Disease Information
General Information of the Disease (ID: DIS00074)
Name |
Pancreatic cancer
|
---|---|
ICD |
ICD-11: 2C10
|
Resistance Map |
Type(s) of Resistant Mechanism of This Disease
ADTT: Aberration of the Drug's Therapeutic Target
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 Drug
Approved Drug(s)
10 drug(s) in total
Cisplatin
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-mir-374b | [1] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Activation | hsa05200 | |
In Vitro Model | BxPC3-R cells | Pancreas | Homo sapiens (Human) | CVCL_XX78 |
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
Resazurin conversion assay | |||
Mechanism Description | The computational prediction that downregulation of miR-374b likely contributed to the acquisition of resistance to cisplatin in BxPC3-R cells was experimentally tested by transfection of miR-374b into BxPC3-R cells and subsequently measuring cisplatin sensitivity of these cells relative to controls. The results demonstrated that miR-374b transfection significantly reduced drug resistance in BxPC3-R cells to levels approaching those of the parental BxPC3 cells. |
Cyclophosphamide
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Drug Inactivation by Structure Modification (DISM) | ||||
Key Molecule: Cytochrome P450 family 3 subfamily A member1 (CYP3A4) | [2] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Cyclophosphamide | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | Panc1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
Sulforhodamine B assay | |||
Mechanism Description | CYP3A4 is the most abundant hepatic and intestinal cytochrome P450 enzyme in humans, contributing to the metabolism of various drugs such as benzodiazepines, HIV antivirals, macrolide antibiotics, and statins. CYP3A4 3'UTR-luciferase activity was significantly decreased in human embryonic kidney 293 cells transfected with plasmid that expressed microRNA-27b (miR-27b) or mouse microRNA-298 (mmu-miR-298), overexpression of miR-27b or mmu-miR-298 in PANC1 cells led to a lower sensitivity to cyclophosphamide. | |||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-mir-27b | [2] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Cyclophosphamide | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | Panc1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Sulforhodamine B assay | |||
Mechanism Description | CYP3A4 is the most abundant hepatic and intestinal cytochrome P450 enzyme in humans, contributing to the metabolism of various drugs such as benzodiazepines, HIV antivirals, macrolide antibiotics, and statins. CYP3A4 3'UTR-luciferase activity was significantly decreased in human embryonic kidney 293 cells transfected with plasmid that expressed microRNA-27b (miR-27b) or mouse microRNA-298 (mmu-miR-298), overexpression of miR-27b or mmu-miR-298 in PANC1 cells led to a lower sensitivity to cyclophosphamide. |
Doxorubicin
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-miR-1291 | [3] | |||
Sensitive Disease | Pancreatic carcinoma [ICD-11: 2C10.2] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Doxorubicin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell growth | Inhibition | hsa05200 | |
In Vitro Model | H69 cells | Lung | Homo sapiens (Human) | CVCL_8121 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Hsa-miR-1291-directed downregulation of ABCC1 led to a greater intracellular drug accumulation and sensitized the cells to doxorubicin. | |||
Irregularity in Drug Uptake and Drug Efflux (IDUE) | ||||
Key Molecule: Multidrug resistance-associated protein 1 (MRP1) | [3] | |||
Sensitive Disease | Pancreatic carcinoma [ICD-11: 2C10.2] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Doxorubicin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell growth | Inhibition | hsa05200 | |
In Vitro Model | H69 cells | Lung | Homo sapiens (Human) | CVCL_8121 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Hsa-miR-1291-directed downregulation of ABCC1 led to a greater intracellular drug accumulation and sensitized the cells to doxorubicin. |
Erlotinib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-mir-124 | [4] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Erlotinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell viability | Inhibition | hsa05200 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
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 | miR-124 overexpression was able to sensitize the response of Capan-1 cells to erlotinib through inhibiting EphA2. | |||
Key Molecule: hsa-mir-497 | [5] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Erlotinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
FGF/FGFR signaling pathway | Inhibition | hsa01521 | ||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | miR-497 suppressed cells proliferation, decreased the percentage of S phase cells, re-sensitized cells to gemcitabine and erlotinib, and attenuated migration and invasion capacities. Furthermore, fibroblast growth factor 2 and fibroblast growth factor receptor 1 were confirmed as miR-497 targets. Overexpression of miR-497 inhibited tumor growth in vivo. Additionally, miR-497 expression was significantly downregulated in pancreatic cancer tissues compared with tumor-adjacent samples. Low expression of miR-497 was an independent adverse prognostic factor of pancreatic cancer. miR-497 plays a role in modulating the malignant phenotype and chemosensitivity of pancreatic cancer cells by directly inhibition of FGF2 and FGFR1 expression. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Ephrin type-A receptor 2 (EPHA2) | [4] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Erlotinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell viability | Inhibition | hsa05200 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
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 | miR-124 overexpression was able to sensitize the response of Capan-1 cells to erlotinib through inhibiting EphA2. | |||
Key Molecule: Fibroblast growth factor 2 (FGF1) | [5] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Erlotinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
FGF/FGFR signaling pathway | Inhibition | hsa01521 | ||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | miR-497 suppressed cells proliferation, decreased the percentage of S phase cells, re-sensitized cells to gemcitabine and erlotinib, and attenuated migration and invasion capacities. Furthermore, fibroblast growth factor 2 and fibroblast growth factor receptor 1 were confirmed as miR-497 targets. Overexpression of miR-497 inhibited tumor growth in vivo. Additionally, miR-497 expression was significantly downregulated in pancreatic cancer tissues compared with tumor-adjacent samples. Low expression of miR-497 was an independent adverse prognostic factor of pancreatic cancer. miR-497 plays a role in modulating the malignant phenotype and chemosensitivity of pancreatic cancer cells by directly inhibition of FGF2 and FGFR1 expression. | |||
Key Molecule: Fibroblast growth factor receptor 1 (FGFR1) | [5] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Erlotinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
FGF/FGFR signaling pathway | Inhibition | hsa01521 | ||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | miR-497 suppressed cells proliferation, decreased the percentage of S phase cells, re-sensitized cells to gemcitabine and erlotinib, and attenuated migration and invasion capacities. Furthermore, fibroblast growth factor 2 and fibroblast growth factor receptor 1 were confirmed as miR-497 targets. Overexpression of miR-497 inhibited tumor growth in vivo. Additionally, miR-497 expression was significantly downregulated in pancreatic cancer tissues compared with tumor-adjacent samples. Low expression of miR-497 was an independent adverse prognostic factor of pancreatic cancer. miR-497 plays a role in modulating the malignant phenotype and chemosensitivity of pancreatic cancer cells by directly inhibition of FGF2 and FGFR1 expression. |
Everolimus
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Eukaryotic translation initiation factor 4E binding protein 1 (EIF4EBP1) | [6] | |||
Resistant Disease | Pancreatic neuroendocrine tumor [ICD-11: 2C10.1] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Everolimus | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | PI3K/AKT/mTOR signaling pathway | Activation | hsa04151 | |
CXCR4-CXCL12-CXCR7 signaling pathway | Activation | hsa04061 | ||
In Vitro Model | A498 cells | Kidney | Homo sapiens (Human) | CVCL_1056 |
SN12C cells | Kidney | Homo sapiens (Human) | CVCL_1705 | |
Experiment for Molecule Alteration |
Western blotting assay | |||
Mechanism Description | When the CXCR4-CXCL12-CXCR7 pathway is activated through CXCL12 in human renal cancer cells were, SN12C and A498, CXCL12 induced the mTOR targets p-P70S6K and p-4EBP1.The combination therapy of mTOR inhibitors with the CXCR4-CXCL12-CXCR7 axis inhibitors in renal cancer tumors could overcome the Everolimus resistance. | |||
Key Molecule: Ribosomal protein S6 kinase beta-1 (RPS6KB1) | [6] | |||
Resistant Disease | Pancreatic neuroendocrine tumor [ICD-11: 2C10.1] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Everolimus | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | PI3K/AKT/mTOR signaling pathway | Activation | hsa04151 | |
CXCR4-CXCL12-CXCR7 signaling pathway | Activation | hsa04061 | ||
In Vitro Model | A498 cells | Kidney | Homo sapiens (Human) | CVCL_1056 |
SN12C cells | Kidney | Homo sapiens (Human) | CVCL_1705 | |
Experiment for Molecule Alteration |
Western blotting assay | |||
