Drug (ID: DG00151) and It's Reported Resistant Information
Name
Paclitaxel
Synonyms
C47H51NO14; weekly paclitaxel; Micellar Paclitaxel; Paclitaxel [USAN:INN:BAN]; SCHEMBL15000506; Benzenepropanoic acid, beta-(benzoylamino)-alpha-hydroxy-, 6,12b-bis(acetyloxy)-12-(benzoyloxy)-2a,3,4,4a,5,6,9,10,11,12,12a,12b-dodecahydro-4,11-dihydroxy-4a,8,13,13-tetramethyl-5-oxo-7,11-methano-1H-cyclodeca(3,4)benz(1,2-b)oxet-9-yl ester, (2aR-(2aalpha,4beta,4abeta,6beta,9alpha(alphaR*,betaS*),11alpha,12alpha,12aalpha,12balpha))-
    Click to Show/Hide
Indication
In total 5 Indication(s)
Breast cancer [ICD-11: 2C60]
Approved
[1]
Solid tumour/cancer [ICD-11: 2A00-2F9Z]
Approved
[1]
Pancreatic cancer [ICD-11: 2C10]
Phase 3
[1]
Ovarian cancer [ICD-11: 2C73]
Phase 2
[1]
Stomach cancer [ICD-11: 2B72]
Phase 2
[1]
Structure
Drug Resistance Disease(s)
Disease(s) with Clinically Reported Resistance for This Drug (6 diseases)
Brain cancer [ICD-11: 2A00]
[2]
Breast cancer [ICD-11: 2C60]
[3]
Cervical cancer [ICD-11: 2C77]
[4]
Head and neck cancer [ICD-11: 2D42]
[5]
Ovarian cancer [ICD-11: 2C73]
[6]
Prostate cancer [ICD-11: 2C82]
[7]
Disease(s) with Resistance Information Discovered by Cell Line Test for This Drug (20 diseases)
Bladder cancer [ICD-11: 2C94]
[8]
Breast cancer [ICD-11: 2C60]
[9]
Cervical cancer [ICD-11: 2C77]
[10]
Colorectal cancer [ICD-11: 2B91]
[11]
Endometrial cancer [ICD-11: 2C76]
[12]
Epithelial ovarian cancer [ICD-11: 2B5D]
[13]
Esophageal cancer [ICD-11: 2B70]
[14]
Gastric cancer [ICD-11: 2B72]
[15]
Liver cancer [ICD-11: 2C12]
[9]
Lung cancer [ICD-11: 2C25]
[16]
Melanoma [ICD-11: 2C30]
[17]
Nasopharyngeal cancer [ICD-11: 2B6B]
[18]
Osteosarcoma [ICD-11: 2B51]
[19]
Ovarian cancer [ICD-11: 2C73]
[20]
Pancreatic cancer [ICD-11: 2C10]
[21]
Pleural mesothelioma [ICD-11: 2C26]
[22]
Prostate cancer [ICD-11: 2C82]
[16]
Skin squamous cell carcinoma [ICD-11: 2C31]
[9]
Testicular cancer [ICD-11: 2C80]
[23]
Uterine sarcoma [ICD-11: 2C72]
[16]
Target Apoptosis regulator Bcl-2 (BCL-2) BCL2_HUMAN [1]
Tubulin beta (TUBB) NOUNIPROTAC [1]
Click to Show/Hide the Molecular Information and External Link(s) of This Drug
Formula
C47H51NO14
IsoSMILES
CC1=C2[C@H](C(=O)[C@@]3([C@H](C[C@@H]4[C@]([C@H]3[C@@H]([C@@](C2(C)C)(C[C@@H]1OC(=O)[C@@H]([C@H](C5=CC=CC=C5)NC(=O)C6=CC=CC=C6)O)O)OC(=O)C7=CC=CC=C7)(CO4)OC(=O)C)O)C)OC(=O)C
InChI
1S/C47H51NO14/c1-25-31(60-43(56)36(52)35(28-16-10-7-11-17-28)48-41(54)29-18-12-8-13-19-29)23-47(57)40(61-42(55)30-20-14-9-15-21-30)38-45(6,32(51)22-33-46(38,24-58-33)62-27(3)50)39(53)37(59-26(2)49)34(25)44(47,4)5/h7-21,31-33,35-38,40,51-52,57H,22-24H2,1-6H3,(H,48,54)/t31-,32-,33+,35-,36+,37+,38-,40-,45+,46-,47+/m0/s1
InChIKey
RCINICONZNJXQF-MZXODVADSA-N
PubChem CID
36314
ChEBI ID
CHEBI:45863
TTD Drug ID
D0C4RB
VARIDT ID
DR00342
INTEDE ID
DR1227
DrugBank ID
DB01229
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
Brain cancer [ICD-11: 2A00]
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 [24]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Glioma [ICD-11: 2A00.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation miR34a/PD-L1 signaling pathway Regulation hsa05206
In Vitro Model U87 cells Brain Homo sapiens (Human) CVCL_0022
U87-P cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Annexin V/PI apoptosis assay; Cell cycle assay; MTT assay
Mechanism Description miR34a attenuates glioma cells progression and chemoresistance via targeting PD-L1.
Key Molecule: hsa-mir-21 [25]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell migration Inhibition hsa04670
EGFR/STAT3 signaling pathway Inhibition hsa01521
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The miR-21 inhibitor could enhance the chemo-sensitivity of human glioblastoma cells to taxol. A combination of miR-21 inhibitor and taxol could be an effective therapeutic strategy for controlling the growth of GBM by inhibiting STAT3 expression and phosphorylation.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Programmed cell death 1 ligand 1 (PD-L1) [24]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Glioma [ICD-11: 2A00.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation miR34a/PD-L1 signaling pathway Regulation hsa05206
In Vitro Model U87 cells Brain Homo sapiens (Human) CVCL_0022
U87-P cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis; Luciferase reporter assay
Experiment for
Drug Resistance
Annexin V/PI apoptosis assay; Cell cycle assay; MTT assay
Mechanism Description miR34a attenuates glioma cells progression and chemoresistance via targeting PD-L1.
Solid tumour/cancer [ICD-11: 2A00-2F9Z]
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-495 [26]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Solid tumour/cancer [ICD-11: 2A00-2F9Z]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A2780 cells Ovary Homo sapiens (Human) CVCL_0134
A2780C cells Ovary Homo sapiens (Human) CVCL_0134
A2780DX5 cells Ovary Homo sapiens (Human) CVCL_4T98
SGC7901R cells Uterus Homo sapiens (Human) CVCL_0520
In Vivo Model Mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
Annexin-V-FITC apoptosis detection assay; Caspase-3 activity assay; MTT assay; Trypan blue exclusion assay
Mechanism Description miR-495 sensitizes MDR cancer cells to the combination of doxorubicin and taxol by inhibiting MDR1 expression, miR-495 was predicted to target ABCB1, which encodes protein MDR1.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [26]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Solid tumour/cancer [ICD-11: 2A00-2F9Z]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A2780 cells Ovary Homo sapiens (Human) CVCL_0134
A2780C cells Ovary Homo sapiens (Human) CVCL_0134
A2780DX5 cells Ovary Homo sapiens (Human) CVCL_4T98
SGC7901R cells Uterus Homo sapiens (Human) CVCL_0520
In Vivo Model Mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Annexin-V-FITC apoptosis detection assay; Caspase-3 activity assay; MTT assay; Trypan blue exclusion assay
Mechanism Description miR-495 sensitizes MDR cancer cells to the combination of doxorubicin and taxol by inhibiting MDR1 expression, miR-495 was predicted to target ABCB1, which encodes protein MDR1.
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: Mitosis associated long intergenic non-coding RNA 1 (MALINC1) [19]
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 proliferation Activation hsa05200
In Vitro Model H1299 cells Lung Homo sapiens (Human) CVCL_0060
U2OS cells Bone Homo sapiens (Human) CVCL_0042
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Silencing of MA-linc1 in unsynchronized cells results in fewer cells in G1 and a concomitant increase in the number of cells in all other stages of the cell cycle, particularly in G2/M. Moreover, its silencing in M phase-arrested cells inhibits mitosis exit. The effect of MA-linc1 on cell cycle progression is mediated, at least in part, by repression of its neighboring gene, Puralpha, a cell cycle regulator whose expression induces cell cycle arrest. Importantly, high levels of MA-linc1 are correlated with decreased survival of breast and lung cancer patients and its silencing sensitizes cancer cells to the apoptotic effect of the M phase specific chemotherapeutic drug, Paclitaxel. This enhancement of Paclitaxel-induced apoptosis is also Puralpha-related.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Transcriptional activator protein Pur-alpha (PURA) [19]
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 proliferation Activation hsa05200
In Vitro Model H1299 cells Lung Homo sapiens (Human) CVCL_0060
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Silencing of MA-linc1 in unsynchronized cells results in fewer cells in G1 and a concomitant increase in the number of cells in all other stages of the cell cycle, particularly in G2/M. Moreover, its silencing in M phase-arrested cells inhibits mitosis exit. The effect of MA-linc1 on cell cycle progression is mediated, at least in part, by repression of its neighboring gene, Puralpha, a cell cycle regulator whose expression induces cell cycle arrest. Importantly, high levels of MA-linc1 are correlated with decreased survival of breast and lung cancer patients and its silencing sensitizes cancer cells to the apoptotic effect of the M phase specific chemotherapeutic drug, Paclitaxel. This enhancement of Paclitaxel-induced apoptosis is also Puralpha-related.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-422a [27]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Osteosarcoma [ICD-11: 2B51.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model MG63 cells Bone marrow Homo sapiens (Human) CVCL_0426
SAOS-2 cells Bone marrow Homo sapiens (Human) CVCL_0548
U2OS cells Bone Homo sapiens (Human) CVCL_0042
HFOB cells Bone Homo sapiens (Human) CVCL_3708
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis of apoptosis; Transwell assay
Mechanism Description Overexpression of miR422a inhibits cell proliferation and invasion, and enhances chemosensitivity by directly targeting TGFbeta2 in osteosarcoma cells.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Transforming growth factor beta-2 proprotein (TGFB2) [27]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Osteosarcoma [ICD-11: 2B51.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model MG63 cells Bone marrow Homo sapiens (Human) CVCL_0426
SAOS-2 cells Bone marrow Homo sapiens (Human) CVCL_0548
U2OS cells Bone Homo sapiens (Human) CVCL_0042
HFOB cells Bone Homo sapiens (Human) CVCL_3708
Experiment for
Molecule Alteration
Western blot analysis; Dual luciferase activity assay
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis of apoptosis; Transwell assay
Mechanism Description Overexpression of miR422a inhibits cell proliferation and invasion, and enhances chemosensitivity by directly targeting TGFbeta2 in osteosarcoma cells.
Epithelial ovarian cancer [ICD-11: 2B5D]
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: Long non-protein coding RNA 1118 (LINC01118) [28]
Molecule Alteration Expression
Up-regulation
Resistant Disease Epithelial ovarian cancer [ICD-11: 2B5D.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
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
COC1 cells Ovary Homo sapiens (Human) CVCL_6891
SkOV3-TR30 cells Ovary Homo sapiens (Human) CVCL_0532
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTS assay; Flow cytometry assay
Mechanism Description LINC01118 Can enhance ABCC1 expression by suppressing miR-134 expression to promote paclitaxel resistance in epithelial ovarian cancer.
Key Molecule: hsa-mir-134 [28]
Molecule Alteration Expression
Down-regulation
Resistant Disease Epithelial ovarian cancer [ICD-11: 2B5D.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
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
COC1 cells Ovary Homo sapiens (Human) CVCL_6891
SkOV3-TR30 cells Ovary Homo sapiens (Human) CVCL_0532
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTS assay; Flow cytometry assay
Mechanism Description LINC01118 Can enhance ABCC1 expression by suppressing miR-134 expression to promote paclitaxel resistance in epithelial ovarian cancer.
Key Molecule: hsa-mir-520g [13]
Molecule Alteration Expression
Up-regulation
Resistant Disease Epithelial ovarian cancer [ICD-11: 2B5D.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell proliferation Activation hsa05200
MAPK/AKT signaling pathway Regulation hsa04010
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
CAOV3 cells Ovary Homo sapiens (Human) CVCL_0201
OVCA433 cells Ovary Homo sapiens (Human) CVCL_0475
OV2008 cells Ovary Homo sapiens (Human) CVCL_0473
ES-2 cells Ovary Homo sapiens (Human) CVCL_3509
MCAS cells Ovary Homo sapiens (Human) CVCL_3020
OVk18 cells Ovary Homo sapiens (Human) CVCL_3770
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-520g expression is significantly increased in EOC and high miR-520g expression promotes tumor development, increases chemoresistance to platinum-based chemotherapy and reduces patient survival. miR-520g directly targets and downregulates DAPk2 by binding the DAPk2 3'UTR. DAPk2 suppression, followed by MAPk and AkT pathway activation, promotes the biological processes mediated by miR-520g in EOC.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance-associated protein 1 (MRP1) [28]
Molecule Alteration Expression
Up-regulation
Resistant Disease Epithelial ovarian cancer [ICD-11: 2B5D.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
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
COC1 cells Ovary Homo sapiens (Human) CVCL_6891
SkOV3-TR30 cells Ovary Homo sapiens (Human) CVCL_0532
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
MTS assay; Flow cytometry assay
Mechanism Description LINC01118 Can enhance ABCC1 expression by suppressing miR-134 expression to promote paclitaxel resistance in epithelial ovarian cancer.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Death-associated protein kinase 2 (DAPK2) [13]
Molecule Alteration Expression
Down-regulation
Resistant Disease Epithelial ovarian cancer [ICD-11: 2B5D.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell proliferation Activation hsa05200
MAPK/AKT signaling pathway Regulation hsa04010
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
CAOV3 cells Ovary Homo sapiens (Human) CVCL_0201
OVCA433 cells Ovary Homo sapiens (Human) CVCL_0475
OV2008 cells Ovary Homo sapiens (Human) CVCL_0473
ES-2 cells Ovary Homo sapiens (Human) CVCL_3509
MCAS cells Ovary Homo sapiens (Human) CVCL_3020
OVk18 cells Ovary Homo sapiens (Human) CVCL_3770
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-520g expression is significantly increased in EOC and high miR-520g expression promotes tumor development, increases chemoresistance to platinum-based chemotherapy and reduces patient survival. miR-520g directly targets and downregulates DAPk2 by binding the DAPk2 3'UTR. DAPk2 suppression, followed by MAPk and AkT pathway activation, promotes the biological processes mediated by miR-520g in EOC.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-146a [29]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Epithelial ovarian cancer [ICD-11: 2B5D.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
miR146a/SOD2/ROS signaling pathway Regulation hsa05206
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
PLC cells Liver Homo sapiens (Human) CVCL_0485
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; CCK8 assay
Mechanism Description miR-146a as a potential tumor suppressor in patients with EOC. miR-146a downregulates expression of SOD2 and enhances ROS generation, leading to increased apoptosis, inhibition of proliferation, and (+) sensitivity to chemotherapy.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Superoxide dismutase Mn (SODM) [29]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Epithelial ovarian cancer [ICD-11: 2B5D.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
miR146a/SOD2/ROS signaling pathway Regulation hsa05206
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
PLC cells Liver Homo sapiens (Human) CVCL_0485
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; CCK8 assay
Mechanism Description miR-146a as a potential tumor suppressor in patients with EOC. miR-146a downregulates expression of SOD2 and enhances ROS generation, leading to increased apoptosis, inhibition of proliferation, and (+) sensitivity to chemotherapy.
Nasopharyngeal cancer [ICD-11: 2B6B]
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: ENSG00000247844 (CCAT1) [18]
Molecule Alteration Expression
Up-regulation
Resistant Disease Nasopharyngeal carcinoma [ICD-11: 2B6B.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation CCAT1/miR181a/CPEB2 signaling pathway Regulation hsa05206
In Vitro Model CNE2 cells Nasopharynx Homo sapiens (Human) CVCL_6889
CNE1 cells Throat Homo sapiens (Human) CVCL_6888
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Upregulated CCAT1 sponges miR181a in NPC cells, and miR181a could directly bind to CCAT1 mRNA in NPC cells. Restoration of miR181a re-sensitized the NPC cells to paclitaxel in vitro, miR181a was a modulator of paclitaxel sensitivity due to its regulative effect on cell apoptosis via targeting CPEB2 in NPC cells.
Key Molecule: ENSG00000247844 (CCAT1) [18]
Molecule Alteration Expression
Up-regulation
Resistant Disease Nasopharyngeal carcinoma [ICD-11: 2B6B.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model CNE2 cells Nasopharynx Homo sapiens (Human) CVCL_6889
CNE1 cells Throat Homo sapiens (Human) CVCL_6888
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description CCAT1 reduced the sensitivity of NPC cells to paclitaxel by suppressing miR181a level and subsequently regulating CPEB2 to monitor NPC cell growth.
Key Molecule: hsa-mir-29a [30]
Molecule Alteration Expression
Down-regulation
Resistant Disease Nasopharyngeal carcinoma [ICD-11: 2B6B.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
JAKT/STAT signaling pathway Regulation hsa04630
Tumorigenesis Activation hsa05206
In Vitro Model NP69 cells Nasopharynx Homo sapiens (Human) CVCL_F755
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR-29a down-regulation is correlated with drug resistance of nasopharyngeal carcinoma cell line CNE-1 and miR-29a up-regulation decreases Taxol resistance of nasopharyngeal carcinoma CNE-1 cells possibly via inhibiting STAT3 and Bcl-2 expression.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Cytoplasmic polyadenylation element-binding protein 2 (CPEB2) [18]
Molecule Alteration Expression
Up-regulation
Resistant Disease Nasopharyngeal carcinoma [ICD-11: 2B6B.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model CNE2 cells Nasopharynx Homo sapiens (Human) CVCL_6889
CNE1 cells Throat Homo sapiens (Human) CVCL_6888
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description CCAT1 reduced the sensitivity of NPC cells to paclitaxel by suppressing miR181a level and subsequently regulating CPEB2 to monitor NPC cell growth.
Key Molecule: Signal transducer activator transcription 3 (STAT3) [30]
Molecule Alteration Expression
Up-regulation
Resistant Disease Nasopharyngeal carcinoma [ICD-11: 2B6B.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
JAKT/STAT signaling pathway Regulation hsa04630
In Vitro Model NP69 cells Nasopharynx Homo sapiens (Human) CVCL_F755
Experiment for
Molecule Alteration
Western blot analysis; RIP assay; Luciferase reporter assay
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR-29a down-regulation is correlated with drug resistance of nasopharyngeal carcinoma cell line CNE-1 and miR-29a up-regulation decreases Taxol resistance of nasopharyngeal carcinoma CNE-1 cells possibly via inhibiting STAT3 and Bcl-2 expression.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: ENSG00000247844 (CCAT1) [18]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Nasopharyngeal carcinoma [ICD-11: 2B6B.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation CCAT1/miR181a/CPEB2 signaling pathway Regulation hsa05206
In Vitro Model CNE2 cells Nasopharynx Homo sapiens (Human) CVCL_6889
CNE1 cells Throat Homo sapiens (Human) CVCL_6888
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Upregulated CCAT1 sponges miR181a in NPC cells, and miR181a could directly bind to CCAT1 mRNA in NPC cells. Restoration of miR181a re-sensitized the NPC cells to paclitaxel in vitro, miR181a was a modulator of paclitaxel sensitivity due to its regulative effect on cell apoptosis via targeting CPEB2 in NPC cells.
Key Molecule: hsa-mir-181a [18]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Nasopharyngeal carcinoma [ICD-11: 2B6B.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation CCAT1/miR181a/CPEB2 signaling pathway Regulation hsa05206
In Vitro Model CNE2 cells Nasopharynx Homo sapiens (Human) CVCL_6889
CNE1 cells Throat Homo sapiens (Human) CVCL_6888
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description LncRNA CCAT1 regulates the sensitivity of paclitaxel in NPC cells via miR181a/CPEB2 axis. miR181a restores CCAT1-induced paclitaxel resistant in NPC cells via targeting CPEB2.
Key Molecule: hsa-mir-29c [31]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Nasopharyngeal carcinoma [ICD-11: 2B6B.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model C666-1 cells Throat Homo sapiens (Human) CVCL_7949
SUNE-1 cells Nasopharynx Homo sapiens (Human) CVCL_6946
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 Overexpression of miR-29c and knockdown of ITGB1 can resensitize drug-resistant NPC cells to Taxol and promote apoptosis.
Key Molecule: hsa-miR-1204 [32]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Nasopharyngeal carcinoma [ICD-11: 2B6B.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model 5-8F cells Nasopharynx Homo sapiens (Human) CVCL_C528
CNE1 cells Throat Homo sapiens (Human) CVCL_6888
HNE-2 cells Nasopharynx Homo sapiens (Human) CVCL_FA07
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Colony formation assay
Mechanism Description miR-1204 sensitizes nasopharyngeal carcinoma cells to paclitaxel both in vitro and in vivo via inhibitsing tumor growth in vivo significantly.
Key Molecule: hsa-miR-634 [33]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Nasopharyngeal carcinoma [ICD-11: 2B6B.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model CNE1 cells Throat Homo sapiens (Human) CVCL_6888
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-634 most significantly downregulated in the paclitaxel-resistant CNE-1/Taxol, in regulating the paclitaxel sensitivity in NPC cells. miR-634 expression in the CNE-1/Taxol cells by lentivirus infection, miR-634 re-sensitized the CNE-1/Taxol cells to paclitaxel in vitro. In xenograft mouse model, miR-634 inhibited tumor growth and (+) paclitaxel sensitivity.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Cytoplasmic polyadenylation element-binding protein 2 (CPEB2) [18]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Nasopharyngeal carcinoma [ICD-11: 2B6B.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation CCAT1/miR181a/CPEB2 signaling pathway Regulation hsa05206
In Vitro Model CNE2 cells Nasopharynx Homo sapiens (Human) CVCL_6889
CNE1 cells Throat Homo sapiens (Human) CVCL_6888
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description LncRNA CCAT1 regulates the sensitivity of paclitaxel in NPC cells via miR181a/CPEB2 axis. miR181a restores CCAT1-induced paclitaxel resistant in NPC cells via targeting CPEB2.
Key Molecule: Integrin beta-1 (ITGB1) [31]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Nasopharyngeal carcinoma [ICD-11: 2B6B.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model C666-1 cells Throat Homo sapiens (Human) CVCL_7949
SUNE-1 cells Nasopharynx Homo sapiens (Human) CVCL_6946
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Overexpression of miR-29c and knockdown of ITGB1 can resensitize drug-resistant NPC cells to Taxol and promote apoptosis.
Oral squamous cell carcinoma [ICD-11: 2B6E]
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: ATP-binding cassette sub-family B5 (ABCB5) [34]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Squamous cell carcinoma [ICD-11: 2B6E.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model KB-3-1 cells Lung Homo sapiens (Human) CVCL_2088
KB-8-5 cells Mouth Homo sapiens (Human) CVCL_5994
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The continuous administration of low dose 5FU with Taxol significantly inhibited the tumor growth. The treatment overcomes drug resistance in tumors by down-regulating multi-drug resistance transporter protein.
Esophageal cancer [ICD-11: 2B70]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-193a-3p [14]
Molecule Alteration Expression
Up-regulation
Resistant Disease Esophageal cancer [ICD-11: 2B70.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model KYSE150 cells Esophagus Homo sapiens (Human) CVCL_1348
KYSE510 cells Esophagus Homo sapiens (Human) CVCL_1354
kYSE410 cells Esophagus Homo sapiens (Human) CVCL_1352
kYSE450 cells Esophagus Homo sapiens (Human) CVCL_1353
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Over-expression of miR-193a-3p increased the radioresistance and chemoresistance of oesophageal squamous cell carcinoma (ESCC) cells. In contrast, the down-regulation of miR-193a-3p decreased the radioresistance and chemoresistance of ESCC cells. In addition, miR-193a-3p inducing DNA damage has also been demonstrated through measuring the level of gamma-H2AX associated with miR-193a-3p. Moreover, a small interfering RNA(siRNA)-induced repression of the PSEN1 gene had an effect similar to that of miR-193a-3p up-regulation. The above processes also inhibited oesophageal cancer cells apoptosis. These findings suggest that miR-193a-3p contributes to the radiation and chemotherapy resistance of oesophageal carcinoma by down-regulating PSEN1.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Presenilin-1 (PSEN1) [14]
Molecule Alteration Expression
Down-regulation
Resistant Disease Esophageal cancer [ICD-11: 2B70.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model KYSE150 cells Esophagus Homo sapiens (Human) CVCL_1348
KYSE510 cells Esophagus Homo sapiens (Human) CVCL_1354
kYSE410 cells Esophagus Homo sapiens (Human) CVCL_1352
kYSE450 cells Esophagus Homo sapiens (Human) CVCL_1353
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Over-expression of miR-193a-3p increased the radioresistance and chemoresistance of oesophageal squamous cell carcinoma (ESCC) cells. In contrast, the down-regulation of miR-193a-3p decreased the radioresistance and chemoresistance of ESCC cells. In addition, miR-193a-3p inducing DNA damage has also been demonstrated through measuring the level of gamma-H2AX associated with miR-193a-3p. Moreover, a small interfering RNA(siRNA)-induced repression of the PSEN1 gene had an effect similar to that of miR-193a-3p up-regulation. The above processes also inhibited oesophageal cancer cells apoptosis. These findings suggest that miR-193a-3p contributes to the radiation and chemotherapy resistance of oesophageal carcinoma by down-regulating PSEN1.
Gastric cancer [ICD-11: 2B72]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-590-5p [15]
Molecule Alteration Expression
Up-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT/ERK signaling pathway Activation hsa04010
In Vitro Model BGC-823 cells Gastric Homo sapiens (Human) CVCL_3360
MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
AGS cells Gastric Homo sapiens (Human) CVCL_0139
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
MkN28 cells Gastric Homo sapiens (Human) CVCL_1416
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay; Matrigel transwell assay
Mechanism Description miR590-5p regulates gastric cancer cell growth and chemosensitivity through RECk and the AkT/ERk pathway. RECk is a direct target of miR590-5p, knockdown of RECk accelerated cell proliferation and motility and decreased the drug sensitivity.The AkT/ERk and STAT3 signaling pathways were activated by miR590-5p overexpression.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Reversion-inducing cysteine-rich protein with Kazal motifs (RECK) [15]
Molecule Alteration Expression
Down-regulation
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT/ERK signaling pathway Activation hsa04010
In Vitro Model BGC-823 cells Gastric Homo sapiens (Human) CVCL_3360
MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
AGS cells Gastric Homo sapiens (Human) CVCL_0139
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
MkN28 cells Gastric Homo sapiens (Human) CVCL_1416
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay; Matrigel transwell assay
Mechanism Description miR590-5p regulates gastric cancer cell growth and chemosensitivity through RECk and the AkT/ERk pathway. RECk is a direct target of miR590-5p, knockdown of RECk accelerated cell proliferation and motility and decreased the drug sensitivity.The AkT/ERk and STAT3 signaling pathways were activated by miR590-5p overexpression.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: Protein lin-28 homolog A (CSDD1) [35]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
Lin28/miR107 pathway Regulation hsa05206
In Vitro Model MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
MkN28 cells Gastric Homo sapiens (Human) CVCL_1416
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Lin28 could inhibit the expression of miR-107, thereby up-regulating C-myc, P-gp and down-regulating Cyclin D1, subsequently result in chemo-resistance of gastric cancer cells. The Lin28/miR-107 pathway might be served as one of many signaling pathways that is associated with gastric cancer chemo-resistance.
Key Molecule: Protein lin-28 homolog B (CSDD2) [35]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
Lin28/miR107 pathway Regulation hsa05206
In Vitro Model MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
MkN28 cells Gastric Homo sapiens (Human) CVCL_1416
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Lin28 could inhibit the expression of miR-107, thereby up-regulating C-myc, P-gp and down-regulating Cyclin D1, subsequently result in chemo-resistance of gastric cancer cells. The Lin28/miR-107 pathway might be served as one of many signaling pathways that is associated with gastric cancer chemo-resistance.
Key Molecule: hsa-miR-107 [35]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
Lin28/miR107 pathway Regulation hsa05206
In Vitro Model MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
MkN28 cells Gastric Homo sapiens (Human) CVCL_1416
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Lin28 could inhibit the expression of miR-107, thereby up-regulating C-myc, P-gp and down-regulating Cyclin D1, subsequently result in chemo-resistance of gastric cancer cells. The Lin28/miR-107 pathway might be served as one of many signaling pathways that is associated with gastric cancer chemo-resistance.
Key Molecule: hsa-miR-34c-5p [36]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model BGC-823 cells Gastric Homo sapiens (Human) CVCL_3360
MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
GES-1 cells Gastric Homo sapiens (Human) CVCL_EQ22
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description MAPT is a microtubule-associated protein which promotes the assembly of tubulin into microtubules to stabilize microtubule structure. Reduced expression of MAPT has been also associated with a better response to paclitaxel in gastric cancer patients, reduced expression of miR-34c-5p, provides a possible mechanism of paclitaxel resistance in gastric cancer, overexpression of miR-34c-5p reduces MAPT expression and restores paclitaxel sensitivity.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: hsa-miR-155-5p [37]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Exosomal delivery of miR 155 5p may induce EMT and chemoresistant phenotypes from paclitaxel resistant gastric cancer cells to the sensitive cells, which may be mediated by GATA3 and TP53INP1 suppression.
Key Molecule: Trans-acting T-cell-specific transcription factor GATA-3 (GATA3) [37]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Exosomal delivery of miR 155 5p may induce EMT and chemoresistant phenotypes from paclitaxel resistant gastric cancer cells to the sensitive cells, which may be mediated by GATA3 and TP53INP1 suppression.
Key Molecule: Tumor protein p53-inducible nuclear protein 1 (TP53INP1) [37]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Exosomal delivery of miR 155 5p may induce EMT and chemoresistant phenotypes from paclitaxel resistant gastric cancer cells to the sensitive cells, which may be mediated by GATA3 and TP53INP1 suppression.
Key Molecule: Catenin beta-1 (CTNNB1) [38]
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 Wnt/Beta-catenin signaling pathway Regulation hsa04310
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Transwell assay; Flow cytometry assay
Mechanism Description Silencing of ZFAS1 augmented the sensitivity to cis-platinum or paclitaxel in gastric cancer cancer cells and silencing of ZFAS1-induced inhibition of malignancies was reversed by beta-catenin.
Key Molecule: ZNFX1 antisense RNA 1 (ZFAS1) [38]
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 Wnt/Beta-catenin signaling pathway Regulation hsa04310
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay; Transwell assay; Flow cytometry assay
Mechanism Description Silencing of ZFAS1 augmented the sensitivity to cis-platinum or paclitaxel in gastric cancer cancer cells and silencing of ZFAS1-induced inhibition of malignancies was reversed by beta-catenin.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Microtubule-associated protein tau (MAPT) [36]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model BGC-823 cells Gastric Homo sapiens (Human) CVCL_3360
MGC-803 cells Gastric Homo sapiens (Human) CVCL_5334
SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
GES-1 cells Gastric Homo sapiens (Human) CVCL_EQ22
MkN-45 cells Gastric Homo sapiens (Human) CVCL_0434
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description MAPT is a microtubule-associated protein which promotes the assembly of tubulin into microtubules to stabilize microtubule structure. Reduced expression of MAPT has been also associated with a better response to paclitaxel in gastric cancer patients, reduced expression of miR-34c-5p, provides a possible mechanism of paclitaxel resistance in gastric cancer, overexpression of miR-34c-5p reduces MAPT expression and restores paclitaxel sensitivity.
