT levels in these two lines. In contrast, anti-miR-21 transfection could reverse invasion and metastasis, and PTEN and pAkT expressions induced by gemcitabine.
Key Molecule: hsa-mir-365 [31]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-365 directly targets the pro-apoptotic molecules SHC1 and BAX, whose reductions contribute to gemcitabine resistance in pancreatic cancer cells.
Key Molecule: hsa-mir-181 [32]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
NF-kappaB signaling pathway Regulation hsa04064
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Panc1 cells Pancreas Homo sapiens (Human) CVCL_0480
PSN1 cells Pancreas Homo sapiens (Human) CVCL_1644
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-181b enhances the activity of NF-kB by inhibiting CYLD, thus leading to the resistance to gemcitabine.
Key Molecule: hsa-miR-320c [33]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell viability Activation hsa05200
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
PSN1 cells Pancreas Homo sapiens (Human) CVCL_1644
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-320c regulates the resistance of pancreatic cancer cells to gemcitabine through SMARCC1.
Key Molecule: hsa-mir-21 [17]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Identified from the Human Clinical Data
Mechanism Description miR-21 is probably the most characterized miRNA associated with gemcitabine resistance. Tissue samples of PDA patients treated with gemcitabine indicate that miR-21 expression is directly correlated with chemotherapy resistance. Patients with high miR-21 expression have significantly shorter overall survival; consistently, overexpression of miR-21 in primary PDA cells in vitro, decreases the anti-proliferative effect of gemcitabine. miR-21 promotes gemcitabine resistance by targeting phosphatase and tensin homologue (PTEN) or by overexpression of matrix metalloproteinases (MMP) 2 and 9, and of vascular endothelial growth factor (VEGF), which in-turn induces the PI3K/AKT pathway.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Solute carrier family 29 member 1 (SLC29A1) [17]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Identified from the Human Clinical Data
Mechanism Description Gemcitabine could be a substrate for several nucleoside transporters (NTs), but its major uptake occurs via the equilibrative and concentrative type NTs (ENTs and CNTs, respectively). ENT1, CNT1 and CNT3 have often been related to gemcitabine transport and resistance in humans. When ENT1 knockout conferred gemcitabine resistance, while its up regulation enhanced its cytotoxic activity. Similarly, retroviral expression of CNT1 renders ovarian cancer cells sensitive to gemcitabine in vitro.
Key Molecule: Solute carrier family 28 member 1 (SLC28A1) [17]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Identified from the Human Clinical Data
Mechanism Description Gemcitabine could be a substrate for several nucleoside transporters (NTs), but its major uptake occurs via the equilibrative and concentrative type NTs (ENTs and CNTs, respectively). ENT1, CNT1 and CNT3 have often been related to gemcitabine transport and resistance in humans. When ENT1 knockout conferred gemcitabine resistance, while its up regulation enhanced its cytotoxic activity. Similarly, retroviral expression of CNT1 renders ovarian cancer cells sensitive to gemcitabine in vitro.
Key Molecule: Solute carrier family 28 member 3 (SLC28A3) [17]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Identified from the Human Clinical Data
Mechanism Description Gemcitabine could be a substrate for several nucleoside transporters (NTs), but its major uptake occurs via the equilibrative and concentrative type NTs (ENTs and CNTs, respectively). ENT1, CNT1 and CNT3 have often been related to gemcitabine transport and resistance in humans. When ENT1 knockout conferred gemcitabine resistance, while its up regulation enhanced its cytotoxic activity. Similarly, retroviral expression of CNT1 renders ovarian cancer cells sensitive to gemcitabine in vitro.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: hsa-mir-221 [34]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
miR221/SOCS3 signaling pathway Regulation hsa05206
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Capan-2 cells Pancreas Homo sapiens (Human) CVCL_0026
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR-221 overexpression can promote proliferation, migration, emt, chemotherapy resistance, and stem cell-like properties in panc-1 cells.
Key Molecule: Transcription factor AP2 gamma (TFAP2C) [35]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
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 viability Activation hsa05200
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
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
Su.86.86 cells Pancreas Homo sapiens (Human) CVCL_3881
T3M4 cells Pancreas Homo sapiens (Human) CVCL_4056
In Vivo Model Nude mouse model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay; Transwell assay
Mechanism Description Transcription factor activating protein 2 gamma (TFAP2C) is a target of miR-10a-5p, and TFAP2C overexpression resensitizes PDAC cells to gemcitabine, which is initiated by miR-10a-5p.
Key Molecule: hsa-miR-10a-5p [35]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell viability Activation hsa05200
Epithelial mesenchymal transition signaling pathway Activation hsa01521
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
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
Su.86.86 cells Pancreas Homo sapiens (Human) CVCL_3881
T3M4 cells Pancreas Homo sapiens (Human) CVCL_4056
In Vivo Model Nude mouse model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay; Transwell assay
Mechanism Description Transcription factor activating protein 2 gamma (TFAP2C) is a target of miR-10a-5p, and TFAP2C overexpression resensitizes PDAC cells to gemcitabine, which is initiated by miR-10a-5p.
Key Molecule: Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) [36]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
CFPAC1 cells Pancreas Homo sapiens (Human) CVCL_1119
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description MALAT-1 could increase the proportion of pancreatic CSCs, maintain self-renewing capacity, decrease the chemosensitivity to anticancer drugs, and accelerate tumor angiogenesis in vitro, and promote tumorigenicity of pancreatic cancer cells in vivo. The underlying mechanisms may involve in increased expression of self-renewal related factors Sox2.
Key Molecule: hsa-mir-100 [37]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues.
Key Molecule: hsa-mir-10a [37]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues.
Key Molecule: hsa-mir-10b [37]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues.
Key Molecule: hsa-mir-134 [37]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues.
Key Molecule: hsa-mir-143 [37]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues.
Key Molecule: hsa-mir-146a [37]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues.
Key Molecule: hsa-mir-15 [37]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues.
Key Molecule: hsa-mir-205 [37]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues.
Key Molecule: hsa-mir-214 [37]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues.
Key Molecule: hsa-mir-32 [37]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues.
Key Molecule: hsa-mir-34 [37]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues.
Key Molecule: hsa-miR-146b-5p [37]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues.
Key Molecule: hsa-mir-7 [37]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues.
Key Molecule: TIMP metallopeptidase inhibitor 2 (TIMP2) [17]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Identified from the Human Clinical Data
Mechanism Description The ECM may provide a mechanical barrier, preventing the tumor from further spread. Disintegration of the ECM by MMPs enables cancer cells to dissociate from the tumor and metastasize. Apart from destabilizing the physical barrier, MMPs overexpression also regulates internal cellular cascades. In response to collagen deposition in the ECM, an MMP dependent ERK-1/2 phosphorylation occurs, triggering the transcription factor HMGA2 and gemcitabine resistance.
Key Molecule: Mucin 4, cell surface associated (MUC4) [17]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Identified from the Human Clinical Data
Mechanism Description Mucin 4 (MUC4) is a membrane-bound O-glycoprotein that is found in the lining of the respiratory tract and GI mucosa, where it enables lubrication and cell-matrix detachment. In PDA, MUC4 expressing cells exhibit greater gemcitabine resistance than do MUC4 negative cells, an effect mediated by interaction with HER2.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Bromodomain-containing protein 4 (BRD4) [18]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model 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
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Luciferase reporter assay; Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Soft agar assay
Mechanism Description Long non-coding RNA LINC00346 promotes pancreatic cancer growth and gemcitabine resistance by sponging miR-188-3p to derepress BRD4 expression.
Key Molecule: Mitogen-activated protein kinase (MAPK) [22]
Molecule Alteration Phosphorylation
Up-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Activation hsa05200
ERK signaling pathway Activation hsa04210
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
PANC-28 cells Pancreatic Homo sapiens (Human) CVCL_3917
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description TUG1 promoted the viability of PDAC cells and enhanced its resistance of gemcitabine and overexpression of TUG1 increased ERk phosphorylation.
Key Molecule: Tumor protein 63 (TP63) [23]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Activation hsa04151
Cell viability Activation hsa05200
In Vitro Model HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
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
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
HPAF-II cells Pancreatic Homo sapiens (Human) CVCL_0313
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR-301a upregulation promoted resistance to gemcitabine under hypoxia through downregulation of TAp63.
Key Molecule: Tafazzin (TAZ) [24]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
In Vitro Model HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
HPDE6-C7 cells Pancreas Homo sapiens (Human) CVCL_0P38
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 Down-regulation of microRNA-455-3p Links to Proliferation and Drug Resistance of Pancreatic Cancer Cells via Targeting TAZ.
Key Molecule: Tumor necrosis factor alpha-induced protein 3 (TNFAIP3) [25]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
In Vitro Model HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-125a may promote chemo-resistance to gemcitabine in pancreatic cell lines through targeting A20, which may provide novel therapeutic targets or molecular biomarkers for cancer therapy and improve tumor diagnosis or predictions of therapeutic responses.
Key Molecule: Serine/threonine-protein kinase LATS2 (LATS2) [26]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Hippo signaling pathway Regulation hsa04392
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-181c directly repressed MST1, LATS2, MOB1 and SAV1 expression in human pancreatic cancer cells. Overexpression of miR-181c induced hyperactivation of the YAP/TAZ and (+) expression of the Hippo signaling downstream genes CTGF, BIRC5 and BLC2L1, leading to pancreatic cancer cell survival and chemoresistance in vitro and in vivo. Importantly, high miR-181c levels were significantly correlated with Hippo signaling inactivation in pancreatic cancer samples, and predicted a poor patient overall survival.
Key Molecule: MOB kinase activator 1A (MOB1A) [26]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Hippo signaling pathway Regulation hsa04392
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-181c directly repressed MST1, LATS2, MOB1 and SAV1 expression in human pancreatic cancer cells. Overexpression of miR-181c induced hyperactivation of the YAP/TAZ and (+) expression of the Hippo signaling downstream genes CTGF, BIRC5 and BLC2L1, leading to pancreatic cancer cell survival and chemoresistance in vitro and in vivo. Importantly, high miR-181c levels were significantly correlated with Hippo signaling inactivation in pancreatic cancer samples, and predicted a poor patient overall survival.
Key Molecule: Serine/threonine-protein kinase 4 (MST1) [26]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Hippo signaling pathway Regulation hsa04392
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-181c directly repressed MST1, LATS2, MOB1 and SAV1 expression in human pancreatic cancer cells. Overexpression of miR-181c induced hyperactivation of the YAP/TAZ and (+) expression of the Hippo signaling downstream genes CTGF, BIRC5 and BLC2L1, leading to pancreatic cancer cell survival and chemoresistance in vitro and in vivo. Importantly, high miR-181c levels were significantly correlated with Hippo signaling inactivation in pancreatic cancer samples, and predicted a poor patient overall survival.
Key Molecule: Protein salvador homolog 1 (SAV1) [26]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Hippo signaling pathway Regulation hsa04392
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-181c directly repressed MST1, LATS2, MOB1 and SAV1 expression in human pancreatic cancer cells. Overexpression of miR-181c induced hyperactivation of the YAP/TAZ and (+) expression of the Hippo signaling downstream genes CTGF, BIRC5 and BLC2L1, leading to pancreatic cancer cell survival and chemoresistance in vitro and in vivo. Importantly, high miR-181c levels were significantly correlated with Hippo signaling inactivation in pancreatic cancer samples, and predicted a poor patient overall survival.
Key Molecule: Homeobox protein Hox-A13 (HOXA13) [27]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
HOTTIP/HOXA13 signaling pathway Activation hsa05202
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-2 cells Pancreas Homo sapiens (Human) CVCL_0026
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description The long non-coding RNA HOTTIP promotes progression and gemcitabine resistance by regulating HOXA13 in pancreatic cancer.Microarray analyses revealed that HOTTIP was one of the most significantly upregulated LncRNAs in PDAC tissues compared with pancreatic tissues.Furthermore, knockdown of HOXA13 by RNA interference (siHOXA13) revealed that HOTTIP promoted PDAC cell proliferation, invasion, and chemoresistance, at least partly through regulating HOXA13. As a crucial tumor promoter, HOTTIP promotes cell proliferation, invasion, and chemoresistance by modulating HOXA13. Therefore, the HOTTIP/HOXA13 axis is a potential therapeutic target and molecular biomarker for PDAC.
Key Molecule: Transcription factor SOX-2 (SOX2) [36]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
CFPAC1 cells Pancreas Homo sapiens (Human) CVCL_1119
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description MALAT-1 could increase the proportion of pancreatic CSCs, maintain self-renewing capacity, decrease the chemosensitivity to anticancer drugs, and accelerate tumor angiogenesis in vitro, and promote tumorigenicity of pancreatic cancer cells in vivo. The underlying mechanisms may involve in increased expression of self-renewal related factors Sox2.
Key Molecule: Cyclin-G2 (CCNG2) [28]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model Panc1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The in vitro drug sensitivity of pancreatic cancer cells was altered according to the miR-1246 expression via CCNG2. In vivo, we found that miR-1246 could increase tumour-initiating potential and induced drug resistance. A high expression level of miR-1246 was correlated with a worse prognosis and CCNG2 expression was significantly lower in those patients. miR-1246 expression was associated with chemoresistance and CSC-like properties via CCNG2, and could predict worse prognosis in pancreatic cancer patients.
Key Molecule: Phosphatase and tensin homolog (PTEN) [30]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell migration Activation hsa04670
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
HPAC cells Pancreas Homo sapiens (Human) CVCL_3517
BxPc3 cells Pancreas Homo sapiens (Human) CVCL_0186
Capan cells Pancreas Homo sapiens (Human) CVCL_0237
HPAF cells Pancreas Homo sapiens (Human) CVCL_B284
PL-45 cells Pancreas Homo sapiens (Human) CVCL_3567
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Histone acetylation levels at miR-21 promoter were increased in PDAC cells after treatment with gemcitabine. Enhanced invasion and metastasis, increased miR-21 expression, decreased PTEN, elevated pAkT level were demonstrated in gemcitabine-resistant HPAC and PANC-1 cells. Pre-miR-21 transfection or TSA treatment further increased invasion and metastasis ability, decreased PTEN, and elevated pAkT levels in these two lines. In contrast, anti-miR-21 transfection could reverse invasion and metastasis, and PTEN and pAkT expressions induced by gemcitabine.