Mechanism Description | When the CXCR4-CXCL12-CXCR7 pathway is activated through CXCL12 in human renal cancer cells were, SN12C and A498, CXCL12 induced the mTOR targets p-P70S6K and p-4EBP1.The combination therapy of mTOR inhibitors with the CXCR4-CXCL12-CXCR7 axis inhibitors in renal cancer tumors could overcome the Everolimus resistance. |
Fluorouracil
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: DiGeorge syndrome critical region gene 5 (DGCR5) | [7] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Fluorouracil | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell migration | Activation | hsa04670 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
HAPC cells | Pancreas | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | DGCR5 and miR320a regulate each other in a reciprocal manner and that DGCR5 reverses the inhibition of PDCD4 by miR320a, which is involved in the regulation of the PDAC cell phenotype and response to 5-FU. miR320a is involved in 5-FU resistance modulated by DGCR5. | |||
Key Molecule: Growth arrest specific 5 (GAS5) | [8] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Fluorouracil | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Hippo signaling pathway | Inhibition | hsa04390 | |
In Vitro Model | SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 |
5-FU cells | Colon | Homo sapiens (Human) | CVCL_1846 | |
PATU8988 | Pancreas | Homo sapiens (Human) | CVCL_1847 | |
PATU8988 cells | Pancreas | Homo sapiens (Human) | CVCL_1846 | |
SW1990/GEM cells | Pancreas | Homo sapiens (Human) | CVCL_ZW98 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | GAS5 regualtes Hippo signaling pathway via miR181c-5p to antagonize the development of multidrug resistance in pancreatic cancer cells. GAS5 regulated chemoresistance and Hippo pathway of pancreatic cancer cells via miR181c-5p/Hippo. | |||
Key Molecule: hsa-miR-221-3p | [9] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Fluorouracil | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
PATU8988 cells | Pancreas | Homo sapiens (Human) | CVCL_1846 | |
293TN cells | Pancreas | Homo sapiens (Human) | CVCL_UL49 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
5-FU and gemcitabine assay; CCK8 assay; Wound healing assay; Transwell chamber invasion assay | |||
Mechanism Description | miRNA-221-3p desensitizes pancreatic cancer cells to 5-fluorouracil by targeting RB1. miR221-3p down-regulated RB1 expression by directly binding to its 3'-UTR and therefore caused increased several aspects of pancreatic cancer pathogenesis, including proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT). | |||
Key Molecule: Urothelial cancer associated 1 (UCA1) | [10] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Fluorouracil | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | AKT signaling pathway | Activation | hsa04151 | |
Cell apoptosis | Inhibition | hsa04210 | ||
Cell invasion | Activation | hsa05200 | ||
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
ERK signaling pathway | Activation | hsa04210 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
CFPAC1 cells | Pancreas | Homo sapiens (Human) | CVCL_1119 | |
HPAC cells | Pancreas | Homo sapiens (Human) | CVCL_3517 | |
In Vivo Model | BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
RT-qPCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay; Wound-healing assay | |||
Mechanism Description | CUDR overexpression inhibits cell apoptosis and promotes drug resistance in PDAC and CUDR overexpression in Panc-1 cells significantly increased phosphorylated (p-) focal adhesion kinase (FAk) and p-AkT levels, whereas the total FAk and AkT were not altered compared with in Panc-1 cells transfected with an empty vector. | |||
Key Molecule: hsa-mir-21 | [11] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Fluorouracil | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Activation | hsa05200 | |
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
PI3K/AKT/mTOR signaling pathway | Regulation | hsa04151 | ||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
PATU8988 cells | Pancreas | Homo sapiens (Human) | CVCL_1846 | |
293TN cells | Pancreas | Homo sapiens (Human) | CVCL_UL49 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | miR-21 regulates 5-FU drug resistance in pancreatic cancer by reducing the expression of its targets, PTEN and PDCD4. And PTEN and PDCD4, as tumor suppressors, not only can inhibit tumor growth and invasion, but also can downregulate the 5-FU resistance induced by miR-21 in pancreatic cancer cells. | |||
Key Molecule: hsa-mir-181c | [12] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Fluorouracil | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Hippo signaling pathway | Regulation | hsa04392 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-181c directly repressed MST1, LATS2, MOB1 and SAV1 expression in human pancreatic cancer cells. Overexpression of miR-181c induced hyperactivation of the YAP/TAZ and (+) expression of the Hippo signaling downstream genes CTGF, BIRC5 and BLC2L1, leading to pancreatic cancer cell survival and chemoresistance in vitro and in vivo. Importantly, high miR-181c levels were significantly correlated with Hippo signaling inactivation in pancreatic cancer samples, and predicted a poor patient overall survival. | |||
Key Molecule: hsa-miR-1246 | [13] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Fluorouracil | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | Panc1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | The in vitro drug sensitivity of pancreatic cancer cells was altered according to the miR-1246 expression via CCNG2. In vivo, we found that miR-1246 could increase tumour-initiating potential and induced drug resistance. A high expression level of miR-1246 was correlated with a worse prognosis and CCNG2 expression was significantly lower in those patients. miR-1246 expression was associated with chemoresistance and CSC-like properties via CCNG2, and could predict worse prognosis in pancreatic cancer patients. | |||
Regulation by the Disease Microenvironment (RTDM) | ||||
Key Molecule: hsa-mir-320 | [14] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Fluorouracil | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell invasion | Activation | hsa05200 | ||
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
PATU8988 cells | Pancreas | Homo sapiens (Human) | CVCL_1846 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Wound Healing assay; Matrigel transmembrane invasion assay | |||
Mechanism Description | miR-320a was up-regulated in 5-FU resistant pancreatic cancer cells and that miR-320a could promote pancreatic cancer cell proliferation, migration and invasion then contributed to the increased 5-FU resistance. Researchers think miR-320a could suppress cell apoptosis by inhibiting PDCD4 and further contribute to drug-resistance, which will be studied in future. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Retinoblastoma-associated protein (RB1) | [9] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Fluorouracil | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
PATU8988 cells | Pancreas | Homo sapiens (Human) | CVCL_1846 | |
293TN cells | Pancreas | Homo sapiens (Human) | CVCL_UL49 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
5-FU and gemcitabine assay; CCK8 assay; Wound healing assay; Transwell chamber invasion assay | |||
Mechanism Description | miRNA-221-3p desensitizes pancreatic cancer cells to 5-fluorouracil by targeting RB1. miR221-3p down-regulated RB1 expression by directly binding to its 3'-UTR and therefore caused increased several aspects of pancreatic cancer pathogenesis, including proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT). | |||
Key Molecule: RAC serine/threonine-protein kinase (AKT) | [10] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Phosphorylation | Up-regulation |
||
Resistant Drug | Fluorouracil | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | AKT signaling pathway | Activation | hsa04151 | |
Cell apoptosis | Inhibition | hsa04210 | ||
Cell invasion | Activation | hsa05200 | ||
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
ERK signaling pathway | Activation | hsa04210 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
CFPAC1 cells | Pancreas | Homo sapiens (Human) | CVCL_1119 | |
HPAC cells | Pancreas | Homo sapiens (Human) | CVCL_3517 | |
In Vivo Model | BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay; Wound-healing assay | |||
Mechanism Description | CUDR overexpression inhibits cell apoptosis and promotes drug resistance in PDAC and CUDR overexpression in Panc-1 cells significantly increased phosphorylated (p-) focal adhesion kinase (FAk) and p-AkT levels, whereas the total FAk and AkT were not altered compared with in Panc-1 cells transfected with an empty vector. | |||
Key Molecule: Mitogen-activated protein kinase (MAPK) | [10] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Phosphorylation | Up-regulation |
||
Resistant Drug | Fluorouracil | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | AKT signaling pathway | Activation | hsa04151 | |
Cell apoptosis | Inhibition | hsa04210 | ||
Cell invasion | Activation | hsa05200 | ||
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
ERK signaling pathway | Activation | hsa04210 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
CFPAC1 cells | Pancreas | Homo sapiens (Human) | CVCL_1119 | |
HPAC cells | Pancreas | Homo sapiens (Human) | CVCL_3517 | |
In Vivo Model | BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay; Wound-healing assay | |||
Mechanism Description | CUDR overexpression inhibits cell apoptosis and promotes drug resistance in PDAC and CUDR overexpression in Panc-1 cells significantly increased phosphorylated (p-) focal adhesion kinase (FAk) and p-AkT levels, whereas the total FAk and AkT were not altered compared with in Panc-1 cells transfected with an empty vector. | |||