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-125a [39]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
In Vivo Model BALB/C nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Overexpression of miR-125a/b significantly inhibited ALDH1A3 and Mcl1 expression, reduced cell survival, and increased cell apoptosis in HT29-taxol cells. Chemoresistance to paclitaxel is initiated by the downregulation of miR-125a/b expression, which subsequently upregulates ALDH1A3 and Mcl1 expression to promote survival of CSCs.
Key Molecule: hsa-mir-125b [39]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
In Vivo Model BALB/C nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Overexpression of miR-125a/b significantly inhibited ALDH1A3 and Mcl1 expression, reduced cell survival, and increased cell apoptosis in HT29-taxol cells. Chemoresistance to paclitaxel is initiated by the downregulation of miR-125a/b expression, which subsequently upregulates ALDH1A3 and Mcl1 expression to promote survival of CSCs.
Key Molecule: hsa-mir-22 [40]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model HT-29 cells Colon Homo sapiens (Human) CVCL_0320
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
Experiment for
Molecule Alteration
Northern blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Overexpression of miR-22 enhanced the anticancer effect of paclitaxel in the p53-mutated cells through increasing cell apoptosis and reducing cell proliferation and survival. The anticancer role of miR-22 was mediated by activation of PTEN signaling, subsequent inhibition of Akt Ser473 phosphorylation and MTDH expression, as well as upregulation of Bax and active caspase-3 levels.
Key Molecule: hsa-mir-203 [41]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
PI3K signaling pathway Regulation hsa04151
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
Experiment for
Molecule Alteration
RT-PCR; Northern blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description the tumor suppressive role of miR-203 was mediated by negatively regulating Akt2 protein expression through mRNA degradation. The inhibition of Akt2 activity downregulated the protein expression of its downstream molecules involved in chemoresistance, such as MTDH and HSP90 genes.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Aldehyde dehydrogenase 6 (ALDH6) [39]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
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 Overexpression of miR-125a/b significantly inhibited ALDH1A3 and Mcl1 expression, reduced cell survival, and increased cell apoptosis in HT29-taxol cells. Chemoresistance to paclitaxel is initiated by the downregulation of miR-125a/b expression, which subsequently upregulates ALDH1A3 and Mcl1 expression to promote survival of CSCs.
Key Molecule: Induced myeloid leukemia cell differentiation protein Mcl-1 (MCL1) [39]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
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 Overexpression of miR-125a/b significantly inhibited ALDH1A3 and Mcl1 expression, reduced cell survival, and increased cell apoptosis in HT29-taxol cells. Chemoresistance to paclitaxel is initiated by the downregulation of miR-125a/b expression, which subsequently upregulates ALDH1A3 and Mcl1 expression to promote survival of CSCs.
Key Molecule: Phosphatase and tensin homolog (PTEN) [40]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model HT-29 cells Colon Homo sapiens (Human) CVCL_0320
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
Experiment for
Molecule Alteration
Northern blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Overexpression of miR-22 enhanced the anticancer effect of paclitaxel in the p53-mutated cells through increasing cell apoptosis and reducing cell proliferation and survival. The anticancer role of miR-22 was mediated by activation of PTEN signaling, subsequent inhibition of Akt Ser473 phosphorylation and MTDH expression, as well as upregulation of Bax and active caspase-3 levels.
Key Molecule: RAC-beta serine/threonine-protein kinase (AKT2) [41]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
PI3K signaling pathway Regulation hsa04151
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
HCT15 cells Colon Homo sapiens (Human) CVCL_0292
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The tumor suppressive role of miR-203 was mediated by negatively regulating Akt2 protein expression through mRNA degradation. The inhibition of Akt2 activity downregulated the protein expression of its downstream molecules involved in chemoresistance, such as MTDH and HSP90 genes.
Colorectal cancer [ICD-11: 2B91]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [11]
Molecule Alteration Expression
Up-regulation
Resistant Disease Colorectal carcinoma [ICD-11: 2B91.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model CaCo2 cells Colon Homo sapiens (Human) CVCL_0025
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description The transient exposure to digoxin for 24 h was found to induce MDR1 mRNA in Caco-2 cells. Here, a digoxin-tolerant Caco-2 subline (Caco/DX) was newly established by the continuous exposure of Caco-2 cells to digoxin, and the effects of continuous exposure to digoxin on MDR1 were examined. The 50% growth inhibitory concentration (IC(50)) values for digoxin in Caco-2 and Caco/DX cells were 17.2 and 81.4 nM, respectively. The IC(50) values for paclitaxel, an MDR1 substrate, were 1.0 and 547 nM, respectively, whereas the cytotoxicity of 5-fluorouracil was comparable in both.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-15 [42]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell viability Inhibition hsa05200
In Vitro Model SW620 cells Colon Homo sapiens (Human) CVCL_0547
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Inhibition of LINC00473 in vivo could overcome the Taxol resistance of CRC cells, could recover the expression of tumor suppressor miR-15a and chemotherapy-induced tumor regression while the BCL-2-related anti-apoptosis pathway was activated and the multidrug-resistant (MDR) genes LRP, MDR1 were up-regulated by LINC00473.
Key Molecule: Long non-protein coding RNA (LINC00473) [42]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell colony Inhibition hsa05200
Cell invasion Inhibition hsa05200
Cell viability Inhibition hsa05200
In Vitro Model SW620 cells Colon Homo sapiens (Human) CVCL_0547
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Inhibition of LINC00473 in vivo could overcome the Taxol resistance of CRC cells, could recover the expression of tumor suppressor miR-15a and chemotherapy-induced tumor regression while the BCL-2-related anti-apoptosis pathway was activated and the multidrug-resistant (MDR) genes LRP, MDR1 were up-regulated by LINC00473.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [42]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal cancer [ICD-11: 2B91.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell colony Inhibition hsa05200
Cell invasion Inhibition hsa05200
Cell viability Inhibition hsa05200
In Vitro Model SW620 cells Colon Homo sapiens (Human) CVCL_0547
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Inhibition of LINC00473 in vivo could overcome the Taxol resistance of CRC cells, could recover the expression of tumor suppressor miR-15a and chemotherapy-induced tumor regression while the BCL-2-related anti-apoptosis pathway was activated and the multidrug-resistant (MDR) genes LRP, MDR1 were up-regulated by LINC00473.
Key Molecule: ATP-binding cassette sub-family B5 (ABCB5) [43]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Colorectal carcinoma [ICD-11: 2B91.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model CaCo2 cells Colon Homo sapiens (Human) CVCL_0025
HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
NIH-G185 cells Ovary Homo sapiens (Human) CVCL_L991
NIH 3T3 cells Colon Homo sapiens (Human) CVCL_0594
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description G185 cells were 27-135 fold more resistant to the cytotoxic drugs doxorubicin, vinblastine, colchicine and paclitaxel than the parental NIH 3T3 cells. Co-administration of TPGS enhanced the cytotoxicity of doxorubicin, vinblastine, paclitaxel, and colchicine in the G185 cells to levels comparable to the parental.
Pancreatic cancer [ICD-11: 2C10]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-181c [21]
Molecule Alteration Expression
Up-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
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.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase LATS2 (LATS2) [21]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
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) [21]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
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) [21]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
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) [21]
Molecule Alteration Expression
Down-regulation
Resistant Disease Pancreatic cancer [ICD-11: 2C10.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
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.
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 [44]
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.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Ribonucleoside-diphosphate reductase subunit M2 (RRM2) [44]
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) [44]
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.
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-16 [45]
Molecule Alteration Expression
Down-regulation
Resistant Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Inhibition hsa05200
NF-kappaB signaling pathway Activation hsa04064
In Vitro Model BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
HCCLM3 cells Liver Homo sapiens (Human) CVCL_6832
BEL-7404 cells Liver Homo sapiens (Human) CVCL_6568
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
PLC cells Liver Homo sapiens (Human) CVCL_0485
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Silencing the expression of miR-16 induced the chemoresistance in HCC by target IkBkB via NF-kB signaling pathway.
Key Molecule: hsa-let-7a [9]
Molecule Alteration Expression
Up-regulation
Resistant Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Caspase-3 is the key executioner caspase in apoptosis. Ectopic expression of let-7adecreased the luciferase activity of a reporter constructcontaining the 30untranslated region of caspase-3. Enforced let-7aexpression increased the resistance in A431 cells andHepG2 cells to apoptosis induced by therapeutic drugs suchas interferon-gamma, doxorubicin and paclitaxel.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: I-kappa-B-kinase beta (IKKB) [45]
Molecule Alteration Expression
Up-regulation
Resistant Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Inhibition hsa05200
NF-kappaB signaling pathway Activation hsa04064
In Vitro Model BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
HCCLM3 cells Liver Homo sapiens (Human) CVCL_6832
BEL-7404 cells Liver Homo sapiens (Human) CVCL_6568
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
PLC cells Liver Homo sapiens (Human) CVCL_0485
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
Mechanism Description Silencing the expression of miR-16 induced the chemoresistance in HCC by target IkBkB via NF-kB signaling pathway.
Key Molecule: Caspase-3 (CASP3) [9]
Molecule Alteration Expression
Down-regulation
Resistant Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Caspase-3 is the key executioner caspase in apoptosis. Ectopic expression of let-7adecreased the luciferase activity of a reporter constructcontaining the 30untranslated region of caspase-3. Enforced let-7aexpression increased the resistance in A431 cells andHepG2 cells to apoptosis induced by therapeutic drugs suchas interferon-gamma, doxorubicin and paclitaxel.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-379 [46]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation IGF1/IGF1R signaling pathway Inhibition hsa05200
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Propidium Iodide (PI) Staining
Mechanism Description IGF1 is a hub gene in HCC and is involved in the p53 signaling pathway regulation. miR379 can sensitize HCC cells to chemotherapeutic reagents via targeting IGF1R and suppressing its expression, and suppressing the IGF1/IGF1R signaling pathway.
Key Molecule: hsa-mir-335 [47]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
miR335/SIAH2/HDAC3 signaling pathway Regulation hsa05206
In Vitro Model SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Trypan blue exclusion assay; Transwell assay
Mechanism Description miR-335-mediated increased sensitivity to anti-cancer drugs was associated with its effect on HDAC3 and SIAH2 expression. miR-335 exerted apoptotic effects and inhibited ubiquitination of HDAC3 in anti-cancer drug-resistant cancer cell lines. miR-335 negatively regulated the invasion, migration, and growth rate of cancer cells. The mouse xenograft model showed that miR-335 negatively regulated the tumorigenic potential of cancer cells. The down-regulation of SIAH2 conferred sensitivity to anti-cancer drugs. The results of the study indicated that the miR-335/SIAH2/HDAC3 axis regulates the response to anti-cancer drugs.
Key Molecule: hsa-mir-223 [48]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
Skhep1 cells Liver Homo sapiens (Human) CVCL_0525
HCC3 cells Liver Homo sapiens (Human) CVCL_0C57
LM-6 cells Liver Homo sapiens (Human) CVCL_7680
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description miR-223 targeted ABCB1 3'UTR directly, and miR-223 down-regulated ABCB1 at both mRNA and protein levels. The over-expression of miR-223 increased the HCC cellsensitivity to anticancer drugs, and the inhibition of miR-223 had the opposite effect. Importantly, the over-expression or silencingof ABCB1 can rescue the cell response to the anticancer drugs mediated by miR-223 over-expression or inhibition.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [48]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
BEL-7402 cells Liver Homo sapiens (Human) CVCL_5492
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Hep3B cells Liver Homo sapiens (Human) CVCL_0326
SMMC7721 cells Uterus Homo sapiens (Human) CVCL_0534
Skhep1 cells Liver Homo sapiens (Human) CVCL_0525
HCC3 cells Liver Homo sapiens (Human) CVCL_0C57
LM-6 cells Liver Homo sapiens (Human) CVCL_7680
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
WST-1 assay
Mechanism Description miR-223 targeted ABCB1 3'UTR directly, and miR-223 down-regulated ABCB1 at both mRNA and protein levels. The over-expression of miR-223 increased the HCC cellsensitivity to anticancer drugs, and the inhibition of miR-223 had the opposite effect. Importantly, the over-expression or silencingof ABCB1 can rescue the cell response to the anticancer drugs mediated by miR-223 over-expression or inhibition.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Insulin-like growth factor 1 receptor (IGF1R) [46]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation IGF1/IGF1R signaling pathway Inhibition hsa05200
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
Experiment for
Molecule Alteration
Dual luciferase assay; Western blot analysis
Experiment for
Drug Resistance
Propidium Iodide (PI) Staining
Mechanism Description IGF1 is a hub gene in HCC and is involved in the p53 signaling pathway regulation. miR379 can sensitize HCC cells to chemotherapeutic reagents via targeting IGF1R and suppressing its expression, and suppressing the IGF1/IGF1R signaling pathway.
Key Molecule: E3 ubiquitin-protein ligase SIAH2 (SIAH2) [47]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Hepatocellular carcinoma [ICD-11: 2C12.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
miR335/SIAH2/HDAC3 signaling pathway Regulation hsa05206
In Vitro Model SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Trypan blue exclusion assay; Transwell assay
Mechanism Description miR-335-mediated increased sensitivity to anti-cancer drugs was associated with its effect on HDAC3 and SIAH2 expression. miR-335 exerted apoptotic effects and inhibited ubiquitination of HDAC3 in anti-cancer drug-resistant cancer cell lines. miR-335 negatively regulated the invasion, migration, and growth rate of cancer cells. The mouse xenograft model showed that miR-335 negatively regulated the tumorigenic potential of cancer cells. The down-regulation of SIAH2 conferred sensitivity to anti-cancer drugs. The results of the study indicated that the miR-335/SIAH2/HDAC3 axis regulates the response to anti-cancer drugs.
Laryngeal cancer [ICD-11: 2C23]
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: Polycomb complex protein BMI-1 (BMI1) [49]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Laryngeal cancer [ICD-11: 2C23.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HEp-2 cells Skin Homo sapiens (Human) CVCL_1906
AMC-HN-8 cells Larynx Homo sapiens (Human) CVCL_5966
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Overexpression of miR128a decreases the expression of BMI1 and suppresses the resistance of laryngeal cancer cells to paclitaxel & cisplatin.
Key Molecule: hsa-mir-128a [49]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Laryngeal cancer [ICD-11: 2C23.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model HEp-2 cells Skin Homo sapiens (Human) CVCL_1906
AMC-HN-8 cells Larynx Homo sapiens (Human) CVCL_5966
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Overexpression of miR128a decreases the expression of BMI1 and suppresses the resistance of laryngeal cancer cells to paclitaxel & cisplatin.
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: Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) [50]
Molecule Alteration Expression
Up-regulation
Resistant Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H460 cells Lung Homo sapiens (Human) CVCL_0459
SPC-A1 cells Lung Homo sapiens (Human) CVCL_6955
H1299 cells Lung Homo sapiens (Human) CVCL_0060
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; CCK8 assay; Colony formation assay; Flow cytometry assay
Mechanism Description MALAT1 could alter chemo-resistance (Cisplatin, Adriamycin, Gefitinib and Paclitaxel) of NSCLC cells by targeting miR-197-3p and regulating p120-ctn expression, which might assist in improvement of chemo-therapies for NSCLC.
Key Molecule: hsa-miR-197-3p [50]
Molecule Alteration Expression
Up-regulation
Resistant Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H460 cells Lung Homo sapiens (Human) CVCL_0459
SPC-A1 cells Lung Homo sapiens (Human) CVCL_6955
H1299 cells Lung Homo sapiens (Human) CVCL_0060
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; CCK8 assay; Colony formation assay; Flow cytometry assay
Mechanism Description MALAT1 could alter chemo-resistance (Cisplatin, Adriamycin, Gefitinib and Paclitaxel) of NSCLC cells by targeting miR-197-3p and regulating p120-ctn expression, which might assist in improvement of chemo-therapies for NSCLC.
Key Molecule: HOXA distal transcript antisense RNA (HOTTIP) [51]
Molecule Alteration Expression
Up-regulation
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Activation hsa04151
Cell proliferation Activation hsa05200
In Vitro Model 16HBE cells Lung Homo sapiens (Human) CVCL_0112
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Overexpression of HOTTIP promotes proliferation and drug resistance of lung adenocarcinoma by regulating AkT signaling pathway.
Key Molecule: CDKN2B antisense RNA 1 (CDKN2B-AS1) [52]
Molecule Alteration Expression
Up-regulation
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
A549/Taxol cells Lung Homo sapiens (Human) CVCL_W218
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Transwell Invasion assay
Mechanism Description ANRIL functioning as a potential oncogene was up-regulated in LAD, and promoted the acquisition of chemo-resistance in paclitaxel partly through the mitochondrial pathway by modulating the expression of apoptosis-related protein cleaved-PARP and Bcl-2. ANRIL decreases Bcl-2 expression and increases PARP expression.
Key Molecule: CDKN2B antisense RNA 1 (CDKN2B-AS1) [52]
Molecule Alteration Expression
Up-regulation
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Mitochondrial signaling pathway Activation hsa04217
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
A549/Taxol cells Lung Homo sapiens (Human) CVCL_W218
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Transwell Invasion assay
Mechanism Description ANRIL, also known as CDkN2B antisense RNA1, was origi.lly identified in the familial melanoma patients, it is located within the CDkN2B-CDkN2A gene cluster at chromosome 9p21. ANRIL decreases Bcl-2 expression and increases PARP expression.
Key Molecule: Mitosis associated long intergenic non-coding RNA 1 (MALINC1) [19]
Molecule Alteration Expression
Up-regulation
Resistant Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
In Vitro Model H1299 cells Lung Homo sapiens (Human) CVCL_0060
U2OS cells Bone Homo sapiens (Human) CVCL_0042
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Silencing of MA-linc1 in unsynchronized cells results in fewer cells in G1 and a concomitant increase in the number of cells in all other stages of the cell cycle, particularly in G2/M. Moreover, its silencing in M phase-arrested cells inhibits mitosis exit. The effect of MA-linc1 on cell cycle progression is mediated, at least in part, by repression of its neighboring gene, Puralpha, a cell cycle regulator whose expression induces cell cycle arrest. Importantly, high levels of MA-linc1 are correlated with decreased survival of breast and lung cancer patients and its silencing sensitizes cancer cells to the apoptotic effect of the M phase specific chemotherapeutic drug, Paclitaxel. This enhancement of Paclitaxel-induced apoptosis is also Puralpha-related.
Key Molecule: hsa-miR-181a-5p [53]
Molecule Alteration Expression
Up-regulation
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
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 proliferation Activation hsa05200
PI3K/AKT signaling pathway Regulation hsa04151
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Transwell assay
Mechanism Description Paclitaxel- and cisplatin-resistant A549 cells acquired metastatic properties and EMT phenotype and had reduced PTEN expression as compared to sensitive cells. miR 181a was identified as a differentially expressed miRNA in drug-resistant A549 cells, and miR-181a mimic and inhibitor were shown to affect migration, invasion, morphology and expression of EMT-associated genes. PTEN was identified as a direct target of miR-181a. Our findings demonstrate that miR-181a expression in lung adenocarcinoma is associated with EMT progression, potentially through targeting of PTEN. Regulation of miR-181a may provide a novel strategy for overcoming resistance to paclitaxel and cisplatin in lung adenocarcinoma.
Key Molecule: hsa-miR-17-5p [54]
Molecule Alteration Expression
Down-regulation
Resistant Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Trypan blue exclusion assay; Flow cytometry assay
Mechanism Description Overexpression of miR-17-5p sensitized paclitaxel resistant lung cancer cells to paclitaxel induced apoptotic cell death. miR-17-5p directly binds to the 3'-UTR of beclin 1 gene, one of the most important autophagy modulator. Overexpression of miR-17-5p into paclitaxel resistant lung cancer cells reduced beclin1 expression and a concordant decease in cellular autophagy. Paclitaxel resistance of lung cancer is associated with downregulation of miR-17-5p expression which might cause upregulation of BECN1 expression.
Key Molecule: Bcl-2-modifying factor (BMF) [55]
Molecule Alteration Expression
Down-regulation
Resistant Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H460 cells Lung Homo sapiens (Human) CVCL_0459
Experiment for
Molecule Alteration
Luciferase assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description Bmf (Bcl-2-modifying factor) is a target of miR-34c-5p, and that its silencing, together with that of c-myc, a known target of miR-34c-5p, contributes to resistance to apoptosis induced by paclitaxel through p53 downregulation.
Key Molecule: hsa-miR-34c-5p [55]
Molecule Alteration Expression
Up-regulation
Resistant Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H460 cells Lung Homo sapiens (Human) CVCL_0459
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Bmf (Bcl-2-modifying factor) is a target of miR-34c-5p, and that its silencing, together with that of c-myc, a known target of miR-34c-5p, contributes to resistance to apoptosis induced by paclitaxel through p53 downregulation.
Key Molecule: hsa-mir-135a [16]
Molecule Alteration Expression
Up-regulation
Resistant Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) is a protein that binds to HIF-1alpha and inhibits its transcriptional activity. HIF1AN is a potential miR-135a target listed in both the TargetScan and PicTar databases. miR-135a-mediated paclitaxel resistance is in part mediated by downregulation of APC.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Catenin delta-1 (CTNND1) [50]
Molecule Alteration Expression
Down-regulation
Resistant Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H460 cells Lung Homo sapiens (Human) CVCL_0459
SPC-A1 cells Lung Homo sapiens (Human) CVCL_6955
H1299 cells Lung Homo sapiens (Human) CVCL_0060
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
MTT assay; CCK8 assay; Colony formation assay; Flow cytometry assay
Mechanism Description MALAT1 could alter chemo-resistance (Cisplatin, Adriamycin, Gefitinib and Paclitaxel) of NSCLC cells by targeting miR-197-3p and regulating p120-ctn expression, which might assist in improvement of chemo-therapies for NSCLC.
Key Molecule: RAC serine/threonine-protein kinase (AKT) [51]
Molecule Alteration Expression
Down-regulation
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Activation hsa04151
Cell proliferation Activation hsa05200
In Vitro Model 16HBE cells Lung Homo sapiens (Human) CVCL_0112
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Overexpression of HOTTIP promotes proliferation and drug resistance of lung adenocarcinoma by regulating AkT signaling pathway.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [52]
Molecule Alteration Expression
Down-regulation
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Mitochondrial signaling pathway Activation hsa04217
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
A549/Taxol cells Lung Homo sapiens (Human) CVCL_W218
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Transwell Invasion assay
Mechanism Description ANRIL, also known as CDkN2B antisense RNA1, was origi.lly identified in the familial melanoma patients, it is located within the CDkN2B-CDkN2A gene cluster at chromosome 9p21. ANRIL decreases Bcl-2 expression and increases PARP expression.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [52]
Molecule Alteration Expression
Down-regulation
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
A549/Taxol cells Lung Homo sapiens (Human) CVCL_W218
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Transwell Invasion assay
Mechanism Description ANRIL functioning as a potential oncogene was up-regulated in LAD, and promoted the acquisition of chemo-resistance in paclitaxel partly through the mitochondrial pathway by modulating the expression of apoptosis-related protein cleaved-PARP and Bcl-2. ANRIL decreases Bcl-2 expression and increases PARP expression.
Key Molecule: Poly[ADP-ribose] synthase 1 (PARP1) [52]
Molecule Alteration Expression
Up-regulation
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Mitochondrial signaling pathway Activation hsa04217
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
A549/Taxol cells Lung Homo sapiens (Human) CVCL_W218
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Transwell Invasion assay
Mechanism Description ANRIL, also known as CDkN2B antisense RNA1, was origi.lly identified in the familial melanoma patients, it is located within the CDkN2B-CDkN2A gene cluster at chromosome 9p21. ANRIL decreases Bcl-2 expression and increases PARP expression.
Key Molecule: Transcriptional activator protein Pur-alpha (PURA) [19]
Molecule Alteration Expression
Down-regulation
Resistant Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
In Vitro Model H1299 cells Lung Homo sapiens (Human) CVCL_0060
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Silencing of MA-linc1 in unsynchronized cells results in fewer cells in G1 and a concomitant increase in the number of cells in all other stages of the cell cycle, particularly in G2/M. Moreover, its silencing in M phase-arrested cells inhibits mitosis exit. The effect of MA-linc1 on cell cycle progression is mediated, at least in part, by repression of its neighboring gene, Puralpha, a cell cycle regulator whose expression induces cell cycle arrest. Importantly, high levels of MA-linc1 are correlated with decreased survival of breast and lung cancer patients and its silencing sensitizes cancer cells to the apoptotic effect of the M phase specific chemotherapeutic drug, Paclitaxel. This enhancement of Paclitaxel-induced apoptosis is also Puralpha-related.
Key Molecule: Phosphatase and tensin homolog (PTEN) [53]
Molecule Alteration Expression
Down-regulation
Resistant Disease Lung adenocarcinoma [ICD-11: 2C25.0]
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 proliferation Activation hsa05200
PI3K/AKT signaling pathway Regulation hsa04151
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Transwell assay
Mechanism Description Paclitaxel- and cisplatin-resistant A549 cells acquired metastatic properties and EMT phenotype and had reduced PTEN expression as compared to sensitive cells. miR 181a was identified as a differentially expressed miRNA in drug-resistant A549 cells, and miR-181a mimic and inhibitor were shown to affect migration, invasion, morphology and expression of EMT-associated genes. PTEN was identified as a direct target of miR-181a. Our findings demonstrate that miR-181a expression in lung adenocarcinoma is associated with EMT progression, potentially through targeting of PTEN. Regulation of miR-181a may provide a novel strategy for overcoming resistance to paclitaxel and cisplatin in lung adenocarcinoma.
Key Molecule: Beclin-1 (BECN1) [54]
Molecule Alteration Expression
Up-regulation
Resistant Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Trypan blue exclusion assay; Flow cytometry assay
Mechanism Description Overexpression of miR-17-5p sensitized paclitaxel resistant lung cancer cells to paclitaxel induced apoptotic cell death. miR-17-5p directly binds to the 3'-UTR of beclin 1 gene, one of the most important autophagy modulator. Overexpression of miR-17-5p into paclitaxel resistant lung cancer cells reduced beclin1 expression and a concordant decease in cellular autophagy. Paclitaxel resistance of lung cancer is associated with downregulation of miR-17-5p expression which might cause upregulation of BECN1 expression.
Key Molecule: Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) [16]
Molecule Alteration Expression
Down-regulation
Resistant Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) is a protein that binds to HIF-1alpha and inhibits its transcriptional activity. HIF1AN is a potential miR-135a target listed in both the TargetScan and PicTar databases. miR-135a-mediated paclitaxel resistance is in part mediated by downregulation of APC.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-128a [56]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
Rapamycin signaling pathway Inhibition hsa04211
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR128 can effectively suppress PTX-resistant lung cancer stem cells via inhibition of BMI-1 and MUC1-C, thus downregulating the PI3k/AkT pathway and the rapamycin pathway.
Key Molecule: hsa-mir-200c [57]
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
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description A miRNA-200c/cathepsin L feedback loop determines paclitaxel resistance in human lung cancer A549 cells in vitro through regulating epithelial-mesenchymal transition.
Key Molecule: hsa-miR-107 [58]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis; Caspase-3 activity assay
Mechanism Description Overexpression of miR107 promotes apoptosis and inhibits proliferation and mobility of A549/Taxol cells under treatment with paclitaxel in vitro. miR107/Bcl-w axis regulates paclitaxel chemoresistance through PI3k-Akt pathway, up-regulation of miR107 resensitizes paclitaxel-resistant NSCLC cells by targeting Bcl-w. Aberrant activation of PI3k-Akt pathway and genetic alterations of its components lead to tumorigenesis.
Key Molecule: hsa-miR-30a-5p [59]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H460 cells Lung Homo sapiens (Human) CVCL_0459
A549/PR cells Lung Homo sapiens (Human) CVCL_0023
H460/PR cells Lung Homo sapiens (Human) CVCL_0459
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometric analysis
Mechanism Description miR30a-5p increases paclitaxel sensitivity by promoting chemotherapy-induced apoptosis via downregulating BCL-2.
Key Molecule: hsa-mir-186 [60]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation p53 signaling pathway Activation hsa04115
In Vitro Model Calu3 cells Lung Homo sapiens (Human) CVCL_0609
H1975 cells Lung Homo sapiens (Human) CVCL_1511
A549 cells Lung Homo sapiens (Human) CVCL_0023
H1299 cells Lung Homo sapiens (Human) CVCL_0060
H4006 cells Lung Homo sapiens (Human) N.A.
293FT cells Kidney Homo sapiens (Human) CVCL_6911
HCC95 cells Lung Homo sapiens (Human) CVCL_5137
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CellTiter-Glo assay
Mechanism Description miR186 regulates chemo-sensitivity to paclitaxel via targeting MAPT in non-small cell lung cancer The chemosensitizing effects of miR186 are partially due to the induction of the p53 mediated apoptotic pathway via MAPT down-regulation.
Key Molecule: Nuclear paraspeckle assembly transcript 1 (NEAT1) [61]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT/mTOR signaling pathway Inhibition hsa04150
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H1299 cells Lung Homo sapiens (Human) CVCL_0060
PC9 cells Lung Homo sapiens (Human) CVCL_B260
H1573 cells Lung Homo sapiens (Human) CVCL_1478
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description NEAT1 was upregulated significantly in paclitaxel-resistant NSCLC cell line while knockdown of NEAT1 could reverse the paclitaxel-resistance through induction of apoptosis by increasing cleaved PARP and cleaved caspase-3 expression.
Key Molecule: Maternally expressed 3 (MEG3) [62]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
MEG3-P53 signaling pathway Activation hsa04115
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description The downregulation of MEG3 attenuated PTX-induced cytotoxicity, whereas upregulation of MEG3 induced cell death and increased P53 expression.
Key Molecule: hsa-miR-935 [63]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
293T cells Breast Homo sapiens (Human) CVCL_0063
Experiment for
Molecule Alteration
PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Knockdown of miR 935 increases paclitaxel sensitivity via regulation of SOX7 in non small cell lung cancer.
Key Molecule: hsa-miR-424-3p [64]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model H1975 cells Lung Homo sapiens (Human) CVCL_1511
A549 cells Lung Homo sapiens (Human) CVCL_0023
NCI-H441 cells Lung Homo sapiens (Human) CVCL_1561
H2172 cells Lung Homo sapiens (Human) CVCL_1537
H827 cells Lung Homo sapiens (Human) N.A.
PC-14 cells Lung Homo sapiens (Human) CVCL_1640
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Wound-healing and Transwell assay
Mechanism Description miR-424-3p was discovered to suppress the level of YAP1 protein by targeting its 3' untranslated region, suggesting that miR-424-3p could be a potential molecular target for treatment of NSCLC with chemoresistance.