Key Molecule: Apoptosis regulator BAX (BAX) [31]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-365 directly targets the pro-apoptotic molecules SHC1 and BAX, whose reductions contribute to gemcitabine resistance in pancreatic cancer cells.
Key Molecule: SHC-transforming protein 1 (SHC1) [31]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-365 directly targets the pro-apoptotic molecules SHC1 and BAX, whose reductions contribute to gemcitabine resistance in pancreatic cancer cells.
Key Molecule: Ubiquitin carboxyl-terminal hydrolase CYLD (CYLD) [32]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
NF-kappaB signaling pathway Regulation hsa04064
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Panc1 cells Pancreas Homo sapiens (Human) CVCL_0480
PSN1 cells Pancreas Homo sapiens (Human) CVCL_1644
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-181b enhances the activity of NF-kB by inhibiting CYLD, thus leading to the resistance to gemcitabine.
Key Molecule: SWI/SNF complex subunit SMARCC1 (SMARCC1) [33]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell viability Activation hsa05200
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
PSN1 cells Pancreas Homo sapiens (Human) CVCL_1644
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-320c regulates the resistance of pancreatic cancer cells to gemcitabine through SMARCC1.
Key Molecule: Tumor necrosis factor ligand superfamily member 6 (FASLG) [29]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
FasL/Fas signaling pathway Inhibition hsa04210
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
WST-8 assay
Mechanism Description Decreased Fas/FasL signaling mediates miR-21-induced chemoresistance in pancreatic cancer, over-expression of miR-21 reduced the endogenous expression of FasL anfd cause resistance to Gemcitabine.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [13]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Upregulation of Bcl-2 expression was detected in cells transfected with miR-21 mimics, accompanied by downregulated Bax expression, less apoptosis, lower caspase-3 activity, decreased chemosensitivity to gemcitabine and increased proliferation.
Key Molecule: RAC serine/threonine-protein kinase (AKT) [5]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AKT signaling pathway Activation hsa04151
Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model LPc006 cells Pancreas Homo sapiens (Human) N.A.
LPc028 cells Pancreas Homo sapiens (Human) N.A.
LPc033 cells Pancreas Homo sapiens (Human) N.A.
LPc067 cells Pancreas Homo sapiens (Human) N.A.
LPc111 cells Pancreas Homo sapiens (Human) N.A.
LPc167 cells Pancreas Homo sapiens (Human) N.A.
PP437 cells Pancreas Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Fluorescence microscopy
Mechanism Description miR-21 regulates expression of PTEN and phosphorylation of its downstream kinase Akt and (b) the reduction of phospho-Akt (pAkt) correlated with the enhancement of gemcitabine-induced apoptosis and antitumor activity in vitro and in vivo, suggesting that Akt pathway plays a significant role in mediating drug resistance in PDAC cells.
Key Molecule: Phosphatase and tensin homolog (PTEN) [5]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AKT signaling pathway Activation hsa04151
Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model LPc006 cells Pancreas Homo sapiens (Human) N.A.
LPc028 cells Pancreas Homo sapiens (Human) N.A.
LPc033 cells Pancreas Homo sapiens (Human) N.A.
LPc067 cells Pancreas Homo sapiens (Human) N.A.
LPc111 cells Pancreas Homo sapiens (Human) N.A.
LPc167 cells Pancreas Homo sapiens (Human) N.A.
PP437 cells Pancreas Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Fluorescence microscopy
Mechanism Description miR-21 regulates expression of PTEN and phosphorylation of its downstream kinase Akt and (b) the reduction of phospho-Akt (pAkt) correlated with the enhancement of gemcitabine-induced apoptosis and antitumor activity in vitro and in vivo, suggesting that Akt pathway plays a significant role in mediating drug resistance in PDAC cells.
Key Molecule: DNA excision repair protein ERCC-1 (ERCC1) [17]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Identified from the Human Clinical Data
Mechanism Description Excision repair cross-complementation 1 (ERCC1) is a DNA repair endonuclease responsible for the incision of DNA cross-link-induced double-strand breaks. ERCC1 can repair gemcitabine-induced strand breaks, and its overexpression is well documented in poor gemcitabine responders.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-663a [38]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
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
T3-M4 cells Pancreas Homo sapiens (Human) CVCL_VQ95
Experiment for
Molecule Alteration
RT-PCR, qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay; Flow cytometry assay
Mechanism Description Upregulated miR-663 expression in PDAC cell lines promotes sensitivity to GEM.
Key Molecule: hsa-mir-200b [19]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic carcinoma [ICD-11: 2C10.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell metastasis Inhibition hsa05205
Cell proliferation Inhibition hsa05200
Chemosensitivity Activation hsa05207
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Capan-2 cells Pancreas Homo sapiens (Human) CVCL_0026
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Colorimetric methylene blue assay; Flow cytometry assay
Mechanism Description Forced expression of miR-200b induces CDH1 expression and promotes gemcitabine sensitivity in Capan-2 and Panc-1 cells.
Key Molecule: hsa-miR-1207-5p [39]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
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
Su.86.86 cells Pancreas Homo sapiens (Human) CVCL_3881
In Vivo Model Engrafted tumor mouse model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Overexpression of the miR-1207 pair improves gemcitabine efficacy in PC cells.
Key Molecule: Pvt1 oncogene (PVT1) [39]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
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
Su.86.86 cells Pancreas Homo sapiens (Human) CVCL_3881
In Vivo Model Engrafted tumor mouse model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description PVT1 inhibition leads to improved efficacy of gemcitabine in PC cells.
Key Molecule: hsa-mir-34 [40]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Slug/PUMA signaling pathway Regulation hsa04390
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Flow Cytometric Analysis, MTT assay; TUNEL staining
Mechanism Description miR34 increases in vitro PANC-1 cell sensitivity to gemcitabine via targeting Slug/PUMA. miR34 enhances sensitization against gemcitabine-mediated apoptosis through the down-regulation of Slug expression, and up-regulation of Slug-dependent PUMA expression.
Key Molecule: hsa-miR-429 [41]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR429 sensitized gemcitabine response in GZ-resistant pancreatic cancer cells via its direct upregulation of PDCD4 expression.
Key Molecule: Pvt1 oncogene (PVT1) [42]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
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
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Muse Cell Cycle Assay; Muse Annexin V and Dead Cell Assay; MTT assay
Mechanism Description Curcumin sensitizes pancreatic cancer cells to gemcitabine by attenuating PRC2 subunit EZH2, and the LncRNA PVT1 expression.
Key Molecule: hsa-let-7a [43]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation CXCR4/let-7a/HMGA2 pathway Regulation hsa05206
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model HPDE6-C7 cells Pancreas Homo sapiens (Human) CVCL_0P38
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Transwell assay; Flow cytometric analysis
Mechanism Description CXCR4/Let-7a axis regulates metastasis and chemoresistance of pancreatic cancer cells through targeting HMGA2. overexpression of HMGA2 restores cell proliferation, metastasis and chemosensitivity of gem inhibited by let-7a.
Key Molecule: CXC chemokine receptor type 4 (CXCR4) [43]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation CXCR4/let-7a/HMGA2 pathway Regulation hsa05206
In Vitro Model HPDE6-C7 cells Pancreas Homo sapiens (Human) CVCL_0P38
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Transwell assay; Flow cytometric analysis
Mechanism Description CXCR4/Let-7a axis regulates metastasis and chemoresistance of pancreatic cancer cells through targeting HMGA2. overexpression of HMGA2 restores cell proliferation, metastasis and chemosensitivity of gem inhibited by let-7a.
Key Molecule: hsa-mir-205 [44]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MIA PaCa-2R cells Pancreas Homo sapiens (Human) CVCL_HA89
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description miR205 resensitizes GEM-resistant pancreatic cancer cells to GEM and acts as a tumor suppressor miRNA.
Key Molecule: hsa-mir-21 [45]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Panc02 cells Pancreas Homo sapiens (Human) CVCL_D627
In Vivo Model Mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Costar Transwell Invasion Assay;
Mechanism Description Upregulating miR21 in CAFs promoted PDAC desmoplasia and increased its drug resistance to gemcitabine treatment by promoting the activation of cancer-associated fibroblasts (CAFs). miR21 mediates activation of CAFs via down-regulating PDCD4.
Key Molecule: Programmed cell death protein 4 (PDCD4) [45]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Panc02 cells Pancreas Homo sapiens (Human) CVCL_D627
In Vivo Model Mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR; Immunofluorescence (IF) staining
Experiment for
Drug Resistance
Costar Transwell Invasion Assay;
Mechanism Description Upregulating miR21 in CAFs promoted PDAC desmoplasia and increased its drug resistance to gemcitabine treatment by promoting the activation of cancer-associated fibroblasts (CAFs). miR21 mediates activation of CAFs via down-regulating PDCD4.
Key Molecule: hsa-mir-153 [46]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
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-2 cells Pancreas Homo sapiens (Human) CVCL_0026
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
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay; Annexin-V/PI Apoptosis assay; TUNEL assay
Mechanism Description miR153 enhanced gemcitabine sensitivity by targeting Snail in pancreatic cancer.
Key Molecule: hsa-mir-101 [47]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Annexin V apoptosis assay; Caspase-3 activity assay
Mechanism Description microRNA-101 silences RNA-Pkcs and sensitizes pancreatic cancer cells to gemcitabine. AntagomiR101 expression causes RNA-Pkcs upregulation and gemcitabine resistance. miR101 expression inhibits Akt activation in PANC-1 cells.
Key Molecule: hsa-mir-210 [48]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
In Vivo Model Chick egg xenograft model Gallus gallus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
RealTime-Glo MT Cell Viability Assay; Caspase-3/7 substrate assay; Colony formation assay
Mechanism Description microRNA-210 overexpression inhibits tumor growth and potentially reverses gemcitabine resistance in pancreatic cancer, miR210 is a direct suppressor of the multidrug efflux transporter ABCC5.
Key Molecule: hsa-mir-145 [49]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic adenocarcinoma [ICD-11: 2C10.4]
Experimental Note Revealed Based on the Cell Line Data
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
RT-PCR; qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Transwell migration assay
Mechanism Description miRNA-145 increases therapeutic sensibility to gemcitabine treatment of pancreatic adenocarcinoma cells, miR145 negatively regulated p70S6k1 expression at the posttranscriptional level in colon cancer.
Key Molecule: hsa-miR-20a-5p [50]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-20a-5p inhibits protein expression of RRM2 and reverses gemcitabine resistance.
Key Molecule: hsa-mir-30a [51]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell colony Inhibition hsa05200
Cell proliferation Inhibition hsa05200
SNAI1/IRS1/AKT signaling pathway Regulation hsa04151
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-30a overexpression suppresses cell proliferation, and sensitizes pancreatic cancer cells to gemcitabine and miR-30a overexpression reduced IRS1 and SNAI1 protein level.
Key Molecule: hsa-miR-373-3p [52]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic carcinoma [ICD-11: 2C10.2]
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 PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-373-3p enhances the chemosensitivity of gemcitabine through cell cycle pathway by downregulating CCND2 in pancreatic carcinoma cells.
Key Molecule: hsa-mir-29c [53]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell colony Inhibition hsa05200
Cell viability Inhibition hsa05200
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-29c targets USP22 and suppresses autophagy-mediated chemoresistance in a xenograft tumor model in vivo.
Key Molecule: hsa-miR-101-3p [54]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
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
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description Long-term treatment of PDA cells with gemcitabine induced pronounced therapy resistance. The RRM1 gene is a major mediator of resistance and its expression is regulated by direct binding of miR-101-3p to two binding sites in the RRM1 3'UTR. The overexpression of miR-101-3p mimics inhibited the expression of RRM1 and partially reversed gemcitabine-resistance.
Key Molecule: hsa-let-7b [55]
Molecule Alteration Expression
Up-regulation
Sensitive Disease KRAS mutant pancreatic ductal adenocarcinoma [ICD-11: 2C10.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
MEK/ERK /PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
NCI-H1975 cells Lung Homo sapiens (Human) CVCL_1511
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Let-7b repletion selectively sensitized kRAS mutant tumor cells to the cytotoxicity of paclitaxel and gemcitabine. Transfection of let-7b mimic downregulated the expression of mutant but not wild-type kRAS. Combination of let-7b mimic with paclitaxel or gemcitabine diminished MEk/ERk and PI3k/AkT signaling concurrently, triggered the onset of apoptosis, and reverted the epithelial-mesenchymal transition in kRAS mutant tumor cells. In addition, let-7b repletion downregulated the expression of beta-tubulin III and ribonucleotide reductase subunit M2, two proteins known to mediate tumor resistance to paclitaxel and gemcitabine, respectively. Let-7 may represent a new class of chemosensitizer for the treatment of kRAS mutant tumors.
Key Molecule: hsa-mir-21 [56]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Hs-578T cells Breast Homo sapiens (Human) CVCL_0332
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Increased p85alpha expression in PDAC TCs results in decreased PI3k-AkT signaling and increased gemcitabine sensitivity. Expression of p85alpha inversely correlates with miR-21 levels in human PDAC. Overexpression of miR-21 results in decreased levels of p85alpha and increased PI3k-AkT activation.