Key Molecule: Programmed cell death protein 4 (PDCD4) | [11], [14] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Fluorouracil | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell invasion | Activation | hsa05200 | ||
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
PI3K/AKT/mTOR signaling pathway | Regulation | hsa04151 | ||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
PATU8988 cells | Pancreas | Homo sapiens (Human) | CVCL_1846 | |
293TN cells | Pancreas | Homo sapiens (Human) | CVCL_UL49 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Wound Healing assay; Matrigel transmembrane invasion assay | |||
Mechanism Description | miR-21 regulates 5-FU drug resistance in pancreatic cancer by reducing the expression of its targets, PTEN and PDCD4. And PTEN and PDCD4, as tumor suppressors, not only can inhibit tumor growth and invasion, but also can downregulate the 5-FU resistance induced by miR-21 in pancreatic cancer cells. | |||
Key Molecule: Phosphatase and tensin homolog (PTEN) | [11] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Fluorouracil | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Activation | hsa05200 | |
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
PI3K/AKT/mTOR signaling pathway | Regulation | hsa04151 | ||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
PATU8988 cells | Pancreas | Homo sapiens (Human) | CVCL_1846 | |
293TN cells | Pancreas | Homo sapiens (Human) | CVCL_UL49 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | miR-21 regulates 5-FU drug resistance in pancreatic cancer by reducing the expression of its targets, PTEN and PDCD4. And PTEN and PDCD4, as tumor suppressors, not only can inhibit tumor growth and invasion, but also can downregulate the 5-FU resistance induced by miR-21 in pancreatic cancer cells. | |||
Key Molecule: Serine/threonine-protein kinase LATS2 (LATS2) | [12] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Fluorouracil | |||
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) | [12] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Fluorouracil | |||
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) | [12] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Fluorouracil | |||
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) | [12] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Fluorouracil | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Hippo signaling pathway | Regulation | hsa04392 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-181c directly repressed MST1, LATS2, MOB1 and SAV1 expression in human pancreatic cancer cells. Overexpression of miR-181c induced hyperactivation of the YAP/TAZ and (+) expression of the Hippo signaling downstream genes CTGF, BIRC5 and BLC2L1, leading to pancreatic cancer cell survival and chemoresistance in vitro and in vivo. Importantly, high miR-181c levels were significantly correlated with Hippo signaling inactivation in pancreatic cancer samples, and predicted a poor patient overall survival. | |||
Key Molecule: Cyclin-G2 (CCNG2) | [13] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Fluorouracil | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | Panc1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | The in vitro drug sensitivity of pancreatic cancer cells was altered according to the miR-1246 expression via CCNG2. In vivo, we found that miR-1246 could increase tumour-initiating potential and induced drug resistance. A high expression level of miR-1246 was correlated with a worse prognosis and CCNG2 expression was significantly lower in those patients. miR-1246 expression was associated with chemoresistance and CSC-like properties via CCNG2, and could predict worse prognosis in pancreatic cancer patients. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-mir-320 | [7] | |||
Sensitive Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Fluorouracil | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | SUDHL-4 cells | Peritoneal effusion | Homo sapiens (Human) | CVCL_0539 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | DGCR5 and miR320a regulate each other in a reciprocal manner and that DGCR5 reverses the inhibition of PDCD4 by miR320a, which is involved in the regulation of the PDAC cell phenotype and response to 5-FU. miR320a is involved in 5-FU resistance modulated by DGCR5. DGCR5 reversed the inhibition of the miR320a target gene PDCD4, which in turn inhibited the proliferation, migration and 5-FU resistance of PDAC cells. | |||
Key Molecule: hsa-miR-181c-5p | [8] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Fluorouracil | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Hippo signaling pathway | Inhibition | hsa04390 | |
In Vitro Model | SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 |
5-FU cells | Colon | Homo sapiens (Human) | CVCL_1846 | |
PATU8988 | Pancreas | Homo sapiens (Human) | CVCL_1847 | |
PATU8988 cells | Pancreas | Homo sapiens (Human) | CVCL_1846 | |
SW1990/GEM cells | Pancreas | Homo sapiens (Human) | CVCL_ZW98 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Long non-coding RNA GAS5 antagonizes the chemoresistance of pancreatic cancer cells through down-regulation of miR181c-5p. GAS5 negatively regulated miR181c-5p, and miR181c-5p dramatically promoted pancreatic cancer cell chemoresistance through inactivating the Hippo signaling. | |||
Key Molecule: hsa-mir-21 | [15] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Fluorouracil | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
HPAC cells | Pancreas | Homo sapiens (Human) | CVCL_3517 | |
HPAF-II cells | Pancreatic | Homo sapiens (Human) | CVCL_0313 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
SRB (sulforhodamine-B) assay | |||
Mechanism Description | Low miR-21 expression was associated with benefit from adjuvant treatment in two independent cohorts of PDAC cases, and anti-miR-21 increased anticancer drug activity in vitro. |
Gemcitabine
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Drug Inactivation by Structure Modification (DISM) | ||||
Key Molecule: Activation induced cytidine deaminase (AICDA) | [16] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | The main mechanism for gemcitabine inactivation is through deamination by cytidine deaminase (CDA) to difluorodeoxyuridine (dFdU). Since dFdU is not a substrate for pyrimidine nucleoside phosphorylases, the drug is degraded and excreted out of the cell. | |||
Key Molecule: Deoxycytidine kinase (DCK) | [16] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | Once taken up into the cell, gemcitabine is phosphorylated by deoxycytidine kinase (dCK) to produce dFdCMP. In turn, dFdCMP is converted by other pyrimidine kinases to its active diphosphate and triphosphate derivatives, dFdCDP and dFdCTP. Due to the central role of dCK in gemcitabine metabolism, its deficiency is a major contributor to gemcitabine resistance. | |||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-miR-188-3p | [17] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay; Soft agar assay | |||
Mechanism Description | Long non-coding RNA LINC00346 promotes pancreatic cancer growth and gemcitabine resistance by sponging miR-188-3p to derepress BRD4 expression. | |||
Key Molecule: P53 regulated carcinoma associated Stat3 activating long intergenic non-protein coding transcript (PRECSIT) | [17] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay; Soft agar assay | |||
Mechanism Description | Long non-coding RNA LINC00346 promotes pancreatic cancer growth and gemcitabine resistance by sponging miR-188-3p to derepress BRD4 expression. | |||
Key Molecule: hsa-mir-301 | [18] | |||
Resistant Disease | Pancreatic carcinoma [ICD-11: 2C10.2] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell invasion | Inhibition | hsa05200 | ||
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Capan-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0026 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
Colorimetric methylene blue assay; Flow cytometry assay | |||
Mechanism Description | Gemcitabine-resistant Capan-2 and Panc-1 cells exhibited increased miR-301 expression, and miR-301 overepression can enhance apoptosis and inhibit cell invasiveness and exhibit strong gemcitabine resistance. | |||
Key Molecule: Maternally expressed 3 (MEG3) | [19] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
COLO357 cells | Pancreas | Homo sapiens (Human) | CVCL_0221 | |
T3M4 cells | Pancreas | Homo sapiens (Human) | CVCL_4056 | |
HTERT-HPNE cells | Pancreas | Homo sapiens (Human) | CVCL_C466 | |
Experiment for Molecule Alteration |
RT-qPCR | |||
Experiment for Drug Resistance |
CCK8 assay; Boyden chamber assay; Sphere formation assay; Flow cytometric analysis | |||
Mechanism Description | Decreased expression of MEG3 could promote PC cell migration and invasion, as well as chemoresistance by regulating the EMT process and CSC properties. | |||
Key Molecule: HOX transcript antisense RNA (HOTAIR) | [20] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Experiment for Molecule Alteration |
RT-qPCR | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | Gemcitabine treatment causes resistance and malignancy of pancreatic cancer stem-like cells via induction of LncRNA HOTAIR. | |||
Key Molecule: Taurine up-regulated 1 (TUG1) | [21] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell colony | Activation | hsa05200 | ||
Cell viability | Activation | hsa05200 | ||
ERK signaling pathway | Activation | hsa04210 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