Key Molecule: hsa-mir-137 [65]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
PI3K/AKT signaling pathway Regulation hsa04151
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
A549/CDDP cells Lung Homo sapiens (Human) CVCL_0023
A549/PTX cells Lung Homo sapiens (Human) CVCL_W218
Experiment for
Molecule Alteration
RT-PCR; qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Overexpression of miR-137 in A549/PTX and A549/CDDP cells inhibited cell proliferation, migration, induced cell apoptosis, arrest the cell cycle in G1 phase and reversed drug resistance to PTX and CDDP in A549/PTX and A549/CDDP cell lines respectively. NUCkS1 is a direct target of miR-137, and is elevated in human lung cancer tissues, which is inversely correlated with miR-137 expression levels. miR-137 enhances the chemosensitivity of paclitaxel and cisplatin in vivo.
Key Molecule: hsa-let-7b [44]
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-16 [66]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
PI3K/AKT/mTOR signaling pathway Regulation hsa04151
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H1299 cells Lung Homo sapiens (Human) CVCL_0060
NCl-H596 cells Lung Homo sapiens (Human) CVCL_1571
NCI-H1734 cells Lung Homo sapiens (Human) CVCL_1491
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-16 was also significantly downregulated in paclitaxel resistant lung cancer cells. anti-apoptotic protein Bcl-2 was directly targeted miR-16 in paclitaxel resistant lung cancer cells. the combined overexpression of miR-16 and miR-17 and subsequent paclitaxel treatment greatly sensitized paclitaxel resistant lung cancer cells to paclitaxel by inducing apoptosis via caspase-3 mediated pathway. Combined overexpression of miR-16 and miR-17 greatly reduced Beclin-1 and Bcl-2 expressions respectively. though miR-17 and miR-16 had no common target, both miR-16 and miR-17 jointly played roles in the development of paclitaxel resistance in lung cancer. miR-17 overexpression reduced cytoprotective autophagy by targeting Beclin-1, whereas overexpression of miR-16 potentiated paclitaxel induced apoptotic cell death by inhibiting anti-apoptotic protein Bcl-2.
Key Molecule: hsa-mir-17 [66]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT/mTOR signaling pathway Regulation hsa04151
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H1299 cells Lung Homo sapiens (Human) CVCL_0060
NCl-H596 cells Lung Homo sapiens (Human) CVCL_1571
NCI-H1734 cells Lung Homo sapiens (Human) CVCL_1491
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-16 was also significantly downregulated in paclitaxel resistant lung cancer cells. anti-apoptotic protein Bcl-2 was directly targeted miR-16 in paclitaxel resistant lung cancer cells. the combined overexpression of miR-16 and miR-17 and subsequent paclitaxel treatment greatly sensitized paclitaxel resistant lung cancer cells to paclitaxel by inducing apoptosis via caspase-3 mediated pathway. Combined overexpression of miR-16 and miR-17 greatly reduced Beclin-1 and Bcl-2 expressions respectively. though miR-17 and miR-16 had no common target, both miR-16 and miR-17 jointly played roles in the development of paclitaxel resistance in lung cancer. miR-17 overexpression reduced cytoprotective autophagy by targeting Beclin-1, whereas overexpression of miR-16 potentiated paclitaxel induced apoptotic cell death by inhibiting anti-apoptotic protein Bcl-2.
Key Molecule: hsa-mir-7 [67]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
95D cells Lung Homo sapiens (Human) CVCL_7110
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Overexpression of miR-7 sensitizes NSCLC cells to PTX and inhibites EGFR expression in A549 cells.
Key Molecule: hsa-miR-337-3p [68]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model NCI-H1155 cells Lung Homo sapiens (Human) CVCL_1456
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description Over-expression of miR-337-3p nor the specific knockdown of STAT3 and RAP1A significantly decrease cell viability or induce G2/M arrest alone, but rather enhance G2/M arrest and cell death only under conditions of paclitaxel treatment.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: CX3C chemokine receptor 1 (CX3CR1) [69]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model Calu3 cells Lung Homo sapiens (Human) CVCL_0609
A549 cells Lung Homo sapiens (Human) CVCL_0023
SPC-A1 cells Lung Homo sapiens (Human) CVCL_6955
HCC827 cells Lung Homo sapiens (Human) CVCL_2063
NCI-H358 cells Lung Homo sapiens (Human) CVCL_1559
H157 cells Lung Homo sapiens (Human) CVCL_2458
D6 cells Lung Homo sapiens (Human) N.A.
LAX cells Lung Homo sapiens (Human) N.A.
LTEP-2 cells Lung Homo sapiens (Human) CVCL_6929
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR296-3p inhibited NSCLC cell proliferation, enhance the drug resistance, and apoptosis. Data of luciferase reporter assays demonstrated that the CX3CR1 gene was a direct regulator of tumorsuppressive miR296-3p. Moreover, overexpressed CX3CR1 was confirmed in NSCLC clinical specimens. Inhibition of CX3CR1 could inhibit cancer cellular survival and increase chemotherapy sensitivity. There was a negative relationship between miR296-3p and CX3CR1 expression in NSCLC tissues.
Key Molecule: hsa-miR-296-3p [69]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model Calu3 cells Lung Homo sapiens (Human) CVCL_0609
A549 cells Lung Homo sapiens (Human) CVCL_0023
SPC-A1 cells Lung Homo sapiens (Human) CVCL_6955
HCC827 cells Lung Homo sapiens (Human) CVCL_2063
NCI-H358 cells Lung Homo sapiens (Human) CVCL_1559
H157 cells Lung Homo sapiens (Human) CVCL_2458
D6 cells Lung Homo sapiens (Human) N.A.
LAX cells Lung Homo sapiens (Human) N.A.
LTEP-2 cells Lung Homo sapiens (Human) CVCL_6929
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR296-3p inhibited NSCLC cell proliferation, enhance the drug resistance, and apoptosis. Data of luciferase reporter assays demonstrated that the CX3CR1 gene was a direct regulator of tumorsuppressive miR296-3p. Moreover, overexpressed CX3CR1 was confirmed in NSCLC clinical specimens. Inhibition of CX3CR1 could inhibit cancer cellular survival and increase chemotherapy sensitivity. There was a negative relationship between miR296-3p and CX3CR1 expression in NSCLC tissues.
Key Molecule: hsa-mir-125b [70]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H460 cells Lung Homo sapiens (Human) CVCL_0459
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-125b was significantly downregulated in A549-PR and H460-PR cells. Notably, ectopic expression of miR-125b led to the reversal of EMT phenotype. Moreover, we found that miR-125b governed PR-induced EMT partly due to down-regulation of its target Sema4C. More importantly, overexpression of miR-125b or depletion of Sema4C sensitized PR cells to paclitaxel. Furthermore, stable overexpression miR-125b in A549-PR cells inhibited tumor xenograft growth in immunodeficient mice. Our study implied that up-regulation of miR-125b could be a novel approach to reverse chemotherapy resistance in lung cancers.
Key Molecule: Semaphorin-4C (SEMA4C) [70]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H460 cells Lung Homo sapiens (Human) CVCL_0459
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-125b was significantly downregulated in A549-PR and H460-PR cells. Notably, ectopic expression of miR-125b led to the reversal of EMT phenotype. Moreover, we found that miR-125b governed PR-induced EMT partly due to down-regulation of its target Sema4C. More importantly, overexpression of miR-125b or depletion of Sema4C sensitized PR cells to paclitaxel. Furthermore, stable overexpression miR-125b in A549-PR cells inhibited tumor xenograft growth in immunodeficient mice. Our study implied that up-regulation of miR-125b could be a novel approach to reverse chemotherapy resistance in lung cancers.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Polycomb complex protein BMI-1 (BMI1) [56]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
Rapamycin signaling pathway Inhibition hsa04211
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Luciferase activity assay
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR128 can effectively suppress PTX-resistant lung cancer stem cells via inhibition of BMI-1 and MUC1-C, thus downregulating the PI3k/AkT pathway and the rapamycin pathway.
Key Molecule: Mucin-1 (MUC1) [56]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
Rapamycin signaling pathway Inhibition hsa04211
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Luciferase activity assay
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR128 can effectively suppress PTX-resistant lung cancer stem cells via inhibition of BMI-1 and MUC1-C, thus downregulating the PI3k/AkT pathway and the rapamycin pathway.
Key Molecule: Procathepsin L (CTSL) [57]
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
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description A miRNA-200c/cathepsin L feedback loop determines paclitaxel resistance in human lung cancer A549 cells in vitro through regulating epithelial-mesenchymal transition.
Key Molecule: Procathepsin L (CTSL) [57]
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
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description A miRNA-200c/cathepsin L feedback loop determines paclitaxel resistance in human lung cancer A549 cells in vitro through regulating epithelial-mesenchymal transition.
Key Molecule: Bcl-2-like protein 2 (BCL2L2) [58]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
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
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Dual-luciferase reporter assay; qRT-PCR; Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis; Caspase-3 activity assay
Mechanism Description Overexpression of miR107 promotes apoptosis and inhibits proliferation and mobility of A549/Taxol cells under treatment with paclitaxel in vitro. miR107/Bcl-w axis regulates paclitaxel chemoresistance through PI3k-Akt pathway, up-regulation of miR107 resensitizes paclitaxel-resistant NSCLC cells by targeting Bcl-w. Aberrant activation of PI3k-Akt pathway and genetic alterations of its components lead to tumorigenesis.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [59]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H460 cells Lung Homo sapiens (Human) CVCL_0459
A549/PR cells Lung Homo sapiens (Human) CVCL_0023
H460/PR cells Lung Homo sapiens (Human) CVCL_0459
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Luciferase reporter assay; Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometric analysis
Mechanism Description miR30a-5p increases paclitaxel sensitivity by promoting chemotherapy-induced apoptosis via downregulating BCL-2.
Key Molecule: Microtubule-associated protein tau (MAPT) [60]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation p53 signaling pathway Activation hsa04115
In Vitro Model Calu3 cells Lung Homo sapiens (Human) CVCL_0609
H1975 cells Lung Homo sapiens (Human) CVCL_1511
A549 cells Lung Homo sapiens (Human) CVCL_0023
H1299 cells Lung Homo sapiens (Human) CVCL_0060
H4006 cells Lung Homo sapiens (Human) N.A.
293FT cells Kidney Homo sapiens (Human) CVCL_6911
HCC95 cells Lung Homo sapiens (Human) CVCL_5137
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Luciferase assay
Experiment for
Drug Resistance
CellTiter-Glo assay
Mechanism Description miR186 regulates chemo-sensitivity to paclitaxel via targeting MAPT in non-small cell lung cancer The chemosensitizing effects of miR186 are partially due to the induction of the p53 mediated apoptotic pathway via MAPT down-regulation.
Key Molecule: RAC serine/threonine-protein kinase (AKT) [61]
Molecule Alteration Phosphorylation
Down-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT/mTOR signaling pathway Inhibition hsa04150
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H1299 cells Lung Homo sapiens (Human) CVCL_0060
PC9 cells Lung Homo sapiens (Human) CVCL_B260
H1573 cells Lung Homo sapiens (Human) CVCL_1478
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description NEAT1 was upregulated significantly in paclitaxel-resistant NSCLC cell line while knockdown of NEAT1 could reverse the paclitaxel-resistance through induction of apoptosis by increasing cleaved PARP and cleaved caspase-3 expression.
Key Molecule: Cellular tumor antigen p53 (TP53) [62]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
MEG3-P53 signaling pathway Activation hsa04115
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description The downregulation of MEG3 attenuated PTX-induced cytotoxicity, whereas upregulation of MEG3 induced cell death and increased P53 expression.
Key Molecule: Transcription factor SOX-7 (SOX7) [63]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
293T cells Breast Homo sapiens (Human) CVCL_0063
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Knockdown of miR 935 increases paclitaxel sensitivity via regulation of SOX7 in non small cell lung cancer.
Key Molecule: Transcriptional coactivator YAP1 (YAP1) [64]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model H1975 cells Lung Homo sapiens (Human) CVCL_1511
A549 cells Lung Homo sapiens (Human) CVCL_0023
NCI-H441 cells Lung Homo sapiens (Human) CVCL_1561
H2172 cells Lung Homo sapiens (Human) CVCL_1537
H827 cells Lung Homo sapiens (Human) N.A.
PC-14 cells Lung Homo sapiens (Human) CVCL_1640
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Wound-healing and Transwell assay
Mechanism Description miR-424-3p was discovered to suppress the level of YAP1 protein by targeting its 3' untranslated region, suggesting that miR-424-3p could be a potential molecular target for treatment of NSCLC with chemoresistance.
Key Molecule: Nuclear ubiquitous casein CDK substrate 1 (NUCKS1) [65]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
PI3K/AKT signaling pathway Regulation hsa04151
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
A549/CDDP cells Lung Homo sapiens (Human) CVCL_0023
A549/PTX cells Lung Homo sapiens (Human) CVCL_W218
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Overexpression of miR-137 in A549/PTX and A549/CDDP cells inhibited cell proliferation, migration, induced cell apoptosis, arrest the cell cycle in G1 phase and reversed drug resistance to PTX and CDDP in A549/PTX and A549/CDDP cell lines respectively. NUCkS1 is a direct target of miR-137, and is elevated in human lung cancer tissues, which is inversely correlated with miR-137 expression levels. miR-137 enhances the chemosensitivity of paclitaxel and cisplatin in vivo.
Key Molecule: Ribonucleoside-diphosphate reductase subunit M2 (RRM2) [44]
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) [44]
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: G1/S-specific cyclin-D1 (CCND1) [66]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT/mTOR signaling pathway Regulation hsa04151
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H1299 cells Lung Homo sapiens (Human) CVCL_0060
NCl-H596 cells Lung Homo sapiens (Human) CVCL_1571
NCI-H1734 cells Lung Homo sapiens (Human) CVCL_1491
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-16 was also significantly downregulated in paclitaxel resistant lung cancer cells. anti-apoptotic protein Bcl-2 was directly targeted miR-16 in paclitaxel resistant lung cancer cells. the combined overexpression of miR-16 and miR-17 and subsequent paclitaxel treatment greatly sensitized paclitaxel resistant lung cancer cells to paclitaxel by inducing apoptosis via caspase-3 mediated pathway. Combined overexpression of miR-16 and miR-17 greatly reduced Beclin-1 and Bcl-2 expressions respectively. though miR-17 and miR-16 had no common target, both miR-16 and miR-17 jointly played roles in the development of paclitaxel resistance in lung cancer. miR-17 overexpression reduced cytoprotective autophagy by targeting Beclin-1, whereas overexpression of miR-16 potentiated paclitaxel induced apoptotic cell death by inhibiting anti-apoptotic protein Bcl-2.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [66]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
PI3K/AKT/mTOR signaling pathway Regulation hsa04151
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H1299 cells Lung Homo sapiens (Human) CVCL_0060
NCl-H596 cells Lung Homo sapiens (Human) CVCL_1571
NCI-H1734 cells Lung Homo sapiens (Human) CVCL_1491
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-16 was also significantly downregulated in paclitaxel resistant lung cancer cells. anti-apoptotic protein Bcl-2 was directly targeted miR-16 in paclitaxel resistant lung cancer cells. the combined overexpression of miR-16 and miR-17 and subsequent paclitaxel treatment greatly sensitized paclitaxel resistant lung cancer cells to paclitaxel by inducing apoptosis via caspase-3 mediated pathway. Combined overexpression of miR-16 and miR-17 greatly reduced Beclin-1 and Bcl-2 expressions respectively. though miR-17 and miR-16 had no common target, both miR-16 and miR-17 jointly played roles in the development of paclitaxel resistance in lung cancer. miR-17 overexpression reduced cytoprotective autophagy by targeting Beclin-1, whereas overexpression of miR-16 potentiated paclitaxel induced apoptotic cell death by inhibiting anti-apoptotic protein Bcl-2.
Key Molecule: Epidermal growth factor receptor (EGFR) [67]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
95D cells Lung Homo sapiens (Human) CVCL_7110
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Overexpression of miR-7 sensitizes NSCLC cells to PTX and inhibites EGFR expression in A549 cells.
Key Molecule: Ras-related protein Rap-1A (RAP1A) [68]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model NCI-H1155 cells Lung Homo sapiens (Human) CVCL_1456
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description Over-expression of miR-337-3p nor the specific knockdown of STAT3 and RAP1A significantly decrease cell viability or induce G2/M arrest alone, but rather enhance G2/M arrest and cell death only under conditions of paclitaxel treatment.
Key Molecule: Signal transducer activator transcription 3 (STAT3) [68]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model NCI-H1155 cells Lung Homo sapiens (Human) CVCL_1456
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description Over-expression of miR-337-3p nor the specific knockdown of STAT3 and RAP1A significantly decrease cell viability or induce G2/M arrest alone, but rather enhance G2/M arrest and cell death only under conditions of paclitaxel treatment.
Pleural mesothelioma [ICD-11: 2C26]
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-149 [22]
Molecule Alteration Expression
Up-regulation
Resistant Disease Malignant pleural mesothelioma [ICD-11: 2C26.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H69 cells Lung Homo sapiens (Human) CVCL_8121
H69AR cells Lung Homo sapiens (Human) CVCL_3513
MSTO-211H cells Lung Homo sapiens (Human) CVCL_1430
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description micro-RNA149 confers taxane resistance to malignant mesothelioma cells via upregulation of P-glycoprotein expression.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: ATP-binding cassette sub-family B5 (ABCB5) [22]
Molecule Alteration Expression
Up-regulation
Resistant Disease Malignant pleural mesothelioma [ICD-11: 2C26.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
H69 cells Lung Homo sapiens (Human) CVCL_8121
H69AR cells Lung Homo sapiens (Human) CVCL_3513
MSTO-211H cells Lung Homo sapiens (Human) CVCL_1430
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description micro-RNA149 confers taxane resistance to malignant mesothelioma cells via upregulation of P-glycoprotein expression.
Melanoma [ICD-11: 2C30]
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-326 [17]
Molecule Alteration Expression
Up-regulation
Resistant Disease Melanoma [ICD-11: 2C30.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
In Vitro Model SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Malme3M cells Skin Homo sapiens (Human) CVCL_1438
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-326, which forms a negative feedback regulatory loop with HDAC3, regulates the invasion and the metastatic potential of cancer cells and tumor-induced angiogenesis in response to anti-cancer drugs. miR-200b, miR-217, and miR-335, which form a positive feedback loop with HDAC3, confer sensitivity to anti-cancer drugs. We show that CAGE, reported to form a feedback loop with miR-200b, serves as a downstream target of HDAC3 and miR-326. In this study, we show that the regulation of the miR-326/HDAC3 axis can be employed for the development of anti-cancer therapeutics.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Histone deacetylase 3 (HDAC3) [17]
Molecule Alteration Expression
Down-regulation
Resistant Disease Melanoma [ICD-11: 2C30.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
In Vitro Model SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Malme3M cells Skin Homo sapiens (Human) CVCL_1438
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-326, which forms a negative feedback regulatory loop with HDAC3, regulates the invasion and the metastatic potential of cancer cells and tumor-induced angiogenesis in response to anti-cancer drugs. miR-200b, miR-217, and miR-335, which form a positive feedback loop with HDAC3, confer sensitivity to anti-cancer drugs. We show that CAGE, reported to form a feedback loop with miR-200b, serves as a downstream target of HDAC3 and miR-326. In this study, we show that the regulation of the miR-326/HDAC3 axis can be employed for the development of anti-cancer therapeutics.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-335 [17], [47]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
miR335/SIAH2/HDAC3 signaling pathway Regulation hsa05206
In Vitro Model Malme3M cells Skin Homo sapiens (Human) CVCL_1438
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Trypan blue exclusion assay; Transwell assay
Mechanism Description miR-335-mediated increased sensitivity to anti-cancer drugs was associated with its effect on HDAC3 and SIAH2 expression. miR-335 exerted apoptotic effects and inhibited ubiquitination of HDAC3 in anti-cancer drug-resistant cancer cell lines. miR-335 negatively regulated the invasion, migration, and growth rate of cancer cells. The mouse xenograft model showed that miR-335 negatively regulated the tumorigenic potential of cancer cells. The down-regulation of SIAH2 conferred sensitivity to anti-cancer drugs. The results of the study indicated that the miR-335/SIAH2/HDAC3 axis regulates the response to anti-cancer drugs.
Key Molecule: hsa-mir-217 [17]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Malme3M cells Skin Homo sapiens (Human) CVCL_1438
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-326, which forms a negative feedback regulatory loop with HDAC3, regulates the invasion and the metastatic potential of cancer cells and tumor-induced angiogenesis in response to anti-cancer drugs. miR-200b, miR-217, and miR-335, which form a positive feedback loop with HDAC3, confer sensitivity to anti-cancer drugs. We show that CAGE, reported to form a feedback loop with miR-200b, serves as a downstream target of HDAC3 and miR-326. In this study, we show that the regulation of the miR-326/HDAC3 axis can be employed for the development of anti-cancer therapeutics.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: hsa-mir-200b [17]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Malme3M cells Skin Homo sapiens (Human) CVCL_1438
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-326, which forms a negative feedback regulatory loop with HDAC3, regulates the invasion and the metastatic potential of cancer cells and tumor-induced angiogenesis in response to anti-cancer drugs. miR-200b, miR-217, and miR-335, which form a positive feedback loop with HDAC3, confer sensitivity to anti-cancer drugs. We show that CAGE, reported to form a feedback loop with miR-200b, serves as a downstream target of HDAC3 and miR-326. In this study, we show that the regulation of the miR-326/HDAC3 axis can be employed for the development of anti-cancer therapeutics.
Key Molecule: hsa-miR-326 [17]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model SNU387 cells Liver Homo sapiens (Human) CVCL_0250
Malme3M cells Skin Homo sapiens (Human) CVCL_1438
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-326, which forms a negative feedback regulatory loop with HDAC3, regulates the invasion and the metastatic potential of cancer cells and tumor-induced angiogenesis in response to anti-cancer drugs. miR-200b, miR-217, and miR-335, which form a positive feedback loop with HDAC3, confer sensitivity to anti-cancer drugs. We show that CAGE, reported to form a feedback loop with miR-200b, serves as a downstream target of HDAC3 and miR-326. In this study, we show that the regulation of the miR-326/HDAC3 axis can be employed for the development of anti-cancer therapeutics.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: E3 ubiquitin-protein ligase SIAH2 (SIAH2) [47]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
miR335/SIAH2/HDAC3 signaling pathway Regulation hsa05206
In Vitro Model Malme3M cells Skin Homo sapiens (Human) CVCL_1438
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Trypan blue exclusion assay; Transwell assay
Mechanism Description miR-335-mediated increased sensitivity to anti-cancer drugs was associated with its effect on HDAC3 and SIAH2 expression. miR-335 exerted apoptotic effects and inhibited ubiquitination of HDAC3 in anti-cancer drug-resistant cancer cell lines. miR-335 negatively regulated the invasion, migration, and growth rate of cancer cells. The mouse xenograft model showed that miR-335 negatively regulated the tumorigenic potential of cancer cells. The down-regulation of SIAH2 conferred sensitivity to anti-cancer drugs. The results of the study indicated that the miR-335/SIAH2/HDAC3 axis regulates the response to anti-cancer drugs.
Skin squamous cell carcinoma [ICD-11: 2C31]
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-let-7a [9]
Molecule Alteration Expression
Up-regulation
Resistant Disease Cutaneous squamous cell carcinoma [ICD-11: 2C31.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model A431 cells Skin Homo sapiens (Human) CVCL_0037
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Caspase-3 is the key executioner caspase in apoptosis. Ectopic expression of let-7adecreased the luciferase activity of a reporter constructcontaining the 30untranslated region of caspase-3. Enforced let-7aexpression increased the resistance in A431 cells andHepG2 cells to apoptosis induced by therapeutic drugs suchas interferon-gamma, doxorubicin and paclitaxel.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Caspase-3 (CASP3) [9]
Molecule Alteration Expression
Down-regulation
Resistant Disease Cutaneous squamous cell carcinoma [ICD-11: 2C31.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model A431 cells Skin Homo sapiens (Human) CVCL_0037
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Caspase-3 is the key executioner caspase in apoptosis. Ectopic expression of let-7adecreased the luciferase activity of a reporter constructcontaining the 30untranslated region of caspase-3. Enforced let-7aexpression increased the resistance in A431 cells andHepG2 cells to apoptosis induced by therapeutic drugs suchas interferon-gamma, doxorubicin and paclitaxel.
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-107 [71]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell apoptosis Inhibition hsa04210
Wnt/Beta-catenin signaling pathway Activation hsa04310
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-107 could enhance PTX sensitivity in breast cancer cells may be targeting TPD52 through Wnt/beta-catenin signaling pathway. And downregualtion of miR-107 ould enhance PTX resistance in BC cells.
Key Molecule: hsa-mir-29c [72]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
T47D cells Breast Homo sapiens (Human) CVCL_0553
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description CDk6 knockdown attenuated the effects of miR-29c inhibition on paclitaxel cytotoxicity in breast cancer cells.
Key Molecule: H19, imprinted maternally expressed transcript (H19) [73]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cullin4A/MDR1 signaling pathway Regulation hsa05206
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
Quantitative real-time RT-PCR
Experiment for
Drug Resistance
CellTiter AQueous One Solution Cell Proliferation Assay
Mechanism Description LncRNA H19 is a major mediator of doxorubicin chemoresistance in breast cancer cells through a cullin4A-MDR1 pathway. H19 overexpression was contributed to cancer cell resistance to anthracyclines and paclitaxel as knockdown of H19 LncRNA by a specific H19 shRNA in Dox-resistant cells significantly improved the cell sensitivity to anthracyclines and paclitaxel.
Key Molecule: Bcl-2-interacting killer (BIK) [74]
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
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
MCF-7R cells Breast Homo sapiens (Human) CVCL_Y493
ZR-75-1R cells Breast Homo sapiens (Human) CVCL_0588
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Annexin V apoptosis assay; Fow cytometry analysis
Mechanism Description LncRNA H19 attenuated cell apoptosis in response to PTX treatment by inhibiting transcription of pro-apoptotic genes BIk and NOXA.
Key Molecule: Estrogen receptor alpha (ESR1) [74]
Molecule Alteration Expression
Up-regulation
Resistant Disease ER positive breast cancer [ICD-11: 2C60.6]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
MCF-7R cells Breast Homo sapiens (Human) CVCL_Y493
ZR-75-1R cells Breast Homo sapiens (Human) CVCL_0588
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
Annexin V apoptosis assay; Fow cytometry analysis
Mechanism Description LncRNA H19 was identified as a powerful factor associated with paclitaxel (PTX) resistance in ERalpha-positive breast cancer cells, but not in ERalpha-negative breast cancer cells. LncRNA H19 was one of the downstream target molecules of ERalpha. Altered ERalpha expression may therefore change H19 levels to modulate the apoptosis response to chemotherapy in breast cancer cells.
Key Molecule: H19, imprinted maternally expressed transcript (H19) [74]
Molecule Alteration Expression
Up-regulation
Resistant Disease ER positive breast cancer [ICD-11: 2C60.6]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
MCF-7R cells Breast Homo sapiens (Human) CVCL_Y493
ZR-75-1R cells Breast Homo sapiens (Human) CVCL_0588
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Annexin V apoptosis assay; Fow cytometry analysis
Mechanism Description LncRNA H19 was identified as a powerful factor associated with paclitaxel (PTX) resistance in ERalpha-positive breast cancer cells, but not in ERalpha-negative breast cancer cells. LncRNA H19 was one of the downstream target molecules of ERalpha. Altered ERalpha expression may therefore change H19 levels to modulate the apoptosis response to chemotherapy in breast cancer cells.
Key Molecule: Phorbol-12-myristate-13-acetate-induced protein 1 (PMAIP1) [74]
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
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
MCF-7R cells Breast Homo sapiens (Human) CVCL_Y493
ZR-75-1R cells Breast Homo sapiens (Human) CVCL_0588
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Annexin V apoptosis assay; Fow cytometry analysis
Mechanism Description LncRNA H19 attenuated cell apoptosis in response to PTX treatment by inhibiting transcription of pro-apoptotic genes BIk and NOXA.
Key Molecule: Phorbol-12-myristate-13-acetate-induced protein 1 (PMAIP1) [74]
Molecule Alteration Expression
Down-regulation
Resistant Disease ER positive breast cancer [ICD-11: 2C60.6]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
MCF-7R cells Breast Homo sapiens (Human) CVCL_Y493
ZR-75-1R cells Breast Homo sapiens (Human) CVCL_0588
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Annexin V apoptosis assay; Fow cytometry analysis
Mechanism Description LncRNA H19 attenuated cell apoptosis in response to PTX treatment by inhibiting transcription of pro-apoptotic genes BIk and NOXA.
Key Molecule: hsa-mir-17 [75]
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
MCF-7/Tax1 cells Breast Homo sapiens (Human) CVCL_IJ26
MCF-7/Tax2 cells Breast Homo sapiens (Human) CVCL_IJ26
MDA-MB-231/Tax1 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-231/Tax2 cells Breast Homo sapiens (Human) CVCL_0062
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Annexin-V-FITC (fluorescein isothiocyanate)/PI (propidium iodide) analysis
Mechanism Description Decreased expression of microRNA-17 and microRNA-20b promotes breast cancer resistance to taxol therapy by upregulation of NCOA3.
Key Molecule: hsa-mir-20b [75]
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
MCF-7/Tax1 cells Breast Homo sapiens (Human) CVCL_IJ26
MCF-7/Tax2 cells Breast Homo sapiens (Human) CVCL_IJ26
MDA-MB-231/Tax1 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-231/Tax2 cells Breast Homo sapiens (Human) CVCL_0062
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Annexin-V-FITC (fluorescein isothiocyanate)/PI (propidium iodide) analysis
Mechanism Description Decreased expression of microRNA-17 and microRNA-20b promotes breast cancer resistance to taxol therapy by upregulation of NCOA3.
Key Molecule: hsa-miR-155-3p [76]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell viability Activation hsa05200
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
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
SRB assay
Mechanism Description miR-155-3p acts as a tumor suppressor and reverses paclitaxel resistance via negative regulation of MYD88 in human breast cancer.
Key Molecule: hsa-miR-1207-5p [77]
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
MDA-MB-436 cells Breast Homo sapiens (Human) CVCL_0623
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-1207-5p induces the resistence of triple-negative breast cancer cells to Taxol treatment via the suppression of LZTS1 expression.
Key Molecule: hsa-miR-200c-3p [78]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Down-regulation of miR 200c 3p contributes to the resistance of breast cancer cells to paclitaxel by targeting SOX2.
Key Molecule: hsa-mir-18a [79]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-18a is an important miRNA that suppresses Dicer expression and increases paclitaxel resistance in triple-negative breast cancer cells.
Key Molecule: hsa-miR-106b~25 [80]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
miR106b~25 cluster/EP300/E-cadherin signaling pathway Regulation hsa05206
In Vitro Model MTMECs cells Breast Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-106b~25 cluster controls transporter-independent MDR by apoptosis evasion via downregulation of EP300.
Key Molecule: hsa-mir-21 [81]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
SkBR3 cells Breast Homo sapiens (Human) CVCL_0033
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-21 inhibitors induced sensitivity of MCF-7/PR and SkBR-3/PR cells to paclitaxel. And miR-21 mimic can increase the expression of MDR1, Bcl-2/Bax and change cell morphology from parental cells to resistant cells.