Key Molecule: hsa-mir-497 [57]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
FGF/FGFR signaling pathway Inhibition hsa01521
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-497 suppressed cells proliferation, decreased the percentage of S phase cells, re-sensitized cells to gemcitabine and erlotinib, and attenuated migration and invasion capacities. Furthermore, fibroblast growth factor 2 and fibroblast growth factor receptor 1 were confirmed as miR-497 targets. Overexpression of miR-497 inhibited tumor growth in vivo. Additionally, miR-497 expression was significantly downregulated in pancreatic cancer tissues compared with tumor-adjacent samples. Low expression of miR-497 was an independent adverse prognostic factor of pancreatic cancer. miR-497 plays a role in modulating the malignant phenotype and chemosensitivity of pancreatic cancer cells by directly inhibition of FGF2 and FGFR1 expression.
Key Molecule: hsa-mir-211 [58]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model Suit2 cells Pancreas Homo sapiens (Human) CVCL_3172
SUIT2-007 cells Pancreas Homo sapiens (Human) CVCL_B279
SUIT2-028 cells Pancreas Homo sapiens (Human) CVCL_B282
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Transwell assay
Mechanism Description The induction of the miR-211 expression in the cells increased the sensitivity to gemcitabine and reduced the expression of its target ribonucleotide reductase subunit 2 (RRM2).
Key Molecule: hsa-mir-29a [59]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
Wnt/Beta-catenin signaling pathway Inhibition hsa04310
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
PSN1 cells Pancreas Homo sapiens (Human) CVCL_1644
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Our findings suggest that miR-29a expression correlates significantly with the growth-inhibitory effect of GEM and that activation of the Wnt/beta-catenin signaling pathway mediated the miR-29a-induced resistance to GEM in pancreatic cancer cell lines.
Key Molecule: hsa-mir-181 [60]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
CFPAC1 cells Pancreas Homo sapiens (Human) CVCL_1119
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description BCL-2 facilitates cell survival against chemotherapy via the blockage of Bax/Bak-induced apoptosis, miRNA-181b sensitizes PDAC cells to gemcitabine by targeting BCL-2.
Key Molecule: hsa-miR-142-5p [61]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model Capan-1 cells Pancreas Homo sapiens (Human) CVCL_0237
SUIT-2 cells Pancreas Homo sapiens (Human) CVCL_3172
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Propidium iodide assay
Mechanism Description High miR-142-5p expression was significantly associated with longer survival times in the gemcitabine group.
Key Molecule: Pvt1 oncogene (PVT1) [62]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
Experiment for
Molecule Alteration
PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Genome-wide screening identified PVT1 as a regulator for Gemcitabine sensitivity in human pancreatic cancer cells.
Key Molecule: hsa-mir-21 [63]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
HPAC cells Pancreas Homo sapiens (Human) CVCL_3517
HPAF-II cells Pancreatic Homo sapiens (Human) CVCL_0313
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
SRB (sulforhodamine-B) assay
Mechanism Description Low miR-21 expression was associated with benefit from adjuvant treatment in two independent cohorts of PDAC cases, and anti-miR-21 increased anticancer drug activity in vitro.
Key Molecule: hsa-mir-21 [64]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Capan-1 cells Pancreas Homo sapiens (Human) CVCL_0237
Capan-2 cells Pancreas Homo sapiens (Human) CVCL_0026
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
Hs-578T cells Breast Homo sapiens (Human) CVCL_0332
CFPAC1 cells Pancreas Homo sapiens (Human) CVCL_1119
SUIT-2 cells Pancreas Homo sapiens (Human) CVCL_3172
H48N cells Pancreas Homo sapiens (Human) CVCL_D554
KP-1N cells Pancreas Homo sapiens (Human) CVCL_3002
KP-2 cells Pancreas Homo sapiens (Human) CVCL_3004
KP-3 cells Pancreas Homo sapiens (Human) CVCL_3005
NOR-P1 cells Pancreas Homo sapiens (Human) CVCL_4716
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Propidium iodide assay
Mechanism Description The cancer cells transfected with the miR-21 precursor showed significantly increased proliferation, Matrigel invasion, and chemoresistance for gemcitabine compared with the control cells. In contrast, inhibition of miR-21 decreased proliferation, Matrigel invasion, and chemoresistance for gemcitabine. Moreover, miR-21 positively correlated with the mRNA expression of invasion-related genes, matrix metalloproteinase-2 and -9, and vascular endothelial growth factor.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: ATP-binding cassette sub-family C5 (ABCC5) [48]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
In Vivo Model Chick egg xenograft model Gallus gallus
Experiment for
Molecule Alteration
Dual luciferase assay; qRT-PCR; Immunofluorescence and immunohistochemistry assay
Experiment for
Drug Resistance
RealTime-Glo MT Cell Viability Assay; Caspase-3/7 substrate assay; Colony formation assay
Mechanism Description microRNA-210 overexpression inhibits tumor growth and potentially reverses gemcitabine resistance in pancreatic cancer, miR210 is a direct suppressor of the multidrug efflux transporter ABCC5.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: Growth arrest specific 5 (GAS5) [34]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
miR221/SOCS3 signaling pathway Regulation hsa05206
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Capan-2 cells Pancreas Homo sapiens (Human) CVCL_0026
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
In Vivo Model Nude 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 Overexpression of GAS5 inhibits proliferation, migration, and chemotherapy resistance by suppressing the emt and tumor stem cell-like properties. LncRNA GAS5 functioned as a competing endogenous RNA for miR-221, and it suppressed cell growth, metastasis, and gemcitabine resistance in PC by regulating the miR-221/SOCS3 pathway mediating EMT and tumor stem cell self-renewal.
Key Molecule: Suppressor of cytokine signaling 3 (SOCS3) [34]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
miR221/SOCS3 signaling pathway Regulation hsa05206
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Capan-2 cells Pancreas Homo sapiens (Human) CVCL_0026
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description SOCS3 overexpression reverses miR-221 overexpression-induced proliferation, migration, emt, chemotherapy resistance, and stem cell-like properties in panc-1 cells.
Key Molecule: hsa-mir-3656 [65]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Epithelial mesenchymal transition signaling pathway Activation hsa01521
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Capan-2 cells Pancreas Homo sapiens (Human) CVCL_0026
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
HPDE6-C7 cells Pancreas Homo sapiens (Human) CVCL_0P38
HTERT-HPNE cells Pancreas Homo sapiens (Human) CVCL_C466
PATU8988 cells Pancreas Homo sapiens (Human) CVCL_1846
CFPAC1 cells Pancreas Homo sapiens (Human) CVCL_1119
HPAC cells Pancreas Homo sapiens (Human) CVCL_3517
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR3656 expression enhances the chemosensitivity of pancreatic cancer to gemcitabine through modulation of the RHOF/EMT axis. miR3656 could target RHOF, a member of the Rho subfamily of small GTPases, and regulate the EMT process, enforced EMT progression via TWIST1 overexpression compromised the chemotherapy-enhancing effects of miR3656. Reduced miR3656 expression levels activated the EMT pathway through upregulation of RHOF, eventually causing drug resistance.
Key Molecule: Rho-related GTP-binding protein RhoF (RHOF) [65]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Epithelial mesenchymal transition signaling pathway Activation hsa01521
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Capan-2 cells Pancreas Homo sapiens (Human) CVCL_0026
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
HPDE6-C7 cells Pancreas Homo sapiens (Human) CVCL_0P38
HTERT-HPNE cells Pancreas Homo sapiens (Human) CVCL_C466
PATU8988 cells Pancreas Homo sapiens (Human) CVCL_1846
CFPAC1 cells Pancreas Homo sapiens (Human) CVCL_1119
HPAC cells Pancreas Homo sapiens (Human) CVCL_3517
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Dual luciferase reporter assay
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR3656 expression enhances the chemosensitivity of pancreatic cancer to gemcitabine through modulation of the RHOF/EMT axis. miR3656 could target RHOF, a member of the Rho subfamily of small GTPases, and regulate the EMT process, enforced EMT progression via TWIST1 overexpression compromised the chemotherapy-enhancing effects of miR3656. Reduced miR3656 expression levels activated the EMT pathway through upregulation of RHOF, eventually causing drug resistance.
Key Molecule: hsa-miR-1243 [66]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Epithelial mesenchymal transition signaling pathway Inhibition hsa01521
TGF-beta signaling pathway Inhibition hsa04350
In Vitro Model MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
Su.86.86 cells Pancreas Homo sapiens (Human) CVCL_3881
CFPAC1 cells Pancreas Homo sapiens (Human) CVCL_1119
KMP3 cells Pancreas Homo sapiens (Human) CVCL_8491
KP4-4 cells Pancreas Homo sapiens (Human) CVCL_Y142
Panc1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
WST-8 assay; Crystal violet staining assay
Mechanism Description Overexpression of miR509-5p and miR1243 increased the expression of E-cadherin through the suppression of EMT-related gene expression and that drug sensitivity increased with a combination of each of these miRNAs and gemcitabine. miR1243 directly regulated SMAD2 and SMAD4, which regulate the TGF-beta signaling pathway, resulting in an induction of the MET phenotype. Suppressing SMADs reduced the effect of TGF-beta.
Key Molecule: hsa-miR-509-5p [66]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Epithelial mesenchymal transition signaling pathway Inhibition hsa01521
TGF-beta signaling pathway Inhibition hsa04350
In Vitro Model MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
Su.86.86 cells Pancreas Homo sapiens (Human) CVCL_3881
CFPAC1 cells Pancreas Homo sapiens (Human) CVCL_1119
KMP3 cells Pancreas Homo sapiens (Human) CVCL_8491
KP4-4 cells Pancreas Homo sapiens (Human) CVCL_Y142
Panc1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
WST-8 assay; Crystal violet staining assay
Mechanism Description miR509-5p and miR1243 increase the sensitivity to gemcitabine by inhibiting epithelial-mesenchymal transition in pancreatic cancer miR509-5p induced an MET phenotype by directly regulating VIM and HMGA2.
Key Molecule: Mothers against decapentaplegic homolog 2 (SMAD2) [66]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Epithelial mesenchymal transition signaling pathway Inhibition hsa01521
TGF-beta signaling pathway Inhibition hsa04350
In Vitro Model MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
Su.86.86 cells Pancreas Homo sapiens (Human) CVCL_3881
CFPAC1 cells Pancreas Homo sapiens (Human) CVCL_1119
KMP3 cells Pancreas Homo sapiens (Human) CVCL_8491
KP4-4 cells Pancreas Homo sapiens (Human) CVCL_Y142
Panc1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
WST-8 assay; Crystal violet staining assay
Mechanism Description Overexpression of miR509-5p and miR1243 increased the expression of E-cadherin through the suppression of EMT-related gene expression and that drug sensitivity increased with a combination of each of these miRNAs and gemcitabine. miR1243 directly regulated SMAD2 and SMAD4, which regulate the TGF-beta signaling pathway, resulting in an induction of the MET phenotype. Suppressing SMADs reduced the effect of TGF-beta.
Key Molecule: Mothers against decapentaplegic homolog 4 (SMAD4) [66]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Epithelial mesenchymal transition signaling pathway Inhibition hsa01521
TGF-beta signaling pathway Inhibition hsa04350
In Vitro Model MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
Su.86.86 cells Pancreas Homo sapiens (Human) CVCL_3881
CFPAC1 cells Pancreas Homo sapiens (Human) CVCL_1119
KMP3 cells Pancreas Homo sapiens (Human) CVCL_8491
KP4-4 cells Pancreas Homo sapiens (Human) CVCL_Y142
Panc1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
WST-8 assay; Crystal violet staining assay
Mechanism Description Overexpression of miR509-5p and miR1243 increased the expression of E-cadherin through the suppression of EMT-related gene expression and that drug sensitivity increased with a combination of each of these miRNAs and gemcitabine. miR1243 directly regulated SMAD2 and SMAD4, which regulate the TGF-beta signaling pathway, resulting in an induction of the MET phenotype. Suppressing SMADs reduced the effect of TGF-beta.
Key Molecule: hsa-mir-155 [67]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Panc1 cells Pancreas Homo sapiens (Human) CVCL_0480
PSN1 cells Pancreas Homo sapiens (Human) CVCL_1644
In Vivo Model Mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The increase of miR155 induced two different functions; exosome secretion and chemoresistance ability via facilitating the anti-apoptotic activity.
Key Molecule: Tyrosine-protein kinase Fyn (FYN) [68]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
Cell viability Inhibition hsa05200
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
PATU8988T cells Pancreatic Homo sapiens (Human) CVCL_1847
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Transwell assay
Mechanism Description miR-125a-3p is responsible for chemosensitivity in PDAC by inhibiting epithelial-mesenchymal transition via Fyn.
Key Molecule: hsa-miR-125a-3p [68]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell viability Inhibition hsa05200
Epithelial mesenchymal transition signaling pathway Inhibition hsa01521
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
PATU8988T cells Pancreatic Homo sapiens (Human) CVCL_1847
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Transwell assay
Mechanism Description miR-125a-3p is responsible for chemosensitivity in PDAC by inhibiting epithelial-mesenchymal transition via Fyn.
Key Molecule: hsa-mir-223 [69]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Transwell migration and invasion assay
Mechanism Description Down-regulation of miR-223 reverses epithelial-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells due to down-regulation of its target Fbw7 and subsequent upregulation of Notch-1, which enhances GR cells to gemcitabine sensitivity.
Key Molecule: hsa-let-7b [70]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description The expression of miR-200b, miR-200c, let-7b, let-7c, let-7d, and let-7e was significantly down-regulated in gemcitabine-resistant cells that showed EMT characteristics such as elongated fibroblastoid morphology, lower expression of epithelial marker E-cadherin, and higher expression of mesenchymal markers such as vimentin and ZEB1.
Key Molecule: hsa-let-7c [70]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description The expression of miR-200b, miR-200c, let-7b, let-7c, let-7d, and let-7e was significantly down-regulated in gemcitabine-resistant cells that showed EMT characteristics such as elongated fibroblastoid morphology, lower expression of epithelial marker E-cadherin, and higher expression of mesenchymal markers such as vimentin and ZEB1.