PANC-28 cells | Pancreatic | Homo sapiens (Human) | CVCL_3917 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | TUG1 promoted the viability of PDAC cells and enhanced its resistance of gemcitabine and overexpression of TUG1 increased ERk phosphorylation. | |||
Key Molecule: hsa-mir-301 | [22] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | AKT signaling pathway | Activation | hsa04151 | |
Cell viability | Activation | hsa05200 | ||
In Vitro Model | HEK293T cells | Kidney | Homo sapiens (Human) | CVCL_0063 |
BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 | |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
HPAF-II cells | Pancreatic | Homo sapiens (Human) | CVCL_0313 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | miR-301a upregulation promoted resistance to gemcitabine under hypoxia through downregulation of TAp63. | |||
Key Molecule: hsa-miR-455-3p | [23] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell viability | Activation | hsa05200 | ||
In Vitro Model | HEK293T cells | Kidney | Homo sapiens (Human) | CVCL_0063 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
HPDE6-C7 cells | Pancreas | Homo sapiens (Human) | CVCL_0P38 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | Down-regulation of microRNA-455-3p Links to Proliferation and Drug Resistance of Pancreatic Cancer Cells via Targeting TAZ. | |||
Key Molecule: hsa-mir-125a | [24] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Activation | hsa05200 | |
In Vitro Model | HEK293T cells | Kidney | Homo sapiens (Human) | CVCL_0063 |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | miR-125a may promote chemo-resistance to gemcitabine in pancreatic cell lines through targeting A20, which may provide novel therapeutic targets or molecular biomarkers for cancer therapy and improve tumor diagnosis or predictions of therapeutic responses. | |||
Key Molecule: hsa-mir-181c | [12] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Hippo signaling pathway | Regulation | hsa04392 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-181c directly repressed MST1, LATS2, MOB1 and SAV1 expression in human pancreatic cancer cells. Overexpression of miR-181c induced hyperactivation of the YAP/TAZ and (+) expression of the Hippo signaling downstream genes CTGF, BIRC5 and BLC2L1, leading to pancreatic cancer cell survival and chemoresistance in vitro and in vivo. Importantly, high miR-181c levels were significantly correlated with Hippo signaling inactivation in pancreatic cancer samples, and predicted a poor patient overall survival. | |||
Key Molecule: HOXA distal transcript antisense RNA (HOTTIP) | [25] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Activation | hsa05200 | |
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
HOTTIP/HOXA13 signaling pathway | Activation | hsa05202 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0026 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | The long non-coding RNA HOTTIP promotes progression and gemcitabine resistance by regulating HOXA13 in pancreatic cancer.Microarray analyses revealed that HOTTIP was one of the most significantly upregulated LncRNAs in PDAC tissues compared with pancreatic tissues.Furthermore, knockdown of HOXA13 by RNA interference (siHOXA13) revealed that HOTTIP promoted PDAC cell proliferation, invasion, and chemoresistance, at least partly through regulating HOXA13. As a crucial tumor promoter, HOTTIP promotes cell proliferation, invasion, and chemoresistance by modulating HOXA13. Therefore, the HOTTIP/HOXA13 axis is a potential therapeutic target and molecular biomarker for PDAC. | |||
Key Molecule: hsa-miR-1246 | [13] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | Panc1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | The in vitro drug sensitivity of pancreatic cancer cells was altered according to the miR-1246 expression via CCNG2. In vivo, we found that miR-1246 could increase tumour-initiating potential and induced drug resistance. A high expression level of miR-1246 was correlated with a worse prognosis and CCNG2 expression was significantly lower in those patients. miR-1246 expression was associated with chemoresistance and CSC-like properties via CCNG2, and could predict worse prognosis in pancreatic cancer patients. | |||
Key Molecule: hsa-mir-21 | [26], [27], [28] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | AKT signaling pathway | Activation | hsa04151 | |
Cell apoptosis | Inhibition | hsa04210 | ||
Cell proliferation | Activation | hsa05200 | ||
FasL/Fas signaling pathway | Inhibition | hsa04210 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
LPc006 cells | Pancreas | Homo sapiens (Human) | N.A. | |
LPc028 cells | Pancreas | Homo sapiens (Human) | N.A. | |
LPc033 cells | Pancreas | Homo sapiens (Human) | N.A. | |
LPc067 cells | Pancreas | Homo sapiens (Human) | N.A. | |
LPc111 cells | Pancreas | Homo sapiens (Human) | N.A. | |
LPc167 cells | Pancreas | Homo sapiens (Human) | N.A. | |
PP437 cells | Pancreas | Homo sapiens (Human) | N.A. | |
In Vivo Model | BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
WST-8 assay; Fluorescence microscopy | |||
Mechanism Description | miR-21 regulates expression of PTEN and phosphorylation of its downstream kinase Akt and (b) the reduction of phospho-Akt (pAkt) correlated with the enhancement of gemcitabine-induced apoptosis and antitumor activity in vitro and in vivo, suggesting that Akt pathway plays a significant role in mediating drug resistance in PDAC cells. | |||
Key Molecule: hsa-mir-21 | [29] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell migration | Activation | hsa04670 | |
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
HPAC cells | Pancreas | Homo sapiens (Human) | CVCL_3517 | |
BxPc3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 | |
Capan cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
HPAF cells | Pancreas | Homo sapiens (Human) | CVCL_B284 | |
PL-45 cells | Pancreas | Homo sapiens (Human) | CVCL_3567 | |
Experiment for Molecule Alteration |
RT-qPCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Histone acetylation levels at miR-21 promoter were increased in PDAC cells after treatment with gemcitabine. Enhanced invasion and metastasis, increased miR-21 expression, decreased PTEN, elevated pAkT level were demonstrated in gemcitabine-resistant HPAC and PANC-1 cells. Pre-miR-21 transfection or TSA treatment further increased invasion and metastasis ability, decreased PTEN, and elevated pAkT levels in these two lines. In contrast, anti-miR-21 transfection could reverse invasion and metastasis, and PTEN and pAkT expressions induced by gemcitabine. | |||
Key Molecule: hsa-mir-365 | [30] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-365 directly targets the pro-apoptotic molecules SHC1 and BAX, whose reductions contribute to gemcitabine resistance in pancreatic cancer cells. | |||
Key Molecule: hsa-mir-181 | [31] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell viability | Activation | hsa05200 | ||
NF-kappaB signaling pathway | Regulation | hsa04064 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
Panc1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
PSN1 cells | Pancreas | Homo sapiens (Human) | CVCL_1644 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-181b enhances the activity of NF-kB by inhibiting CYLD, thus leading to the resistance to gemcitabine. | |||
Key Molecule: hsa-miR-320c | [32] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell viability | Activation | hsa05200 | |
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
PSN1 cells | Pancreas | Homo sapiens (Human) | CVCL_1644 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-320c regulates the resistance of pancreatic cancer cells to gemcitabine through SMARCC1. | |||
Key Molecule: hsa-mir-21 | [16] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | miR-21 is probably the most characterized miRNA associated with gemcitabine resistance. Tissue samples of PDA patients treated with gemcitabine indicate that miR-21 expression is directly correlated with chemotherapy resistance. Patients with high miR-21 expression have significantly shorter overall survival; consistently, overexpression of miR-21 in primary PDA cells in vitro, decreases the anti-proliferative effect of gemcitabine. miR-21 promotes gemcitabine resistance by targeting phosphatase and tensin homologue (PTEN) or by overexpression of matrix metalloproteinases (MMP) 2 and 9, and of vascular endothelial growth factor (VEGF), which in-turn induces the PI3K/AKT pathway. | |||
Irregularity in Drug Uptake and Drug Efflux (IDUE) | ||||
Key Molecule: Solute carrier family 29 member 1 (SLC29A1) | [16] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | Gemcitabine could be a substrate for several nucleoside transporters (NTs), but its major uptake occurs via the equilibrative and concentrative type NTs (ENTs and CNTs, respectively). ENT1, CNT1 and CNT3 have often been related to gemcitabine transport and resistance in humans. When ENT1 knockout conferred gemcitabine resistance, while its up regulation enhanced its cytotoxic activity. Similarly, retroviral expression of CNT1 renders ovarian cancer cells sensitive to gemcitabine in vitro. | |||
Key Molecule: Solute carrier family 28 member 1 (SLC28A1) | [16] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | Gemcitabine could be a substrate for several nucleoside transporters (NTs), but its major uptake occurs via the equilibrative and concentrative type NTs (ENTs and CNTs, respectively). ENT1, CNT1 and CNT3 have often been related to gemcitabine transport and resistance in humans. When ENT1 knockout conferred gemcitabine resistance, while its up regulation enhanced its cytotoxic activity. Similarly, retroviral expression of CNT1 renders ovarian cancer cells sensitive to gemcitabine in vitro. | |||