Key Molecule: Mitosis associated long intergenic non-coding RNA 1 (MALINC1) [19]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
In Vitro Model H1299 cells Lung Homo sapiens (Human) CVCL_0060
U2OS cells Bone Homo sapiens (Human) CVCL_0042
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Silencing of MA-linc1 in unsynchronized cells results in fewer cells in G1 and a concomitant increase in the number of cells in all other stages of the cell cycle, particularly in G2/M. Moreover, its silencing in M phase-arrested cells inhibits mitosis exit. The effect of MA-linc1 on cell cycle progression is mediated, at least in part, by repression of its neighboring gene, Puralpha, a cell cycle regulator whose expression induces cell cycle arrest. Importantly, high levels of MA-linc1 are correlated with decreased survival of breast and lung cancer patients and its silencing sensitizes cancer cells to the apoptotic effect of the M phase specific chemotherapeutic drug, Paclitaxel. This enhancement of Paclitaxel-induced apoptosis is also Puralpha-related.
Key Molecule: hsa-miR-520h [82]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
T47D cells Breast Homo sapiens (Human) CVCL_0553
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
Hs-578T cells Breast Homo sapiens (Human) CVCL_0332
HBL-100 cells Breast Homo sapiens (Human) CVCL_4362
BT483 cells Breast Homo sapiens (Human) CVCL_2319
MDA-MB-361 cells Breast Homo sapiens (Human) CVCL_0620
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTS assay; Flow cytometry assay
Mechanism Description Through protecting cells from paclitaxel-induced apoptosis, expression of miR-520h promoted the drug resistance of human breast cancer cells. Bioinformatics prediction, compensatory mutation and functional validation further confirmed the essential role of miR-520h-suppressed Death-associated protein kinase 2 (DAPk2) expression, as restoring DAPk2 abolished miR-520h-promoted drug resistance, and knockdown of DAPk2 mitigated cell death caused by the depletion of miR-520h.
Key Molecule: hsa-mir-149 [83]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
HS signaling pathway Inhibition hsa00534
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR; RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-149 modulated chemoresistance through targeting the expression of GlcNAc N-deacetylase/N-sulfotransferase-1 (NDST1). With downregulated miR-149, NDST1 expression was increased in chemoresistant MCF-7/ADM cells versus control MCF-7 wild-type cells. The increased NDST1 then activated a heparan sulfate-related pathway involving activation of heparanase. Finally, expression of miR-149 and NDST1 was confirmed in clinical chemoresistant samples of breast cancers receiving anthracycline/taxane-based chemotherapies. The high expression of NDST1 was also an unfavorable predictor for distant relapse-free survival in Her2 and basal breast cancers.
Key Molecule: hsa-mir-137 [84]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Elevated miR-137 expression could sensitize breast cancer cells to chemotherapeutic agents (like Vincristine) through modulating the expression of P-gp by targeting YB-1.
Key Molecule: hsa-mir-135a [16]
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 A549 cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) is a protein that binds to HIF-1alpha and inhibits its transcriptional activity. HIF1AN is a potential miR-135a target listed in both the TargetScan and PicTar databases. miR-135a-mediated paclitaxel resistance is in part mediated by downregulation of APC.
Key Molecule: hsa-mir-125b [85]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
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
BT474 cells Breast Homo sapiens (Human) CVCL_0179
MDA-MB-436 cells Breast Homo sapiens (Human) CVCL_0623
MDA-MB-435 cells Breast Homo sapiens (Human) CVCL_0417
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Celltiter 96 aqueous one solution cell proliferation assay
Mechanism Description miR-125b was up-regulated in Taxol-resistant cells, causing a marked inhibition of Taxol-induced cytotoxicity and apoptosis and a subsequent increase in the resistance to Taxol in cancer cells. The pro-apoptotic Bcl-2 antagonist killer 1 (Bak1) is a direct target of miR-125b. Down-regulation of Bak1 suppressed Taxol-induced apoptosis and led to an increased resistance to Taxol.
Key Molecule: hsa-mir-155 [3]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
TGF-beta/Smad signaling pathway Regulation hsa04350
In Vitro Model Breast cancer cell lines Colon Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qRT-PCR; Northern blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Loss of FOXO3a is often linked to a decline in apoptotic activity and increased chemoresistance in cancer cells. miR-155 directly interacts with 3'-UTR of FOXO3a and blocks FOXO3a translation. knockdown of miR-155 renders cells to apoptosis and enhances chemosensitivity.
Key Molecule: hsa-let-7a [9]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast adenocarcinoma [ICD-11: 2C60.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Caspase-3 is the key executioner caspase in apoptosis. Ectopic expression of let-7adecreased the luciferase activity of a reporter constructcontaining the 30untranslated region of caspase-3. Enforced let-7aexpression increased the resistance in A431 cells andHepG2 cells to apoptosis induced by therapeutic drugs suchas interferon-gamma, doxorubicin and paclitaxel.
Key Molecule: Structural maintenance of chromosomes protein 4 (SMC4) [86]
Molecule Alteration Missense mutation
p.I1000S
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
Circulating-free DNA assay; Whole exome sequencing assay
Mechanism Description Quantification of allele fractions in plasma identified increased representation of mutant alleles in association with emergence of therapy resistance.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: ATP-binding cassette sub-family B5 (ABCB5) [81]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
SkBR3 cells Breast Homo sapiens (Human) CVCL_0033
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-21 inhibitors induced sensitivity of MCF-7/PR and SkBR-3/PR cells to paclitaxel. And miR-21 mimic can increase the expression of MDR1, Bcl-2/Bax and change cell morphology from parental cells to resistant cells.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: hsa-mir-155 [87]
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
PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Exosome-mediated breast cancer chemoresistance via miR155 transfeRNA Increased miR155 expression increases miR155 content of exosomes, leading to EMT-associated chemoresistance.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Tumor protein D52 (TPD52) [71]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Wnt/Beta-catenin signaling pathway Activation hsa04310
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-107 could enhance PTX sensitivity in breast cancer cells may be targeting TPD52 through Wnt/beta-catenin signaling pathway. And downregualtion of miR-107 ould enhance PTX resistance in BC cells.
Key Molecule: Cyclin-dependent kinase 6 (CDK6) [72]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
T47D cells Breast Homo sapiens (Human) CVCL_0553
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description CDk6 knockdown attenuated the effects of miR-29c inhibition on paclitaxel cytotoxicity in breast cancer cells.
Key Molecule: Histone-lysine N-methyltransferase EZH2 (EZH2) [74]
Molecule Alteration Expression
Down-regulation
Resistant Disease ER positive breast cancer [ICD-11: 2C60.6]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
MCF-7R cells Breast Homo sapiens (Human) CVCL_Y493
ZR-75-1R cells Breast Homo sapiens (Human) CVCL_0588
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Annexin V apoptosis assay; Fow cytometry analysis
Mechanism Description H19 was confirmed to suppress the promoter activity of BIk by recruiting EZH2 and by trimethylating the histone H3 at lysine 27. H19 bound to EZH2 in breast cancer cells, epigenetic silencing of BIk by H19 was dependent on EZH2.
Key Molecule: Nuclear receptor coactivator 3 (NCOA3) [75]
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
MCF-7/Tax1 cells Breast Homo sapiens (Human) CVCL_IJ26
MCF-7/Tax2 cells Breast Homo sapiens (Human) CVCL_IJ26
MDA-MB-231/Tax1 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-231/Tax2 cells Breast Homo sapiens (Human) CVCL_0062
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR; Western blot analysis; Dual-luciferase activity assay
Experiment for
Drug Resistance
MTT assay; Annexin-V-FITC (fluorescein isothiocyanate)/PI (propidium iodide) analysis
Mechanism Description Decreased expression of microRNA-17 and microRNA-20b promotes breast cancer resistance to taxol therapy by upregulation of NCOA3.
Key Molecule: Myeloid differentiation primary response protein MyD88 (MYD88) [76]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell viability Activation hsa05200
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
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
SRB assay
Mechanism Description miR-155-3p acts as a tumor suppressor and reverses paclitaxel resistance via negative regulation of MYD88 in human breast cancer.
Key Molecule: Leucine zipper putative tumor suppressor 1 (LZTS1) [77]
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
MDA-MB-436 cells Breast Homo sapiens (Human) CVCL_0623
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-1207-5p induces the resistence of triple-negative breast cancer cells to Taxol treatment via the suppression of LZTS1 expression.
Key Molecule: Transcription factor SOX-2 (SOX2) [78]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Down-regulation of miR 200c 3p contributes to the resistance of breast cancer cells to paclitaxel by targeting SOX2.
Key Molecule: Endoribonuclease Dicer (DICER1) [79]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell 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
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-18a is an important miRNA that suppresses Dicer expression and increases paclitaxel resistance in triple-negative breast cancer cells.
Key Molecule: Histone acetyltransferase p300 (EP300) [80]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
miR106b~25 cluster/EP300/E-cadherin signaling pathway Regulation hsa05206
In Vitro Model MTMECs cells Breast Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-106b~25 cluster controls transporter-independent MDR by apoptosis evasion via downregulation of EP300.
Key Molecule: Transcriptional activator protein Pur-alpha (PURA) [19]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
In Vitro Model H1299 cells Lung Homo sapiens (Human) CVCL_0060
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Silencing of MA-linc1 in unsynchronized cells results in fewer cells in G1 and a concomitant increase in the number of cells in all other stages of the cell cycle, particularly in G2/M. Moreover, its silencing in M phase-arrested cells inhibits mitosis exit. The effect of MA-linc1 on cell cycle progression is mediated, at least in part, by repression of its neighboring gene, Puralpha, a cell cycle regulator whose expression induces cell cycle arrest. Importantly, high levels of MA-linc1 are correlated with decreased survival of breast and lung cancer patients and its silencing sensitizes cancer cells to the apoptotic effect of the M phase specific chemotherapeutic drug, Paclitaxel. This enhancement of Paclitaxel-induced apoptosis is also Puralpha-related.
Key Molecule: Death-associated protein kinase 2 (DAPK2) [82]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
T47D cells Breast Homo sapiens (Human) CVCL_0553
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
Hs-578T cells Breast Homo sapiens (Human) CVCL_0332
HBL-100 cells Breast Homo sapiens (Human) CVCL_4362
BT483 cells Breast Homo sapiens (Human) CVCL_2319
MDA-MB-361 cells Breast Homo sapiens (Human) CVCL_0620
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay; Flow cytometry assay
Mechanism Description Through protecting cells from paclitaxel-induced apoptosis, expression of miR-520h promoted the drug resistance of human breast cancer cells. Bioinformatics prediction, compensatory mutation and functional validation further confirmed the essential role of miR-520h-suppressed Death-associated protein kinase 2 (DAPk2) expression, as restoring DAPk2 abolished miR-520h-promoted drug resistance, and knockdown of DAPk2 mitigated cell death caused by the depletion of miR-520h.
Key Molecule: Bifunctional heparan sulfate N-deacetylase/sulfotransferase 1 (NDST1) [83]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
HS signaling pathway Inhibition hsa00534
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-149 modulated chemoresistance through targeting the expression of GlcNAc N-deacetylase/N-sulfotransferase-1 (NDST1). With downregulated miR-149, NDST1 expression was increased in chemoresistant MCF-7/ADM cells versus control MCF-7 wild-type cells. The increased NDST1 then activated a heparan sulfate-related pathway involving activation of heparanase. Finally, expression of miR-149 and NDST1 was confirmed in clinical chemoresistant samples of breast cancers receiving anthracycline/taxane-based chemotherapies. The high expression of NDST1 was also an unfavorable predictor for distant relapse-free survival in Her2 and basal breast cancers.
Key Molecule: Y-box-binding protein 1 (YBX1) [84]
Molecule Alteration Expression
Up-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Elevated miR-137 expression could sensitize breast cancer cells to chemotherapeutic agents (like Vincristine) through modulating the expression of P-gp by targeting YB-1.
Key Molecule: Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) [16]
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 A549 cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) is a protein that binds to HIF-1alpha and inhibits its transcriptional activity. HIF1AN is a potential miR-135a target listed in both the TargetScan and PicTar databases. miR-135a-mediated paclitaxel resistance is in part mediated by downregulation of APC.
Key Molecule: Bcl-2 homologous antagonist/killer (BAK1) [85]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
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
BT474 cells Breast Homo sapiens (Human) CVCL_0179
MDA-MB-436 cells Breast Homo sapiens (Human) CVCL_0623
MDA-MB-435 cells Breast Homo sapiens (Human) CVCL_0417
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Celltiter 96 aqueous one solution cell proliferation assay
Mechanism Description miR-125b was up-regulated in Taxol-resistant cells, causing a marked inhibition of Taxol-induced cytotoxicity and apoptosis and a subsequent increase in the resistance to Taxol in cancer cells. The pro-apoptotic Bcl-2 antagonist killer 1 (Bak1) is a direct target of miR-125b. Down-regulation of Bak1 suppressed Taxol-induced apoptosis and led to an increased resistance to Taxol.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [85]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
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
BT474 cells Breast Homo sapiens (Human) CVCL_0179
MDA-MB-436 cells Breast Homo sapiens (Human) CVCL_0623
MDA-MB-435 cells Breast Homo sapiens (Human) CVCL_0417
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Celltiter 96 aqueous one solution cell proliferation assay
Mechanism Description miR-125b was up-regulated in Taxol-resistant cells, causing a marked inhibition of Taxol-induced cytotoxicity and apoptosis and a subsequent increase in the resistance to Taxol in cancer cells. The pro-apoptotic Bcl-2 antagonist killer 1 (Bak1) is a direct target of miR-125b. Down-regulation of Bak1 suppressed Taxol-induced apoptosis and led to an increased resistance to Taxol.
Key Molecule: Forkhead box protein O3 (FOXO3) [3]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
TGF-beta/Smad signaling pathway Regulation hsa04350
In Vitro Model Breast cancer cell lines Colon Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Loss of FOXO3a is often linked to a decline in apoptotic activity and increased chemoresistance in cancer cells. miR-155 directly interacts with 3'-UTR of FOXO3a and blocks FOXO3a translation. knockdown of miR-155 renders cells to apoptosis and enhances chemosensitivity.
Key Molecule: Caspase-3 (CASP3) [9]
Molecule Alteration Expression
Down-regulation
Resistant Disease Breast adenocarcinoma [ICD-11: 2C60.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Caspase-3 is the key executioner caspase in apoptosis. Ectopic expression of let-7adecreased the luciferase activity of a reporter constructcontaining the 30untranslated region of caspase-3. Enforced let-7aexpression increased the resistance in A431 cells andHepG2 cells to apoptosis induced by therapeutic drugs suchas interferon-gamma, doxorubicin and paclitaxel.
Key Molecule: Structural maintenance of chromosomes protein 4 (SMC4) [86]
Molecule Alteration Missense mutation
p.I1000S
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AXLK signaling pathway Activation hsa01521
In Vitro Model Plasma Blood Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Circulating-free DNA assay; Whole exome sequencing assay
Mechanism Description Quantification of allele fractions in plasma identified increased representation of mutant alleles in association with emergence of therapy resistance.
Key Molecule: PI3-kinase alpha (PIK3CA) [86]
Molecule Alteration Missense mutation
p.E545K
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
Circulating-free DNA assay; Whole exome sequencing assay
Mechanism Description Quantification of allele fractions in plasma identified increased representation of mutant alleles in association with emergence of therapy resistance.
Key Molecule: Polycomb complex protein BMI-1 (BMI1) [86]
Molecule Alteration Missense mutation
p.S324Y
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
Circulating-free DNA assay; Whole exome sequencing assay
Mechanism Description Quantification of allele fractions in plasma identified increased representation of mutant alleles in association with emergence of therapy 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-29c [72]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
T47D cells Breast Homo sapiens (Human) CVCL_0553
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description LINC00511 knockdown enhanced paclitaxel cytotoxicity in breast cancer cells by upregulating miR-29c.
Key Molecule: Long non-protein coding RNA 511 (LINC00511) [72]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
T47D cells Breast Homo sapiens (Human) CVCL_0553
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description LINC00511 knockdown enhanced paclitaxel cytotoxicity in breast cancer cells by upregulating miR-29c.
Key Molecule: Eosinophil granule ontogeny transcript (EGOT) [88]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell autophagy Activation hsa04140
In Vitro Model UACC-812 cells Breast Homo sapiens (Human) CVCL_1781
In Vivo Model Athymic BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description EGOT induces autophagy to enhance paclitaxel sensitivity through ITPR1.
Key Molecule: Long non-protein coding RNA 968 (LINC00968) [89]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Wnt2/Beta-catenin signaling pathway Inhibition hsa04310
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
KPL-4 cells Breast Homo sapiens (Human) CVCL_5310
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR,Northern blot
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay
Mechanism Description Long non-coding RNA LINC00968 attenuates drug resistance of breast cancer cells through inhibiting the Wnt2/beta-catenin signaling pathway by regulating WNT2.
Key Molecule: hsa-mir-324 [90]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
In Vivo Model Mouse xenograft models Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description PIWI-interacting RNA-36712 restrains breast cancer progression and chemoresistance by interaction with SEPW1 pseudogene SEPW1P RNA.
Key Molecule: hsa-mir-7 [90]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
In Vivo Model Mouse xenograft models Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description PIWI-interacting RNA-36712 restrains breast cancer progression and chemoresistance by interaction with SEPW1 pseudogene SEPW1P RNA.
Key Molecule: piR-hsa-36712 [90]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
In Vivo Model Mouse xenograft models Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description PIWI-interacting RNA-36712 restrains breast cancer progression and chemoresistance by interaction with SEPW1 pseudogene SEPW1P RNA.
Key Molecule: TTC36 and KMT2A antisense RNA 1 (TTC36-AS1) [91]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MCF-7/PR cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Sulforhodamine B assay
Mechanism Description Down-regulation of LncRNA RP11-770J1.3 and TMEM25 enhanced the sensitivity of MCF-7/PR cells to paclitaxel, and inhibited the expression of MRP, BCRP and MDR1/P-gp.
Key Molecule: Transmembrane protein 25 (TMEM25) [91]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MCF-7/PR cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Sulforhodamine B assay
Mechanism Description Down-regulation of LncRNA RP11-770J1.3 and TMEM25 enhanced the sensitivity of MCF-7/PR cells to paclitaxel, and inhibited the expression of MRP, BCRP and MDR1/P-gp.
Key Molecule: Insulin receptor substrate 1 (IRS1) [92]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT/ERK signaling pathway Inhibition hsa04010
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
MCF10A cells Breast Homo sapiens (Human) CVCL_0598
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Dual-luciferase reporter assay
Experiment for
Drug Resistance
CCK8 assay; Wound healing assay; Flow cytometry assay; Caspase-3 Activity Assay
Mechanism Description miR30e inhibits tumor growth and chemoresistance via targeting IRS1 in Breast Cancer, by targeting IRS1, miR30e is able to inhibit AkT and ERk1/2 pathways.
Key Molecule: hsa-mir-30e [92]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT/ERK signaling pathway Inhibition hsa04010
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
MCF10A cells Breast Homo sapiens (Human) CVCL_0598
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Wound healing assay; Flow cytometry assay; Caspase-3 Activity Assay
Mechanism Description miR30e inhibits tumor growth and chemoresistance via targeting IRS1 in Breast Cancer, by targeting IRS1, miR30e is able to inhibit AkT and ERk1/2 pathways.
Key Molecule: hsa-mir-451 [93]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Beta-catenin signaling pathway Inhibition hsa04520
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Chemosensitivity Activation hsa05207
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
SkBR3 cells Breast Homo sapiens (Human) CVCL_0033
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Muse Cell Cycle Assay
Mechanism Description miR451 suppresses cell migration, invasion and induces cell-cycle arrest and apoptosis in breast cancermiR451 decreases the mRNA and protein expression level of beta-catenin and relative genes of beta-catenin signaling pathway in vitro.
Key Molecule: hsa-mir-503 [94]
Molecule Alteration Expression
Up-regulation
Sensitive 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
MCF-7/ADR cells Breast Homo sapiens (Human) CVCL_1452
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description Down-regulation of eIF4G by microRNA-503 enhances drug sensitivity of MCF-7/ADR cells through suppressing the expression of ABC transport proteins.
Key Molecule: hsa-mir-143 [95]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation miR143/CIAPIN1 signaling pathway Regulation hsa05206
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
HEK293 cells Kidney Homo sapiens (Human) CVCL_0045
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Upregulation of microRNA-143 reverses drug resistance in human breast cancer cells via inhibition of cytokine-induced apoptosis inhibitor 1.
Key Molecule: hsa-miR-17-5p [96]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation STAT3/p53 pathway Inhibition hsa05212
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
TUNEL assaay; Sulphorhodamine B assay
Mechanism Description miR17-5p promoted apoptosis by increasing p53 expression, which was inhibited by STAT3, miR17-5p inhibits STAT3 and increases p53 expression to promote apoptosis in breast cancer cells. miR17-5p directly targets STAT3 and induces apoptosis in breast cancer cells by inhibiting the STAT3/p53 pathway.
Key Molecule: hsa-mir-24 [97]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast carcinoma [ICD-11: 2C60.2]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
SkBR3 cells Breast Homo sapiens (Human) CVCL_0033
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; TUNEL assay
Mechanism Description Overexpression of microRNA-24 increases the sensitivity to paclitaxel in drug-resistant breast carcinoma cell lines via targeting ABCB9.
Key Molecule: hsa-miR-129-5p [98]
Molecule Alteration Expression
Up-regulation
Sensitive 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
MCF-7/ADM cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description There is a reciprocal regulation between miR129-5p and SOX4 via the SOX4/EZH2 complex mediated H3k27me3 modification in breast cancer cells. miR129-5p is an important miRNA modulating EMT and MDR in breast cancer cells.
Key Molecule: hsa-mir-22 [99]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDM231 cells Breast Homo sapiens (Human) CVCL_5T76
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description microRNA-22 sensitized breast cancer cells to paclitaxel by downregulation of NRAS.
Key Molecule: H19, imprinted maternally expressed transcript (H19) [100]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Inhibition hsa04151
Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
MDA-MB-157 cells Breast Homo sapiens (Human) CVCL_0618
In Vivo Model BALB/c nude mouse xenograft mode Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Knockdown of LncRNA H19 restores chemo-sensitivity in paclitaxel-resistant triple-negative breast cancer through triggering apoptosis and regulating Akt signaling pathway.
Key Molecule: hsa-mir-199a [101]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
miR199a/MRP1 signaling pathway Regulation hsa05206
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MCF-7/ADR cells Breast Homo sapiens (Human) CVCL_1452
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
XTT assay; Flow cytometry assay; Caspase 9 activity assay
Mechanism Description Linc00518 downregulation reduced MDR by upregulating miR-199a which downregulates MRP1 in breast cancer.
Key Molecule: Long non-protein coding RNA 518 (LINC00518) [101]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
miR199a/MRP1 signaling pathway Regulation hsa05206
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MCF-7/ADR cells Breast Homo sapiens (Human) CVCL_1452
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
XTT assay; Flow cytometry assay; Caspase 9 activity assay
Mechanism Description Linc00518 downregulation reduced MDR by upregulating miR-199a which downregulates MRP1 in breast cancer.
Key Molecule: hsa-miR-485-5p [102]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Transwell assay
Mechanism Description miR-485-5p suppresses breast cancer progression and enhances chemosensitivity through down-regulation of survivin expression.
Key Molecule: hsa-miR-27b-3p [103]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
MAPK/ERK signaling pathway Inhibition hsa04010
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model BCap37 cells Breast Homo sapiens (Human) CVCL_0164
Bads-200 cells Breast Homo sapiens (Human) N.A.
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-27b inhibits proliferation and resistance to PTX of breast cancer cell by repressing CBLB and GRB2 and suppresses activities of PI3k/AkT and MAPk/ERk signaling pathways through downregulation of CBLB and GRB2.
Key Molecule: hsa-mir-375 [104]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7/PTX cells Breast Homo sapiens (Human) CVCL_4V97
Experiment for
Molecule Alteration
qRT-PCR; MSP assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-375 is downregulated in MCF-7/ADM and MCF-7/PTX cells, and its downregulation is a result of promoter methylation. miR-375 can directly target 3'UTR of YBX1 and, thereby, decrease its expression, which might be an important mechanism of MDR in breast cancer cells. miR-375 is downregulated in MCF-7/ADM and MCF-7/PTX cells, and its downregulation is a result of promoter methylation. miR-375 can directly target 3'UTR of YBX1 and thereby decrease its expression, which might be an important mechanism of MDR in breast cancer cells.
Key Molecule: hsa-mir-16 [105]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
NF-kappaB signaling pathway Regulation hsa04064
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Ectopic expression of miR-16 promoted Taxol-induced cytotoxicity and apoptosis in breast cancer cells. Furthermore, IkBkB was identified to be a direct target of miR-16, restoring the expression of IkBkB counteracted miR-16-mediated Taxol sensitivity. Moreover, miR-16 was highly expressed in Taxol-sensitive breast cancer patients and negatively associated with T stages, whereas IkBkB was lowly expressed in Taxol-sensitive breast cancer and positively correlated with T, N and clinical stages.
Key Molecule: hsa-mir-100 [106]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
Homologous recombination-mediated repair pathway Inhibition hsa03440
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
T47D cells Breast Homo sapiens (Human) CVCL_0553
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
BT549 cells Breast Homo sapiens (Human) CVCL_1092
Hs-578T cells Breast Homo sapiens (Human) CVCL_0332
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-100 expression was significantly downregulated in breast cancer, and the downregulation was more extensive in luminal A breast cancers and was associated with worse patient survival. Ectopic expression of miR-100 sensitized, while inhibition of miR-100 expression desensitized, breast cancer cells to the effect of paclitaxel on cell cycle arrest, multinucleation, apoptosis and tumorigenesis. Expression of genes that are part of a known signature of paclitaxel sensitivity in breast cancer significantly correlated with miR-100 expression. Mechanistically, targeting mTOR appeared to mediate miR-100's function in sensitizing breast cancer cells to paclitaxel, but other mechanisms also seem to be involved, including targeting other molecules such as PLk1.
Key Molecule: hsa-mir-21 [81]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
SkBR3 cells Breast Homo sapiens (Human) CVCL_0033
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-21 inhibitors induced sensitivity of MCF-7/PR and SkBR-3/PR cells to paclitaxel. And miR-21 mimic can increase the expression of MDR1, Bcl-2/Bax and change cell morphology from parental cells to resistant cells.
Key Molecule: hsa-let-7b [44]
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.
Key Molecule: hsa-mir-34 [107]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
Notch1 signaling pathway Inhibition hsa04330
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR-34a negatively regulated cell proliferation, migration, and invasion and breast cancer stem cell propagation by downregulating Notch1. The expression of miR-34a was negatively correlated with tumor stages, metastasis, and Notch1 expression in breast cancer tissues. Furthermore, overexpression of miR-34a increased chemosensitivity of breast cancer cells to paclitaxel (PTX) by downregulating the Notch1 pathway. Mammosphere formation and expression of the stemness factor ALDH1 were also reduced in the cells treated with miR-34a and PTX compared to those treated with PTX alone. miR-34a inhibited breast cancer stemness and increased the chemosensitivity to PTX partially by downregulating the Notch1 pathway, suggesting that miR-34a/Notch1 play an important role in regulating breast cancer stem cells.
Key Molecule: hsa-mir-320 [108]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
TRPC5 signaling pathway Regulation hsa05206
In Vitro Model MCF-7/ADM cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The overexpression of miR-320a, which downregulated TRPC5 and NFATC3, colud inreduce chemoresistance.
Key Molecule: hsa-miR-342-3p [109]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Triple-negative breast cancer with high expression of miR-342-3p is more sensitive to chemotherapy drugs, and miR-342-3p can regulate the chemotherapy sensitivity of breast cancer cell line MDA-MB-231 to paclitaxel and cisplatin.
Key Molecule: hsa-mir-223 [110]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell migration Inhibition hsa04670
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
Cell Death assay
Mechanism Description miR-223 Impairs Tumor Cell Migration and Invasion, miR-223 Expression Enhances Cell Death in Anoikis Conditions or in Presence of Chemotherapeutic Drugs (Doxorubicin and Paclitaxel), miR-223 Affects Signal Transduction Pathways Involved in Cell Death and Directly Targets STAT5A, Down-modulation of STAT5A Accounts for miR-223 Biological Effects.
Key Molecule: hsa-mir-26a [111]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
Cell viability Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
MCF10A cells Breast Homo sapiens (Human) CVCL_0598
MDA-MB-435 cells Breast Homo sapiens (Human) CVCL_0417
184A1 cells Breast Homo sapiens (Human) CVCL_3040
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description MCL-1(myeloid cell leukemia 1), a pro-survival member of the Bcl-2(B-cell CLL/lymphoma 2) family, several miRNAs induces apoptosis by targeting MCL-1, miR-26a Inhibits MCL-1 expression, increased sensitivity of breast cancer cells to paclitaxel.
Key Molecule: hsa-mir-21 [112]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast carcinoma [ICD-11: 2C60.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Inhibition hsa04151
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Treatment of the miR-21 inhibitor-transfected cells with the anti-cancer drugs taxol resulted in signifi-cantly reduced cell viability and invasiveness compared with control cells.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: ATP-binding cassette sub-family G2 (ABCG2) [91]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MCF-7/PR cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Sulforhodamine B assay
Mechanism Description Down-regulation of LncRNA RP11-770J1.3 and TMEM25 enhanced the sensitivity of MCF-7/PR cells to paclitaxel, and inhibited the expression of MRP, BCRP and MDR1/P-gp.
Key Molecule: Multidrug resistance protein 1 (ABCB1) [91]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MCF-7/PR cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Sulforhodamine B assay
Mechanism Description Down-regulation of LncRNA RP11-770J1.3 and TMEM25 enhanced the sensitivity of MCF-7/PR cells to paclitaxel, and inhibited the expression of MRP, BCRP and MDR1/P-gp.
Key Molecule: Multidrug resistance-associated protein 1 (MRP1) [91]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MCF-7/PR cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Sulforhodamine B assay
Mechanism Description Down-regulation of LncRNA RP11-770J1.3 and TMEM25 enhanced the sensitivity of MCF-7/PR cells to paclitaxel, and inhibited the expression of MRP, BCRP and MDR1/P-gp.
Key Molecule: ATP-binding cassette sub-family B5 (ABCB5) [91]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MCF-7/PR cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Sulforhodamine B assay
Mechanism Description Down-regulation of LncRNA RP11-770J1.3 and TMEM25 enhanced the sensitivity of MCF-7/PR cells to paclitaxel, and inhibited the expression of MRP, BCRP and MDR1/P-gp.
Key Molecule: ABC-type oligopeptide transporter ABCB9 (ABCB9) [97]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast carcinoma [ICD-11: 2C60.2]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
SkBR3 cells Breast Homo sapiens (Human) CVCL_0033
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Luciferase assay; Western blot analysis
Experiment for
Drug Resistance
MTT assay; TUNEL assay
Mechanism Description Overexpression of microRNA-24 increases the sensitivity to paclitaxel in drug-resistant breast carcinoma cell lines via targeting ABCB9.
Key Molecule: Multidrug resistance-associated protein 1 (MRP1) [101]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
miR199a/MRP1 signaling pathway Regulation hsa05206
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MCF-7/ADR cells Breast Homo sapiens (Human) CVCL_1452
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
XTT assay; Flow cytometry assay; Caspase 9 activity assay
Mechanism Description Linc00518 downregulation reduced MDR by upregulating miR-199a which downregulates MRP1 in breast cancer.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: Interleukin-11 (IL11) [113]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
G418 cells Breast Homo sapiens (Human) N.A.