Key Molecule: hsa-let-7d [70]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description The expression of miR-200b, miR-200c, let-7b, let-7c, let-7d, and let-7e was significantly down-regulated in gemcitabine-resistant cells that showed EMT characteristics such as elongated fibroblastoid morphology, lower expression of epithelial marker E-cadherin, and higher expression of mesenchymal markers such as vimentin and ZEB1.
Key Molecule: hsa-let-7e [70]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description The expression of miR-200b, miR-200c, let-7b, let-7c, let-7d, and let-7e was significantly down-regulated in gemcitabine-resistant cells that showed EMT characteristics such as elongated fibroblastoid morphology, lower expression of epithelial marker E-cadherin, and higher expression of mesenchymal markers such as vimentin and ZEB1.
Key Molecule: hsa-mir-200b [70]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description Re-expression of miR-200 in gemcitabine-resistant cells showed partial reversal of EMT characteristics as documented by increased expression of E-cadherin and decreased expression of vimentin, ZEB1, and slug. These results suggest that miR-200 family regulates the expression of ZEB1, slug, E-cadherin, and vimentin and that the re-expression of miR-200 could be useful for the reversal of EMT phenotype to mesenchymal-epithelial transition (MET). re-expression of miR-200 by transfection studies or treatment of gemcitabine-resistant cells with either DIM or isoflavone resulted in the down-regulation of ZEB1, slug, and vimentin, which was consistent with morphological reversal of EMT phenotype leading to epithelial morphology.
Key Molecule: hsa-mir-200c [70]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description Re-expression of miR-200 in gemcitabine-resistant cells showed partial reversal of EMT characteristics as documented by increased expression of E-cadherin and decreased expression of vimentin, ZEB1, and slug. These results suggest that miR-200 family regulates the expression of ZEB1, slug, E-cadherin, and vimentin and that the re-expression of miR-200 could be useful for the reversal of EMT phenotype to mesenchymal-epithelial transition (MET). re-expression of miR-200 by transfection studies or treatment of gemcitabine-resistant cells with either DIM or isoflavone resulted in the down-regulation of ZEB1, slug, and vimentin, which was consistent with morphological reversal of EMT phenotype leading to epithelial morphology.
Key Molecule: Zinc finger E-box-binding homeobox 1 (ZEB1) [70]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description Re-expression of miR-200 in gemcitabine-resistant cells showed partial reversal of EMT characteristics as documented by increased expression of E-cadherin and decreased expression of vimentin, ZEB1, and slug. These results suggest that miR-200 family regulates the expression of ZEB1, slug, E-cadherin, and vimentin and that the re-expression of miR-200 could be useful for the reversal of EMT phenotype to mesenchymal-epithelial transition (MET). re-expression of miR-200 by transfection studies or treatment of gemcitabine-resistant cells with either DIM or isoflavone resulted in the down-regulation of ZEB1, slug, and vimentin, which was consistent with morphological reversal of EMT phenotype leading to epithelial morphology.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Cadherin-1 (CDH1) [19]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic carcinoma [ICD-11: 2C10.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Capan-2 cells Pancreas Homo sapiens (Human) CVCL_0026
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Colorimetric methylene blue assay; Flow cytometry assay
Mechanism Description Forced expression of miR-200b induces CDH1 expression and promotes gemcitabine sensitivity in Capan-2 and Panc-1 cells.
Key Molecule: Transforming protein RhoA (RHOA) [39]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
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
Su.86.86 cells Pancreas Homo sapiens (Human) CVCL_3881
In Vivo Model Engrafted tumor mouse model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; qRT-PCR; IHC analyses
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description RhoA inhibition leads to improved efficacy of gemcitabine in PC cells.
Key Molecule: Proto-oncogene tyrosine-protein kinase Src (SRC) [39]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
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
Su.86.86 cells Pancreas Homo sapiens (Human) CVCL_3881
In Vivo Model Engrafted tumor mouse model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; qRT-PCR; IHC analyses
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description SRC inhibition leads to improved efficacy of gemcitabine in PC cells.
Key Molecule: High mobility group protein HMGI-C (HMGA2) [66], [43]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation CXCR4/let-7a/HMGA2 pathway Regulation hsa05206
In Vitro Model HPDE6-C7 cells Pancreas Homo sapiens (Human) CVCL_0P38
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR; Western blot analysis; Luciferase reporter assay
Experiment for
Drug Resistance
MTT assay; Transwell assay; Flow cytometric analysis
Mechanism Description CXCR4/Let-7a axis regulates metastasis and chemoresistance of pancreatic cancer cells through targeting HMGA2. overexpression of HMGA2 restores cell proliferation, metastasis and chemosensitivity of gem inhibited by let-7a.
Key Molecule: Bcl-2-binding component 3 (BBC3) [40]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Slug/PUMA signaling pathway Activation hsa04390
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
RT-PCR; Western blot analysis
Experiment for
Drug Resistance
Flow Cytometric Analysis, MTT assay; TUNEL staining
Mechanism Description miR34 induces Slug-mediated upregulation of PUMA expression. miR34 sensitizes to gemcitabine-mediated apoptosis by PUMA upregulation.
Key Molecule: Zinc finger protein SNAI2 (SNAI2) [40]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Slug/PUMA signaling pathway Regulation hsa04390
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
RT-PCR; Western blot analysis
Experiment for
Drug Resistance
Flow Cytometric Analysis, MTT assay; TUNEL staining
Mechanism Description miR34 increases in vitro PANC-1 cell sensitivity to gemcitabine via targeting Slug/PUMA. miR34 enhances sensitization against gemcitabine-mediated apoptosis through the down-regulation of Slug expression, and up-regulation of Slug-dependent PUMA expression.
Key Molecule: Programmed cell death protein 4 (PDCD4) [41]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR429 sensitized gemcitabine response in GZ-resistant pancreatic cancer cells via its direct upregulation of PDCD4 expression.
Key Molecule: Zinc finger protein SNAI1 (SNAI1) [46]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
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-2 cells Pancreas Homo sapiens (Human) CVCL_0026
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
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay; Annexin-V/PI Apoptosis assay; TUNEL assay
Mechanism Description miR153 enhanced gemcitabine sensitivity by targeting Snail in pancreatic cancer.
Key Molecule: DNA-dependent catalytic protein kinase (PRKDC) [47]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Annexin V apoptosis assay; Caspase-3 activity assay
Mechanism Description microRNA-101 silences RNA-Pkcs and sensitizes pancreatic cancer cells to gemcitabine. AntagomiR101 expression causes RNA-Pkcs upregulation and gemcitabine resistance. miR101 expression inhibits Akt activation in PANC-1 cells.
Key Molecule: Ribosomal protein S6 kinase beta-1 (RPS6KB1) [49]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic adenocarcinoma [ICD-11: 2C10.4]
Experimental Note Revealed Based on the Cell Line Data
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
CCK8 assay; Transwell migration assay
Mechanism Description miRNA-145 increases therapeutic sensibility to gemcitabine treatment of pancreatic adenocarcinoma cells, miR145 negatively regulated p70S6k1 expression at the posttranscriptional level in colon cancer.
Key Molecule: Ribonucleoside-diphosphate reductase subunit M2 (RRM2) [50]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-20a-5p inhibits protein expression of RRM2 and reverses gemcitabine resistance.
Key Molecule: Insulin receptor substrate 1 (IRS1) [51]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell colony Inhibition hsa05200
Cell proliferation Inhibition hsa05200
SNAI1/IRS1/AKT signaling pathway Regulation hsa04151
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-30a overexpression suppresses cell proliferation, and sensitizes pancreatic cancer cells to gemcitabine and miR-30a overexpression reduced IRS1 and SNAI1 protein level.
Key Molecule: Zinc finger protein SNAI1 (SNAI1) [51]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell colony Inhibition hsa05200
Cell proliferation Inhibition hsa05200
SNAI1/IRS1/AKT signaling pathway Regulation hsa04151
In Vitro Model BxPC-3 cells Pancreas Homo sapiens (Human) CVCL_0186
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-30a overexpression suppresses cell proliferation, and sensitizes pancreatic cancer cells to gemcitabine and miR-30a overexpression reduced IRS1 and SNAI1 protein level.
Key Molecule: G1/S-specific cyclin-D2 (CCND2) [52]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic carcinoma [ICD-11: 2C10.2]
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 viability Inhibition hsa05200
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-373-3p enhances the chemosensitivity of gemcitabine through cell cycle pathway by downregulating CCND2 in pancreatic carcinoma cells.
Key Molecule: Ubiquitin carboxyl-terminal hydrolase 22 (USP22) [53]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell colony Inhibition hsa05200
Cell viability Inhibition hsa05200
In Vitro Model PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; RIP assay; Luciferase reporter assay
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-29c targets USP22 and suppresses autophagy-mediated chemoresistance in a xenograft tumor model in vivo.
Key Molecule: Ribonucleoside-diphosphate reductase large subunit (RRM1) [54]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
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
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description Long-term treatment of PDA cells with gemcitabine induced pronounced therapy resistance. The RRM1 gene is a major mediator of resistance and its expression is regulated by direct binding of miR-101-3p to two binding sites in the RRM1 3'UTR. The overexpression of miR-101-3p mimics inhibited the expression of RRM1 and partially reversed gemcitabine-resistance.
Key Molecule: Ribonucleoside-diphosphate reductase subunit M2 (RRM2) [55]
Molecule Alteration Expression
Down-regulation
Sensitive Disease KRAS mutant pancreatic ductal adenocarcinoma [ICD-11: 2C10.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
MEK/ERK /PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Let-7b repletion selectively sensitized kRAS mutant tumor cells to the cytotoxicity of paclitaxel and gemcitabine. Transfection of let-7b mimic downregulated the expression of mutant but not wild-type kRAS. Combination of let-7b mimic with paclitaxel or gemcitabine diminished MEk/ERk and PI3k/AkT signaling concurrently, triggered the onset of apoptosis, and reverted the epithelial-mesenchymal transition in kRAS mutant tumor cells. In addition, let-7b repletion downregulated the expression of beta-tubulin III and ribonucleotide reductase subunit M2, two proteins known to mediate tumor resistance to paclitaxel and gemcitabine, respectively. Let-7 may represent a new class of chemosensitizer for the treatment of kRAS mutant tumors.
Key Molecule: Tubulin beta-3 chain (TUBB3) [55]
Molecule Alteration Expression
Down-regulation
Sensitive Disease KRAS mutant pancreatic ductal adenocarcinoma [ICD-11: 2C10.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
MEK/ERK /PI3K/AKT signaling pathway Inhibition hsa04151
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 Let-7b repletion selectively sensitized kRAS mutant tumor cells to the cytotoxicity of paclitaxel and gemcitabine. Transfection of let-7b mimic downregulated the expression of mutant but not wild-type kRAS. Combination of let-7b mimic with paclitaxel or gemcitabine diminished MEk/ERk and PI3k/AkT signaling concurrently, triggered the onset of apoptosis, and reverted the epithelial-mesenchymal transition in kRAS mutant tumor cells. In addition, let-7b repletion downregulated the expression of beta-tubulin III and ribonucleotide reductase subunit M2, two proteins known to mediate tumor resistance to paclitaxel and gemcitabine, respectively. Let-7 may represent a new class of chemosensitizer for the treatment of kRAS mutant tumors.
Key Molecule: PI3-kinase regulatory subunit alpha (PIK3R1) [56]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
Hs-578T cells Breast Homo sapiens (Human) CVCL_0332
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Increased p85alpha expression in PDAC TCs results in decreased PI3k-AkT signaling and increased gemcitabine sensitivity. Expression of p85alpha inversely correlates with miR-21 levels in human PDAC. Overexpression of miR-21 results in decreased levels of p85alpha and increased PI3k-AkT activation.
Key Molecule: F-box/WD repeat-containing protein 7 (FBXW7) [69]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
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 PANC-1 cells Pancreas Homo sapiens (Human) CVCL_0480
AsPC-1 cells Pancreas Homo sapiens (Human) CVCL_0152
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Transwell migration and invasion assay
Mechanism Description Down-regulation of miR-223 reverses epithelial-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells due to down-regulation of its target Fbw7 and subsequent upregulation of Notch-1, which enhances GR cells to gemcitabine sensitivity.
Key Molecule: Fibroblast growth factor 2 (FGF1) [57]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
FGF/FGFR signaling pathway Inhibition hsa01521
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-497 suppressed cells proliferation, decreased the percentage of S phase cells, re-sensitized cells to gemcitabine and erlotinib, and attenuated migration and invasion capacities. Furthermore, fibroblast growth factor 2 and fibroblast growth factor receptor 1 were confirmed as miR-497 targets. Overexpression of miR-497 inhibited tumor growth in vivo. Additionally, miR-497 expression was significantly downregulated in pancreatic cancer tissues compared with tumor-adjacent samples. Low expression of miR-497 was an independent adverse prognostic factor of pancreatic cancer. miR-497 plays a role in modulating the malignant phenotype and chemosensitivity of pancreatic cancer cells by directly inhibition of FGF2 and FGFR1 expression.
Key Molecule: Fibroblast growth factor receptor 1 (FGFR1) [57]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
FGF/FGFR signaling pathway Inhibition hsa01521
In Vitro Model MIA PaCa-2 cells Pancreas Homo sapiens (Human) CVCL_0428
SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-497 suppressed cells proliferation, decreased the percentage of S phase cells, re-sensitized cells to gemcitabine and erlotinib, and attenuated migration and invasion capacities. Furthermore, fibroblast growth factor 2 and fibroblast growth factor receptor 1 were confirmed as miR-497 targets. Overexpression of miR-497 inhibited tumor growth in vivo. Additionally, miR-497 expression was significantly downregulated in pancreatic cancer tissues compared with tumor-adjacent samples. Low expression of miR-497 was an independent adverse prognostic factor of pancreatic cancer. miR-497 plays a role in modulating the malignant phenotype and chemosensitivity of pancreatic cancer cells by directly inhibition of FGF2 and FGFR1 expression.