Key Molecule: Solute carrier family 28 member 3 (SLC28A3) | [16] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | Gemcitabine could be a substrate for several nucleoside transporters (NTs), but its major uptake occurs via the equilibrative and concentrative type NTs (ENTs and CNTs, respectively). ENT1, CNT1 and CNT3 have often been related to gemcitabine transport and resistance in humans. When ENT1 knockout conferred gemcitabine resistance, while its up regulation enhanced its cytotoxic activity. Similarly, retroviral expression of CNT1 renders ovarian cancer cells sensitive to gemcitabine in vitro. | |||
Regulation by the Disease Microenvironment (RTDM) | ||||
Key Molecule: hsa-mir-221 | [33] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
miR221/SOCS3 signaling pathway | Regulation | hsa05206 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0026 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | miR-221 overexpression can promote proliferation, migration, emt, chemotherapy resistance, and stem cell-like properties in panc-1 cells. | |||
Key Molecule: Transcription factor AP2 gamma (TFAP2C) | [34] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell invasion | Activation | hsa05200 | ||
Cell migration | Activation | hsa04670 | ||
Cell viability | Activation | hsa05200 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
Su.86.86 cells | Pancreas | Homo sapiens (Human) | CVCL_3881 | |
T3M4 cells | Pancreas | Homo sapiens (Human) | CVCL_4056 | |
In Vivo Model | Nude mouse model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay; Transwell assay | |||
Mechanism Description | Transcription factor activating protein 2 gamma (TFAP2C) is a target of miR-10a-5p, and TFAP2C overexpression resensitizes PDAC cells to gemcitabine, which is initiated by miR-10a-5p. | |||
Key Molecule: hsa-miR-10a-5p | [34] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell invasion | Activation | hsa05200 | ||
Cell viability | Activation | hsa05200 | ||
Epithelial mesenchymal transition signaling pathway | Activation | hsa01521 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
Su.86.86 cells | Pancreas | Homo sapiens (Human) | CVCL_3881 | |
T3M4 cells | Pancreas | Homo sapiens (Human) | CVCL_4056 | |
In Vivo Model | Nude mouse model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay; Transwell assay | |||
Mechanism Description | Transcription factor activating protein 2 gamma (TFAP2C) is a target of miR-10a-5p, and TFAP2C overexpression resensitizes PDAC cells to gemcitabine, which is initiated by miR-10a-5p. | |||
Key Molecule: Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) | [35] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Activation | hsa04670 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 |
CFPAC1 cells | Pancreas | Homo sapiens (Human) | CVCL_1119 | |
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay | |||
Mechanism Description | MALAT-1 could increase the proportion of pancreatic CSCs, maintain self-renewing capacity, decrease the chemosensitivity to anticancer drugs, and accelerate tumor angiogenesis in vitro, and promote tumorigenicity of pancreatic cancer cells in vivo. The underlying mechanisms may involve in increased expression of self-renewal related factors Sox2. | |||
Key Molecule: hsa-mir-100 | [36] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues. | |||
Key Molecule: hsa-mir-10a | [36] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues. | |||
Key Molecule: hsa-mir-10b | [36] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues. | |||
Key Molecule: hsa-mir-134 | [36] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues. | |||
Key Molecule: hsa-mir-143 | [36] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues. | |||
Key Molecule: hsa-mir-146a | [36] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues. | |||
Key Molecule: hsa-mir-15 | [36] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues. | |||
Key Molecule: hsa-mir-205 | [36] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues. | |||
Key Molecule: hsa-mir-214 | [36] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues. | |||
Key Molecule: hsa-mir-32 | [36] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues. | |||
Key Molecule: hsa-mir-34 | [36] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues. | |||
Key Molecule: hsa-miR-146b-5p | [36] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues. | |||
Key Molecule: hsa-mir-7 | [36] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Pancreatic cancers relapse due to small but distinct population of cancer stem cells (CSCs) which are in turn regulated by miRNAs. Those miRNA were either upregulated (e.g. miR-146) or downregulated (e.g. miRNA-205, miRNA-7) in gemcitabine resistant MIA PaCa-2 cancer cells and clinical metastatic pancreatic cancer tissues. | |||
Key Molecule: TIMP metallopeptidase inhibitor 2 (TIMP2) | [16] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | The ECM may provide a mechanical barrier, preventing the tumor from further spread. Disintegration of the ECM by MMPs enables cancer cells to dissociate from the tumor and metastasize. Apart from destabilizing the physical barrier, MMPs overexpression also regulates internal cellular cascades. In response to collagen deposition in the ECM, an MMP dependent ERK-1/2 phosphorylation occurs, triggering the transcription factor HMGA2 and gemcitabine resistance. | |||
Key Molecule: Mucin 4, cell surface associated (MUC4) | [16] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | Mucin 4 (MUC4) is a membrane-bound O-glycoprotein that is found in the lining of the respiratory tract and GI mucosa, where it enables lubrication and cell-matrix detachment. In PDA, MUC4 expressing cells exhibit greater gemcitabine resistance than do MUC4 negative cells, an effect mediated by interaction with HER2. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Bromodomain-containing protein 4 (BRD4) | [17] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Luciferase reporter assay; Western blot analysis; RT-qPCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay; Soft agar assay | |||
Mechanism Description | Long non-coding RNA LINC00346 promotes pancreatic cancer growth and gemcitabine resistance by sponging miR-188-3p to derepress BRD4 expression. | |||
Key Molecule: Mitogen-activated protein kinase (MAPK) | [21] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Phosphorylation | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell viability | Activation | hsa05200 | |
ERK signaling pathway | Activation | hsa04210 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
PANC-28 cells | Pancreatic | Homo sapiens (Human) | CVCL_3917 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | TUG1 promoted the viability of PDAC cells and enhanced its resistance of gemcitabine and overexpression of TUG1 increased ERk phosphorylation. | |||
Key Molecule: Tumor protein 63 (TP63) | [22] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | AKT signaling pathway | Activation | hsa04151 | |
Cell viability | Activation | hsa05200 | ||
In Vitro Model | HEK293T cells | Kidney | Homo sapiens (Human) | CVCL_0063 |
BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 | |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
HPAF-II cells | Pancreatic | Homo sapiens (Human) | CVCL_0313 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | miR-301a upregulation promoted resistance to gemcitabine under hypoxia through downregulation of TAp63. | |||
Key Molecule: Tafazzin (TAZ) | [23] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell viability | Activation | hsa05200 | ||
In Vitro Model | HEK293T cells | Kidney | Homo sapiens (Human) | CVCL_0063 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
HPDE6-C7 cells | Pancreas | Homo sapiens (Human) | CVCL_0P38 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | Down-regulation of microRNA-455-3p Links to Proliferation and Drug Resistance of Pancreatic Cancer Cells via Targeting TAZ. | |||
Key Molecule: Tumor necrosis factor alpha-induced protein 3 (TNFAIP3) | [24] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Activation | hsa05200 | |
In Vitro Model | HEK293T cells | Kidney | Homo sapiens (Human) | CVCL_0063 |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | miR-125a may promote chemo-resistance to gemcitabine in pancreatic cell lines through targeting A20, which may provide novel therapeutic targets or molecular biomarkers for cancer therapy and improve tumor diagnosis or predictions of therapeutic responses. | |||
Key Molecule: Serine/threonine-protein kinase LATS2 (LATS2) | [12] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
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) | [12] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
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) | [12] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
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) | [12] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Hippo signaling pathway | Regulation | hsa04392 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-181c directly repressed MST1, LATS2, MOB1 and SAV1 expression in human pancreatic cancer cells. Overexpression of miR-181c induced hyperactivation of the YAP/TAZ and (+) expression of the Hippo signaling downstream genes CTGF, BIRC5 and BLC2L1, leading to pancreatic cancer cell survival and chemoresistance in vitro and in vivo. Importantly, high miR-181c levels were significantly correlated with Hippo signaling inactivation in pancreatic cancer samples, and predicted a poor patient overall survival. | |||