In Vivo Model NOD/SCID nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-30c plays a pivotal role in Paclitaxel and Doxorubicin chemo-resistance by a direct targeting of TWF1, which encodes an actin-binding protein and promotes EMT.
Key Molecule: hsa-mir-30c [113]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
G418 cells Breast Homo sapiens (Human) N.A.
In Vivo Model NOD/SCID nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-30c plays a pivotal role in Paclitaxel and Doxorubicin chemo-resistance by a direct targeting of TWF1, which encodes an actin-binding protein and promotes EMT.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Cyclin-dependent kinase 6 (CDK6) [72]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
T47D cells Breast Homo sapiens (Human) CVCL_0553
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description LINC00511 positively regulated CDk6 expression in breast cancer cells. And LINC00511 knockdown enhanced paclitaxel cytotoxicity in breast cancer cells by upregulating miR-29c.
Key Molecule: Inositol 1,4,5-trisphosphate receptor type 1 (ITPR1) [88]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell autophagy Activation hsa04140
In Vitro Model UACC-812 cells Breast Homo sapiens (Human) CVCL_1781
In Vivo Model Athymic BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description EGOT induces autophagy to enhance paclitaxel sensitivity through ITPR1.
Key Molecule: Hairy/enhancer-of-split related with YRPW motif protein 1 (HEY1) [89]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Wnt2/Beta-catenin signaling pathway Inhibition hsa04310
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
KPL-4 cells Breast Homo sapiens (Human) CVCL_5310
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RIP assay; ChIP assay; Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay
Mechanism Description Long non-coding RNA LINC00968 attenuates drug resistance of breast cancer cells through inhibiting the Wnt2/beta-catenin signaling pathway by regulating WNT2.
Key Molecule: Selenoprotein W (SEPW1) [90]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
In Vivo Model Mouse xenograft models Mus musculus
Experiment for
Molecule Alteration
Luciferase reporter assay; Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description PIWI-interacting RNA-36712 restrains breast cancer progression and chemoresistance by interaction with SEPW1 pseudogene SEPW1P RNA.
Key Molecule: Protein zeta/delta 14-3-3 (YWHAZ) [93]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Beta-catenin signaling pathway Inhibition hsa04520
Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
SkBR3 cells Breast Homo sapiens (Human) CVCL_0033
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Luciferase reporter assay; RT-PCR; Western blot analysis
Experiment for
Drug Resistance
Muse Cell Cycle Assay
Mechanism Description miR451 suppresses cell migration, invasion and induces cell-cycle arrest and apoptosis in breast cancermiR451 decreases the mRNA and protein expression level of beta-catenin and relative genes of beta-catenin signaling pathway in vitro.
Key Molecule: Eukaryotic translation initiation factor 4 gamma 1 (EIF4G1) [94]
Molecule Alteration Expression
Down-regulation
Sensitive 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
MCF-7/ADR cells Breast Homo sapiens (Human) CVCL_1452
Experiment for
Molecule Alteration
Western blot analysis; Luciferase reporter assay
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description Down-regulation of eIF4G by microRNA-503 enhances drug sensitivity of MCF-7/ADR cells through suppressing the expression of ABC transport proteins.
Key Molecule: Anamorsin (CIAPIN1) [95]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation miR143/CIAPIN1 signaling pathway Regulation hsa05206
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
HEK293 cells Kidney Homo sapiens (Human) CVCL_0045
Experiment for
Molecule Alteration
Western blot analysis; Luciferase reporter assay
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Upregulation of microRNA-143 reverses drug resistance in human breast cancer cells via inhibition of cytokine-induced apoptosis inhibitor 1.
Key Molecule: Signal transducer activator transcription 3 (STAT3) [96]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation STAT3/p53 pathway Inhibition hsa05212
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
TUNEL assaay; Sulphorhodamine B assay
Mechanism Description miR17-5p promoted apoptosis by increasing p53 expression, which was inhibited by STAT3, miR17-5p inhibits STAT3 and increases p53 expression to promote apoptosis in breast cancer cells. miR17-5p directly targets STAT3 and induces apoptosis in breast cancer cells by inhibiting the STAT3/p53 pathway.
Key Molecule: Transcription factor SOX-4 (SOX4) [98]
Molecule Alteration Expression
Down-regulation
Sensitive 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
MCF-7/ADM cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
IP assay; ChIP assay; Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description There is a reciprocal regulation between miR129-5p and SOX4 via the SOX4/EZH2 complex mediated H3k27me3 modification in breast cancer cells. miR129-5p is an important miRNA modulating EMT and MDR in breast cancer cells.
Key Molecule: GTPase Nras (NRAS) [99]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDM231 cells Breast Homo sapiens (Human) CVCL_5T76
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description microRNA-22 sensitized breast cancer cells to paclitaxel by downregulation of NRAS.
Key Molecule: RAC serine/threonine-protein kinase (AKT) [100]
Molecule Alteration Phosphorylation
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Inhibition hsa04151
Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
MDA-MB-157 cells Breast Homo sapiens (Human) CVCL_0618
In Vivo Model BALB/c nude mouse xenograft mode Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Knockdown of LncRNA H19 restores chemo-sensitivity in paclitaxel-resistant triple-negative breast cancer through triggering apoptosis and regulating Akt signaling pathway.
Key Molecule: Baculoviral IAP repeat-containing protein 5 (BIRC5) [102]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Transwell assay
Mechanism Description miR-485-5p suppresses breast cancer progression and enhances chemosensitivity through down-regulation of survivin expression.
Key Molecule: E3 ubiquitin-protein ligase CBL-B (CBLB) [103]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
MAPK/ERK signaling pathway Inhibition hsa04010
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model BCap37 cells Breast Homo sapiens (Human) CVCL_0164
Bads-200 cells Breast Homo sapiens (Human) N.A.
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-27b inhibits proliferation and resistance to PTX of breast cancer cell by repressing CBLB and GRB2 and suppresses activities of PI3k/AkT and MAPk/ERk signaling pathways through downregulation of CBLB and GRB2.
Key Molecule: Growth factor receptor-bound protein 2 (GRB2) [103]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
MAPK/ERK signaling pathway Inhibition hsa04010
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model BCap37 cells Breast Homo sapiens (Human) CVCL_0164
Bads-200 cells Breast Homo sapiens (Human) N.A.
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-27b inhibits proliferation and resistance to PTX of breast cancer cell by repressing CBLB and GRB2 and suppresses activities of PI3k/AkT and MAPk/ERk signaling pathways through downregulation of CBLB and GRB2.
Key Molecule: Y-box-binding protein 1 (YBX1) [104]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7/PTX cells Breast Homo sapiens (Human) CVCL_4V97
Experiment for
Molecule Alteration
Dual luciferase assay; Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-375 is downregulated in MCF-7/ADM and MCF-7/PTX cells, and its downregulation is a result of promoter methylation. miR-375 can directly target 3'UTR of YBX1 and, thereby, decrease its expression, which might be an important mechanism of MDR in breast cancer cells. miR-375 is downregulated in MCF-7/ADM and MCF-7/PTX cells, and its downregulation is a result of promoter methylation. miR-375 can directly target 3'UTR of YBX1 and thereby decrease its expression, which might be an important mechanism of MDR in breast cancer cells.
Key Molecule: I-kappa-B-kinase beta (IKKB) [105]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
NF-kappaB signaling pathway Regulation hsa04064
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
MTT assay
Mechanism Description Ectopic expression of miR-16 promoted Taxol-induced cytotoxicity and apoptosis in breast cancer cells. Furthermore, IkBkB was identified to be a direct target of miR-16, restoring the expression of IkBkB counteracted miR-16-mediated Taxol sensitivity. Moreover, miR-16 was highly expressed in Taxol-sensitive breast cancer patients and negatively associated with T stages, whereas IkBkB was lowly expressed in Taxol-sensitive breast cancer and positively correlated with T, N and clinical stages.
Key Molecule: Serine/threonine-protein kinase mTOR (mTOR) [106]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
T47D cells Breast Homo sapiens (Human) CVCL_0553
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
BT549 cells Breast Homo sapiens (Human) CVCL_1092
Hs-578T cells Breast Homo sapiens (Human) CVCL_0332
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-100 expression was significantly downregulated in breast cancer, and the downregulation was more extensive in luminal A breast cancers and was associated with worse patient survival. Ectopic expression of miR-100 sensitized, while inhibition of miR-100 expression desensitized, breast cancer cells to the effect of paclitaxel on cell cycle arrest, multinucleation, apoptosis and tumorigenesis. Expression of genes that are part of a known signature of paclitaxel sensitivity in breast cancer significantly correlated with miR-100 expression. Mechanistically, targeting mTOR appeared to mediate miR-100's function in sensitizing breast cancer cells to paclitaxel, but other mechanisms also seem to be involved, including targeting other molecules such as PLk1.
Key Molecule: Apoptosis regulator BAX (BAX) [81]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
SkBR3 cells Breast Homo sapiens (Human) CVCL_0033
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-21 inhibitors induced sensitivity of MCF-7/PR and SkBR-3/PR cells to paclitaxel. And miR-21 mimic can increase the expression of MDR1, Bcl-2/Bax and change cell morphology from parental cells to resistant cells.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [81]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
SkBR3 cells Breast Homo sapiens (Human) CVCL_0033
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-21 inhibitors induced sensitivity of MCF-7/PR and SkBR-3/PR cells to paclitaxel. And miR-21 mimic can increase the expression of MDR1, Bcl-2/Bax and change cell morphology from parental cells to resistant cells.
Key Molecule: Ribonucleoside-diphosphate reductase subunit M2 (RRM2) [44]
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) [44]
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.
Key Molecule: Neurogenic locus notch homolog protein 1 (NOTCH1) [107]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
Notch1 signaling pathway Inhibition hsa04330
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR-34a negatively regulated cell proliferation, migration, and invasion and breast cancer stem cell propagation by downregulating Notch1. The expression of miR-34a was negatively correlated with tumor stages, metastasis, and Notch1 expression in breast cancer tissues. Furthermore, overexpression of miR-34a increased chemosensitivity of breast cancer cells to paclitaxel (PTX) by downregulating the Notch1 pathway. Mammosphere formation and expression of the stemness factor ALDH1 were also reduced in the cells treated with miR-34a and PTX compared to those treated with PTX alone. miR-34a inhibited breast cancer stemness and increased the chemosensitivity to PTX partially by downregulating the Notch1 pathway, suggesting that miR-34a/Notch1 play an important role in regulating breast cancer stem cells.
Key Molecule: Short transient receptor potential channel 5 (TRPC5) [108]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
TRPC5 signaling pathway Regulation hsa05206
In Vitro Model MCF-7/ADM cells Breast Homo sapiens (Human) CVCL_0031
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The overexpression of miR-320a, which downregulated TRPC5 and NFATC3, colud inreduce chemoresistance.
Key Molecule: Signal transducer activator transcription 5A (STAT5A) [110]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell migration Inhibition hsa04670
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
Luciferase assay
Experiment for
Drug Resistance
Cell Death assay
Mechanism Description miR-223 Impairs Tumor Cell Migration and Invasion, miR-223 Expression Enhances Cell Death in Anoikis Conditions or in Presence of Chemotherapeutic Drugs (Doxorubicin and Paclitaxel), miR-223 Affects Signal Transduction Pathways Involved in Cell Death and Directly Targets STAT5A, Down-modulation of STAT5A Accounts for miR-223 Biological Effects.
Key Molecule: Induced myeloid leukemia cell differentiation protein Mcl-1 (MCL1) [111]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
Tumorigenesis Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
MCF10A cells Breast Homo sapiens (Human) CVCL_0598
MDA-MB-435 cells Breast Homo sapiens (Human) CVCL_0417
184A1 cells Breast Homo sapiens (Human) CVCL_3040
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description MCL-1(myeloid cell leukemia 1), a pro-survival member of the Bcl-2(B-cell CLL/lymphoma 2) family, several miRNAs induces apoptosis by targeting MCL-1, miR-26a Inhibits MCL-1 expression, increased sensitivity of breast cancer cells to paclitaxel.
Key Molecule: Twinfilin-1 (TWF1) [113]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
G418 cells Breast Homo sapiens (Human) N.A.
In Vivo Model NOD/SCID nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-30c plays a pivotal role in Paclitaxel and Doxorubicin chemo-resistance by a direct targeting of TWF1, which encodes an actin-binding protein and promotes EMT.
Uterine sarcoma [ICD-11: 2C72]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-135a [16]
Molecule Alteration Expression
Up-regulation
Resistant Disease Uterine sarcoma [ICD-11: 2C72.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) is a protein that binds to HIF-1alpha and inhibits its transcriptional activity. HIF1AN is a potential miR-135a target listed in both the TargetScan and PicTar databases. miR-135a-mediated paclitaxel resistance is in part mediated by downregulation of APC.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) [16]
Molecule Alteration Expression
Down-regulation
Resistant Disease Uterine sarcoma [ICD-11: 2C72.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) is a protein that binds to HIF-1alpha and inhibits its transcriptional activity. HIF1AN is a potential miR-135a target listed in both the TargetScan and PicTar databases. miR-135a-mediated paclitaxel resistance is in part mediated by downregulation of APC.
Ovarian cancer [ICD-11: 2C73]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Drug Inactivation by Structure Modification (DISM) Click to Show/Hide
Key Molecule: Glutathione S-transferase P (GSTP1) [114]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model A2780 cells Ovary Homo sapiens (Human) CVCL_0134
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description The upregulation of GST-Pi cause excessive intensity of detoxification of cytostatics, affect drug metabolism and influence the effects of chemotherapy, which results in resistance for paclitaxel in the ovarian cancer cells.
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-654-5p [1]
Molecule Alteration Expression
Down-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
MYC/WNT/AKT signaling pathway Regulation hsa04217
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
In Vivo Model NMRI-nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Time course proliferation assay; Flow cytometry assay
Mechanism Description CDCP1 and PLAGL2 oncogenes were found to be the most relevant direct miR-654-5p targets and both genes convey in a molecular signature associated with key cancer pathways relevant to ovarian tumorigenesis, such as MYC, WNT and AkT pathways.
Key Molecule: hsa-mir-194 [115]
Molecule Alteration Expression
Down-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
Hey A8 cells Ovary Homo sapiens (Human) CVCL_8878
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description LncRNA NEAT1 contributes to paclitaxel resistance of ovarian cancer cells by regulating ZEB1 expression via miR194. NEAT1 contributed to PTX resistance of ovarian cancer cells at least partly through upregulating ZEB1 expression by sponging miR194.
Key Molecule: Nuclear paraspeckle assembly transcript 1 (NEAT1) [115]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
Hey A8 cells Ovary Homo sapiens (Human) CVCL_8878
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description LncRNA NEAT1 contributes to paclitaxel resistance of ovarian cancer cells by regulating ZEB1 expression via miR194. NEAT1 contributed to PTX resistance of ovarian cancer cells at least partly through upregulating ZEB1 expression by sponging miR194.
Key Molecule: hsa-mir-1307 [116]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
A2780/Taxol cells Ovary Homo sapiens (Human) CVCL_IJ13
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Colony formation assay; Apoptosis analysis by FITC immunofluorescence
Mechanism Description miR1307 promotes ovarian cancer cell chemoresistance by targeting the ING5 expression.
Key Molecule: hsa-miR-194-5p [117]
Molecule Alteration Expression
Down-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Activation hsa05200
In Vitro Model Hey A8 cells Ovary Homo sapiens (Human) CVCL_8878
SkVO3ip1 cells Ovary Homo sapiens (Human) CVCL_0C84
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description Down-regulation of miR-194-5p induces paclitaxel resistance in ovarian cancer cells by altering MDM2 expression.
Key Molecule: Urothelial cancer associated 1 (UCA1) [118]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
UCA1/miR129/ABCB1 signaling pathway Regulation hsa05206
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
Hey A8 cells Ovary Homo sapiens (Human) CVCL_8878
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description ABCB1 up-regulated by UCA1/miR-129 axis contributed to PTX resistance in PTX-resistant OC cells.
Key Molecule: hsa-mir-129 [118]
Molecule Alteration Expression
Down-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
UCA1/miR129/ABCB1 signaling pathway Regulation hsa05206
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
Hey A8 cells Ovary Homo sapiens (Human) CVCL_8878
Experiment for
Molecule Alteration
RT-qPCR; RIP assay; Luciferase reporter assay
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description ABCB1 up-regulated by UCA1/miR-129 axis contributed to PTX resistance in PTX-resistant OC cells.
Key Molecule: hsa-mir-21 [119]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
TGF signaling pathway Regulation hsa04350
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
Hey A8 cells Ovary Homo sapiens (Human) CVCL_8878
OVCA433 cells Ovary Homo sapiens (Human) CVCL_0475
ALST cells Ovary Homo sapiens (Human) CVCL_W778
HeyA8-MDR cells Ovary Homo sapiens (Human) CVCL_8879
OVCA432 cells Ovary Homo sapiens (Human) CVCL_3769
OVCAR5 cells Ovary Homo sapiens (Human) CVCL_1628
SkOV3-TR cells Ovary Homo sapiens (Human) CVCL_HF69
SkOV3ip cells Ovary Homo sapiens (Human) CVCL_0C84
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The identification of APAF1 as a direct target of miR21 and APAF1 as a mediator of miR21 for conferring chemoresistance in ovarian cancer suggests that strategies based on the upregulation of APAF1 in ovarian cancer cells can be used to sensitize ovarian cancer cells to paclitaxel treatment.ovarian cancer suggests that strategies based on the upregulation of APAF1 in ovarian cancer cells can be used to sensitize ovarian cancer cells to paclitaxel treatment.
Key Molecule: hsa-miR-17-5p [120]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
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 proliferation Activation hsa05200
PTEN/AKT signaling pathway Regulation hsa05235
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
OVCAR3 cells Ovary Homo sapiens (Human) CVCL_0465
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description microRNA-17-5p induces drug resistance and invasion of ovarian carcinoma cells by targeting PTEN signaling.
Key Molecule: hsa-mir-149 [121]
Molecule Alteration Expression
Down-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
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 proliferation Activation hsa05200
TLR/MyD88 signaling pathway Regulation hsa04620
In Vitro Model A2780 cells Ovary Homo sapiens (Human) CVCL_0134
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 In the present study, flow cytometric assays were used to detect the apoptosis of A2780 cells after down-regulation of miRNA-149. We found that down-regulation of miRNA-149 decreased the apoptosis induced by paclitaxel when compared to the control group. Furthermore, we showed that down-regulation of miRNA-149 in A2780 cells (+) the expression of the anti-apoptotic protein Bcl-2 and inhibited the expression of the pro-apoptotic protein bax, which may have led to paclitaxel resistance.
Key Molecule: hsa-miR-490-3p [114]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model A2780 cells Ovary Homo sapiens (Human) CVCL_0134
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description microRNA 490-3P was involved in the development of drug resistance through regulating MDR1/P-gp and GST-Pi expression in ovarian cancer cells.
Key Molecule: hsa-mir-106a [122]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
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 SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
TUNEL assay
Mechanism Description miR-106a and miR-591 have important roles in conferring PTX resistance to ovarian cancer cells. Modulation of these microRNAs resensitizes PTX-resistant cancer cells by targeting BCL10, caspase-7, and ZEB1.
Key Molecule: hsa-miR-591 [122]
Molecule Alteration Expression
Down-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
TUNEL assay
Mechanism Description miR-106a and miR-591 have important roles in conferring PTX resistance to ovarian cancer cells. Modulation of these microRNAs resensitizes PTX-resistant cancer cells by targeting BCL10, caspase-7, and ZEB1.
Key Molecule: hsa-let-7g [6]
Molecule Alteration Expression
Down-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model T47D cells Breast Homo sapiens (Human) CVCL_0553
IGROV1 cells Ovary Homo sapiens (Human) CVCL_1304
OVCAR8 cells Ovary Homo sapiens (Human) CVCL_1629
LOX-IMVI cells Ovary Homo sapiens (Human) CVCL_1381
NCI/ADR-RES cells Ovary Homo sapiens (Human) CVCL_1452
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
SRB cytotoxicity assay
Mechanism Description IMP-1 is an RNA binding protein that acts by stabilizing the mRNA of a number of target genes. In addition, IMP-1 was shown to protect the mRNA of MDR1 from endonucleolytic attack in an in vitro RNA stability assay. Introducing let-7g into ADR-RES cells expressing both IMP-1 and MDR1 reduced expression of both proteins rendering the cells more sensitive to treatment with either Taxol or vinblastine without affecting the sensitivity of the cells to carboplatin.
Key Molecule: hsa-mir-135a [16]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) is a protein that binds to HIF-1alpha and inhibits its transcriptional activity. HIF1AN is a potential miR-135a target listed in both the TargetScan and PicTar databases. miR-135a-mediated paclitaxel resistance is in part mediated by downregulation of APC.
Key Molecule: hsa-mir-27a [123]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
miRNAs/HIPk2/MDR1/P-gp signaling pathway Regulation hsa05206
In Vitro Model A2780 cells Ovary Homo sapiens (Human) CVCL_0134
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Transfection of A2780/Taxol cells with the inhibitors of miR-27a decreased the expression of MDR1 mRNA and P-gp protein, increased HIPk2 protein expression, enhanced the sensitivity of A2780/taxol cells to paclitaxel, increased paclitaxel-induced apoptosis and the fluorescence intensity of intracellular Rh-123. The deregulation of miR-27a may be involved in the development of drug resistance, regulating the expression of MDR1/P-gp, at least in part, by targeting HIPk2 in ovarian cancer cells.
Key Molecule: hsa-mir-130a [20]
Molecule Alteration Expression
Down-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A2780 cells Ovary Homo sapiens (Human) CVCL_0134
A2780CIS cells Ovary Homo sapiens (Human) CVCL_1942
Experiment for
Molecule Alteration
qRT-PCR; Northern blotting analysis
Experiment for
Drug Resistance
Clonogenic assay
Mechanism Description Finally downstreamtarget validation was proven for the miR-130a, whose downregulation was linked to the translational activation of the M-CSF gene, a knownresistance factor for ovarian cancer.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [118]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
UCA1/miR129/ABCB1 signaling pathway Regulation hsa05206
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
Hey A8 cells Ovary Homo sapiens (Human) CVCL_8878
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description ABCB1 up-regulated by UCA1/miR-129 axis contributed to PTX resistance in PTX-resistant OC cells.
Key Molecule: ATP-binding cassette sub-family B5 (ABCB5) [124]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-181a level in chemoresistant (CR) cancer tissues were significantly higher than in chemosensitive (CS) cancer tissues and in normal tissue. SkOV3/PTX cells had significantly higher expression of miR-181a and N-cadherin than SkOV3 cells. SkOV3 cells had decreased E-cadherin expression and increased N-cadherin expression after enforced miR-181a expression, while SkOV3/PTX cells had increased E-cadherin expression and decreased N-cadherin expression after miR-181a knockdown. SkOV3 cells had increased P-gp expression after enforced miR-181a expression. Following MTT assay and flow cytometry analysis both confirmed that miR-181a overexpression decreased the PTX sensitivity of SkOV3 cells and while miR-181a inhibition increased the sensitivity of SkOV3/PTX cells. miR-181a is an important oncomiR significantly increased in chemoresistant ovarian cancer. Its upregulation is associated with increased level of EMT and decreased cell apoptosis induced by PTX treatment.
Key Molecule: Multidrug resistance protein 1 (ABCB1) [114]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model A2780 cells Ovary Homo sapiens (Human) CVCL_0134
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description The upregulation of P-gp cause ovarian cancer cells pumping drug substance outside to reduce cytotoxicity presented and enhances the resistance of paclitaxe.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: hsa-mir-181a [124]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
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-181a level in chemoresistant (CR) cancer tissues were significantly higher than in chemosensitive (CS) cancer tissues and in normal tissue. SkOV3/PTX cells had significantly higher expression of miR-181a and N-cadherin than SkOV3 cells. SkOV3 cells had decreased E-cadherin expression and increased N-cadherin expression after enforced miR-181a expression, while SkOV3/PTX cells had increased E-cadherin expression and decreased N-cadherin expression after miR-181a knockdown. SkOV3 cells had increased P-gp expression after enforced miR-181a expression. Following MTT assay and flow cytometry analysis both confirmed that miR-181a overexpression decreased the PTX sensitivity of SkOV3 cells and while miR-181a inhibition increased the sensitivity of SkOV3/PTX cells. miR-181a is an important oncomiR significantly increased in chemoresistant ovarian cancer. Its upregulation is associated with increased level of EMT and decreased cell apoptosis induced by PTX treatment.
Key Molecule: hsa-mir-141 [125]
Molecule Alteration Expression
Down-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model OVCAR3 cells Ovary Homo sapiens (Human) CVCL_0465
MES-OV cells Ovary Homo sapiens (Human) CVCL_CZ92
In Vivo Model Mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
SRB colorimetric assay; Flow cytometry assay
Mechanism Description The miR-200 family has major roles in EMT and taxane resistance in taxane selected ovarian cancer cell variants, and that re-introduction of miR-200s was not sufficient to fully reverse the mesenchymal phenotype in these variants. Although miR-200s were able to restore paclitaxel sensitivity in one of the variants, they did not do so in the other, and produced resistance to carboplatin in both. The divergent effects of miR-200s on taxane and carboplatin cytotoxicity should be further investigated in ovarian cancers. miR-200c and miR-141 mimics conferred resistance to carboplatin in MES-OV/TP cells, similar to OVCAR-3/TP, but sensitized MES-OV to paclitaxel. Several genes involved in balancing oxidative stress were altered in OVCAR-3/TP 200c141 cells compared to controls. The miR-200 family plays major, cell-context dependent roles in regulating EMT and sensitivity to carboplatin and paclitaxel in OVCAR-3 and MES-OV cells.
Key Molecule: hsa-mir-200c [125]
Molecule Alteration Expression
Down-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model OVCAR3 cells Ovary Homo sapiens (Human) CVCL_0465
MES-OV cells Ovary Homo sapiens (Human) CVCL_CZ92
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
SRB colorimetric assay; Flow cytometry assay
Mechanism Description The miR-200 family has major roles in EMT and taxane resistance in taxane selected ovarian cancer cell variants, and that re-introduction of miR-200s was not sufficient to fully reverse the mesenchymal phenotype in these variants. Although miR-200s were able to restore paclitaxel sensitivity in one of the variants, they did not do so in the other, and produced resistance to carboplatin in both. The divergent effects of miR-200s on taxane and carboplatin cytotoxicity should be further investigated in ovarian cancers. miR-200c and miR-141 mimics conferred resistance to carboplatin in MES-OV/TP cells, similar to OVCAR-3/TP, but sensitized MES-OV to paclitaxel. Several genes involved in balancing oxidative stress were altered in OVCAR-3/TP 200c141 cells compared to controls. The miR-200 family plays major, cell-context dependent roles in regulating EMT and sensitivity to carboplatin and paclitaxel in OVCAR-3 and MES-OV cells.
Key Molecule: Tubulin beta-3 chain (TUBB3) [125]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model OVCAR3 cells Ovary Homo sapiens (Human) CVCL_0465
MES-OV cells Ovary Homo sapiens (Human) CVCL_CZ92
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
SRB colorimetric assay; Flow cytometry assay
Mechanism Description The miR-200 family has major roles in EMT and taxane resistance in taxane selected ovarian cancer cell variants, and that re-introduction of miR-200s was not sufficient to fully reverse the mesenchymal phenotype in these variants. Although miR-200s were able to restore paclitaxel sensitivity in one of the variants, they did not do so in the other, and produced resistance to carboplatin in both. The divergent effects of miR-200s on taxane and carboplatin cytotoxicity should be further investigated in ovarian cancers. miR-200c and miR-141 mimics conferred resistance to carboplatin in MES-OV/TP cells, similar to OVCAR-3/TP, but sensitized MES-OV to paclitaxel. Several genes involved in balancing oxidative stress were altered in OVCAR-3/TP 200c141 cells compared to controls. The miR-200 family plays major, cell-context dependent roles in regulating EMT and sensitivity to carboplatin and paclitaxel in OVCAR-3 and MES-OV cells.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: CUB domain-containing protein 1 (CDCP1) [1]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
MYC/WNT/AKT signaling pathway Regulation hsa04217
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
In Vivo Model NMRI-nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Time course proliferation assay; Flow cytometry assay
Mechanism Description CDCP1 and PLAGL2 oncogenes were found to be the most relevant direct miR-654-5p targets and both genes convey in a molecular signature associated with key cancer pathways relevant to ovarian tumorigenesis, such as MYC, WNT and AkT pathways.
Key Molecule: Zinc finger protein PLAGL2 (PLAGL2) [1]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
MYC/WNT/AKT signaling pathway Regulation hsa04217
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
In Vivo Model NMRI-nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Time course proliferation assay; Flow cytometry assay
Mechanism Description CDCP1 and PLAGL2 oncogenes were found to be the most relevant direct miR-654-5p targets and both genes convey in a molecular signature associated with key cancer pathways relevant to ovarian tumorigenesis, such as MYC, WNT and AkT pathways.
Key Molecule: Growth protein 5 inhibitor (ING5) [116]
Molecule Alteration Expression
Down-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
A2780/Taxol cells Ovary Homo sapiens (Human) CVCL_IJ13
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Colony formation assay; Apoptosis analysis by FITC immunofluorescence
Mechanism Description miR1307 promotes ovarian cancer cell chemoresistance by targeting the ING5 expression.
Key Molecule: E3 ubiquitin-protein ligase Mdm2 (MDM2) [117]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Activation hsa05200
In Vitro Model Hey A8 cells Ovary Homo sapiens (Human) CVCL_8878
SkVO3ip1 cells Ovary Homo sapiens (Human) CVCL_0C84
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description Down-regulation of miR-194-5p induces paclitaxel resistance in ovarian cancer cells by altering MDM2 expression.
Key Molecule: Apoptotic protease-activating factor 1 (APAF1) [119]
Molecule Alteration Expression
Down-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Epithelial mesenchymal transition signaling pathway Activation hsa01521
TGF signaling pathway Regulation hsa04350
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
Hey A8 cells Ovary Homo sapiens (Human) CVCL_8878
OVCA433 cells Ovary Homo sapiens (Human) CVCL_0475
ALST cells Ovary Homo sapiens (Human) CVCL_W778
HeyA8-MDR cells Ovary Homo sapiens (Human) CVCL_8879
OVCA432 cells Ovary Homo sapiens (Human) CVCL_3769
OVCAR5 cells Ovary Homo sapiens (Human) CVCL_1628
SkOV3-TR cells Ovary Homo sapiens (Human) CVCL_HF69
SkOV3ip cells Ovary Homo sapiens (Human) CVCL_0C84
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Flow cytometric assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description The identification of APAF1 as a direct target of miR21 and APAF1 as a mediator of miR21 for conferring chemoresistance in ovarian cancer suggests that strategies based on the upregulation of APAF1 in ovarian cancer cells can be used to sensitize ovarian cancer cells to paclitaxel treatment.ovarian cancer suggests that strategies based on the upregulation of APAF1 in ovarian cancer cells can be used to sensitize ovarian cancer cells to paclitaxel treatment.