Key Molecule: Ribonucleoside-diphosphate reductase subunit M2 (RRM2) [58]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model Suit2 cells Pancreas Homo sapiens (Human) CVCL_3172
SUIT2-007 cells Pancreas Homo sapiens (Human) CVCL_B279
SUIT2-028 cells Pancreas Homo sapiens (Human) CVCL_B282
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Transwell assay
Mechanism Description The induction of the miR-211 expression in the cells increased the sensitivity to gemcitabine and reduced the expression of its target ribonucleotide reductase subunit 2 (RRM2).
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [60]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model SW1990 cells Pancreas Homo sapiens (Human) CVCL_1723
CFPAC1 cells Pancreas Homo sapiens (Human) CVCL_1119
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description BCL-2 facilitates cell survival against chemotherapy via the blockage of Bax/Bak-induced apoptosis, miRNA-181b sensitizes PDAC cells to gemcitabine by targeting BCL-2.
Liver cancer [ICD-11: 2C12]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-130a-3p [10]
Molecule Alteration Expression
Up-regulation
Resistant Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model CCLP-1 cells Liver Homo sapiens (Human) CVCL_0205
MzChA-1 cells Liver Homo sapiens (Human) CVCL_6932
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Transfection of miR130a-3p mimic suppressed the expression of PPARG and increased gemcitabine resistance.
Key Molecule: Peroxisome proliferator-activated receptor gamma (PPARG) [10]
Molecule Alteration Expression
Down-regulation
Resistant Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model CCLP-1 cells Liver Homo sapiens (Human) CVCL_0205
MzChA-1 cells Liver Homo sapiens (Human) CVCL_6932
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Transfection of miR130a-3p mimic suppressed the expression of PPARG and increased gemcitabine resistance.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-205 [71]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HuCCT1 cells Bile duct Homo sapiens (Human) CVCL_0324
HuH28 cells Bile duct Homo sapiens (Human) CVCL_2955
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-205 could conferred Gem sensitivity to innately Gem-resistant CCA cells.
Key Molecule: hsa-mir-221 [71]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HuCCT1 cells Bile duct Homo sapiens (Human) CVCL_0324
HuH28 cells Bile duct Homo sapiens (Human) CVCL_2955
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-221 was downregulated in Gem-resistant HuH28 cells, and that it acted as a potent enhancer of Gem sensitivity, at least partly, by downregulating PIk3R1 expression.
Key Molecule: hsa-mir-29b [71]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HuCCT1 cells Bile duct Homo sapiens (Human) CVCL_0324
HuH28 cells Bile duct Homo sapiens (Human) CVCL_2955
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Two miR-29b target genes, PIk3R1 and MMP-2, that are, at least partly, responsible for the resistance of CCA Gem treatment. PIk3R1 encodes phosphoinositide 3-kinase (PI3k) regulatory subunit designated p85 alpha; p85 alpha is regarded as integrator of multiple signaling pathways that together promote cell proliferation, cell survival, and carcinogenesis.
Key Molecule: hsa-mir-200b [72]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
In Vitro Model H69 cells Lung Homo sapiens (Human) CVCL_8121
KMCH-1 cells Gallbladder Homo sapiens (Human) CVCL_7970
Mz-ChA-1 cells Gallbladder Homo sapiens (Human) CVCL_6932
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Northern blotting analysis
Experiment for
Drug Resistance
Celltiter 96 aqueous one solution cell proliferation assay
Mechanism Description PTPN12 can bind and dephosphorylate the product ofoncogenes such as c-Abl or Src and inactivate the Raspathway. Thus, deregulation of PTPN12 expressionmay contribute to tumor cell survival and oncogenesis. In cells transfected with anti-miR-200b, PTPN12 ex-pression was increased to 132.2%+/-7.2% of controlafter 48 hours and 147.3%+/-12.8% of control after 72hours. Moreover, inhibition of miR-200b significantlyreduced the tyrosine phosphorylation of a downstreamtarget Src, a key mediator of tumor cell proliferation anddifferentiation.
Key Molecule: hsa-mir-21 [72]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
PI3K signaling pathway Activation hsa04151
In Vitro Model H69 cells Lung Homo sapiens (Human) CVCL_8121
KMCH-1 cells Gallbladder Homo sapiens (Human) CVCL_7970
Mz-ChA-1 cells Gallbladder Homo sapiens (Human) CVCL_6932
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Northern blotting analysis
Experiment for
Drug Resistance
Celltiter 96 aqueous one solution cell proliferation assay
Mechanism Description miR-21, miR-141, and miR-200b werehighly over-expressed in malignant cholangiocytes. Inhibi-tion of miR-21 and miR-200b increased sensitivity to gem-citabine, whereas inhibition of miR-141 decreased cellgrowth. miR-21 modulates gemcitabine-induced apo-ptosis by phosphatase and tensin homolog deleted onchromosome 10 (PTEN) -dependent activation of PI 3-ki-nase signaling.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: Collagenase 72 kDa type IV collagenase (MMP2) [71]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HuCCT1 cells Bile duct Homo sapiens (Human) CVCL_0324
HuH28 cells Bile duct Homo sapiens (Human) CVCL_2955
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Two miR-29b target genes, PIk3R1 and MMP-2, that are, at least partly, responsible for the resistance of CCA Gem treatment. PIk3R1 encodes phosphoinositide 3-kinase (PI3k) regulatory subunit designated p85 alpha; p85 alpha is regarded as integrator of multiple signaling pathways that together promote cell proliferation, cell survival, and carcinogenesis.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: PI3-kinase regulatory subunit alpha (PIK3R1) [71]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HuCCT1 cells Bile duct Homo sapiens (Human) CVCL_0324
HuH28 cells Bile duct Homo sapiens (Human) CVCL_2955
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Two miR-29b target genes, PIk3R1 and MMP-2, that are, at least partly, responsible for the resistance of CCA Gem treatment. PIk3R1 encodes phosphoinositide 3-kinase (PI3k) regulatory subunit designated p85 alpha; p85 alpha is regarded as integrator of multiple signaling pathways that together promote cell proliferation, cell survival, and carcinogenesis.
Key Molecule: Phosphatase and tensin homolog (PTEN) [72]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
PI3K signaling pathway Activation hsa04151
In Vitro Model H69 cells Lung Homo sapiens (Human) CVCL_8121
KMCH-1 cells Gallbladder Homo sapiens (Human) CVCL_7970
Mz-ChA-1 cells Gallbladder Homo sapiens (Human) CVCL_6932
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Celltiter 96 aqueous one solution cell proliferation assay
Mechanism Description miR-21, miR-141, and miR-200b werehighly over-expressed in malignant cholangiocytes. Inhibi-tion of miR-21 and miR-200b increased sensitivity to gem-citabine, whereas inhibition of miR-141 decreased cellgrowth. miR-21 modulates gemcitabine-induced apo-ptosis by phosphatase and tensin homolog deleted onchromosome 10 (PTEN) -dependent activation of PI 3-ki-nase signaling.
Key Molecule: Tyrosine-protein phosphatase non-receptor type 12 (PTPN12) [72]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
In Vitro Model H69 cells Lung Homo sapiens (Human) CVCL_8121
KMCH-1 cells Gallbladder Homo sapiens (Human) CVCL_7970
Mz-ChA-1 cells Gallbladder Homo sapiens (Human) CVCL_6932
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Celltiter 96 aqueous one solution cell proliferation assay
Mechanism Description PTPN12 can bind and dephosphorylate the product ofoncogenes such as c-Abl or Src and inactivate the Raspathway. Thus, deregulation of PTPN12 expressionmay contribute to tumor cell survival and oncogenesis. In cells transfected with anti-miR-200b, PTPN12 ex-pression was increased to 132.2%+/-7.2% of controlafter 48 hours and 147.3%+/-12.8% of control after 72hours. Moreover, inhibition of miR-200b significantlyreduced the tyrosine phosphorylation of a downstreamtarget Src, a key mediator of tumor cell proliferation anddifferentiation.
Gallbladder cancer [ICD-11: 2C13]
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-218-5p [73]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Gallbladder cancer [ICD-11: 2C13.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PRKCE/MDR1 signaling pathway Inhibition hsa05206
In Vitro Model GBC-SD cells Gallbladder Homo sapiens (Human) CVCL_6903
NOZ cells Gallbladder Homo sapiens (Human) CVCL_3079
SGC-996 cells Gallbladder Homo sapiens (Human) CVCL_M737
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay; Annexin V assay
Mechanism Description miR218-5p restores sensitivity to gemcitabine through PRkCE/MDR1 axis in gallbladder cancer, miR218-5p promotes sensitivity of gemcitabine by abolishing PRkCE-induced upregulation of MDR1/P-gp.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Protein kinase C epsilon type (PRKCE) [73]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gallbladder cancer [ICD-11: 2C13.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PRKCE/MDR1 signaling pathway Inhibition hsa05206
In Vitro Model GBC-SD cells Gallbladder Homo sapiens (Human) CVCL_6903
NOZ cells Gallbladder Homo sapiens (Human) CVCL_3079
SGC-996 cells Gallbladder Homo sapiens (Human) CVCL_M737
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Immunoblot assay; Dual-luciferase reporter assay
Experiment for
Drug Resistance
MTS assay; Annexin V assay
Mechanism Description miR218-5p restores sensitivity to gemcitabine through PRkCE/MDR1 axis in gallbladder cancer, miR218-5p promotes sensitivity of gemcitabine by abolishing PRkCE-induced upregulation of MDR1/P-gp.
Biliary tract cancer [ICD-11: 2C15]
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: Midkine (MDK) [7]
Molecule Alteration Expression
Up-regulation
Resistant Disease Biliary tract cancer [ICD-11: 2C15.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model GBC-SD cells Gallbladder Homo sapiens (Human) CVCL_6903
RBE cells Liver Homo sapiens (Human) CVCL_4896
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Mechanism Description BTC cell lines were more resistant to gemcitabine plus MDK compared with gemcitabine alone. In terms of the underlying mechanism, MDK promoted the epithelial to mesenchymal transition (EMT) of BTC cells and the enhancing effect of MDK on gemcitabine resistance was abrogated when the EMT was blocked with small interfering (si)RNA targeting Twist. In addition, MDK promoted the expression of Notch-1, while knockdown of Notch-1 by siRNA blocked the EMT process in the BTC cell lines.
Lung cancer [ICD-11: 2C25]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-223 [3]
Molecule Alteration Expression
Down-regulation
Resistant Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
IGF1R/AKT/PI3K signaling pathway Activation hsa05224
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H1299 cells Lung Homo sapiens (Human) CVCL_0060
NCI-H358 cells Lung Homo sapiens (Human) CVCL_1559
NCI-H292 cells Lung Homo sapiens (Human) CVCL_0455
NCI-H460 cells Lung Homo sapiens (Human) CVCL_0459
NCI-H838 cells Lung Homo sapiens (Human) CVCL_1594
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Long non-coding RNA EGFR-AS1 Can enhance IGF1R expression by suppressing miR-223 expression to promotes gemcitabine resistance in the non-small cell lung cancer.
Key Molecule: EGFR antisense RNA 1 (EGFR-AS1) [3]
Molecule Alteration Expression
Up-regulation
Resistant Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
IGF1R/AKT/PI3K signaling pathway Activation hsa05224
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H1299 cells Lung Homo sapiens (Human) CVCL_0060
NCI-H358 cells Lung Homo sapiens (Human) CVCL_1559
NCI-H292 cells Lung Homo sapiens (Human) CVCL_0455
NCI-H460 cells Lung Homo sapiens (Human) CVCL_0459
NCI-H838 cells Lung Homo sapiens (Human) CVCL_1594
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Long non-coding RNA EGFR-AS1 Can enhance IGF1R expression by suppressing miR-223 expression to promotes gemcitabine resistance in the non-small cell lung cancer.
Key Molecule: Insulin-like growth factor 1 receptor (IGF1R) [3]
Molecule Alteration Expression
Up-regulation
Resistant Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
IGF1R/AKT/PI3K signaling pathway Activation hsa05224
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H1299 cells Lung Homo sapiens (Human) CVCL_0060
NCI-H358 cells Lung Homo sapiens (Human) CVCL_1559
NCI-H292 cells Lung Homo sapiens (Human) CVCL_0455
NCI-H460 cells Lung Homo sapiens (Human) CVCL_0459
NCI-H838 cells Lung Homo sapiens (Human) CVCL_1594
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Long non-coding RNA EGFR-AS1 Can enhance IGF1R expression by suppressing miR-223 expression to promotes gemcitabine resistance in the non-small cell lung cancer.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: ATPase H+ transporting V0 subunit d1 (ATP6V0D1) [74]
Molecule Alteration Expression
Up-regulation
Resistant Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Identified from the Human Clinical Data
In Vitro Model Non-small cell lung cancer isolates Lung Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Immunofluorescence assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description The drug resistance of cancer cells is likely to be related to the changes in pH gradient between the extracellular environment and the cytoplasm.Vacuolar-H+ -ATPase(V-ATPase) plays a major role in the regulation of cellular pH conditions.The expression of V-ATPase was shown to be related to the pathological type and grade of the cancer and might be associated with the chemotherapy drug resistance in NSCLC.
Key Molecule: ATPase H+ transporting V0 subunit d1 (ATP6V0D1) [74]
Molecule Alteration Expression
Up-regulation
Resistant Disease Lung squamous cell carcinoma [ICD-11: 2C25.3]
Experimental Note Identified from the Human Clinical Data
In Vitro Model Non-small cell lung cancer isolates Lung Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Immunofluorescence assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description The drug resistance of cancer cells is likely to be related to the changes in pH gradient between the extracellular environment and the cytoplasm.Vacuolar-H+ -ATPase(V-ATPase) plays a major role in the regulation of cellular pH conditions.The expression of V-ATPase was shown to be related to the pathological type and grade of the cancer and might be associated with the chemotherapy drug resistance in NSCLC.