Key Molecule: Homeobox protein Hox-A13 (HOXA13) | [25] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Activation | hsa05200 | |
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
HOTTIP/HOXA13 signaling pathway | Activation | hsa05202 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0026 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | The long non-coding RNA HOTTIP promotes progression and gemcitabine resistance by regulating HOXA13 in pancreatic cancer.Microarray analyses revealed that HOTTIP was one of the most significantly upregulated LncRNAs in PDAC tissues compared with pancreatic tissues.Furthermore, knockdown of HOXA13 by RNA interference (siHOXA13) revealed that HOTTIP promoted PDAC cell proliferation, invasion, and chemoresistance, at least partly through regulating HOXA13. As a crucial tumor promoter, HOTTIP promotes cell proliferation, invasion, and chemoresistance by modulating HOXA13. Therefore, the HOTTIP/HOXA13 axis is a potential therapeutic target and molecular biomarker for PDAC. | |||
Key Molecule: Transcription factor SOX-2 (SOX2) | [35] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Activation | hsa04670 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 |
CFPAC1 cells | Pancreas | Homo sapiens (Human) | CVCL_1119 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay | |||
Mechanism Description | MALAT-1 could increase the proportion of pancreatic CSCs, maintain self-renewing capacity, decrease the chemosensitivity to anticancer drugs, and accelerate tumor angiogenesis in vitro, and promote tumorigenicity of pancreatic cancer cells in vivo. The underlying mechanisms may involve in increased expression of self-renewal related factors Sox2. | |||
Key Molecule: Cyclin-G2 (CCNG2) | [13] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | Panc1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | The in vitro drug sensitivity of pancreatic cancer cells was altered according to the miR-1246 expression via CCNG2. In vivo, we found that miR-1246 could increase tumour-initiating potential and induced drug resistance. A high expression level of miR-1246 was correlated with a worse prognosis and CCNG2 expression was significantly lower in those patients. miR-1246 expression was associated with chemoresistance and CSC-like properties via CCNG2, and could predict worse prognosis in pancreatic cancer patients. | |||
Key Molecule: Phosphatase and tensin homolog (PTEN) | [29] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell migration | Activation | hsa04670 | |
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
HPAC cells | Pancreas | Homo sapiens (Human) | CVCL_3517 | |
BxPc3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 | |
Capan cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
HPAF cells | Pancreas | Homo sapiens (Human) | CVCL_B284 | |
PL-45 cells | Pancreas | Homo sapiens (Human) | CVCL_3567 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Histone acetylation levels at miR-21 promoter were increased in PDAC cells after treatment with gemcitabine. Enhanced invasion and metastasis, increased miR-21 expression, decreased PTEN, elevated pAkT level were demonstrated in gemcitabine-resistant HPAC and PANC-1 cells. Pre-miR-21 transfection or TSA treatment further increased invasion and metastasis ability, decreased PTEN, and elevated pAkT levels in these two lines. In contrast, anti-miR-21 transfection could reverse invasion and metastasis, and PTEN and pAkT expressions induced by gemcitabine. | |||
Key Molecule: Apoptosis regulator BAX (BAX) | [30] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-365 directly targets the pro-apoptotic molecules SHC1 and BAX, whose reductions contribute to gemcitabine resistance in pancreatic cancer cells. | |||
Key Molecule: SHC-transforming protein 1 (SHC1) | [30] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-365 directly targets the pro-apoptotic molecules SHC1 and BAX, whose reductions contribute to gemcitabine resistance in pancreatic cancer cells. | |||
Key Molecule: Ubiquitin carboxyl-terminal hydrolase CYLD (CYLD) | [31] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell viability | Activation | hsa05200 | ||
NF-kappaB signaling pathway | Regulation | hsa04064 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
Panc1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
PSN1 cells | Pancreas | Homo sapiens (Human) | CVCL_1644 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-181b enhances the activity of NF-kB by inhibiting CYLD, thus leading to the resistance to gemcitabine. | |||
Key Molecule: SWI/SNF complex subunit SMARCC1 (SMARCC1) | [32] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell viability | Activation | hsa05200 | |
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
PSN1 cells | Pancreas | Homo sapiens (Human) | CVCL_1644 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-320c regulates the resistance of pancreatic cancer cells to gemcitabine through SMARCC1. | |||
Key Molecule: Tumor necrosis factor ligand superfamily member 6 (FASLG) | [28] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
FasL/Fas signaling pathway | Inhibition | hsa04210 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
In Vivo Model | BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
WST-8 assay | |||
Mechanism Description | Decreased Fas/FasL signaling mediates miR-21-induced chemoresistance in pancreatic cancer, over-expression of miR-21 reduced the endogenous expression of FasL anfd cause resistance to Gemcitabine. | |||
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) | [27] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | Upregulation of Bcl-2 expression was detected in cells transfected with miR-21 mimics, accompanied by downregulated Bax expression, less apoptosis, lower caspase-3 activity, decreased chemosensitivity to gemcitabine and increased proliferation. | |||
Key Molecule: RAC serine/threonine-protein kinase (AKT) | [26] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | AKT signaling pathway | Activation | hsa04151 | |
Cell apoptosis | Inhibition | hsa04210 | ||
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | LPc006 cells | Pancreas | Homo sapiens (Human) | N.A. |
LPc028 cells | Pancreas | Homo sapiens (Human) | N.A. | |
LPc033 cells | Pancreas | Homo sapiens (Human) | N.A. | |
LPc067 cells | Pancreas | Homo sapiens (Human) | N.A. | |
LPc111 cells | Pancreas | Homo sapiens (Human) | N.A. | |
LPc167 cells | Pancreas | Homo sapiens (Human) | N.A. | |
PP437 cells | Pancreas | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Fluorescence microscopy | |||
Mechanism Description | miR-21 regulates expression of PTEN and phosphorylation of its downstream kinase Akt and (b) the reduction of phospho-Akt (pAkt) correlated with the enhancement of gemcitabine-induced apoptosis and antitumor activity in vitro and in vivo, suggesting that Akt pathway plays a significant role in mediating drug resistance in PDAC cells. | |||
Key Molecule: Phosphatase and tensin homolog (PTEN) | [26] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | AKT signaling pathway | Activation | hsa04151 | |
Cell apoptosis | Inhibition | hsa04210 | ||
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | LPc006 cells | Pancreas | Homo sapiens (Human) | N.A. |
LPc028 cells | Pancreas | Homo sapiens (Human) | N.A. | |
LPc033 cells | Pancreas | Homo sapiens (Human) | N.A. | |
LPc067 cells | Pancreas | Homo sapiens (Human) | N.A. | |
LPc111 cells | Pancreas | Homo sapiens (Human) | N.A. | |
LPc167 cells | Pancreas | Homo sapiens (Human) | N.A. | |
PP437 cells | Pancreas | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Fluorescence microscopy | |||
Mechanism Description | miR-21 regulates expression of PTEN and phosphorylation of its downstream kinase Akt and (b) the reduction of phospho-Akt (pAkt) correlated with the enhancement of gemcitabine-induced apoptosis and antitumor activity in vitro and in vivo, suggesting that Akt pathway plays a significant role in mediating drug resistance in PDAC cells. | |||
Key Molecule: DNA excision repair protein ERCC-1 (ERCC1) | [16] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | Excision repair cross-complementation 1 (ERCC1) is a DNA repair endonuclease responsible for the incision of DNA cross-link-induced double-strand breaks. ERCC1 can repair gemcitabine-induced strand breaks, and its overexpression is well documented in poor gemcitabine responders. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-miR-663a | [37] | |||
Sensitive Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
T3-M4 cells | Pancreas | Homo sapiens (Human) | CVCL_VQ95 | |
Experiment for Molecule Alteration |
RT-PCR, qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Colony formation assay; Flow cytometry assay | |||
Mechanism Description | Upregulated miR-663 expression in PDAC cell lines promotes sensitivity to GEM. | |||
Key Molecule: hsa-mir-200b | [18] | |||
Sensitive Disease | Pancreatic carcinoma [ICD-11: 2C10.2] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell invasion | Inhibition | hsa05200 | ||
Cell metastasis | Inhibition | hsa05205 | ||
Cell proliferation | Inhibition | hsa05200 | ||
Chemosensitivity | Activation | hsa05207 | ||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Capan-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0026 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
Colorimetric methylene blue assay; Flow cytometry assay | |||
Mechanism Description | Forced expression of miR-200b induces CDH1 expression and promotes gemcitabine sensitivity in Capan-2 and Panc-1 cells. | |||