Key Molecule: Phosphatase and tensin homolog (PTEN) [120]
Molecule Alteration Expression
Down-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
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 proliferation Activation hsa05200
PTEN/AKT signaling pathway Regulation hsa05235
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
OVCAR3 cells Ovary Homo sapiens (Human) CVCL_0465
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description microRNA-17-5p induces drug resistance and invasion of ovarian carcinoma cells by targeting PTEN signaling.
Key Molecule: Myeloid differentiation primary response protein MyD88 (MYD88) [121]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
TLR/MyD88 signaling pathway Regulation hsa04620
In Vitro Model A2780 cells Ovary Homo sapiens (Human) CVCL_0134
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Transwell assay
Mechanism Description In the present study, flow cytometric assays were used to detect the apoptosis of A2780 cells after down-regulation of miRNA-149. We found that down-regulation of miRNA-149 decreased the apoptosis induced by paclitaxel when compared to the control group. Furthermore, we showed that down-regulation of miRNA-149 in A2780 cells (+) the expression of the anti-apoptotic protein Bcl-2 and inhibited the expression of the pro-apoptotic protein bax, which may have led to paclitaxel resistance.
Key Molecule: B-cell lymphoma/leukemia 10 (BCL10) [122]
Molecule Alteration Expression
Down-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
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 SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
Experiment for
Molecule Alteration
Immunoblotting analysis
Experiment for
Drug Resistance
TUNEL assay
Mechanism Description miR-106a and miR-591 have important roles in conferring PTX resistance to ovarian cancer cells. Modulation of these microRNAs resensitizes PTX-resistant cancer cells by targeting BCL10, caspase-7, and ZEB1.
Key Molecule: Caspase-7 (CASP7) [122]
Molecule Alteration Expression
Down-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
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 SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
Experiment for
Molecule Alteration
Immunoblotting analysis
Experiment for
Drug Resistance
TUNEL assay
Mechanism Description miR-106a and miR-591 have important roles in conferring PTX resistance to ovarian cancer cells. Modulation of these microRNAs resensitizes PTX-resistant cancer cells by targeting BCL10, caspase-7, and ZEB1.
Key Molecule: Zinc finger E-box-binding homeobox 1 (ZEB1) [122]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
Experiment for
Molecule Alteration
Immunoblotting analysis
Experiment for
Drug Resistance
TUNEL assay
Mechanism Description miR-106a and miR-591 have important roles in conferring PTX resistance to ovarian cancer cells. Modulation of these microRNAs resensitizes PTX-resistant cancer cells by targeting BCL10, caspase-7, and ZEB1.
Key Molecule: Insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) [6]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model T47D cells Breast Homo sapiens (Human) CVCL_0553
IGROV1 cells Ovary Homo sapiens (Human) CVCL_1304
OVCAR8 cells Ovary Homo sapiens (Human) CVCL_1629
LOX-IMVI cells Ovary Homo sapiens (Human) CVCL_1381
NCI/ADR-RES cells Ovary Homo sapiens (Human) CVCL_1452
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
SRB cytotoxicity assay
Mechanism Description IMP-1 is an RNA binding protein that acts by stabilizing the mRNA of a number of target genes. In addition, IMP-1 was shown to protect the mRNA of MDR1 from endonucleolytic attack in an in vitro RNA stability assay. Introducing let-7g into ADR-RES cells expressing both IMP-1 and MDR1 reduced expression of both proteins rendering the cells more sensitive to treatment with either Taxol or vinblastine without affecting the sensitivity of the cells to carboplatin.
Key Molecule: Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) [16]
Molecule Alteration Expression
Down-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) is a protein that binds to HIF-1alpha and inhibits its transcriptional activity. HIF1AN is a potential miR-135a target listed in both the TargetScan and PicTar databases. miR-135a-mediated paclitaxel resistance is in part mediated by downregulation of APC.
Key Molecule: Homeodomain-interacting protein kinase 2 (HIPK2) [123]
Molecule Alteration Expression
Down-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
miRNAs/HIPk2/MDR1/P-gp signaling pathway Regulation hsa05206
In Vitro Model A2780 cells Ovary Homo sapiens (Human) CVCL_0134
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Transfection of A2780/Taxol cells with the inhibitors of miR-27a decreased the expression of MDR1 mRNA and P-gp protein, increased HIPk2 protein expression, enhanced the sensitivity of A2780/taxol cells to paclitaxel, increased paclitaxel-induced apoptosis and the fluorescence intensity of intracellular Rh-123. The deregulation of miR-27a may be involved in the development of drug resistance, regulating the expression of MDR1/P-gp, at least in part, by targeting HIPk2 in ovarian cancer cells.
Key Molecule: Macrophage colony-stimulating factor 1 (MCSF) [20]
Molecule Alteration Expression
Up-regulation
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A2780 cells Ovary Homo sapiens (Human) CVCL_0134
A2780CIS cells Ovary Homo sapiens (Human) CVCL_1942
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Clonogenic assay
Mechanism Description Finally downstreamtarget validation was proven for the miR-130a, whose downregulation was linked to the translational activation of the M-CSF gene, a knownresistance factor for ovarian cancer.
Key Molecule: Carboxylesterase 4A (CES4A) [86]
Molecule Alteration Missense mutation
p.P55S
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AXLK signaling pathway Activation hsa01521
In Vitro Model Plasma Blood Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Circulating-free DNA assay; Whole exome sequencing assay
Mechanism Description Quantification of allele fractions in plasma identified increased representation of mutant alleles in association with emergence of therapy resistance.
Key Molecule: Mitotic checkpoint serine/threonine-protein kinase BUB1 (BUB1) [86]
Molecule Alteration Missense mutation
p.M889K
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AXLK signaling pathway Activation hsa01521
In Vitro Model Plasma Blood Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Circulating-free DNA assay; Whole exome sequencing assay
Mechanism Description Quantification of allele fractions in plasma identified increased representation of mutant alleles in association with emergence of therapy resistance.
Key Molecule: Serine/threonine-protein kinase mTOR (mTOR) [86]
Molecule Alteration Missense mutation
p.K1655N
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AXLK signaling pathway Activation hsa01521
In Vitro Model Plasma Blood Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Circulating-free DNA assay; Whole exome sequencing assay
Mechanism Description Quantification of allele fractions in plasma identified increased representation of mutant alleles in association with emergence of therapy resistance.
Key Molecule: Serine/threonine-protein kinase mTOR (mTOR) [86]
Molecule Alteration Missense mutation
p.K1655N
Resistant Disease Ovarian serous carcinoma [ICD-11: 2C73.2]
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
Circulating-free DNA assay; Whole exome sequencing assay
Mechanism Description Quantification of allele fractions in plasma identified increased representation of mutant alleles in association with emergence of therapy resistance.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Drug Inactivation by Structure Modification (DISM) Click to Show/Hide
Key Molecule: Glutathione S-transferase P (GSTP1) [126]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model A2780 cells Ovary Homo sapiens (Human) CVCL_0134
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Overexpression of miR-133b increases ovarian cancer cell sensitivity to cisplatin and paclitaxel by decreasing GST-Pi and MDR1 expression.
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-146a [29]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation miR146a/SOD2/ROS signaling pathway Regulation hsa05206
In Vitro Model HEY cells Ovary Homo sapiens (Human) CVCL_0297
OVCAR3 cells Ovary Homo sapiens (Human) CVCL_0465
CAOV3 cells Ovary Homo sapiens (Human) CVCL_0201
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay; TUNEL Assay
Mechanism Description miR146a downregulates the expression of SOD2 and enhances ROS generation, leading to increased apoptosis, inhibition of proliferation, and enhanced sensitivity to chemotherapy.
Key Molecule: hsa-mir-136 [127]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
WST assay; Spheroid formation assay; Colony-forming assay; TUNEL assay; Wound healing assay
Mechanism Description microRNA-136 inhibits cancer stem cell activity and enhances the anti-tumor effect of paclitaxel against chemoresistant ovarian cancer cells by targeting Notch3.
Key Molecule: hsa-miR-383-5p [128]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
PI3K/AKT signaling pathway Regulation hsa04151
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
OVCAR3 cells Ovary Homo sapiens (Human) CVCL_0465
CAOV3 cells Ovary Homo sapiens (Human) CVCL_0201
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Hoechst 33
Mechanism Description Up-regulation of miR-383-5p inhibited cell proliferation, tumor growth and enhanced chemosensitivity of ovarian cancer cells through suppressing TRIM27 expression.
Key Molecule: hsa-mir-129 [118]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
UCA1/miR129/ABCB1 signaling pathway Regulation hsa05206
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
Hey A8 cells Ovary Homo sapiens (Human) CVCL_8878
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description ABCB1 up-regulated by UCA1/miR-129 axis contributed to PTX resistance in PTX-resistant OC cells.
Key Molecule: hsa-miR-630 [129]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Activation hsa04670
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Wound healing assay; Invasion assay; CCK8 assay; Flow cytometry assay
Mechanism Description miR-630 inhibitor attenuated chemoresistant epithelial ovarian cancer proliferation and invasion, probably by targeting APAF-1, re-sensitizing the cells to chemotherapy.
Key Molecule: hsa-mir-186 [130]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model A2780 cells Ovary Homo sapiens (Human) CVCL_0134
OVCAR3 cells Ovary Homo sapiens (Human) CVCL_0465
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Both A2780/DDP and A2780/Taxol cells expressed miR-186 at lower levels than A2780. miR-186 overexpression increased the sensitivity of ovarian cancer cell lines to paclitaxel and cisplatin compared with the negative control or mock cells, miR-186 transfection induced cell apoptosis while anti-miR-186 transfection reduced cell apoptosis, suggesting that miR-186 may inhibit the development of drug resistance in ovarian cancer cells. miR-186 overexpression may increase the sensitivity of ovarian cancer cells to paclitaxel by targeting ABCB1 and modulating GST-Pi.
Key Molecule: hsa-miR-133b [126]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model A2780 cells Ovary Homo sapiens (Human) CVCL_0134
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Overexpression of miR-133b increases ovarian cancer cell sensitivity to cisplatin and paclitaxel by decreasing GST-Pi and MDR1 expression.
Key Molecule: hsa-mir-29b [131]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation miR29b signaling pathway Regulation hsa05206
In Vitro Model OVCAR3 cells Ovary Homo sapiens (Human) CVCL_0465
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
H&E staining assay
Mechanism Description The ATG9A down expression due to miR-29b increasing could significantly promote Ovarian carcinoma drug sensitivity on different chemotherapeutic drugs (Cisplatin, Paclitaxel, Platinum, Cyclophosphamide).
Key Molecule: hsa-mir-145 [132]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
MCF-7/ADM cells Breast Homo sapiens (Human) CVCL_0031
A2780/PTX cells Ovary Homo sapiens (Human) CVCL_IJ13
HOEC cells Ovary Homo sapiens (Human) N.A.
SkOV3/PTX cells Ovary Homo sapiens (Human) CVCL_HF69
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-145 modulates the cellular response to anticancer drugs, Down-regulation of miR-145 is correlated with overexpression of Sp1 and Cdk6, Sp1 and Cdk6 are targets of miR-145, miR-145 downregulated P-gp and pRb through inhibition of Sp1 and Cdk6, miR-145 sensitized EOC cells to paclitaxel via Sp1 and Cdk6 inhibition, Overexpression of miR-145 enhanced paclitaxel sensitivity in vivo.
Key Molecule: hsa-mir-200c [133]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell adhesion Inhibition hsa04514
Cell apoptosis Inhibition hsa04210
Cell migration Activation hsa04670
In Vitro Model HEY cells Ovary Homo sapiens (Human) CVCL_0297
SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
OVCA433 cells Ovary Homo sapiens (Human) CVCL_0475
OV 1847 cells Breast Homo sapiens (Human) CVCL_D703
OVCA 420 cells Breast Homo sapiens (Human) CVCL_3935
In Vivo Model (NOD) /SCID nude mouse xenograft model .
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Overexpression of TUBB3 is thought to result in resistance to taxanes is by enhancement of the dynamic instability of microtubules, thereby counteracting the activity of microtubule targeting agents. Transient restoration of miR-200c using miRNA mimics cause a significant decrease in TUBB3 levels, thus results in the resistance to taxanes.
Key Molecule: hsa-mir-199a [134]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model CD44+/CD117+ ovarian CICs cells Ovary 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
MTT assay
Mechanism Description CD44 plays an important role in cellular adhesion, lymphocyte activation/migration, tumorigenesis, and the formation of metastases, endogenous mature miR-199a may prevent the growth of human ovarian CICs via decreasing the expression of CD44.
Key Molecule: hsa-mir-130b [135]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description CSF-1 expression was negatively associated with miR-130b level in ovarian tissues and cell lines. miR-130b modulates MDR by targeting CSF-1, Down-regulation of miR-130b promotes the development of multidrug resistant ovarian cancer partially by targeting the 3'-UTR of CSF-1.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [118]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
UCA1/miR129/ABCB1 signaling pathway Regulation hsa05206
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
Hey A8 cells Ovary Homo sapiens (Human) CVCL_8878
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description ABCB1 up-regulated by UCA1/miR-129 axis contributed to PTX resistance in PTX-resistant OC cells.
Key Molecule: Multidrug resistance protein 1 (ABCB1) [130]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model A2780 cells Ovary Homo sapiens (Human) CVCL_0134
OVCAR3 cells Ovary Homo sapiens (Human) CVCL_0465
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Both A2780/DDP and A2780/Taxol cells expressed miR-186 at lower levels than A2780. miR-186 overexpression increased the sensitivity of ovarian cancer cell lines to paclitaxel and cisplatin compared with the negative control or mock cells, miR-186 transfection induced cell apoptosis while anti-miR-186 transfection reduced cell apoptosis, suggesting that miR-186 may inhibit the development of drug resistance in ovarian cancer cells. miR-186 overexpression may increase the sensitivity of ovarian cancer cells to paclitaxel by targeting ABCB1 and modulating GST-Pi.
Key Molecule: Multidrug resistance protein 1 (ABCB1) [126]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model A2780 cells Ovary Homo sapiens (Human) CVCL_0134
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Overexpression of miR-133b increases ovarian cancer cell sensitivity to cisplatin and paclitaxel by decreasing GST-Pi and MDR1 expression.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Superoxide dismutase Mn (SODM) [29]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation miR146a/SOD2/ROS signaling pathway Regulation hsa05206
In Vitro Model HEY cells Ovary Homo sapiens (Human) CVCL_0297
OVCAR3 cells Ovary Homo sapiens (Human) CVCL_0465
CAOV3 cells Ovary Homo sapiens (Human) CVCL_0201
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; TUNEL Assay
Mechanism Description miR146a downregulates the expression of SOD2 and enhances ROS generation, leading to increased apoptosis, inhibition of proliferation, and enhanced sensitivity to chemotherapy.
Key Molecule: Neurogenic locus notch homolog protein 3 (NOTCH3) [127]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
WST assay; Spheroid formation assay; Colony-forming assay; TUNEL assay; Wound healing assay
Mechanism Description microRNA-136 inhibits cancer stem cell activity and enhances the anti-tumor effect of paclitaxel against chemoresistant ovarian cancer cells by targeting Notch3.
Key Molecule: Zinc finger protein RFP (TRIM27) [128]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
PI3K/AKT signaling pathway Regulation hsa04151
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
OVCAR3 cells Ovary Homo sapiens (Human) CVCL_0465
CAOV3 cells Ovary Homo sapiens (Human) CVCL_0201
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Hoechst 33
Mechanism Description Up-regulation of miR-383-5p inhibited cell proliferation, tumor growth and enhanced chemosensitivity of ovarian cancer cells through suppressing TRIM27 expression.
Key Molecule: Apoptotic protease-activating factor 1 (APAF1) [129]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Wound healing assay; Invasion assay; CCK8 assay; Flow cytometry assay
Mechanism Description miR-630 inhibitor attenuated chemoresistant epithelial ovarian cancer proliferation and invasion, probably by targeting APAF-1, re-sensitizing the cells to chemotherapy.
Key Molecule: Autophagy-related protein 9A (ATG9A) [131]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation miR29b signaling pathway Regulation hsa05206
In Vitro Model OVCAR3 cells Ovary Homo sapiens (Human) CVCL_0465
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
H&E staining assay
Mechanism Description The ATG9A down expression due to miR-29b increasing could significantly promote Ovarian carcinoma drug sensitivity on different chemotherapeutic drugs (Cisplatin, Paclitaxel, Platinum, Cyclophosphamide).
Key Molecule: Cyclin-dependent kinase 6 (CDK6) [132]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
MCF-7/ADM cells Breast Homo sapiens (Human) CVCL_0031
A2780/PTX cells Ovary Homo sapiens (Human) CVCL_IJ13
HOEC cells Ovary Homo sapiens (Human) N.A.
SkOV3/PTX cells Ovary Homo sapiens (Human) CVCL_HF69
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-145 modulates the cellular response to anticancer drugs, Down-regulation of miR-145 is correlated with overexpression of Sp1 and Cdk6, Sp1 and Cdk6 are targets of miR-145, miR-145 downregulated P-gp and pRb through inhibition of Sp1 and Cdk6, miR-145 sensitized EOC cells to paclitaxel via Sp1 and Cdk6 inhibition, Overexpression of miR-145 enhanced paclitaxel sensitivity in vivo.
Key Molecule: Transcription factor Sp1 (SP1) [132]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
MCF-7/ADM cells Breast Homo sapiens (Human) CVCL_0031
A2780/PTX cells Ovary Homo sapiens (Human) CVCL_IJ13
HOEC cells Ovary Homo sapiens (Human) N.A.
SkOV3/PTX cells Ovary Homo sapiens (Human) CVCL_HF69
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-145 modulates the cellular response to anticancer drugs, Down-regulation of miR-145 is correlated with overexpression of Sp1 and Cdk6, Sp1 and Cdk6 are targets of miR-145, miR-145 downregulated P-gp and pRb through inhibition of Sp1 and Cdk6, miR-145 sensitized EOC cells to paclitaxel via Sp1 and Cdk6 inhibition, Overexpression of miR-145 enhanced paclitaxel sensitivity in vivo.
Key Molecule: Tubulin beta-3 chain (TUBB3) [133]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell adhesion Inhibition hsa04514
Cell apoptosis Inhibition hsa04210
In Vitro Model HEY cells Ovary Homo sapiens (Human) CVCL_0297
SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
OVCA433 cells Ovary Homo sapiens (Human) CVCL_0475
OV 1847 cells Breast Homo sapiens (Human) CVCL_D703
OVCA 420 cells Breast Homo sapiens (Human) CVCL_3935
In Vivo Model (NOD) /SCID nude mouse xenograft model .
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Overexpression of TUBB3 is thought to result in resistance to taxanes is by enhancement of the dynamic instability of microtubules, thereby counteracting the activity of microtubule targeting agents. Transient restoration of miR-200c using miRNA mimics cause a significant decrease in TUBB3 levels, thus results in the resistance to taxanes.
Key Molecule: Extracellular matrix receptor III (CD44) [134]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
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 CD44+/CD117+ ovarian CICs cells Ovary Homo sapiens (Human) N.A.
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 CD44 plays an important role in cellular adhesion, lymphocyte activation/migration, tumorigenesis, and the formation of metastases, endogenous mature miR-199a may prevent the growth of human ovarian CICs via decreasing the expression of CD44.
Key Molecule: Macrophage colony-stimulating factor 1 (MCSF) [135]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
A2780 cells Ovary Homo sapiens (Human) CVCL_0134
Experiment for
Molecule Alteration
Luciferase reporter assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description CSF-1 expression was negatively associated with miR-130b level in ovarian tissues and cell lines. miR-130b modulates MDR by targeting CSF-1, Down-regulation of miR-130b promotes the development of multidrug resistant ovarian cancer partially by targeting the 3'-UTR of CSF-1.
Endometrial cancer [ICD-11: 2C76]
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: Long non-protein coding RNA 672 (LINC00672) [12]
Molecule Alteration Expression
Down-regulation
Resistant Disease Endometrial cancer [ICD-11: 2C76.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
Ishikawa cells Endometrium Homo sapiens (Human) CVCL_2529
HEC-1A cells Uterus Homo sapiens (Human) CVCL_0293
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Transwell migration assay; Matrigel invasion assay; Flow cytometry assay; TUNEL assay; Wound healing assay; Colony formation assay
Mechanism Description LINC00672 can down-regulate LASP1 expression as a locus-restricted cofactor for p53-mediated gene suppression, thus impacting EC malig.ncies and chemosensitivity to paclitaxel.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: LIM and SH3 domain protein 1 (LASP1) [12]
Molecule Alteration Expression
Up-regulation
Resistant Disease Endometrial cancer [ICD-11: 2C76.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
Ishikawa cells Endometrium Homo sapiens (Human) CVCL_2529
HEC-1A cells Uterus Homo sapiens (Human) CVCL_0293
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Transwell migration assay; Matrigel invasion assay; Flow cytometry assay; TUNEL assay; Wound healing assay; Colony formation assay
Mechanism Description LINC00672 can down-regulate LASP1 expression as a locus-restricted cofactor for p53-mediated gene suppression, thus impacting EC malig.ncies and chemosensitivity to paclitaxel.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-23b [136]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Endometrial carcinoma [ICD-11: 2C76.2]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model HEC1A cells Uterus Homo sapiens (Human) CVCL_0293
Human normal endometrial epithelial cell line Uterus Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
RNA pull-down assay; qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometric analysis
Mechanism Description Long non-coding RNA TUSC7 acted as a potential tumor suppressor gene to inhibit cell growth as well as advance the chemotherapy sensitivity through targeted silencing of miR23b.
Key Molecule: Tumor suppressor candidate 7 (TUSC7) [136]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Endometrial carcinoma [ICD-11: 2C76.2]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model HEC1A cells Uterus Homo sapiens (Human) CVCL_0293
Human normal endometrial epithelial cell line Uterus Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometric analysis
Mechanism Description Long non-coding RNA TUSC7 acted as a potential tumor suppressor gene to inhibit cell growth as well as advance the chemotherapy sensitivity through targeted silencing of miR23b.
Key Molecule: hsa-miR-29c-3p [137]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Endometrial carcinoma [ICD-11: 2C76.2]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell colony Inhibition hsa05200
Cell invasion Inhibition hsa05200
Cell viability Inhibition hsa05200
miR125a-5p/BCL2/MRP4 signaling pathway Regulation hsa05206
In Vitro Model Ishikawa cells Endometrium Homo sapiens (Human) CVCL_2529
HEC-1A cells Uterus Homo sapiens (Human) CVCL_0293
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The up-regulation of miR-29c-3p using exogenous mimic molecules markedly increased PTX sensitivity in both cell lines and reduced expression of kDM5B while the inhibitor of miR-29-3p resulted in the opposite effects.
Key Molecule: hsa-mir-24 [138]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Endometrial carcinoma [ICD-11: 2C76.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model HEC-1A cells Uterus Homo sapiens (Human) CVCL_0293
In Vivo Model Crl:NU-Foxn1nu nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-24, which is under-expressed in EC, functions as a tumor-suppressing gene to inhibit malignant proliferation and advance chemotherapy sensitivity to paclitaxel in EC by targeted silencing of S100A8.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: hsa-mir-200c [139]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Endometrial cancer [ICD-11: 2C76.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
In Vitro Model Hec50 cells Endometrium Homo sapiens (Human) CVCL_2929
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
ELISA assay
Mechanism Description Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3.
Key Molecule: BDNF/NT-3 growth factors receptor (NTRK2) [139]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Endometrial cancer [ICD-11: 2C76.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
In Vitro Model Hec50 cells Endometrium Homo sapiens (Human) CVCL_2929
Experiment for
Molecule Alteration
Immunoblotting analysis
Experiment for
Drug Resistance
ELISA assay
Mechanism Description Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3.
Key Molecule: Protein quaking (QKI) [139]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Endometrial cancer [ICD-11: 2C76.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
In Vitro Model Hec50 cells Endometrium Homo sapiens (Human) CVCL_2929
Experiment for
Molecule Alteration
Immunoblotting analysis
Experiment for
Drug Resistance
ELISA assay
Mechanism Description Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3.
Key Molecule: Zinc finger E-box-binding homeobox 1 (ZEB1) [139]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Endometrial cancer [ICD-11: 2C76.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
In Vitro Model Hec50 cells Endometrium Homo sapiens (Human) CVCL_2929
Experiment for
Molecule Alteration
Immunoblotting analysis
Experiment for
Drug Resistance
ELISA assay
Mechanism Description Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3.
Key Molecule: Zinc finger E-box-binding homeobox 2 (ZEB2) [139]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Endometrial cancer [ICD-11: 2C76.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
In Vitro Model Hec50 cells Endometrium Homo sapiens (Human) CVCL_2929
Experiment for
Molecule Alteration
Immunoblotting analysis
Experiment for
Drug Resistance
ELISA assay
Mechanism Description Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Lysine-specific demethylase 5B (KDM5B) [137]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Endometrial carcinoma [ICD-11: 2C76.2]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell colony Inhibition hsa05200
Cell invasion Inhibition hsa05200
Cell viability Inhibition hsa05200
miR125a-5p/BCL2/MRP4 signaling pathway Regulation hsa05206
In Vitro Model Ishikawa cells Endometrium Homo sapiens (Human) CVCL_2529
HEC-1A cells Uterus Homo sapiens (Human) CVCL_0293
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The up-regulation of miR-29c-3p using exogenous mimic molecules markedly increased PTX sensitivity in both cell lines and reduced expression of kDM5B while the inhibitor of miR-29-3p resulted in the opposite effects.
Key Molecule: Protein S100-A8 (S100A8) [138]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Endometrial carcinoma [ICD-11: 2C76.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model HEC-1A cells Uterus Homo sapiens (Human) CVCL_0293
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-24, which is under-expressed in EC, functions as a tumor-suppressing gene to inhibit malignant proliferation and advance chemotherapy sensitivity to paclitaxel in EC by targeted silencing of S100A8.
Key Molecule: Fibronectin (FN1) [139]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Endometrial cancer [ICD-11: 2C76.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
In Vitro Model Hec50 cells Endometrium Homo sapiens (Human) CVCL_2929
Experiment for
Molecule Alteration
Immunoblotting analysis
Experiment for
Drug Resistance
ELISA assay
Mechanism Description Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3.
Key Molecule: Tubulin beta-3 chain (TUBB3) [139]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Endometrial cancer [ICD-11: 2C76.1]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
In Vitro Model Hec50 cells Endometrium Homo sapiens (Human) CVCL_2929
Experiment for
Molecule Alteration
Immunoblotting analysis
Experiment for
Drug Resistance
ELISA assay
Mechanism Description Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3.
Cervical cancer [ICD-11: 2C77]
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-375 [140]
Molecule Alteration Expression
Up-regulation
Resistant Disease Cervical cancer [ICD-11: 2C77.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model Siha cells Cervix uteri Homo sapiens (Human) CVCL_0032
Caski cells Uterus Homo sapiens (Human) CVCL_1100
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description Paclitaxel transiently induced up-regulation of miR-375 expression, proliferation inhibition, transition from epithelial to mesenchymal phenotype, and consequently impaired paclitaxel sensitivity. Forced over-expression of miR-375 may suppress Ecadherin expression by a directly targeting pathway, which led to paclitaxel resistance. Contrarily, re-expression of Ecadherin partly reversed epithelial-mesenchymal transition phenotype and miR-375 induced paclitaxel-resistance. Our findings suggest that paclitaxel-induced miR-375 over-expression facilitates epithelial-mesenchymal transition process via directly targeting Ecadherin, proliferation inhibition, and consequently results in chemo-resistance in cervical cancer cells.
Key Molecule: hsa-mir-375 [4]
Molecule Alteration Expression
Up-regulation
Resistant Disease Cervical cancer [ICD-11: 2C77.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
Cell viability Activation hsa05200
In Vitro Model Siha cells Cervix uteri Homo sapiens (Human) CVCL_0032
Caski cells Uterus Homo sapiens (Human) CVCL_1100
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description Paclitaxel induced upregulated miR-375 expression in a clear dose-dependent manner. Forced overexpression of miR-375 in cervical cancer cells decreased paclitaxel sensitivity in vitro and in vivo.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: hsa-mir-195 [10]
Molecule Alteration Expression
Up-regulation
Resistant Disease Cervical cancer [ICD-11: 2C77.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model Siha cells Cervix uteri Homo sapiens (Human) CVCL_0032
Caski cells Uterus Homo sapiens (Human) CVCL_1100
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometric analysis; CCK8 assay
Mechanism Description LncRNA PVT1 epigenetically silences miR195 and modulates EMT and chemoresistance in cervical cancer cells. PVT1 could decrease miR195 expression via enhancing histone H3k27me3 in the miR195 promoter region and also via direct sponging of miR195.
Key Molecule: Pvt1 oncogene (PVT1) [10]
Molecule Alteration Expression
Up-regulation
Resistant Disease Cervical cancer [ICD-11: 2C77.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Epithelial mesenchymal transition signaling pathway Inhibition hsa01521
In Vitro Model Siha cells Cervix uteri Homo sapiens (Human) CVCL_0032
Caski cells Uterus Homo sapiens (Human) CVCL_1100
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
Flow cytometric analysis; CCK8 assay
Mechanism Description LncRNA PVT1 epigenetically silences miR195 and modulates EMT and chemoresistance in cervical cancer cells. PVT1 could decrease miR195 expression via enhancing histone H3k27me3 in the miR195 promoter region and also via direct sponging of miR195.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Cadherin-1 (CDH1) [140]
Molecule Alteration Expression
Down-regulation
Resistant Disease Cervical cancer [ICD-11: 2C77.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
In Vitro Model Siha cells Cervix uteri Homo sapiens (Human) CVCL_0032
Caski cells Uterus Homo sapiens (Human) CVCL_1100
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description Paclitaxel transiently induced up-regulation of miR-375 expression, proliferation inhibition, transition from epithelial to mesenchymal phenotype, and consequently impaired paclitaxel sensitivity. Forced over-expression of miR-375 may suppress Ecadherin expression by a directly targeting pathway, which led to paclitaxel resistance. Contrarily, re-expression of Ecadherin partly reversed epithelial-mesenchymal transition phenotype and miR-375 induced paclitaxel-resistance. Our findings suggest that paclitaxel-induced miR-375 over-expression facilitates epithelial-mesenchymal transition process via directly targeting Ecadherin, proliferation inhibition, and consequently results in chemo-resistance in cervical cancer cells.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: Long non-protein coding RNA 511 (LINC00511) [141]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Cervical cancer [ICD-11: 2C77.0]
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model Hela cells Cervix uteri Homo sapiens (Human) CVCL_0030
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description LINC00511 knockdown prevents cervical cancer cell proliferation and reduces resistance to paclitaxel.