Key Molecule: ATPase H+ transporting V0 subunit d1 (ATP6V0D1) [74]
Molecule Alteration Expression
Up-regulation
Resistant Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Identified from the Human Clinical Data
In Vitro Model Non-small cell lung cancer isolates Lung Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Immunofluorescence assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description The drug resistance of cancer cells is likely to be related to the changes in pH gradient between the extracellular environment and the cytoplasm.Vacuolar-H+ -ATPase(V-ATPase) plays a major role in the regulation of cellular pH conditions.The expression of V-ATPase was shown to be related to the pathological type and grade of the cancer and might be associated with the chemotherapy drug resistance in NSCLC.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-let-7b [55]
Molecule Alteration Expression
Up-regulation
Sensitive Disease kRAS mutant non-small cell lung cancer [ICD-11: 2C25.9]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
MEK/ERK /PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
NCI-H1975 cells Lung Homo sapiens (Human) CVCL_1511
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Let-7b repletion selectively sensitized kRAS mutant tumor cells to the cytotoxicity of paclitaxel and gemcitabine. Transfection of let-7b mimic downregulated the expression of mutant but not wild-type kRAS. Combination of let-7b mimic with paclitaxel or gemcitabine diminished MEk/ERk and PI3k/AkT signaling concurrently, triggered the onset of apoptosis, and reverted the epithelial-mesenchymal transition in kRAS mutant tumor cells. In addition, let-7b repletion downregulated the expression of beta-tubulin III and ribonucleotide reductase subunit M2, two proteins known to mediate tumor resistance to paclitaxel and gemcitabine, respectively. Let-7 may represent a new class of chemosensitizer for the treatment of kRAS mutant tumors.
Key Molecule: hsa-mir-330 [16]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H460 cells Lung Homo sapiens (Human) CVCL_0459
SW1573 cells Lung Homo sapiens (Human) CVCL_1720
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Sulforhodamide B (SRB) test assay
Mechanism Description Deoxycytidine kinase (dCk) is essential for phosphorylation of natural deoxynucleosides andanalogs, such as gemcitabine and cytarabine, two widely used anticancer compounds. miR-330 expression negatively correlated withdCk mRNA expression, suggesting a role of miR-330 in post-transcriptional regulationof dCk. Expression of miR-330 in various colon and lung cancer cell lines,as measured by QRT-PCR, varied five-fold between samples and correlated with in-vitro gemcitabineresistance.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Ribonucleoside-diphosphate reductase subunit M2 (RRM2) [55]
Molecule Alteration Expression
Down-regulation
Sensitive Disease kRAS mutant non-small cell lung cancer [ICD-11: 2C25.9]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
MEK/ERK /PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Let-7b repletion selectively sensitized kRAS mutant tumor cells to the cytotoxicity of paclitaxel and gemcitabine. Transfection of let-7b mimic downregulated the expression of mutant but not wild-type kRAS. Combination of let-7b mimic with paclitaxel or gemcitabine diminished MEk/ERk and PI3k/AkT signaling concurrently, triggered the onset of apoptosis, and reverted the epithelial-mesenchymal transition in kRAS mutant tumor cells. In addition, let-7b repletion downregulated the expression of beta-tubulin III and ribonucleotide reductase subunit M2, two proteins known to mediate tumor resistance to paclitaxel and gemcitabine, respectively. Let-7 may represent a new class of chemosensitizer for the treatment of kRAS mutant tumors.
Key Molecule: Tubulin beta-3 chain (TUBB3) [55]
Molecule Alteration Expression
Down-regulation
Sensitive Disease kRAS mutant non-small cell lung cancer [ICD-11: 2C25.9]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
MEK/ERK /PI3K/AKT signaling pathway Inhibition hsa04151
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 Let-7b repletion selectively sensitized kRAS mutant tumor cells to the cytotoxicity of paclitaxel and gemcitabine. Transfection of let-7b mimic downregulated the expression of mutant but not wild-type kRAS. Combination of let-7b mimic with paclitaxel or gemcitabine diminished MEk/ERk and PI3k/AkT signaling concurrently, triggered the onset of apoptosis, and reverted the epithelial-mesenchymal transition in kRAS mutant tumor cells. In addition, let-7b repletion downregulated the expression of beta-tubulin III and ribonucleotide reductase subunit M2, two proteins known to mediate tumor resistance to paclitaxel and gemcitabine, respectively. Let-7 may represent a new class of chemosensitizer for the treatment of kRAS mutant tumors.
Key Molecule: Deoxycytidine kinase (DCK) [16]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H460 cells Lung Homo sapiens (Human) CVCL_0459
SW1573 cells Lung Homo sapiens (Human) CVCL_1720
Experiment for
Molecule Alteration
qRT -PCR
Experiment for
Drug Resistance
Sulforhodamide B (SRB) test assay
Mechanism Description Deoxycytidine kinase (dCk) is essential for phosphorylation of natural deoxynucleosides andanalogs, such as gemcitabine and cytarabine, two widely used anticancer compounds. miR-330 expression negatively correlated withdCk mRNA expression, suggesting a role of miR-330 in post-transcriptional regulationof dCk. Expression of miR-330 in various colon and lung cancer cell lines,as measured by QRT-PCR, varied five-fold between samples and correlated with in-vitro gemcitabineresistance.
Pleural mesothelioma [ICD-11: 2C26]
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-16 [75]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Malignant pleural mesothelioma [ICD-11: 2C26.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MET-5A cells Lung Homo sapiens (Human) CVCL_3749
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Colony formation assay
Mechanism Description Growth inhibition caused by miR-16 correlated with downregulation of target genes including Bcl-2 and CCND1, and miR-16 re-expression sensitised MPM cells to pemetrexed and gemcitabine.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [75]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Malignant pleural mesothelioma [ICD-11: 2C26.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MET-5A cells Lung Homo sapiens (Human) CVCL_3749
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay; Colony formation assay
Mechanism Description Growth inhibition caused by miR-16 correlated with downregulation of target genes including Bcl-2 and CCND1, and miR-16 re-expression sensitised MPM cells to pemetrexed and gemcitabine.
Key Molecule: G1/S-specific cyclin-D1 (CCND1) [75]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Malignant pleural mesothelioma [ICD-11: 2C26.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MET-5A cells Lung Homo sapiens (Human) CVCL_3749
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay; Colony formation assay
Mechanism Description Growth inhibition caused by miR-16 correlated with downregulation of target genes including Bcl-2 and CCND1, and miR-16 re-expression sensitised MPM cells to pemetrexed and gemcitabine.
Breast cancer [ICD-11: 2C60]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-620 [1]
Molecule Alteration Expression
Up-regulation
Resistant Disease Triple negative breast cancer [ICD-11: 2C60.9]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Overexpression of microRNA-620 facilitates the resistance of triple negative breast cancer cells to gemcitabine treatment by targeting DCTD.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: Cyclin-G2 (CCNG2) [9]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Exosomal microRNA miR1246 promotes cell proliferation, invasion and drug resistance by suppressing the expression level of CCNG2 in Breast Cancer.
Key Molecule: hsa-miR-1246 [9]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Exosomal microRNA miR1246 promotes cell proliferation, invasion and drug resistance by suppressing the expression level of CCNG2 in Breast Cancer.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Deoxycytidylate deaminase (DCTD) [1]
Molecule Alteration Expression
Down-regulation
Resistant Disease Triple negative breast cancer [ICD-11: 2C60.9]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Overexpression of microRNA-620 facilitates the resistance of triple negative breast cancer cells to gemcitabine treatment by targeting DCTD.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-200a [76]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation p73-mediated apoptosis signaling pathway Inhibition hsa04210
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
SkBR3 cells Breast Homo sapiens (Human) CVCL_0033
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
T47D cells Breast Homo sapiens (Human) CVCL_0553
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
BT-549 Breast Homo sapiens (Human) CVCL_1092
MCF-10A Breast Homo sapiens (Human) CVCL_0598
MDA-MB-436 cells Breast Homo sapiens (Human) CVCL_0623
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
ZR-75-30 cells Breast Homo sapiens (Human) CVCL_1661
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
TUNEL assays
Mechanism Description microRNA-200a confers chemoresistance by antagonizing TP53INP1 and YAP1 in human breast cancer Inhibition of miR200a enhances gemcitabine chemosensitivity in resistance cancer cells. TP53INP1 and YAP1 are involved in the RNA damage-induced p73-mediated apoptosis.
Key Molecule: hsa-let-7b [55]
Molecule Alteration Expression
Up-regulation
Sensitive Disease KRAS mutant breast cancer [ICD-11: 2C60.10]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
MEK/ERK /PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
NCI-H1975 cells Lung Homo sapiens (Human) CVCL_1511
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Let-7b repletion selectively sensitized kRAS mutant tumor cells to the cytotoxicity of paclitaxel and gemcitabine. Transfection of let-7b mimic downregulated the expression of mutant but not wild-type kRAS. Combination of let-7b mimic with paclitaxel or gemcitabine diminished MEk/ERk and PI3k/AkT signaling concurrently, triggered the onset of apoptosis, and reverted the epithelial-mesenchymal transition in kRAS mutant tumor cells. In addition, let-7b repletion downregulated the expression of beta-tubulin III and ribonucleotide reductase subunit M2, two proteins known to mediate tumor resistance to paclitaxel and gemcitabine, respectively. Let-7 may represent a new class of chemosensitizer for the treatment of kRAS mutant tumors.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Tumor protein p53-inducible nuclear protein 1 (TP53INP1) [76]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation p73-mediated apoptosis signaling pathway Inhibition hsa04210
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
SkBR3 cells Breast Homo sapiens (Human) CVCL_0033
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
T47D cells Breast Homo sapiens (Human) CVCL_0553
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
BT-549 Breast Homo sapiens (Human) CVCL_1092
MCF-10A Breast Homo sapiens (Human) CVCL_0598
MDA-MB-436 cells Breast Homo sapiens (Human) CVCL_0623
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
ZR-75-30 cells Breast Homo sapiens (Human) CVCL_1661
Experiment for
Molecule Alteration
Immunoblotting assay
Experiment for
Drug Resistance
TUNEL assays
Mechanism Description microRNA-200a confers chemoresistance by antagonizing TP53INP1 and YAP1 in human breast cancer Inhibition of miR200a enhances gemcitabine chemosensitivity in resistance cancer cells. TP53INP1 and YAP1 are involved in the RNA damage-induced p73-mediated apoptosis.
Key Molecule: Transcriptional coactivator YAP1 (YAP1) [76]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation p73-mediated apoptosis signaling pathway Inhibition hsa04210
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
SkBR3 cells Breast Homo sapiens (Human) CVCL_0033
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
T47D cells Breast Homo sapiens (Human) CVCL_0553
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
BT-549 Breast Homo sapiens (Human) CVCL_1092
MCF-10A Breast Homo sapiens (Human) CVCL_0598
MDA-MB-436 cells Breast Homo sapiens (Human) CVCL_0623
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
ZR-75-30 cells Breast Homo sapiens (Human) CVCL_1661
Experiment for
Molecule Alteration
Immunoblotting assay
Experiment for
Drug Resistance
TUNEL assays
Mechanism Description microRNA-200a confers chemoresistance by antagonizing TP53INP1 and YAP1 in human breast cancer Inhibition of miR200a enhances gemcitabine chemosensitivity in resistance cancer cells. TP53INP1 and YAP1 are involved in the RNA damage-induced p73-mediated apoptosis.
Key Molecule: Ribonucleoside-diphosphate reductase subunit M2 (RRM2) [55]
Molecule Alteration Expression
Down-regulation
Sensitive Disease KRAS mutant breast cancer [ICD-11: 2C60.10]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
MEK/ERK /PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Let-7b repletion selectively sensitized kRAS mutant tumor cells to the cytotoxicity of paclitaxel and gemcitabine. Transfection of let-7b mimic downregulated the expression of mutant but not wild-type kRAS. Combination of let-7b mimic with paclitaxel or gemcitabine diminished MEk/ERk and PI3k/AkT signaling concurrently, triggered the onset of apoptosis, and reverted the epithelial-mesenchymal transition in kRAS mutant tumor cells. In addition, let-7b repletion downregulated the expression of beta-tubulin III and ribonucleotide reductase subunit M2, two proteins known to mediate tumor resistance to paclitaxel and gemcitabine, respectively. Let-7 may represent a new class of chemosensitizer for the treatment of kRAS mutant tumors.
Key Molecule: Tubulin beta-3 chain (TUBB3) [55]
Molecule Alteration Expression
Down-regulation
Sensitive Disease KRAS mutant breast cancer [ICD-11: 2C60.10]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
MEK/ERK /PI3K/AKT signaling pathway Inhibition hsa04151
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 Let-7b repletion selectively sensitized kRAS mutant tumor cells to the cytotoxicity of paclitaxel and gemcitabine. Transfection of let-7b mimic downregulated the expression of mutant but not wild-type kRAS. Combination of let-7b mimic with paclitaxel or gemcitabine diminished MEk/ERk and PI3k/AkT signaling concurrently, triggered the onset of apoptosis, and reverted the epithelial-mesenchymal transition in kRAS mutant tumor cells. In addition, let-7b repletion downregulated the expression of beta-tubulin III and ribonucleotide reductase subunit M2, two proteins known to mediate tumor resistance to paclitaxel and gemcitabine, respectively. Let-7 may represent a new class of chemosensitizer for the treatment of kRAS mutant tumors.
Bladder cancer [ICD-11: 2C94]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-196a-5p [2]
Molecule Alteration Expression
Up-regulation
Resistant Disease Bladder cancer [ICD-11: 2C94.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
UMUC-2 cells Bladder Homo sapiens (Human) CVCL_8155
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Annexin V-FITC/PI Apoptosis assay
Mechanism Description Long non-coding RNA UCA1 promotes cisplatin/gemcitabine resistance through CREB modulating miR196a-5p in bladder cancer cells. UCA1 upregulates miR196a-5p through transcription factor CREB.
