Drug Information
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)
|
||||
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 | |||||
ChEBI ID | |||||
TTD Drug ID | |||||
VARIDT ID | |||||
INTEDE ID | |||||
DrugBank ID |
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
Brain cancer [ICD-11: 2A00]
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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]
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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) | ||||
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) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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) | ||||
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) | ||||
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]
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Irregularity in Drug Uptake and Drug Efflux (IDUE) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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) | ||||
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) | ||||
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) | ||||
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]
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Irregularity in Drug Uptake and Drug Efflux (IDUE) | ||||
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) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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) | ||||
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) | ||||
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) | ||||
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]
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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) | ||||
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) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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) | ||||
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) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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) | ||||
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) | ||||
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) | ||||
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) | ||||
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) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Drug Inactivation by Structure Modification (DISM) | ||||
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) | ||||
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) | ||||
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) | ||||
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) | ||||
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) | ||||
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) | ||||
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) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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) | ||||
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) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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) | ||||
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) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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) | ||||
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) | ||||
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]
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
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) | ||||
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) | ||||
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) | ||||
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]
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
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) | ||||
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
If you find any error in data or bug in web service, please kindly report it to Dr. Sun and Dr. Zhang.