Key Molecule: hsa-mir-21 [142]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Cervical cancer [ICD-11: 2C77.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
PTEN/AKT signaling pathway Regulation hsa05235
In Vitro Model Hela cells Cervix uteri Homo sapiens (Human) CVCL_0030
Siha cells Cervix uteri Homo sapiens (Human) CVCL_0032
Caski cells Uterus Homo sapiens (Human) CVCL_1100
ME-180 cells Uterus Homo sapiens (Human) CVCL_1401
C33A cells Uterus Homo sapiens (Human) CVCL_1094
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
Annexin V-FITC/PI staining for cell apoptosis assay; Hoechst 33258 staining for cell apoptosis assay; MTT assay
Mechanism Description miR21 inhibitor suppresses cell proliferation and colony formation through regulating the PTEN/AkT pathway and improves paclitaxel sensitivity in cervical cancer cells.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Phosphatase and tensin homolog (PTEN) [142]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Cervical cancer [ICD-11: 2C77.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
PTEN/AKT signaling pathway Regulation hsa05235
In Vitro Model Hela cells Cervix uteri Homo sapiens (Human) CVCL_0030
Siha cells Cervix uteri Homo sapiens (Human) CVCL_0032
Caski cells Uterus Homo sapiens (Human) CVCL_1100
ME-180 cells Uterus Homo sapiens (Human) CVCL_1401
C33A cells Uterus Homo sapiens (Human) CVCL_1094
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Annexin V-FITC/PI staining for cell apoptosis assay; Hoechst 33258 staining for cell apoptosis assay; MTT assay
Mechanism Description miR21 inhibitor suppresses cell proliferation and colony formation through regulating the PTEN/AkT pathway and improves paclitaxel sensitivity in cervical cancer cells.
Prostate cancer [ICD-11: 2C82]
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-216b-5p [143]
Molecule Alteration Expression
Down-regulation
Resistant Disease Prostate cancer [ICD-11: 2C82.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model DU-145 cells Prostate Homo sapiens (Human) CVCL_0105
LNCaP cells Prostate Homo sapiens (Human) CVCL_0395
PC3 cells Prostate Homo sapiens (Human) CVCL_0035
RWPE-1 cells Prostate Homo sapiens (Human) CVCL_3791
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Long non-coding RNA Linc00518 Can enhance GATA6 expression by suppressing miR-216b-5p expression to promotes paclitaxel resistance in the human prostate cancer.
Key Molecule: Long non-protein coding RNA 518 (LINC00518) [143]
Molecule Alteration Expression
Up-regulation
Resistant Disease Prostate cancer [ICD-11: 2C82.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model DU-145 cells Prostate Homo sapiens (Human) CVCL_0105
LNCaP cells Prostate Homo sapiens (Human) CVCL_0395
PC3 cells Prostate Homo sapiens (Human) CVCL_0035
RWPE-1 cells Prostate Homo sapiens (Human) CVCL_3791
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Long non-coding RNA Linc00518 Can enhance GATA6 expression by suppressing miR-216b-5p expression to promotes paclitaxel resistance in the human prostate cancer.
Key Molecule: hsa-mir-199a [7]
Molecule Alteration Expression
Down-regulation
Resistant Disease Prostate cancer [ICD-11: 2C82.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model PC3/TXR cells Prostate Homo sapiens (Human) CVCL_0035
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Annexin V-FITC and PI Flow cytometry assay
Mechanism Description Overexpression of miR199a inhibited PTX resistance. YES1 was a target of miR199a, and overexpression of YES1 reversed the effect of miR199a in suppressing PTX resistance. In vivo, miR199a increased tumor PTX sensitivity.
Key Molecule: hsa-mir-130a [144]
Molecule Alteration Expression
Down-regulation
Resistant Disease Prostate cancer [ICD-11: 2C82.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Caspase-3 signaling pathway Activation hsa04210
Cell apoptosis Inhibition hsa04210
In Vitro Model PC3 cells Prostate Homo sapiens (Human) CVCL_0035
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CellTiter-Glo luminescent cell viability assay
Mechanism Description Restoration of miR-130a activated caspase-8 and increased the drug sensitivity in taxane-resistant prostate cancer cells, suggesting that miR-130a may become a potential target for therapy of taxane-resistant CRPC. Since the mechanism of the action of miR-130a was different from that of paclitaxel, a combination therapy of paclitaxel and miR-130a mimic may be effective in treatment of CRPC. Furthermore, it was reported that miR-130a expression was decreased in prostate cancer tissues. It is therefore possible that the restoration of miR-130a could be an effective approach for treating not only taxane-resistant prostate cancer but also prostate cancer with reduced expression of miR-130a.
Key Molecule: hsa-mir-135a [16]
Molecule Alteration Expression
Up-regulation
Resistant Disease Prostate cancer [ICD-11: 2C82.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) is a protein that binds to HIF-1alpha and inhibits its transcriptional activity. HIF1AN is a potential miR-135a target listed in both the TargetScan and PicTar databases. miR-135a-mediated paclitaxel resistance is in part mediated by downregulation of APC.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Transcription factor GATA6 (GATA6) [143]
Molecule Alteration Expression
Up-regulation
Resistant Disease Prostate cancer [ICD-11: 2C82.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model DU-145 cells Prostate Homo sapiens (Human) CVCL_0105
LNCaP cells Prostate Homo sapiens (Human) CVCL_0395
PC3 cells Prostate Homo sapiens (Human) CVCL_0035
RWPE-1 cells Prostate Homo sapiens (Human) CVCL_3791
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Long non-coding RNA Linc00518 Can enhance GATA6 expression by suppressing miR-216b-5p expression to promotes paclitaxel resistance in the human prostate cancer.
Key Molecule: Tyrosine-protein kinase Yes (YES1) [7]
Molecule Alteration Expression
Up-regulation
Resistant Disease Prostate cancer [ICD-11: 2C82.0]
Experimental Note Identified from the Human Clinical Data
In Vitro Model PC3/TXR cells Prostate Homo sapiens (Human) CVCL_0035
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Luciferase reporter assay
Experiment for
Drug Resistance
MTT assay; Annexin V-FITC and PI Flow cytometry assay
Mechanism Description Overexpression of miR199a inhibited PTX resistance. YES1 was a target of miR199a, and overexpression of YES1 reversed the effect of miR199a in suppressing PTX resistance. In vivo, miR199a increased tumor PTX sensitivity.
Key Molecule: SLAIN motif-containing protein 1 (SLAIN1) [144]
Molecule Alteration Expression
Up-regulation
Resistant Disease Prostate cancer [ICD-11: 2C82.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Caspase-3 signaling pathway Activation hsa04210
Cell apoptosis Inhibition hsa04210
In Vitro Model PC3 cells Prostate Homo sapiens (Human) CVCL_0035
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CellTiter-Glo luminescent cell viability assay
Mechanism Description Restoration of miR-130a activated caspase-8 and increased the drug sensitivity in taxane-resistant prostate cancer cells, suggesting that miR-130a may become a potential target for therapy of taxane-resistant CRPC. Since the mechanism of the action of miR-130a was different from that of paclitaxel, a combination therapy of paclitaxel and miR-130a mimic may be effective in treatment of CRPC. Furthermore, it was reported that miR-130a expression was decreased in prostate cancer tissues. It is therefore possible that the restoration of miR-130a could be an effective approach for treating not only taxane-resistant prostate cancer but also prostate cancer with reduced expression of miR-130a.
Key Molecule: Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) [16]
Molecule Alteration Expression
Down-regulation
Resistant Disease Prostate cancer [ICD-11: 2C82.0]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) is a protein that binds to HIF-1alpha and inhibits its transcriptional activity. HIF1AN is a potential miR-135a target listed in both the TargetScan and PicTar databases. miR-135a-mediated paclitaxel resistance is in part mediated by downregulation of APC.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-148a [145]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Prostate cancer [ICD-11: 2C82.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
MSK1 signaling pathway Inhibition hsa04010
In Vitro Model PC3PR cells Prostate Homo sapiens (Human) CVCL_0035
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Cell Growth Assay
Mechanism Description MSk1 is a novel target gene of miR-148a in both PC3 and PC3PR cells and miR-148 attenuates paclitaxel-resistance of PC3PR cells by modulating MSk1 expression.
Key Molecule: hsa-mir-34 [146]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Prostate cancer [ICD-11: 2C82.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
Notch1 signaling pathway Inhibition hsa04330
In Vitro Model PC3 cells Prostate Homo sapiens (Human) CVCL_0035
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description microRNA-34a Attenuates Paclitaxel Resistance in Prostate Cancer Cells via Direct Suppression of JAG1/Notch1 Axis.
Key Molecule: hsa-mir-34 [147]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Prostate cancer [ICD-11: 2C82.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Inhibition hsa05200
In Vitro Model PC3 cells Prostate Homo sapiens (Human) CVCL_0035
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Trypan blue dye exclusion assay
Mechanism Description SIRT1 plays crucial roles in various cellular processes including cell survival under genotoxic and oxidative stresses. Bcl2I is an anti-apoptotic factor. In PC3PR cells, reduced expression of miR-34a confers paclitaxel resistance via up-regulating SIRT1 and Bcl2 expression.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Ribosomal protein S6 kinase alpha-5 (RPS6KA5) [145]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Prostate cancer [ICD-11: 2C82.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
MSK1 signaling pathway Inhibition hsa04010
In Vitro Model PC3PR cells Prostate Homo sapiens (Human) CVCL_0035
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Cell Growth Assay
Mechanism Description MSk1 is a novel target gene of miR-148a in both PC3 and PC3PR cells and miR-148 attenuates paclitaxel-resistance of PC3PR cells by modulating MSk1 expression.
Key Molecule: Protein jagged-1 (JAG1) [146]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Prostate cancer [ICD-11: 2C82.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell colony Inhibition hsa05200
Cell proliferation Inhibition hsa05200
Notch1 signaling pathway Inhibition hsa04330
In Vitro Model PC3 cells Prostate Homo sapiens (Human) CVCL_0035
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 microRNA-34a Attenuates Paclitaxel Resistance in Prostate Cancer Cells via Direct Suppression of JAG1/Notch1 Axis.
Key Molecule: Neurogenic locus notch homolog protein 1 (NOTCH1) [146]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Prostate cancer [ICD-11: 2C82.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell colony Inhibition hsa05200
Cell proliferation Inhibition hsa05200
Notch1 signaling pathway Inhibition hsa04330
In Vitro Model PC3 cells Prostate Homo sapiens (Human) CVCL_0035
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 microRNA-34a Attenuates Paclitaxel Resistance in Prostate Cancer Cells via Direct Suppression of JAG1/Notch1 Axis.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [147]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Prostate cancer [ICD-11: 2C82.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Inhibition hsa05200
In Vitro Model PC3 cells Prostate Homo sapiens (Human) CVCL_0035
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Trypan blue dye exclusion assay
Mechanism Description SIRT1 plays crucial roles in various cellular processes including cell survival under genotoxic and oxidative stresses. Bcl2I is an anti-apoptotic factor. In PC3PR cells, reduced expression of miR-34a confers paclitaxel resistance via up-regulating SIRT1 and Bcl2 expression.
Key Molecule: NAD-dependent protein deacetylase sirtuin-1 (SIRT1) [147]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Prostate cancer [ICD-11: 2C82.0]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Inhibition hsa05200
In Vitro Model PC3 cells Prostate Homo sapiens (Human) CVCL_0035
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Trypan blue dye exclusion assay
Mechanism Description SIRT1 plays crucial roles in various cellular processes including cell survival under genotoxic and oxidative stresses. Bcl2I is an anti-apoptotic factor. In PC3PR cells, reduced expression of miR-34a confers paclitaxel resistance via up-regulating SIRT1 and Bcl2 expression.
Kidney cancer [ICD-11: 2C90]
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-21 [148]
Molecule Alteration Expression
Down-regulation
Sensitive Disease Renal carcinoma [ICD-11: 2C90.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model 786-O cells Kidney Homo sapiens (Human) CVCL_1051
ACHN cells Pleural effusion Homo sapiens (Human) CVCL_1067
HK-2 cells Kidney Homo sapiens (Human) CVCL_0302
RCC10 cells Kidney Homo sapiens (Human) CVCL_6265
RCC4 cells Kidney Homo sapiens (Human) CVCL_0498
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
Celltiter96 Aqueous Non Radioactive Cell Proliferation Assay
Mechanism Description Tumor suppressor genes like PTEN, PDCD4 and TIMP3, are target genes of miR21. PTEN is a potent inhibitor of PI3k/Akt pathway, as well as a direct target of miR21.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Programmed cell death protein 4 (PDCD4) [148]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Renal carcinoma [ICD-11: 2C90.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model 786-O cells Kidney Homo sapiens (Human) CVCL_1051
ACHN cells Pleural effusion Homo sapiens (Human) CVCL_1067
HK-2 cells Kidney Homo sapiens (Human) CVCL_0302
RCC10 cells Kidney Homo sapiens (Human) CVCL_6265
RCC4 cells Kidney Homo sapiens (Human) CVCL_0498
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Celltiter96 Aqueous Non Radioactive Cell Proliferation Assay
Mechanism Description Tumor suppressor genes like PTEN, PDCD4 and TIMP3, are target genes of miR21. PTEN is a potent inhibitor of PI3k/Akt pathway, as well as a direct target of miR21.
Key Molecule: Phosphatase and tensin homolog (PTEN) [148]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Renal carcinoma [ICD-11: 2C90.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model 786-O cells Kidney Homo sapiens (Human) CVCL_1051
ACHN cells Pleural effusion Homo sapiens (Human) CVCL_1067
HK-2 cells Kidney Homo sapiens (Human) CVCL_0302
RCC10 cells Kidney Homo sapiens (Human) CVCL_6265
RCC4 cells Kidney Homo sapiens (Human) CVCL_0498
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Celltiter96 Aqueous Non Radioactive Cell Proliferation Assay
Mechanism Description Tumor suppressor genes like PTEN, PDCD4 and TIMP3, are target genes of miR21. PTEN is a potent inhibitor of PI3k/Akt pathway, as well as a direct target of miR21.
Key Molecule: Metalloproteinase inhibitor 3 (TIMP3) [148]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Renal carcinoma [ICD-11: 2C90.2]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model 786-O cells Kidney Homo sapiens (Human) CVCL_1051
ACHN cells Pleural effusion Homo sapiens (Human) CVCL_1067
HK-2 cells Kidney Homo sapiens (Human) CVCL_0302
RCC10 cells Kidney Homo sapiens (Human) CVCL_6265
RCC4 cells Kidney Homo sapiens (Human) CVCL_0498
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Celltiter96 Aqueous Non Radioactive Cell Proliferation Assay
Mechanism Description Tumor suppressor genes like PTEN, PDCD4 and TIMP3, are target genes of miR21. PTEN is a potent inhibitor of PI3k/Akt pathway, as well as a direct target of miR21.
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-34b-3p [149]
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
Notch/PkC/Ca++ signaling pathway Inhibition hsa04330
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
EJ cells Bladder Homo sapiens (Human) CVCL_UI82
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR-34b-3p Represses the Multidrug-Chemoresistance of Bladder Cancer Cells by Regulating the CCND2 and P2RY1 Genes.
Key Molecule: hsa-miR-22-3p [150]
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: hsa-miR-193a-3p [8], [151], [152]
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
DNA damage repair signaling pathway Inhibition hsa03410
DNA damage response/Oxidative stress signaling pathway Inhibition hsa04218
Myc/Max signaling pathway Inhibition hsa04218
NF-kappaB signaling pathway Inhibition hsa04064
Notch signaling pathway Activation hsa04330
Oxidative stress signaling pathway Regulation hsa00190
Oxidative stress signaling pathway Activation hsa00190
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
BIU87 cells Bladder Homo sapiens (Human) CVCL_6881
H-bc cells Bladder Homo sapiens (Human) CVCL_BT00
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-193a-3p promotes the BCa multi-drug resistance phenotype via its repression of the lysyl oxidase-like 4 (LOXL4) gene, a newly identified direct target of miR-193a-3p. The LOXL4 protein is an important member of the lysyl oxidase (an extracellular copper-dependent amine oxidase) family that catalyzes the first step of the crosslinks between collagens and elastin during the biogenesis of connective tissue and is frequently deregulated in cancer. The Oxidative stress (OS) pathway is the predominant pathway affected by miR-193a-3p via its repression of LOXL4 expression.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: G1/S-specific cyclin-D2 (CCND2) [149]
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
Notch/PkC/Ca++ signaling pathway Inhibition hsa04330
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
EJ cells Bladder Homo sapiens (Human) CVCL_UI82
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 miR-34b-3p Represses the Multidrug-Chemoresistance of Bladder Cancer Cells by Regulating the CCND2 and P2RY1 Genes.
Key Molecule: P2Y purinoceptor 1 (P2RY1) [149]
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
Notch/PkC/Ca++ signaling pathway Inhibition hsa04330
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
EJ cells Bladder Homo sapiens (Human) CVCL_UI82
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 miR-34b-3p Represses the Multidrug-Chemoresistance of Bladder Cancer Cells by Regulating the CCND2 and P2RY1 Genes.
Key Molecule: Neuroepithelial cell-transforming gene 1 protein (NET1) [150]
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.
Key Molecule: Homeobox protein Hox-C9 (HOXC9) [152]
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
DNA damage response/Oxidative stress signaling pathway Inhibition hsa04218
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
H-bc cells Bladder Homo sapiens (Human) CVCL_BT00
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-193a-3p promotes the multi-chemoresistance of bladder cancer by targeting the HOXC9 gene.
Key Molecule: Lysyl oxidase homolog 4 (LOXL4) [151]
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
Oxidative stress signaling pathway Regulation hsa00190
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
BIU87 cells Bladder Homo sapiens (Human) CVCL_6881
H-bc cells Bladder Homo sapiens (Human) CVCL_BT00
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-193a-3p promotes the BCa multi-drug resistance phenotype via its repression of the lysyl oxidase-like 4 (LOXL4) gene, a newly identified direct target of miR-193a-3p. The LOXL4 protein is an important member of the lysyl oxidase (an extracellular copper-dependent amine oxidase) family that catalyzes the first step of the crosslinks between collagens and elastin during the biogenesis of connective tissue and is frequently deregulated in cancer. The Oxidative stress (OS) pathway is the predominant pathway affected by miR-193a-3p via its repression of LOXL4 expression.
Key Molecule: Hypermethylated in cancer 2 protein (HIC2) [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 proliferation Inhibition hsa05200
DNA damage repair signaling pathway Inhibition hsa03410
Myc/Max signaling pathway Inhibition hsa04218
NF-kappaB signaling pathway Inhibition hsa04064
Notch signaling pathway Activation hsa04330
Oxidative stress signaling pathway Activation hsa00190
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
T24 cells Bladder Homo sapiens (Human) CVCL_0554
BIU87 cells Bladder Homo sapiens (Human) CVCL_6881
H-bc cells Bladder Homo sapiens (Human) CVCL_BT00
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The DNA methylation-regulated miR-193a-3p dictates the multi-chemoresistance of bladder cancer via repression of SRSF2/PLAU/HIC2 expression.
Key Molecule: Urokinase-type plasminogen activator (PLAU) [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 proliferation Inhibition hsa05200
DNA damage repair signaling pathway Inhibition hsa03410
Myc/Max signaling pathway Inhibition hsa04218
NF-kappaB signaling pathway Inhibition hsa04064
Notch signaling pathway Activation hsa04330
Oxidative stress signaling pathway Activation hsa00190
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
T24 cells Bladder Homo sapiens (Human) CVCL_0554
BIU87 cells Bladder Homo sapiens (Human) CVCL_6881
H-bc cells Bladder Homo sapiens (Human) CVCL_BT00
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The DNA methylation-regulated miR-193a-3p dictates the multi-chemoresistance of bladder cancer via repression of SRSF2/PLAU/HIC2 expression.
Key Molecule: Serine/arginine-rich splicing factor 2 (SRSF2) [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 proliferation Inhibition hsa05200
DNA damage repair signaling pathway Inhibition hsa03410
Myc/Max signaling pathway Inhibition hsa04218
NF-kappaB signaling pathway Inhibition hsa04064
Notch signaling pathway Activation hsa04330
Oxidative stress signaling pathway Activation hsa00190
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
T24 cells Bladder Homo sapiens (Human) CVCL_0554
BIU87 cells Bladder Homo sapiens (Human) CVCL_6881
H-bc cells Bladder Homo sapiens (Human) CVCL_BT00
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The DNA methylation-regulated miR-193a-3p dictates the multi-chemoresistance of bladder cancer via repression of SRSF2/PLAU/HIC2 expression.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-193a-3p [153]
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
DNA damage response signaling pathway Activation hsa04218
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
H-bc cells Bladder Homo sapiens (Human) CVCL_BT00
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Among the differentially expressed genes between the chemosensitive (5637) and chemoresistant (H-bc) bladder cancer cell lines, the expression level of the PSEN1 gene (presenilin 1), a key component of the Gamma-secretase, is negatively correlated with chemoresistance. A small interfering RNA mediated repression of the PSEN1 gene suppresses cell apoptosis and de-sensitizes 5637 cells, while overexpression of the presenilin 1 sensitizes H-bc cells to the drug-triggered cell death. As a direct target of microRNA-193a-3p that promotes the multi-chemoresistance of the bladder cancer cell, PSEN1 acts as an important executor for the microRNA-193a-3p's positive impact on the multi-chemoresistance of bladder cancer, probably via its activating effect on DNA damage response pathway. In addition to the mechanistic insights, the key players in this microRNA-193a-3p/PSEN1 axis are likely the diagnostic and/or therapeutic targets for an effective chemotherapy of bladder cancer.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Presenilin-1 (PSEN1) [153]
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
DNA damage response signaling pathway Activation hsa04218
In Vitro Model 5637 cells Bladder Homo sapiens (Human) CVCL_0126
H-bc cells Bladder Homo sapiens (Human) CVCL_BT00
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Among the differentially expressed genes between the chemosensitive (5637) and chemoresistant (H-bc) bladder cancer cell lines, the expression level of the PSEN1 gene (presenilin 1), a key component of the Gamma-secretase, is negatively correlated with chemoresistance. A small interfering RNA mediated repression of the PSEN1 gene suppresses cell apoptosis and de-sensitizes 5637 cells, while overexpression of the presenilin 1 sensitizes H-bc cells to the drug-triggered cell death. As a direct target of microRNA-193a-3p that promotes the multi-chemoresistance of the bladder cancer cell, PSEN1 acts as an important executor for the microRNA-193a-3p's positive impact on the multi-chemoresistance of bladder cancer, probably via its activating effect on DNA damage response pathway. In addition to the mechanistic insights, the key players in this microRNA-193a-3p/PSEN1 axis are likely the diagnostic and/or therapeutic targets for an effective chemotherapy of bladder cancer.
Head and neck cancer [ICD-11: 2D42]
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: Interleukin 2 receptor subunit alpha (IL2RA) [5]
Molecule Alteration Expression
Up-regulation
Resistant Disease Head and neck squamous cell carcinoma [ICD-11: 2D42.1]
Experimental Note Identified from the Human Clinical Data
In Vitro Model PCI-13 cells Ovary Homo sapiens (Human) CVCL_C182
Experiment for
Molecule Alteration
Western blotting assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description IL-2Ralpha-expressing cells were significantly more resistant to apoptosis induction by a tripeptidyl proteasome inhibitor (ALLN) and two chemotherapeutic drugs (VP-16 and taxol) than the control or IL-2Rgamma+ cells.IL-2Ralpha overexpression increases cell proliferation rate associated with increasing levels of cell cycle regulatory proteins.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-let-7d [154]
Molecule Alteration Expression
Up-regulation
Sensitive Disease Head and neck squamous cell carcinoma [ICD-11: 2D42.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
FaDu cells Pharynx Homo sapiens (Human) CVCL_1218
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Clonogenic assay; MTT assay
Mechanism Description The level of let-7d expression is an important factor for cell response to irradiation and chemotherapeutics. Overexpressed let-7d inhibited chemoresistance to cisplatin and paclitaxel in OSCC-ALDH1+ cells.
References
Ref 1 MicroRNA-654-5p suppresses ovarian cancer development impacting on MYC, WNT and AKT pathways. Oncogene. 2019 Aug;38(32):6035-6050. doi: 10.1038/s41388-019-0860-0. Epub 2019 Jul 5.
Ref 2 In vitro drug response and molecular markers associated with drug resistance in malignant gliomas .Clin Cancer Res. 2006 Aug 1;12(15):4523-32. doi: 10.1158/1078-0432.CCR-05-1830. 10.1158/1078-0432.CCR-05-1830
Ref 3 MicroRNA-155 regulates cell survival, growth, and chemosensitivity by targeting FOXO3a in breast cancer. J Biol Chem. 2010 Jun 4;285(23):17869-79. doi: 10.1074/jbc.M110.101055. Epub 2010 Apr 6.
Ref 4 miR-375 is upregulated in acquired paclitaxel resistance in cervical cancer. Br J Cancer. 2013 Jul 9;109(1):92-9. doi: 10.1038/bjc.2013.308. Epub 2013 Jun 18.
Ref 5 Overexpression of interleukin-2 receptor alpha in a human squamous cell carcinoma of the head and neck cell line is associated with increased proliferation, drug resistance, and transforming ability .J Cell Biochem. 2003 Jul 1;89(4):824-36. doi: 10.1002/jcb.10557. 10.1002/jcb.10557
Ref 6 Let-7 modulates acquired resistance of ovarian cancer to Taxanes via IMP-1-mediated stabilization of multidrug resistance 1. Int J Cancer. 2012 Apr 15;130(8):1787-97. doi: 10.1002/ijc.26190. Epub 2011 Aug 16.
Ref 7 MiR-199a suppresses prostate cancer paclitaxel resistance by targeting YES1. World J Urol. 2018 Mar;36(3):357-365. doi: 10.1007/s00345-017-2143-0. Epub 2017 Dec 4.
Ref 8 The DNA methylation-regulated miR-193a-3p dictates the multi-chemoresistance of bladder cancer via repression of SRSF2/PLAU/HIC2 expression. Cell Death Dis. 2014 Sep 4;5(9):e1402. doi: 10.1038/cddis.2014.367.
Ref 9 Let-7a microRNA suppresses therapeutics-induced cancer cell death by targeting caspase-3. Apoptosis. 2008 Oct;13(10):1215-22. doi: 10.1007/s10495-008-0256-z.
Ref 10 LncRNA PVT1 epigenetically silences miR-195 and modulates EMT and chemoresistance in cervical cancer cells. J Drug Target. 2017 Aug;25(7):637-644. doi: 10.1080/1061186X.2017.1307379. Epub 2017 Apr 3.
Ref 11 Effects of continuous exposure to digoxin on MDR1 function and expression in Caco-2 cells. J Pharm Pharmacol. 2003 May;55(5):675-81. doi: 10.1211/002235703765344595.
Ref 12 Long non-coding RNA LINC00672 contributes to p53 protein-mediated gene suppression and promotes endometrial cancer chemosensitivity. J Biol Chem. 2017 Apr 7;292(14):5801-5813. doi: 10.1074/jbc.M116.758508. Epub 2017 Feb 23.
Ref 13 MicroRNA-520g promotes epithelial ovarian cancer progression and chemoresistance via DAPK2 repression. Oncotarget. 2016 May 3;7(18):26516-34. doi: 10.18632/oncotarget.8530.
Ref 14 miR-193a-3p regulation of chemoradiation resistance in oesophageal cancer cells via the PSEN1 gene. Gene. 2016 Apr 1;579(2):139-45. doi: 10.1016/j.gene.2015.12.060. Epub 2015 Dec 29.
Ref 15 miR-590-5p regulates gastric cancer cell growth and chemosensitivity through RECK and the AKT/ERK pathway. Onco Targets Ther. 2016 Oct 3;9:6009-6019. doi: 10.2147/OTT.S110923. eCollection 2016.
Ref 16 miR-135a contributes to paclitaxel resistance in tumor cells both in vitro and in vivo. Oncogene. 2011 Oct 27;30(43):4386-98. doi: 10.1038/onc.2011.148. Epub 2011 May 9.
Ref 17 miR-326-histone deacetylase-3 feedback loop regulates the invasion and tumorigenic and angiogenic response to anti-cancer drugs. J Biol Chem. 2014 Oct 3;289(40):28019-39. doi: 10.1074/jbc.M114.578229. Epub 2014 Aug 19.
Ref 18 LncRNA CCAT1 modulates the sensitivity of paclitaxel in nasopharynx cancers cells via miR-181a/CPEB2 axis. Cell Cycle. 2017 Apr 18;16(8):795-801. doi: 10.1080/15384101.2017.1301334. Epub 2017 Mar 30.
Ref 19 A novel mitosis-associated lncRNA, MA-linc1, is required for cell cycle progression and sensitizes cancer cells to Paclitaxel. Oncotarget. 2015 Sep 29;6(29):27880-90. doi: 10.18632/oncotarget.4944.
Ref 20 Role of microRNAs in drug-resistant ovarian cancer cells. Gynecol Oncol. 2008 Dec;111(3):478-86. doi: 10.1016/j.ygyno.2008.08.017. Epub 2008 Sep 26.
Ref 21 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 22 Micro-RNA149 confers taxane resistance to malignant mesothelioma cells via regulation of P-glycoprotein expression. Cancer Biol Ther. 2018 Mar 4;19(3):181-187. doi: 10.1080/15384047.2017.1415677. Epub 2018 Jan 15.
Ref 23 Pre-clinical activity of taxol in non-seminomatous germ cell tumor cell lines and nude mouse xenografts .Invest New Drugs. 1997;15(2):91-8. doi: 10.1023/a:1005852521656. 10.1023/a:1005852521656
Ref 24 miR-34a attenuates glioma cells progression and chemoresistance via targeting PD-L1. Biotechnol Lett. 2017 Oct;39(10):1485-1492. doi: 10.1007/s10529-017-2397-z. Epub 2017 Jul 18.
Ref 25 MicroRNA-21 inhibitor sensitizes human glioblastoma cells U251 (PTEN-mutant) and LN229 (PTEN-wild type) to taxol. BMC Cancer. 2010 Jan 31;10:27. doi: 10.1186/1471-2407-10-27.
Ref 26 miR-495 sensitizes MDR cancer cells to the combination of doxorubicin and taxol by inhibiting MDR1 expression. J Cell Mol Med. 2017 Sep;21(9):1929-1943. doi: 10.1111/jcmm.13114. Epub 2017 Apr 14.
Ref 27 Overexpression of miR-422a inhibits cell proliferation and invasion, and enhances chemosensitivity in osteosarcoma cells. Oncol Rep. 2016 Dec;36(6):3371-3378. doi: 10.3892/or.2016.5182. Epub 2016 Oct 19.
Ref 28 LINC01118 Modulates Paclitaxel Resistance of Epithelial Ovarian Cancer by Regulating miR-134/ABCC1. Med Sci Monit. 2018 Dec 6;24:8831-8839. doi: 10.12659/MSM.910932.
Ref 29 miR-146a Inhibits Proliferation and Enhances Chemosensitivity in Epithelial Ovarian Cancer via Reduction of SOD2. Oncol Res. 2016;23(6):275-82. doi: 10.3727/096504016X14562725373798.
Ref 30 Targeted regulationof STAT3 by miR-29a in mediating Taxol resistance of nasopharyngeal carcinoma cell line CNE-1. Cancer Biomark. 2018;22(4):641-648. doi: 10.3233/CBM-170964.
Ref 31 miR-29c regulates resistance to paclitaxel in nasopharyngeal cancer by targeting ITGB1. Exp Cell Res. 2019 May 1;378(1):1-10. doi: 10.1016/j.yexcr.2019.02.012. Epub 2019 Feb 16.
Ref 32 MiR-1204 sensitizes nasopharyngeal carcinoma cells to paclitaxel both in vitro and in vivo. Cancer Biol Ther. 2015;16(2):261-7. doi: 10.1080/15384047.2014.1001287.