Key Molecule: hsa-miR-22-3p [8]
Molecule Alteration Expression
Up-regulation
Resistant Disease Bladder cancer [ICD-11: 2C94.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
T24 cells Bladder Homo sapiens (Human) CVCL_0554
UM-UC-3 cells Bladder Homo sapiens (Human) CVCL_1783
H-bc cells Bladder Homo sapiens (Human) CVCL_BT00
HTB-1 cells Bladder Homo sapiens (Human) CVCL_0359
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR 22 3p enhances multi chemoresistance by targeting NET1 in bladder cancer cells.
Key Molecule: Golgi phosphoprotein 3 (GOLPH3) [77]
Molecule Alteration Expression
Up-regulation
Resistant Disease Bladder urothelial carcinoma [ICD-11: 2C94.2]
Experimental Note Identified from the Human Clinical Data
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
T24 cells Bladder Homo sapiens (Human) CVCL_0554
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR; Western blotting assay
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description The expression levels of miR34a were decreased and GOLPH3 were increased in GC chemoresistant UBC cell lines. Down-regulation of miR34a resulted in the overexpression of GOLPH3.The ectopic expression of miR34a decreased the stem cell properties of chemoresistant UBC cells and re-sensitized these cells to GC treatment in vitro and in vivo.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: Transcription factor SOX-2 (SOX2) [78]
Molecule Alteration Expression
Up-regulation
Resistant Disease Bladder urothelial carcinoma [ICD-11: 2C94.2]
Experimental Note Identified from the Human Clinical Data
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
J82 cells Bladder Homo sapiens (Human) CVCL_0359
T24 cells Bladder Homo sapiens (Human) CVCL_0554
BFTC 909 cells Kidney Homo sapiens (Human) CVCL_1084
BFTC 905 cells Urinary bladder Homo sapiens (Human) CVCL_1083
HT-1376 cells Urinary bladder Homo sapiens (Human) CVCL_1292
SCaBER cells Urinary bladder Homo sapiens (Human) CVCL_3599
RT-4 cells Urinary bladder Homo sapiens (Human) CVCL_0036
UM-UC3 cells Urinary bladder Homo sapiens (Human) CVCL_1783
In Vivo Model Athymic (nu+/nu+) mouse xenograft model; NOD/SCID/IL2Rgamma -/- mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting assay
Mechanism Description Chemotherapy-induced COX2 and YAP1 signaling may promote CSC expansion via SOX2 overexpression and subsequent chemotherapy resistance.The YAP1-SOX2 axis, via re-activated PI3K/AKT signaling, may also be relevant to an acquired resistance to the EGFR inhibitor, as demonstrated by our findings that the resistant tumors again became sensitive to the EGFR inhibitor in combination with the YAP1 inhibitor.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Cyclin-dependent kinase inhibitor 1B (CDKN1B) [2]
Molecule Alteration Expression
Down-regulation
Resistant Disease Bladder cancer [ICD-11: 2C94.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
UMUC-2 cells Bladder Homo sapiens (Human) CVCL_8155
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Annexin V-FITC/PI Apoptosis assay
Mechanism Description miR196a-5p is involved in UCA1-mediated cisplatin/gemcitabine resistance via targeting p27kip1.
Key Molecule: Neuroepithelial cell-transforming gene 1 protein (NET1) [8]
Molecule Alteration Expression
Down-regulation
Resistant Disease Bladder cancer [ICD-11: 2C94.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
T24 cells Bladder Homo sapiens (Human) CVCL_0554
UM-UC-3 cells Bladder Homo sapiens (Human) CVCL_1783
H-bc cells Bladder Homo sapiens (Human) CVCL_BT00
HTB-1 cells Bladder Homo sapiens (Human) CVCL_0359
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR 22 3p enhances multi chemoresistance by targeting NET1 in bladder cancer cells.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-143 [79]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Bladder cancer [ICD-11: 2C94.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
IGF1R signaling pathway Inhibition hsa05200
MAPK sigaling pathway Inhibition hsahsa04
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
SV-HUC-1 cells Bladder Homo sapiens (Human) CVCL_3798
T24 cells Bladder Homo sapiens (Human) CVCL_0554
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR143 inhibits bladder cancer cell proliferation and enhances their sensitivity to gemcitabine by repressing IGF-1R signaling. Down-regulation of miR143 in bladder cancer may be involved in tumor development via the activation of IGF-1R and other downstream pathways like PI3k/Akt and MAPk.
Key Molecule: Urothelial cancer associated 1 (UCA1) [2]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Bladder cancer [ICD-11: 2C94.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
UMUC-2 cells Bladder Homo sapiens (Human) CVCL_8155
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Annexin V-FITC/PI Apoptosis assay
Mechanism Description Long non-coding RNA UCA1 promotes cisplatin/gemcitabine resistance through CREB modulating miR196a-5p in bladder cancer cells. UCA1 upregulates miR196a-5p through transcription factor CREB.
Key Molecule: hsa-miR-129-5p [80]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Bladder cancer [ICD-11: 2C94.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model SW780 cells Bladder Homo sapiens (Human) CVCL_1728
UM-UC-3 cells Bladder Homo sapiens (Human) CVCL_1783
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-129-5p inhibits gemcitabine resistance and promotes cell apoptosis of bladder cancer cells by downregulating Wnt5a.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Insulin-like growth factor 1 receptor (IGF1R) [79]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Bladder cancer [ICD-11: 2C94.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
IGF1R signaling pathway Inhibition hsa05200
MAPK sigaling pathway Inhibition hsahsa04
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
SV-HUC-1 cells Bladder Homo sapiens (Human) CVCL_3798
T24 cells Bladder Homo sapiens (Human) CVCL_0554
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR143 inhibits bladder cancer cell proliferation and enhances their sensitivity to gemcitabine by repressing IGF-1R signaling. Down-regulation of miR143 in bladder cancer may be involved in tumor development via the activation of IGF-1R and other downstream pathways like PI3k/Akt and MAPk.
Key Molecule: Protein Wnt-5a (WNT5A) [80]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Bladder cancer [ICD-11: 2C94.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model SW780 cells Bladder Homo sapiens (Human) CVCL_1728
UM-UC-3 cells Bladder Homo sapiens (Human) CVCL_1783
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-129-5p inhibits gemcitabine resistance and promotes cell apoptosis of bladder cancer cells by downregulating Wnt5a.
References
Ref 1 Overexpression of microRNA-620 facilitates the resistance of triple negative breast cancer cells to gemcitabine treatment by targeting DCTD. Exp Ther Med. 2019 Jul;18(1):550-558. doi: 10.3892/etm.2019.7601. Epub 2019 May 23.
Ref 2 Long non-coding RNA UCA1 promotes cisplatin/gemcitabine resistance through CREB modulating miR-196a-5p in bladder cancer cells. Cancer Lett. 2016 Nov 1;382(1):64-76. doi: 10.1016/j.canlet.2016.08.015. Epub 2016 Aug 31.
Ref 3 Overexpression of lncRNA EGFR AS1 is associated with a poor prognosis and promotes chemotherapy resistance in non small cell lung cancer. Int J Oncol. 2019 Jan;54(1):295-305. doi: 10.3892/ijo.2018.4629. Epub 2018 Nov 9.
Ref 4 MicroRNA187 overexpression is related to tumor progression and determines sensitivity to bortezomib in peripheral T-cell lymphoma. Leukemia. 2014 Apr;28(4):880-7. doi: 10.1038/leu.2013.291. Epub 2013 Oct 9.
Ref 5 MicroRNA-21 in pancreatic cancer: correlation with clinical outcome and pharmacologic aspects underlying its role in the modulation of gemcitabine activity. Cancer Res. 2010 Jun 1;70(11):4528-38. doi: 10.1158/0008-5472.CAN-09-4467. Epub 2010 May 11.
Ref 6 Low drug resistance to both platinum and taxane chemotherapy on an in vitro drug resistance assay predicts improved survival in patients with advanced epithelial ovarian, fallopian and peritoneal cancer .Int J Cancer. 2009 Dec 1;125(11):2721-7. doi: 10.1002/ijc.24654. 10.1002/ijc.24654
Ref 7 Effect of midkine on gemcitabine resistance in biliary tract cancer .Int J Mol Med. 2018 Apr;41(4):2003-2011. doi: 10.3892/ijmm.2018.3399. Epub 2018 Jan 18. 10.3892/ijmm.2018.3399
Ref 8 miR 22 3p enhances multi chemoresistance by targeting NET1 in bladder cancer cells. Oncol Rep. 2018 Jun;39(6):2731-2740. doi: 10.3892/or.2018.6355. Epub 2018 Apr 4.
Ref 9 Exosomal MicroRNA MiR-1246 Promotes Cell Proliferation, Invasion and Drug Resistance by Targeting CCNG2 in Breast Cancer. Cell Physiol Biochem. 2017;44(5):1741-1748. doi: 10.1159/000485780. Epub 2017 Dec 6.
Ref 10 Micro-RNA-130a-3p Regulates Gemcitabine Resistance via PPARG in Cholangiocarcinoma. Ann Surg Oncol. 2017 Aug;24(8):2344-2352. doi: 10.1245/s10434-017-5871-x. Epub 2017 May 30.
Ref 11 [14-3-3Zeta protein mediates gemcitabine resistance in NK/T-cell lymphoma] .Zhonghua Xue Ye Xue Za Zhi. 2019 Nov 14;40(11):906-911. doi: 10.3760/cma.j.issn.0253-2727.2019.11.004. 10.3760/cma.j.issn.0253-2727.2019.11.004
Ref 12 lncRNAPVT1 targets miR-152 to enhance chemoresistance of osteosarcoma to gemcitabine through activating c-MET/PI3K/AKT pathway. Pathol Res Pract. 2019 Mar;215(3):555-563. doi: 10.1016/j.prp.2018.12.013. Epub 2018 Dec 12.
Ref 13 Bcl-2 upregulation induced by miR-21 via a direct interaction is associated with apoptosis and chemoresistance in MIA PaCa-2 pancreatic cancer cells. Arch Med Res. 2011 Jan;42(1):8-14. doi: 10.1016/j.arcmed.2011.01.006.
Ref 14 sATP binding cassette subfamily G member 2 enhances the multidrug resistance properties of human nasal natural killer/T cell lymphoma side population cells .Oncol Rep. 2020 Oct;44(4):1467-1478. doi: 10.3892/or.2020.7722. Epub 2020 Aug 10. 10.3892/or.2020.7722
Ref 15 Restoration of tumor suppressor miR-34 inhibits human p53-mutant gastric cancer tumorspheres. BMC Cancer. 2008 Sep 21;8:266. doi: 10.1186/1471-2407-8-266.
Ref 16 Regulation of deoxycytidine kinase expression and sensitivity to gemcitabine by micro-RNA 330 and promoter methylation in cancer cells. Nucleosides Nucleotides Nucleic Acids. 2011 Dec;30(12):1214-22. doi: 10.1080/15257770.2011.629271.
Ref 17 Gemcitabine resistance in pancreatic ductal adenocarcinoma .Drug Resist Updat. 2015 Nov;23:55-68. doi: 10.1016/j.drup.2015.10.002. Epub 2015 Nov 3. 10.1016/j.drup.2015.10.002
Ref 18 Long non-coding RNA LINC00346 promotes pancreatic cancer growth and gemcitabine resistance by sponging miR-188-3p to derepress BRD4 expression. J Exp Clin Cancer Res. 2019 Feb 6;38(1):60. doi: 10.1186/s13046-019-1055-9.
Ref 19 MicroRNA-200b and -301 are associated with gemcitabine response as biomarkers in pancreatic carcinoma cells. Int J Oncol. 2019 Mar;54(3):991-1000. doi: 10.3892/ijo.2019.4676. Epub 2019 Jan 7.
Ref 20 Long non-coding RNA MEG3 functions as a tumour suppressor and has prognostic predictive value in human pancreatic cancer. Oncol Rep. 2018 Mar;39(3):1132-1140. doi: 10.3892/or.2018.6178. Epub 2018 Jan 2.
Ref 21 Gemcitabine treatment causes resistance and malignancy of pancreatic cancer stem-like cells via induction of lncRNA HOTAIR. Exp Ther Med. 2017 Nov;14(5):4773-4780. doi: 10.3892/etm.2017.5151. Epub 2017 Sep 20.
Ref 22 LncRNA TUG1 promoted viability and associated with gemcitabine resistant in pancreatic ductal adenocarcinoma. J Pharmacol Sci. 2018 Jun;137(2):116-121. doi: 10.1016/j.jphs.2018.06.002. Epub 2018 Jun 7.
Ref 23 miR-301a plays a pivotal role in hypoxia-induced gemcitabine resistance in pancreatic cancer. Exp Cell Res. 2018 Aug 1;369(1):120-128. doi: 10.1016/j.yexcr.2018.05.013. Epub 2018 May 23.
Ref 24 Downregulation of MicroRNA-455-3p Links to Proliferation and Drug Resistance of Pancreatic Cancer Cells via Targeting TAZ. Mol Ther Nucleic Acids. 2018 Mar 2;10:215-226. doi: 10.1016/j.omtn.2017.12.002. Epub 2017 Dec 9.
Ref 25 MiR-125a regulates chemo-sensitivity to gemcitabine in human pancreatic cancer cells through targeting A20. Acta Biochim Biophys Sin (Shanghai). 2016 Feb;48(2):202-8. doi: 10.1093/abbs/gmv129. Epub 2016 Jan 11.
Ref 26 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 27 The long non-coding RNA HOTTIP promotes progression and gemcitabine resistance by regulating HOXA13 in pancreatic cancer. J Transl Med. 2015 Mar 12;13:84. doi: 10.1186/s12967-015-0442-z.