Key Molecule: hsa-miR-1207-5p | [38] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
Su.86.86 cells | Pancreas | Homo sapiens (Human) | CVCL_3881 | |
In Vivo Model | Engrafted tumor mouse model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | Overexpression of the miR-1207 pair improves gemcitabine efficacy in PC cells. | |||
Key Molecule: Pvt1 oncogene (PVT1) | [38] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
Su.86.86 cells | Pancreas | Homo sapiens (Human) | CVCL_3881 | |
In Vivo Model | Engrafted tumor mouse model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | PVT1 inhibition leads to improved efficacy of gemcitabine in PC cells. | |||
Key Molecule: hsa-mir-34 | [39] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Slug/PUMA signaling pathway | Regulation | hsa04390 | |
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
Flow Cytometric Analysis, MTT assay; TUNEL staining | |||
Mechanism Description | miR34 increases in vitro PANC-1 cell sensitivity to gemcitabine via targeting Slug/PUMA. miR34 enhances sensitization against gemcitabine-mediated apoptosis through the down-regulation of Slug expression, and up-regulation of Slug-dependent PUMA expression. | |||
Key Molecule: hsa-miR-429 | [40] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR429 sensitized gemcitabine response in GZ-resistant pancreatic cancer cells via its direct upregulation of PDCD4 expression. | |||
Key Molecule: Pvt1 oncogene (PVT1) | [41] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Muse Cell Cycle Assay; Muse Annexin V and Dead Cell Assay; MTT assay | |||
Mechanism Description | Curcumin sensitizes pancreatic cancer cells to gemcitabine by attenuating PRC2 subunit EZH2, and the LncRNA PVT1 expression. | |||
Key Molecule: hsa-let-7a | [42] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | CXCR4/let-7a/HMGA2 pathway | Regulation | hsa05206 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | HPDE6-C7 cells | Pancreas | Homo sapiens (Human) | CVCL_0P38 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay; Transwell assay; Flow cytometric analysis | |||
Mechanism Description | CXCR4/Let-7a axis regulates metastasis and chemoresistance of pancreatic cancer cells through targeting HMGA2. overexpression of HMGA2 restores cell proliferation, metastasis and chemosensitivity of gem inhibited by let-7a. | |||
Key Molecule: CXC chemokine receptor type 4 (CXCR4) | [42] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | CXCR4/let-7a/HMGA2 pathway | Regulation | hsa05206 | |
In Vitro Model | HPDE6-C7 cells | Pancreas | Homo sapiens (Human) | CVCL_0P38 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay; Transwell assay; Flow cytometric analysis | |||
Mechanism Description | CXCR4/Let-7a axis regulates metastasis and chemoresistance of pancreatic cancer cells through targeting HMGA2. overexpression of HMGA2 restores cell proliferation, metastasis and chemosensitivity of gem inhibited by let-7a. | |||
Key Molecule: hsa-mir-205 | [43] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | MIA PaCa-2R cells | Pancreas | Homo sapiens (Human) | CVCL_HA89 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometric analysis | |||
Mechanism Description | miR205 resensitizes GEM-resistant pancreatic cancer cells to GEM and acts as a tumor suppressor miRNA. | |||
Key Molecule: hsa-mir-21 | [44] | |||
Sensitive Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Panc02 cells | Pancreas | Homo sapiens (Human) | CVCL_D627 | |
In Vivo Model | Mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Costar Transwell Invasion Assay; | |||
Mechanism Description | Upregulating miR21 in CAFs promoted PDAC desmoplasia and increased its drug resistance to gemcitabine treatment by promoting the activation of cancer-associated fibroblasts (CAFs). miR21 mediates activation of CAFs via down-regulating PDCD4. | |||
Key Molecule: Programmed cell death protein 4 (PDCD4) | [44] | |||
Sensitive Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Panc02 cells | Pancreas | Homo sapiens (Human) | CVCL_D627 | |
In Vivo Model | Mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR; Immunofluorescence (IF) staining | |||
Experiment for Drug Resistance |
Costar Transwell Invasion Assay; | |||
Mechanism Description | Upregulating miR21 in CAFs promoted PDAC desmoplasia and increased its drug resistance to gemcitabine treatment by promoting the activation of cancer-associated fibroblasts (CAFs). miR21 mediates activation of CAFs via down-regulating PDCD4. | |||
Key Molecule: hsa-mir-153 | [45] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0026 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
CFPAC1 cells | Pancreas | Homo sapiens (Human) | CVCL_1119 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTS assay; Annexin-V/PI Apoptosis assay; TUNEL assay | |||
Mechanism Description | miR153 enhanced gemcitabine sensitivity by targeting Snail in pancreatic cancer. | |||
Key Molecule: hsa-mir-101 | [46] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Experiment for Molecule Alteration |
RT-qPCR | |||
Experiment for Drug Resistance |
MTT assay; Annexin V apoptosis assay; Caspase-3 activity assay | |||
Mechanism Description | microRNA-101 silences RNA-Pkcs and sensitizes pancreatic cancer cells to gemcitabine. AntagomiR101 expression causes RNA-Pkcs upregulation and gemcitabine resistance. miR101 expression inhibits Akt activation in PANC-1 cells. | |||
Key Molecule: hsa-mir-210 | [47] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
In Vivo Model | Chick egg xenograft model | Gallus gallus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
RealTime-Glo MT Cell Viability Assay; Caspase-3/7 substrate assay; Colony formation assay | |||
Mechanism Description | microRNA-210 overexpression inhibits tumor growth and potentially reverses gemcitabine resistance in pancreatic cancer, miR210 is a direct suppressor of the multidrug efflux transporter ABCC5. | |||
Key Molecule: hsa-mir-145 | [48] | |||
Sensitive Disease | Pancreatic adenocarcinoma [ICD-11: 2C10.4] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Experiment for Molecule Alteration |
RT-PCR; qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Transwell migration assay | |||
Mechanism Description | miRNA-145 increases therapeutic sensibility to gemcitabine treatment of pancreatic adenocarcinoma cells, miR145 negatively regulated p70S6k1 expression at the posttranscriptional level in colon cancer. | |||
Key Molecule: hsa-miR-20a-5p | [49] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell viability | Inhibition | hsa05200 | ||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay | |||
Mechanism Description | miR-20a-5p inhibits protein expression of RRM2 and reverses gemcitabine resistance. | |||
Key Molecule: hsa-mir-30a | [50] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell colony | Inhibition | hsa05200 | ||
Cell proliferation | Inhibition | hsa05200 | ||
SNAI1/IRS1/AKT signaling pathway | Regulation | hsa04151 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
In Vivo Model | BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTS assay | |||
Mechanism Description | miR-30a overexpression suppresses cell proliferation, and sensitizes pancreatic cancer cells to gemcitabine and miR-30a overexpression reduced IRS1 and SNAI1 protein level. | |||
Key Molecule: hsa-miR-373-3p | [51] | |||
Sensitive Disease | Pancreatic carcinoma [ICD-11: 2C10.2] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
Cell viability | Inhibition | hsa05200 | ||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay | |||
Mechanism Description | miR-373-3p enhances the chemosensitivity of gemcitabine through cell cycle pathway by downregulating CCND2 in pancreatic carcinoma cells. | |||
Key Molecule: hsa-mir-29c | [52] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell colony | Inhibition | hsa05200 | ||
Cell viability | Inhibition | hsa05200 | ||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay | |||
Mechanism Description | miR-29c targets USP22 and suppresses autophagy-mediated chemoresistance in a xenograft tumor model in vivo. | |||
Key Molecule: hsa-miR-101-3p | [53] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTS assay | |||
Mechanism Description | Long-term treatment of PDA cells with gemcitabine induced pronounced therapy resistance. The RRM1 gene is a major mediator of resistance and its expression is regulated by direct binding of miR-101-3p to two binding sites in the RRM1 3'UTR. The overexpression of miR-101-3p mimics inhibited the expression of RRM1 and partially reversed gemcitabine-resistance. | |||
Key Molecule: hsa-let-7b | [54] | |||
Sensitive Disease | KRAS mutant pancreatic ductal adenocarcinoma [ICD-11: 2C10.5] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell invasion | Inhibition | hsa05200 | ||
Cell migration | Inhibition | hsa04670 | ||
MEK/ERK /PI3K/AKT signaling pathway | Inhibition | hsa04151 | ||
In Vitro Model | A549 cells | Lung | Homo sapiens (Human) | CVCL_0023 |
NCI-H1975 cells | Lung | Homo sapiens (Human) | CVCL_1511 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | Let-7b repletion selectively sensitized kRAS mutant tumor cells to the cytotoxicity of paclitaxel and gemcitabine. Transfection of let-7b mimic downregulated the expression of mutant but not wild-type kRAS. Combination of let-7b mimic with paclitaxel or gemcitabine diminished MEk/ERk and PI3k/AkT signaling concurrently, triggered the onset of apoptosis, and reverted the epithelial-mesenchymal transition in kRAS mutant tumor cells. In addition, let-7b repletion downregulated the expression of beta-tubulin III and ribonucleotide reductase subunit M2, two proteins known to mediate tumor resistance to paclitaxel and gemcitabine, respectively. Let-7 may represent a new class of chemosensitizer for the treatment of kRAS mutant tumors. | |||