Ref 33 MiR-634 sensitizes nasopharyngeal carcinoma cells to paclitaxel and inhibits cell growth both in vitro and in vivo. Int J Clin Exp Pathol. 2014 Sep 15;7(10):6784-91. eCollection 2014.
Ref 34 Extreme low dose of 5-fluorouracil reverses MDR in cancer by sensitizing cancer associated fibroblasts and down-regulating P-gp. PLoS One. 2017 Jun 29;12(6):e0180023. doi: 10.1371/journal.pone.0180023. eCollection 2017.
Ref 35 Overexpression of Lin28 Decreases the Chemosensitivity of Gastric Cancer Cells to Oxaliplatin, Paclitaxel, Doxorubicin, and Fluorouracil in Part via microRNA-107. PLoS One. 2015 Dec 4;10(12):e0143716. doi: 10.1371/journal.pone.0143716. eCollection 2015.
Ref 36 Regulation of microtubule-associated protein tau (MAPT) by miR-34c-5p determines the chemosensitivity of gastric cancer to paclitaxel. Cancer Chemother Pharmacol. 2013 May;71(5):1159-71. doi: 10.1007/s00280-013-2108-y. Epub 2013 Feb 20.
Ref 37 Paclitaxel resistant gastric cancer MGC 803 cells promote epithelial to mesenchymal transition and chemoresistance in paclitaxel sensitive cells via exosomal delivery of miR 155 5p. Int J Oncol. 2019 Jan;54(1):326-338. doi: 10.3892/ijo.2018.4601. Epub 2018 Oct 22.
Ref 38 Silencing of lncRNA ZFAS1 inhibits malignancies by blocking Wnt/Beta-catenin signaling in gastric cancer cells. Biosci Biotechnol Biochem. 2018 Mar;82(3):456-465. doi: 10.1080/09168451.2018.1431518. Epub 2018 Feb 9.
Ref 39 MiR-125a/b regulates the activation of cancer stem cells in paclitaxel-resistant colon cancer. Cancer Invest. 2013 Jan;31(1):17-23. doi: 10.3109/07357907.2012.743557.
Ref 40 Overexpression of miR-22 reverses paclitaxel-induced chemoresistance through activation of PTEN signaling in p53-mutated colon cancer cells. Mol Cell Biochem. 2011 Nov;357(1-2):31-8. doi: 10.1007/s11010-011-0872-8. Epub 2011 May 19.
Ref 41 miR-203 reverses chemoresistance in p53-mutated colon cancer cells through downregulation of Akt2 expression. Cancer Lett. 2011 May 1;304(1):52-9. doi: 10.1016/j.canlet.2011.02.003. Epub 2011 Feb 26.
Ref 42 LINC00473 promotes the Taxol resistance via miR-15a in colorectal cancer. Biosci Rep. 2018 Sep 20;38(5):BSR20180790. doi: 10.1042/BSR20180790. Print 2018 Oct 31.
Ref 43 Inhibition of P-glycoprotein by D-alpha-tocopheryl polyethylene glycol 1000 succinate (TPGS). Pharm Res. 1999 Oct;16(10):1550-6. doi: 10.1023/a:1015000503629.
Ref 44 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 45 Inhibition of microRNA-16 facilitates the paclitaxel resistance by targeting IKBKB via NF-kB signaling pathway in hepatocellular carcinoma. Biochem Biophys Res Commun. 2018 Sep 5;503(2):1035-1041. doi: 10.1016/j.bbrc.2018.06.113. Epub 2018 Aug 2.
Ref 46 Bioinformatic identification of IGF1 as a hub gene in hepatocellular carcinoma (HCC) and in-vitro analysis of the chemosensitizing effect of miR-379 via suppressing the IGF1/IGF1R signaling pathway. Eur Rev Med Pharmacol Sci. 2016 Dec;20(24):5098-5106.
Ref 47 miR-335 Targets SIAH2 and Confers Sensitivity to Anti-Cancer Drugs by Increasing the Expression of HDAC3. Mol Cells. 2015 Jun;38(6):562-72. doi: 10.14348/molcells.2015.0051. Epub 2015 May 22.
Ref 48 MiR-223 modulates multidrug resistance via downregulation of ABCB1 in hepatocellular carcinoma cells. Exp Biol Med (Maywood). 2013 Sep;238(9):1024-32. doi: 10.1177/1535370213497321. Epub 2013 Aug 7.
Ref 49 Overexpressed miR-128a enhances chemoradiotherapy to laryngeal cancer cells and its correlation with BMI1. Future Oncol. 2018 Mar;14(7):611-620. doi: 10.2217/fon-2017-0542. Epub 2017 Nov 30.
Ref 50 LncRNA MALAT1 Depressed Chemo-Sensitivity of NSCLC Cells through Directly Functioning on miR-197-3p/p120 Catenin Axis. Mol Cells. 2019 Mar 31;42(3):270-283. doi: 10.14348/molcells.2019.2364. Epub 2019 Feb 19.
Ref 51 Overexpression of HOTTIP promotes proliferation and drug resistance of lung adenocarcinoma by regulating AKT signaling pathway. Eur Rev Med Pharmacol Sci. 2017 Dec;21(24):5683-5690. doi: 10.26355/eurrev_201712_14013.
Ref 52 The long noncoding RNA ANRIL acts as an oncogene and contributes to paclitaxel resistance of lung adenocarcinoma A549 cells. Oncotarget. 2017 Jun 13;8(24):39177-39184. doi: 10.18632/oncotarget.16640.
Ref 53 MicroRNA-181a regulates epithelial-mesenchymal transition by targeting PTEN in drug-resistant lung adenocarcinoma cells. Int J Oncol. 2015 Oct;47(4):1379-92. doi: 10.3892/ijo.2015.3144. Epub 2015 Aug 31.
Ref 54 miR-17-5p downregulation contributes to paclitaxel resistance of lung cancer cells through altering beclin1 expression. PLoS One. 2014 Apr 22;9(4):e95716. doi: 10.1371/journal.pone.0095716. eCollection 2014.
Ref 55 miR-34c may protect lung cancer cells from paclitaxel-induced apoptosis. Oncogene. 2013 Jan 17;32(3):341-51. doi: 10.1038/onc.2012.51. Epub 2012 Feb 27.
Ref 56 MicroRNA-128 suppresses paclitaxel-resistant lung cancer by inhibiting MUC1-C and BMI-1 in cancer stem cells. Oncotarget. 2017 Nov 30;8(66):110540-110551. doi: 10.18632/oncotarget.22818. eCollection 2017 Dec 15.
Ref 57 A miRNA-200c/cathepsin L feedback loop determines paclitaxel resistance in human lung cancer A549 cells in vitro through regulating epithelial-mesenchymal transition. Acta Pharmacol Sin. 2018 Jun;39(6):1034-1047. doi: 10.1038/aps.2017.164. Epub 2017 Dec 7.
Ref 58 MiRNA-107 enhances chemosensitivity to paclitaxel by targeting antiapoptotic factor Bcl-w in non small cell lung cancer. Am J Cancer Res. 2017 Sep 1;7(9):1863-1873. eCollection 2017.
Ref 59 miR-30a-5p enhances paclitaxel sensitivity in non-small cell lung cancer through targeting BCL-2 expression. J Mol Med (Berl). 2017 Aug;95(8):861-871. doi: 10.1007/s00109-017-1539-z. Epub 2017 May 9.
Ref 60 miR-186 regulates chemo-sensitivity to paclitaxel via targeting MAPT in non-small cell lung cancer (NSCLC). Mol Biosyst. 2016 Oct 18;12(11):3417-3424. doi: 10.1039/c6mb00576d.
Ref 61 NEAT1 mediates paclitaxel-resistance of non-small cell of lung cancer through activation of Akt/mTOR signalling pathway. J Drug Target. 2019 Dec;27(10):1061-1067. doi: 10.1080/1061186X.2019.1585437. Epub 2019 Mar 20.
Ref 62 Paclitaxel promotes lung cancer cell apoptosis via MEG3-P53 pathway activation. Biochem Biophys Res Commun. 2018 Sep 26;504(1):123-128. doi: 10.1016/j.bbrc.2018.08.142. Epub 2018 Aug 31.
Ref 63 Knockdown of miR 935 increases paclitaxel sensitivity via regulation of SOX7 in non small cell lung cancer. Mol Med Rep. 2018 Sep;18(3):3397-3402. doi: 10.3892/mmr.2018.9330. Epub 2018 Jul 27.
Ref 64 Loss of MiR-424-3p, not miR-424-5p, confers chemoresistance through targeting YAP1 in non-small cell lung cancer. Mol Carcinog. 2017 Mar;56(3):821-832. doi: 10.1002/mc.22536. Epub 2016 Aug 26.
Ref 65 MicroRNA-137 inhibits tumor growth and sensitizes chemosensitivity to paclitaxel and cisplatin in lung cancer. Oncotarget. 2016 Apr 12;7(15):20728-42. doi: 10.18632/oncotarget.8011.
Ref 66 MiR-16 targets Bcl-2 in paclitaxel-resistant lung cancer cells and overexpression of miR-16 along with miR-17 causes unprecedented sensitivity by simultaneously modulating autophagy and apoptosis. Cell Signal. 2015 Feb;27(2):189-203. doi: 10.1016/j.cellsig.2014.11.023. Epub 2014 Nov 27.
Ref 67 MicroRNA-7 sensitizes non-small cell lung cancer cells to paclitaxel. Oncol Lett. 2014 Nov;8(5):2193-2200. doi: 10.3892/ol.2014.2500. Epub 2014 Sep 4.
Ref 68 miR-337-3p and its targets STAT3 and RAP1A modulate taxane sensitivity in non-small cell lung cancers. PLoS One. 2012;7(6):e39167. doi: 10.1371/journal.pone.0039167. Epub 2012 Jun 18.
Ref 69 miRNA-296-3p modulates chemosensitivity of lung cancer cells by targeting CX3CR1. Am J Transl Res. 2016 Apr 15;8(4):1848-56. eCollection 2016.
Ref 70 Up-regulation of miR-125b reverses epithelial-mesenchymal transition in paclitaxel-resistant lung cancer cells. Biol Chem. 2015 Aug 20:/j/bchm.just-accepted/hsz-2015-0153/hsz-2015-0153.xml. doi: 10.1515/hsz-2015-0153. Online ahead of print.
Ref 71 miR-107 Enhances the Sensitivity of Breast Cancer Cells to Paclitaxel. Open Med (Wars). 2019 Jun 1;14:456-466. doi: 10.1515/med-2019-0049. eCollection 2019.
Ref 72 LINC00511 knockdown enhances paclitaxel cytotoxicity in breast cancer via regulating miR-29c/CDK6 axis. Life Sci. 2019 Jul 1;228:135-144. doi: 10.1016/j.lfs.2019.04.063. Epub 2019 May 7.
Ref 73 LncRNA H19 is a major mediator of doxorubicin chemoresistance in breast cancer cells through a cullin4A-MDR1 pathway. Oncotarget. 2017 Sep 21;8(54):91990-92003. doi: 10.18632/oncotarget.21121. eCollection 2017 Nov 3.
Ref 74 LncRNA H19 confers chemoresistance in ERAlpha-positive breast cancer through epigenetic silencing of the pro-apoptotic gene BIK. Oncotarget. 2016 Dec 6;7(49):81452-81462. doi: 10.18632/oncotarget.13263.
Ref 75 Decreased expression of microRNA-17 and microRNA-20b promotes breast cancer resistance to taxol therapy by upregulation of NCOA3. Cell Death Dis. 2016 Nov 10;7(11):e2463. doi: 10.1038/cddis.2016.367.
Ref 76 MiR-155-3p acts as a tumor suppressor and reverses paclitaxel resistance via negative regulation of MYD88 in human breast cancer. Gene. 2019 Jun 5;700:85-95. doi: 10.1016/j.gene.2019.02.066. Epub 2019 Mar 13.
Ref 77 miR-1207-5p regulates the sensitivity of triple-negative breast cancer cells to Taxol treatment via the suppression of LZTS1 expression. Oncol Lett. 2019 Jan;17(1):990-998. doi: 10.3892/ol.2018.9687. Epub 2018 Nov 12.
Ref 78 Downregulation of miR 200c 3p contributes to the resistance of breast cancer cells to paclitaxel by targeting SOX2. Oncol Rep. 2018 Dec;40(6):3821-3829. doi: 10.3892/or.2018.6735. Epub 2018 Sep 26.
Ref 79 MiR-18a upregulation decreases Dicer expression and confers paclitaxel resistance in triple negative breast cancer. Eur Rev Med Pharmacol Sci. 2016 Jun;20(11):2201-8.
Ref 80 MiR-106b~25 cluster regulates multidrug resistance in an ABC transporter-independent manner via downregulation of EP300. Oncol Rep. 2016 Feb;35(2):1170-8. doi: 10.3892/or.2015.4412. Epub 2015 Nov 12.
Ref 81 [Effects of miRNA-21 on paclitaxel-resistance in human breast cancer cells]. Zhejiang Da Xue Xue Bao Yi Xue Ban. 2015 Jul;44(4):400-9. doi: 10.3785/j.issn.1008-9292.2015.07.09.
Ref 82 miR-520h is crucial for DAPK2 regulation and breast cancer progression. Oncogene. 2016 Mar 3;35(9):1134-42. doi: 10.1038/onc.2015.168. Epub 2015 May 18.
Ref 83 Methylation-regulated miR-149 modulates chemoresistance by targeting GlcNAc N-deacetylase/N-sulfotransferase-1 in human breast cancer. FEBS J. 2014 Oct;281(20):4718-30. doi: 10.1111/febs.13012. Epub 2014 Sep 30.
Ref 84 miR-137 restoration sensitizes multidrug-resistant MCF-7/ADM cells to anticancer agents by targeting YB-1. Acta Biochim Biophys Sin (Shanghai). 2013 Feb;45(2):80-6. doi: 10.1093/abbs/gms099. Epub 2012 Nov 23.
Ref 85 MicroRNA-125b confers the resistance of breast cancer cells to paclitaxel through suppression of pro-apoptotic Bcl-2 antagonist killer 1 (Bak1) expression. J Biol Chem. 2010 Jul 9;285(28):21496-507. doi: 10.1074/jbc.M109.083337. Epub 2010 May 11.
Ref 86 Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013 May 2;497(7447):108-12. doi: 10.1038/nature12065. Epub 2013 Apr 7.
Ref 87 Exosome-mediated breast cancer chemoresistance via miR-155 transfer. Sci Rep. 2018 Jan 16;8(1):829. doi: 10.1038/s41598-018-19339-5.
Ref 88 Ai-lncRNA EGOT enhancing autophagy sensitizes paclitaxel cytotoxicity via upregulation of ITPR1 expression by RNA-RNA and RNA-protein interactions in human cancer. Mol Cancer. 2019 Apr 18;18(1):89. doi: 10.1186/s12943-019-1017-z.
Ref 89 Long non-coding RNA LINC00968 attenuates drug resistance of breast cancer cells through inhibiting the Wnt2/Beta-catenin signaling pathway by regulating WNT2. J Exp Clin Cancer Res. 2019 Feb 21;38(1):94. doi: 10.1186/s13046-019-1100-8.
Ref 90 PIWI-interacting RNA-36712 restrains breast cancer progression and chemoresistance by interaction with SEPW1 pseudogene SEPW1P RNA. Mol Cancer. 2019 Jan 12;18(1):9. doi: 10.1186/s12943-019-0940-3.
Ref 91 [Effects of lncRNA RP11-770J1.3 and TMEM25 expression on paclitaxel resistance in human breast cancer cells]. Zhejiang Da Xue Xue Bao Yi Xue Ban. 2017 Jul 25;46(4):364-370. doi: 10.3785/j.issn.1008-9292.2017.08.04.
Ref 92 MiR-30e inhibits tumor growth and chemoresistance via targeting IRS1 in Breast Cancer. Sci Rep. 2017 Nov 21;7(1):15929. doi: 10.1038/s41598-017-16175-x.
Ref 93 Involvement of miR-451 in resistance to paclitaxel by regulating YWHAZ in breast cancer. Cell Death Dis. 2017 Oct 5;8(10):e3071. doi: 10.1038/cddis.2017.460.
Ref 94 Downregulation of eIF4G by microRNA-503 enhances drug sensitivity of MCF-7/ADR cells through suppressing the expression of ABC transport proteins. Oncol Lett. 2017 Jun;13(6):4785-4793. doi: 10.3892/ol.2017.6049. Epub 2017 Apr 19.
Ref 95 Upregulation of microRNA-143 reverses drug resistance in human breast cancer cells via inhibition of cytokine-induced apoptosis inhibitor 1. Oncol Lett. 2017 Jun;13(6):4695-4700. doi: 10.3892/ol.2017.6078. Epub 2017 Apr 24.
Ref 96 STAT3 is required for MiR-17-5p-mediated sensitization to chemotherapy-induced apoptosis in breast cancer cells. Oncotarget. 2017 Feb 28;8(9):15763-15774. doi: 10.18632/oncotarget.15000.
Ref 97 Overexpression of microRNA-24 increases the sensitivity to paclitaxel in drug-resistant breast carcinoma cell lines via targeting ABCB9. Oncol Lett. 2016 Nov;12(5):3905-3911. doi: 10.3892/ol.2016.5139. Epub 2016 Sep 15.
Ref 98 MiR-129-5p is downregulated in breast cancer cells partly due to promoter H3K27m3 modification and regulates epithelial-mesenchymal transition and multi-drug resistance. Eur Rev Med Pharmacol Sci. 2016 Oct;20(20):4257-4265.
Ref 99 MicroRNA-22 Suppresses Breast Cancer Cell Growth and Increases Paclitaxel Sensitivity by Targeting NRAS. Technol Cancer Res Treat. 2018 Jan 1;17:1533033818809997. doi: 10.1177/1533033818809997.
Ref 100 Knockdown of lncRNA H19 restores chemo-sensitivity in paclitaxel-resistant triple-negative breast cancer through triggering apoptosis and regulating Akt signaling pathway. Toxicol Appl Pharmacol. 2018 Nov 15;359:55-61. doi: 10.1016/j.taap.2018.09.018. Epub 2018 Sep 20.
Ref 101 Linc00518 Contributes to Multidrug Resistance Through Regulating the MiR-199a/MRP1 Axis in Breast Cancer. Cell Physiol Biochem. 2018;48(1):16-28. doi: 10.1159/000491659. Epub 2018 Jul 12.
Ref 102 miR-485-5p suppresses breast cancer progression and chemosensitivity by targeting survivin. Biochem Biophys Res Commun. 2018 Jun 18;501(1):48-54. doi: 10.1016/j.bbrc.2018.04.129. Epub 2018 Apr 22.
Ref 103 miR-27b-3p inhibits proliferation and potentially reverses multi-chemoresistance by targeting CBLB/GRB2 in breast cancer cells. Cell Death Dis. 2018 Feb 7;9(2):188. doi: 10.1038/s41419-017-0211-4.
Ref 104 MiR-375 is epigenetically downregulated due to promoter methylation and modulates multi-drug resistance in breast cancer cells via targeting YBX1. Eur Rev Med Pharmacol Sci. 2016 Jul;20(15):3223-9.
Ref 105 MicroRNA-16 sensitizes breast cancer cells to paclitaxel through suppression of IKBKB expression. Oncotarget. 2016 Apr 26;7(17):23668-83. doi: 10.18632/oncotarget.8056.
Ref 106 MicroRNA 100 sensitizes luminal A breast cancer cells to paclitaxel treatment in part by targeting mTOR. Oncotarget. 2016 Feb 2;7(5):5702-14. doi: 10.18632/oncotarget.6790.
Ref 107 MicroRNA-34a suppresses the breast cancer stem cell-like characteristics by downregulating Notch1 pathway. Cancer Sci. 2015 Jun;106(6):700-8. doi: 10.1111/cas.12656.
Ref 108 A methylation-based regulatory network for microRNA 320a in chemoresistant breast cancer. Mol Pharmacol. 2014 Nov;86(5):536-47. doi: 10.1124/mol.114.092759. Epub 2014 Aug 26.
Ref 109 [Effect of miR-342-3p on chemotherapy sensitivity in triple-negative breast cancer]. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2014 May;39(5):488-95. doi: 10.3969/j.issn.1672-7347.2014.05.009.
Ref 110 miR-223 is a coordinator of breast cancer progression as revealed by bioinformatics predictions. PLoS One. 2014 Jan 6;9(1):e84859. doi: 10.1371/journal.pone.0084859. eCollection 2014.
Ref 111 MiR-26a inhibits proliferation and migration of breast cancer through repression of MCL-1. PLoS One. 2013 Jun 4;8(6):e65138. doi: 10.1371/journal.pone.0065138. Print 2013.
Ref 112 Downregulation of miR-21 enhances chemotherapeutic effect of taxol in breast carcinoma cells. Technol Cancer Res Treat. 2010 Feb;9(1):77-86. doi: 10.1177/153303461000900109.
Ref 113 MicroRNA-30c inhibits human breast tumour chemotherapy resistance by regulating TWF1 and IL-11. Nat Commun. 2013;4:1393. doi: 10.1038/ncomms2393.
Ref 114 microRNA 490-3P enhances the drug-resistance of human ovarian cancer cells. J Ovarian Res. 2014 Aug 31;7:84. doi: 10.1186/s13048-014-0084-4.
Ref 115 LncRNA NEAT1 contributes to paclitaxel resistance of ovarian cancer cells by regulating ZEB1 expression via miR-194. Onco Targets Ther. 2017 Nov 10;10:5377-5390. doi: 10.2147/OTT.S147586. eCollection 2017.
Ref 116 MiR-1307 promotes ovarian cancer cell chemoresistance by targeting the ING5 expression. J Ovarian Res. 2017 Jan 11;10(1):1. doi: 10.1186/s13048-016-0301-4.
Ref 117 Downregulation of miR-194-5p induces paclitaxel resistance in ovarian cancer cells by altering MDM2 expression. Oncotarget. 2019 Jan 18;10(6):673-683. doi: 10.18632/oncotarget.26586. eCollection 2019 Jan 18.
Ref 118 UCA1 confers paclitaxel resistance to ovarian cancer through miR-129/ABCB1 axis. Biochem Biophys Res Commun. 2018 Jul 2;501(4):1034-1040. doi: 10.1016/j.bbrc.2018.05.104. Epub 2018 May 19.
Ref 119 Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat Commun. 2016 Mar 29;7:11150. doi: 10.1038/ncomms11150.
Ref 120 MicroRNA-17-5p induces drug resistance and invasion of ovarian carcinoma cells by targeting PTEN signaling. J Biol Res (Thessalon). 2015 Oct 22;22:12. doi: 10.1186/s40709-015-0035-2. eCollection 2015 Dec.
Ref 121 MiRNA-149 modulates chemosensitivity of ovarian cancer A2780 cells to paclitaxel by targeting MyD88. J Ovarian Res. 2015 Jul 30;8:48. doi: 10.1186/s13048-015-0178-7.
Ref 122 Dysregulation of miR-106a and miR-591 confers paclitaxel resistance to ovarian cancer. Br J Cancer. 2013 Jul 23;109(2):452-61. doi: 10.1038/bjc.2013.305. Epub 2013 Jun 27.
Ref 123 MiR-27a modulates MDR1/P-glycoprotein expression by targeting HIPK2 in human ovarian cancer cells. Gynecol Oncol. 2010 Oct;119(1):125-30. doi: 10.1016/j.ygyno.2010.06.004. Epub 2010 Jul 10.
Ref 124 MiR-181a upregulation is associated with epithelial-to-mesenchymal transition (EMT) and multidrug resistance (MDR) of ovarian cancer cells. Eur Rev Med Pharmacol Sci. 2016 May;20(10):2004-10.
Ref 125 The miR-200 family differentially regulates sensitivity to paclitaxel and carboplatin in human ovarian carcinoma OVCAR-3 and MES-OV cells. Mol Oncol. 2015 Oct;9(8):1678-93. doi: 10.1016/j.molonc.2015.04.015. Epub 2015 May 16.
Ref 126 MicroRNA-133b targets glutathione S-transferase Pi expression to increase ovarian cancer cell sensitivity to chemotherapy drugs. Drug Des Devel Ther. 2015 Sep 16;9:5225-35. doi: 10.2147/DDDT.S87526. eCollection 2015.
Ref 127 MicroRNA-136 inhibits cancer stem cell activity and enhances the anti-tumor effect of paclitaxel against chemoresistant ovarian cancer cells by targeting Notch3. Cancer Lett. 2017 Feb 1;386:168-178. doi: 10.1016/j.canlet.2016.11.017. Epub 2016 Nov 22.
Ref 128 Up-regulation of miR-383-5p suppresses proliferation and enhances chemosensitivity in ovarian cancer cells by targeting TRIM27. Biomed Pharmacother. 2019 Jan;109:595-601. doi: 10.1016/j.biopha.2018.10.148. Epub 2018 Nov 3.
Ref 129 MicroRNA-630 inhibitor sensitizes chemoresistant ovarian cancer to chemotherapy by enhancing apoptosis. Biochem Biophys Res Commun. 2018 Mar 4;497(2):513-520. doi: 10.1016/j.bbrc.2018.02.062. Epub 2018 Feb 13.
Ref 130 MicroRNA-186 induces sensitivity of ovarian cancer cells to paclitaxel and cisplatin by targeting ABCB1. J Ovarian Res. 2015 Dec 2;8:80. doi: 10.1186/s13048-015-0207-6.
Ref 131 Involvement of miR-29b signaling in the sensitivity to chemotherapy in patients with ovarian carcinoma. Hum Pathol. 2014 Jun;45(6):1285-93. doi: 10.1016/j.humpath.2014.02.008. Epub 2014 Feb 28.
Ref 132 miR-145 sensitizes ovarian cancer cells to paclitaxel by targeting Sp1 and Cdk6. Int J Cancer. 2014 Sep 15;135(6):1286-96. doi: 10.1002/ijc.28774. Epub 2014 Apr 28.
Ref 133 Restoration of miR-200c to ovarian cancer reduces tumor burden and increases sensitivity to paclitaxel. Mol Cancer Ther. 2012 Dec;11(12):2556-65. doi: 10.1158/1535-7163.MCT-12-0463. Epub 2012 Oct 16.
Ref 134 MicroRNA-199a targets CD44 to suppress the tumorigenicity and multidrug resistance of ovarian cancer-initiating cells. FEBS J. 2012 Jun;279(11):2047-59. doi: 10.1111/j.1742-4658.2012.08589.x. Epub 2012 Apr 24.
Ref 135 Epigenetic silencing of miR-130b in ovarian cancer promotes the development of multidrug resistance by targeting colony-stimulating factor 1. Gynecol Oncol. 2012 Feb;124(2):325-34. doi: 10.1016/j.ygyno.2011.10.013. Epub 2011 Oct 15.
Ref 136 Long non-coding RNA tumor suppressor candidate 7 advances chemotherapy sensitivity of endometrial carcinoma through targeted silencing of miR-23b. Tumour Biol. 2017 Jun;39(6):1010428317707883. doi: 10.1177/1010428317707883.
Ref 137 Investigation of the potential theranostic role of KDM5B/miR-29c signaling axis in paclitaxel resistant endometrial carcinoma. Gene. 2019 Apr 30;694:76-82. doi: 10.1016/j.gene.2018.12.076. Epub 2019 Jan 16.
Ref 138 Targeted Silencing of S100A8 Gene by miR-24 to Increase Chemotherapy Sensitivity of Endometrial Carcinoma Cells to Paclitaxel. Med Sci Monit. 2016 Jun 9;22:1953-8. doi: 10.12659/msm.899179.
Ref 139 MicroRNA-200c mitigates invasiveness and restores sensitivity to microtubule-targeting chemotherapeutic agents. Mol Cancer Ther. 2009 May;8(5):1055-66. doi: 10.1158/1535-7163.MCT-08-1046. Epub 2009 May 12.
Ref 140 miR-375 mediated acquired chemo-resistance in cervical cancer by facilitating EMT. PLoS One. 2014 Oct 16;9(10):e109299. doi: 10.1371/journal.pone.0109299. eCollection 2014.
Ref 141 LINC00511 knockdown prevents cervical cancer cell proliferation and reduces resistance to paclitaxel. J Biosci. 2019 Jun;44(2):44.
Ref 142 miR-21 inhibitor suppresses cell proliferation and colony formation through regulating the PTEN/AKT pathway and improves paclitaxel sensitivity in cervical cancer cells. Mol Med Rep. 2017 May;15(5):2713-2719. doi: 10.3892/mmr.2017.6340. Epub 2017 Mar 16.
Ref 143 Long non-coding RNA Linc00518 promotes paclitaxel resistance of the human prostate cancer by sequestering miR-216b-5p. Biol Cell. 2019 Feb;111(2):39-50. doi: 10.1111/boc.201800054. Epub 2018 Dec 7.
Ref 144 miR-130a activates apoptotic signaling through activation of caspase-8 in taxane-resistant prostate cancer cells. Prostate. 2015 Oct;75(14):1568-78. doi: 10.1002/pros.23031. Epub 2015 Jun 12.
Ref 145 MiR-148a attenuates paclitaxel resistance of hormone-refractory, drug-resistant prostate cancer PC3 cells by regulating MSK1 expression. J Biol Chem. 2010 Jun 18;285(25):19076-84. doi: 10.1074/jbc.M109.079525. Epub 2010 Apr 20.
Ref 146 MicroRNA-34a Attenuates Paclitaxel Resistance in Prostate Cancer Cells via Direct Suppression of JAG1/Notch1 Axis. Cell Physiol Biochem. 2018;50(1):261-276. doi: 10.1159/000494004. Epub 2018 Oct 3.
Ref 147 MiR-34a attenuates paclitaxel-resistance of hormone-refractory prostate cancer PC3 cells through direct and indirect mechanisms. Prostate. 2010 Oct 1;70(14):1501-12. doi: 10.1002/pros.21185.
Ref 148 Targeting miR-21 decreases expression of multi-drug resistant genes and promotes chemosensitivity of renal carcinoma. Tumour Biol. 2017 Jul;39(7):1010428317707372. doi: 10.1177/1010428317707372.
Ref 149 MiR-34b-3p Represses the Multidrug-Chemoresistance of Bladder Cancer Cells by Regulating the CCND2 and P2RY1 Genes. Med Sci Monit. 2019 Feb 19;25:1323-1335. doi: 10.12659/MSM.913746.
Ref 150 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 151 miR-193a-3p regulates the multi-drug resistance of bladder cancer by targeting the LOXL4 gene and the oxidative stress pathway. Mol Cancer. 2014 Oct 14;13:234. doi: 10.1186/1476-4598-13-234.
Ref 152 MiR-193a-3p promotes the multi-chemoresistance of bladder cancer by targeting the HOXC9 gene. Cancer Lett. 2015 Feb 1;357(1):105-113. doi: 10.1016/j.canlet.2014.11.002. Epub 2014 Nov 11.
Ref 153 The miR-193a-3p regulated PSEN1 gene suppresses the multi-chemoresistance of bladder cancer. Biochim Biophys Acta. 2015 Mar;1852(3):520-8. doi: 10.1016/j.bbadis.2014.12.014. Epub 2014 Dec 24.
Ref 154 Different levels of let-7d expression modulate response of FaDu cells to irradiation and chemotherapeutics. PLoS One. 2017 Jun 30;12(6):e0180265. doi: 10.1371/journal.pone.0180265. eCollection 2017.

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