Ref 28 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 29 The serum miR-21 level serves as a predictor for the chemosensitivity of advanced pancreatic cancer, and miR-21 expression confers chemoresistance by targeting FasL. Mol Oncol. 2013 Jun;7(3):334-45. doi: 10.1016/j.molonc.2012.10.011. Epub 2012 Nov 7.
Ref 30 MiR-21 upregulation induced by promoter zone histone acetylation is associated with chemoresistance to gemcitabine and enhanced malignancy of pancreatic cancer cells. Asian Pac J Cancer Prev. 2013;14(12):7529-36. doi: 10.7314/apjcp.2013.14.12.7529.
Ref 31 MiR-365 induces gemcitabine resistance in pancreatic cancer cells by targeting the adaptor protein SHC1 and pro-apoptotic regulator BAX. Cell Signal. 2014 Feb;26(2):179-85. doi: 10.1016/j.cellsig.2013.11.003. Epub 2013 Nov 9.
Ref 32 Involvement of microRNA-181b in the gemcitabine resistance of pancreatic cancer cells. Pancreatology. 2013 Sep-Oct;13(5):517-23. doi: 10.1016/j.pan.2013.06.007. Epub 2013 Jun 28.
Ref 33 miR-320c regulates gemcitabine-resistance in pancreatic cancer via SMARCC1. Br J Cancer. 2013 Jul 23;109(2):502-11. doi: 10.1038/bjc.2013.320. Epub 2013 Jun 25.
Ref 34 lncRNA GAS5 Reverses EMT and Tumor Stem Cell-Mediated Gemcitabine Resistance and Metastasis by Targeting miR-221/SOCS3 in Pancreatic Cancer. Mol Ther Nucleic Acids. 2018 Dec 7;13:472-482. doi: 10.1016/j.omtn.2018.09.026. Epub 2018 Oct 6.
Ref 35 MiR-10a-5p targets TFAP2C to promote gemcitabine resistance in pancreatic ductal adenocarcinoma. J Exp Clin Cancer Res. 2018 Apr 3;37(1):76. doi: 10.1186/s13046-018-0739-x.
Ref 36 Long noncoding RNA MALAT-1 enhances stem cell-like phenotypes in pancreatic cancer cells. Int J Mol Sci. 2015 Mar 24;16(4):6677-93. doi: 10.3390/ijms16046677.
Ref 37 miRNA profiling in pancreatic cancer and restoration of chemosensitivity. Cancer Lett. 2013 Jul 1;334(2):211-20. doi: 10.1016/j.canlet.2012.10.008. Epub 2012 Oct 13.
Ref 38 Gemcitabine enhances OSI-027 cytotoxicity by upregulation of miR-663a in pancreatic ductal adenocarcinoma cells. Am J Transl Res. 2019 Jan 15;11(1):473-485. eCollection 2019.
Ref 39 Gemcitabine exhibits a suppressive effect on pancreatic cancer cell growth by regulating processing of PVT1 to miR1207. Mol Oncol. 2018 Dec;12(12):2147-2164. doi: 10.1002/1878-0261.12393. Epub 2018 Oct 30.
Ref 40 miR-34 increases in vitro PANC-1 cell sensitivity to gemcitabine via targeting Slug/PUMA. Cancer Biomark. 2018;21(4):755-762. doi: 10.3233/CBM-170289.
Ref 41 MicroRNA-429 sensitizes pancreatic cancer cells to gemcitabine through regulation of PDCD4. Am J Transl Res. 2017 Nov 15;9(11):5048-5055. eCollection 2017.
Ref 42 Curcumin sensitizes pancreatic cancer cells to gemcitabine by attenuating PRC2 subunit EZH2, and the lncRNA PVT1 expression. Carcinogenesis. 2017 Oct 1;38(10):1036-1046. doi: 10.1093/carcin/bgx065.
Ref 43 CXCR4/Let-7a Axis Regulates Metastasis and Chemoresistance of Pancreatic Cancer Cells Through Targeting HMGA2. Cell Physiol Biochem. 2017;43(2):840-851. doi: 10.1159/000481610. Epub 2017 Sep 28.
Ref 44 Chemosensitization and inhibition of pancreatic cancer stem cell proliferation by overexpression of microRNA-205. Cancer Lett. 2017 Aug 28;402:1-8. doi: 10.1016/j.canlet.2017.05.007. Epub 2017 May 20.
Ref 45 Micro-RNA-21 Regulates Cancer-Associated Fibroblast-Mediated Drug Resistance in Pancreatic Cancer. Oncol Res. 2018 Jul 5;26(6):827-835. doi: 10.3727/096504017X14934840662335. Epub 2017 May 5.
Ref 46 miR-153 enhances the therapeutic effect of gemcitabine by targeting Snail in pancreatic cancer. Acta Biochim Biophys Sin (Shanghai). 2017 Jun 1;49(6):520-529. doi: 10.1093/abbs/gmx039.
Ref 47 micorRNA-101 silences DNA-PKcs and sensitizes pancreatic cancer cells to gemcitabine. Biochem Biophys Res Commun. 2017 Jan 29;483(1):725-731. doi: 10.1016/j.bbrc.2016.12.074. Epub 2016 Dec 14.
Ref 48 microRNA-210 overexpression inhibits tumor growth and potentially reverses gemcitabine resistance in pancreatic cancer. Cancer Lett. 2017 Mar 1;388:107-117. doi: 10.1016/j.canlet.2016.11.035. Epub 2016 Dec 7.
Ref 49 MiRNA-145 increases therapeutic sensibility to gemcitabine treatment of pancreatic adenocarcinoma cells. Oncotarget. 2016 Oct 25;7(43):70857-70868. doi: 10.18632/oncotarget.12268.
Ref 50 MiR-20a-5p regulates gemcitabine chemosensitivity by targeting RRM2 in pancreatic cancer cells and serves as a predictor for gemcitabine-based chemotherapy. Biosci Rep. 2019 May 7;39(5):BSR20181374. doi: 10.1042/BSR20181374. Print 2019 May 31.
Ref 51 MiR-30a regulates cancer cell response to chemotherapy through SNAI1/IRS1/AKT pathway. Cell Death Dis. 2019 Feb 15;10(3):153. doi: 10.1038/s41419-019-1326-6.
Ref 52 MiR-373-3p enhances the chemosensitivity of gemcitabine through cell cycle pathway by targeting CCND2 in pancreatic carcinoma cells. Biomed Pharmacother. 2018 Sep;105:887-898. doi: 10.1016/j.biopha.2018.05.091. Epub 2018 Jun 19.
Ref 53 MicroRNA-29c Increases the Chemosensitivity of Pancreatic Cancer Cells by Inhibiting USP22 Mediated Autophagy. Cell Physiol Biochem. 2018;47(2):747-758. doi: 10.1159/000490027. Epub 2018 May 22.
Ref 54 MicroRNA-101-3p reverses gemcitabine resistance by inhibition of ribonucleotide reductase M1 in pancreatic cancer. Cancer Lett. 2016 Apr 1;373(1):130-137. doi: 10.1016/j.canlet.2016.01.038. Epub 2016 Jan 28.
Ref 55 Let-7 Sensitizes KRAS Mutant Tumor Cells to Chemotherapy. PLoS One. 2015 May 6;10(5):e0126653. doi: 10.1371/journal.pone.0126653. eCollection 2015.
Ref 56 p85Alpha is a microRNA target and affects chemosensitivity in pancreatic cancer. J Surg Res. 2015 Jun 15;196(2):285-293. doi: 10.1016/j.jss.2015.02.071. Epub 2015 Mar 6.
Ref 57 MiR-497 downregulation contributes to the malignancy of pancreatic cancer and associates with a poor prognosis. Oncotarget. 2014 Aug 30;5(16):6983-93. doi: 10.18632/oncotarget.2184.
Ref 58 miR-211 modulates gemcitabine activity through downregulation of ribonucleotide reductase and inhibits the invasive behavior of pancreatic cancer cells. Nucleosides Nucleotides Nucleic Acids. 2014;33(4-6):384-93. doi: 10.1080/15257770.2014.891741.
Ref 59 MicroRNA-29a induces resistance to gemcitabine through the Wnt/Beta-catenin signaling pathway in pancreatic cancer cells. Int J Oncol. 2013 Oct;43(4):1066-72. doi: 10.3892/ijo.2013.2037. Epub 2013 Jul 24.
Ref 60 miRNA-181b increases the sensitivity of pancreatic ductal adenocarcinoma cells to gemcitabine in vitro and in nude mice by targeting BCL-2. Oncol Rep. 2013 May;29(5):1769-76. doi: 10.3892/or.2013.2297. Epub 2013 Feb 21.
Ref 61 MicroRNA expression as a predictive marker for gemcitabine response after surgical resection of pancreatic cancer. Ann Surg Oncol. 2011 Aug;18(8):2381-7. doi: 10.1245/s10434-011-1602-x. Epub 2011 Feb 23.
Ref 62 Genome-wide screen identifies PVT1 as a regulator of Gemcitabine sensitivity in human pancreatic cancer cells. Biochem Biophys Res Commun. 2011 Apr 1;407(1):1-6. doi: 10.1016/j.bbrc.2011.02.027. Epub 2011 Feb 18.
Ref 63 Identification of microRNA-21 as a biomarker for chemoresistance and clinical outcome following adjuvant therapy in resectable pancreatic cancer. PLoS One. 2010 May 14;5(5):e10630. doi: 10.1371/journal.pone.0010630.
Ref 64 MicroRNA-21 modulates biological functions of pancreatic cancer cells including their proliferation, invasion, and chemoresistance. Mol Cancer Ther. 2009 May;8(5):1067-74. doi: 10.1158/1535-7163.MCT-08-0592. Epub 2009 May 12.
Ref 65 miR-3656 expression enhances the chemosensitivity of pancreatic cancer to gemcitabine through modulation of the RHOF/EMT axis. Cell Death Dis. 2017 Oct 19;8(10):e3129. doi: 10.1038/cddis.2017.530.
Ref 66 miR-509-5p and miR-1243 increase the sensitivity to gemcitabine by inhibiting epithelial-mesenchymal transition in pancreatic cancer. Sci Rep. 2017 Jun 21;7(1):4002. doi: 10.1038/s41598-017-04191-w.
Ref 67 MicroRNA-155 Controls Exosome Synthesis and Promotes Gemcitabine Resistance in Pancreatic Ductal Adenocarcinoma. Sci Rep. 2017 Feb 15;7:42339. doi: 10.1038/srep42339.
Ref 68 miR-125a-3p is responsible for chemosensitivity in PDAC by inhibiting epithelial-mesenchymal transition via Fyn. Biomed Pharmacother. 2018 Oct;106:523-531. doi: 10.1016/j.biopha.2018.06.114. Epub 2018 Jul 11.
Ref 69 Down-regulation of miR-223 reverses epithelial-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells. Oncotarget. 2015 Jan 30;6(3):1740-9. doi: 10.18632/oncotarget.2714.
Ref 70 Up-regulation of miR-200 and let-7 by natural agents leads to the reversal of epithelial-to-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells. Cancer Res. 2009 Aug 15;69(16):6704-12. doi: 10.1158/0008-5472.CAN-09-1298. Epub 2009 Aug 4.
Ref 71 miR-29b, miR-205 and miR-221 enhance chemosensitivity to gemcitabine in HuH28 human cholangiocarcinoma cells. PLoS One. 2013 Oct 17;8(10):e77623. doi: 10.1371/journal.pone.0077623. eCollection 2013.
Ref 72 Involvement of human micro-RNA in growth and response to chemotherapy in human cholangiocarcinoma cell lines. Gastroenterology. 2006 Jun;130(7):2113-29. doi: 10.1053/j.gastro.2006.02.057.
Ref 73 miR-218-5p restores sensitivity to gemcitabine through PRKCE/MDR1 axis in gallbladder cancer. Cell Death Dis. 2017 May 11;8(5):e2770. doi: 10.1038/cddis.2017.178.
Ref 74 The expression of V-ATPase is associated with drug resistance and pathology of non-small-cell lung cancer .Diagn Pathol. 2013 Aug 28;8:145. doi: 10.1186/1746-1596-8-145. 10.1186/1746-1596-8-145
Ref 75 Restoring expression of miR-16: a novel approach to therapy for malignant pleural mesothelioma. Ann Oncol. 2013 Dec;24(12):3128-35. doi: 10.1093/annonc/mdt412. Epub 2013 Oct 22.
Ref 76 MicroRNA-200a confers chemoresistance by antagonizing TP53INP1 and YAP1 in human breast cancer. BMC Cancer. 2018 Jan 12;18(1):74. doi: 10.1186/s12885-017-3930-0.
Ref 77 miR34a/GOLPH3 Axis abrogates Urothelial Bladder Cancer Chemoresistance via Reduced Cancer Stemness .Theranostics. 2017 Oct 17;7(19):4777-4790. doi: 10.7150/thno.21713. eCollection 2017. 10.7150/thno.21713
Ref 78 YAP1 and COX2 Coordinately Regulate Urothelial Cancer Stem-like Cells .Cancer Res. 2018 Jan 1;78(1):168-181. doi: 10.1158/0008-5472.CAN-17-0836. Epub 2017 Nov 27. 10.1158/0008-5472.CAN-17-0836
Ref 79 miR-143 inhibits bladder cancer cell proliferation and enhances their sensitivity to gemcitabine by repressing IGF-1R signaling. Oncol Lett. 2017 Jan;13(1):435-440. doi: 10.3892/ol.2016.5388. Epub 2016 Nov 16.
Ref 80 miR-129-5p inhibits gemcitabine resistance and promotes cell apoptosis of bladder cancer cells by targeting Wnt5a. Int Urol Nephrol. 2018 Oct;50(10):1811-1819. doi: 10.1007/s11255-018-1959-x. Epub 2018 Aug 16.

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