Key Molecule: hsa-mir-21 | [55] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | PI3K/AKT signaling pathway | Inhibition | hsa04151 | |
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Hs-578T cells | Breast | Homo sapiens (Human) | CVCL_0332 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Increased p85alpha expression in PDAC TCs results in decreased PI3k-AkT signaling and increased gemcitabine sensitivity. Expression of p85alpha inversely correlates with miR-21 levels in human PDAC. Overexpression of miR-21 results in decreased levels of p85alpha and increased PI3k-AkT activation. | |||
Key Molecule: hsa-mir-497 | [5] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
FGF/FGFR signaling pathway | Inhibition | hsa01521 | ||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | miR-497 suppressed cells proliferation, decreased the percentage of S phase cells, re-sensitized cells to gemcitabine and erlotinib, and attenuated migration and invasion capacities. Furthermore, fibroblast growth factor 2 and fibroblast growth factor receptor 1 were confirmed as miR-497 targets. Overexpression of miR-497 inhibited tumor growth in vivo. Additionally, miR-497 expression was significantly downregulated in pancreatic cancer tissues compared with tumor-adjacent samples. Low expression of miR-497 was an independent adverse prognostic factor of pancreatic cancer. miR-497 plays a role in modulating the malignant phenotype and chemosensitivity of pancreatic cancer cells by directly inhibition of FGF2 and FGFR1 expression. | |||
Key Molecule: hsa-mir-211 | [56] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
In Vitro Model | Suit2 cells | Pancreas | Homo sapiens (Human) | CVCL_3172 |
SUIT2-007 cells | Pancreas | Homo sapiens (Human) | CVCL_B279 | |
SUIT2-028 cells | Pancreas | Homo sapiens (Human) | CVCL_B282 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Transwell assay | |||
Mechanism Description | The induction of the miR-211 expression in the cells increased the sensitivity to gemcitabine and reduced the expression of its target ribonucleotide reductase subunit 2 (RRM2). | |||
Key Molecule: hsa-mir-29a | [57] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell proliferation | Inhibition | hsa05200 | ||
Wnt/Beta-catenin signaling pathway | Inhibition | hsa04310 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
PSN1 cells | Pancreas | Homo sapiens (Human) | CVCL_1644 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Our findings suggest that miR-29a expression correlates significantly with the growth-inhibitory effect of GEM and that activation of the Wnt/beta-catenin signaling pathway mediated the miR-29a-induced resistance to GEM in pancreatic cancer cell lines. | |||
Key Molecule: hsa-mir-181 | [58] | |||
Sensitive Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
In Vitro Model | SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 |
CFPAC1 cells | Pancreas | Homo sapiens (Human) | CVCL_1119 | |
In Vivo Model | BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | BCL-2 facilitates cell survival against chemotherapy via the blockage of Bax/Bak-induced apoptosis, miRNA-181b sensitizes PDAC cells to gemcitabine by targeting BCL-2. | |||
Key Molecule: hsa-miR-142-5p | [59] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 |
SUIT-2 cells | Pancreas | Homo sapiens (Human) | CVCL_3172 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Propidium iodide assay | |||
Mechanism Description | High miR-142-5p expression was significantly associated with longer survival times in the gemcitabine group. | |||
Key Molecule: Pvt1 oncogene (PVT1) | [60] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 |
Experiment for Molecule Alteration |
PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Genome-wide screening identified PVT1 as a regulator for Gemcitabine sensitivity in human pancreatic cancer cells. | |||
Key Molecule: hsa-mir-21 | [15] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
HPAC cells | Pancreas | Homo sapiens (Human) | CVCL_3517 | |
HPAF-II cells | Pancreatic | Homo sapiens (Human) | CVCL_0313 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
SRB (sulforhodamine-B) assay | |||
Mechanism Description | Low miR-21 expression was associated with benefit from adjuvant treatment in two independent cohorts of PDAC cases, and anti-miR-21 increased anticancer drug activity in vitro. | |||
Key Molecule: hsa-mir-21 | [61] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
Capan-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0026 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
Hs-578T cells | Breast | Homo sapiens (Human) | CVCL_0332 | |
CFPAC1 cells | Pancreas | Homo sapiens (Human) | CVCL_1119 | |
SUIT-2 cells | Pancreas | Homo sapiens (Human) | CVCL_3172 | |
H48N cells | Pancreas | Homo sapiens (Human) | CVCL_D554 | |
KP-1N cells | Pancreas | Homo sapiens (Human) | CVCL_3002 | |
KP-2 cells | Pancreas | Homo sapiens (Human) | CVCL_3004 | |
KP-3 cells | Pancreas | Homo sapiens (Human) | CVCL_3005 | |
NOR-P1 cells | Pancreas | Homo sapiens (Human) | CVCL_4716 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Propidium iodide assay | |||
Mechanism Description | The cancer cells transfected with the miR-21 precursor showed significantly increased proliferation, Matrigel invasion, and chemoresistance for gemcitabine compared with the control cells. In contrast, inhibition of miR-21 decreased proliferation, Matrigel invasion, and chemoresistance for gemcitabine. Moreover, miR-21 positively correlated with the mRNA expression of invasion-related genes, matrix metalloproteinase-2 and -9, and vascular endothelial growth factor. | |||
Irregularity in Drug Uptake and Drug Efflux (IDUE) | ||||
Key Molecule: ATP-binding cassette sub-family C5 (ABCC5) | [47] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
In Vivo Model | Chick egg xenograft model | Gallus gallus | ||
Experiment for Molecule Alteration |
Dual luciferase assay; qRT-PCR; Immunofluorescence and immunohistochemistry assay | |||
Experiment for Drug Resistance |
RealTime-Glo MT Cell Viability Assay; Caspase-3/7 substrate assay; Colony formation assay | |||
Mechanism Description | microRNA-210 overexpression inhibits tumor growth and potentially reverses gemcitabine resistance in pancreatic cancer, miR210 is a direct suppressor of the multidrug efflux transporter ABCC5. | |||
Regulation by the Disease Microenvironment (RTDM) | ||||
Key Molecule: Growth arrest specific 5 (GAS5) | [33] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
miR221/SOCS3 signaling pathway | Regulation | hsa05206 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0026 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis; RT-qPCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | Overexpression of GAS5 inhibits proliferation, migration, and chemotherapy resistance by suppressing the emt and tumor stem cell-like properties. LncRNA GAS5 functioned as a competing endogenous RNA for miR-221, and it suppressed cell growth, metastasis, and gemcitabine resistance in PC by regulating the miR-221/SOCS3 pathway mediating EMT and tumor stem cell self-renewal. | |||
Key Molecule: Suppressor of cytokine signaling 3 (SOCS3) | [33] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
miR221/SOCS3 signaling pathway | Regulation | hsa05206 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0026 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
Experiment for Molecule Alteration |
Western blot analysis; RT-qPCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | SOCS3 overexpression reverses miR-221 overexpression-induced proliferation, migration, emt, chemotherapy resistance, and stem cell-like properties in panc-1 cells. | |||
Key Molecule: hsa-mir-3656 | [62] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Epithelial mesenchymal transition signaling pathway | Activation | hsa01521 | |
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0026 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
HPDE6-C7 cells | Pancreas | Homo sapiens (Human) | CVCL_0P38 | |
HTERT-HPNE cells | Pancreas | Homo sapiens (Human) | CVCL_C466 | |
PATU8988 cells | Pancreas | Homo sapiens (Human) | CVCL_1846 | |
CFPAC1 cells | Pancreas | Homo sapiens (Human) | CVCL_1119 | |
HPAC cells | Pancreas | Homo sapiens (Human) | CVCL_3517 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
MTS assay | |||
Mechanism Description | miR3656 expression enhances the chemosensitivity of pancreatic cancer to gemcitabine through modulation of the RHOF/EMT axis. miR3656 could target RHOF, a member of the Rho subfamily of small GTPases, and regulate the EMT process, enforced EMT progression via TWIST1 overexpression compromised the chemotherapy-enhancing effects of miR3656. Reduced miR3656 expression levels activated the EMT pathway through upregulation of RHOF, eventually causing drug resistance. | |||
Key Molecule: Rho-related GTP-binding protein RhoF (RHOF) | [62] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Epithelial mesenchymal transition signaling pathway | Activation | hsa01521 | |
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0026 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
HPDE6-C7 cells | Pancreas | Homo sapiens (Human) | CVCL_0P38 | |
HTERT-HPNE cells | Pancreas | Homo sapiens (Human) | CVCL_C466 | |
PATU8988 cells | Pancreas | Homo sapiens (Human) | CVCL_1846 | |
CFPAC1 cells | Pancreas | Homo sapiens (Human) | CVCL_1119 | |
HPAC cells | Pancreas | Homo sapiens (Human) | CVCL_3517 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis; Dual luciferase reporter assay | |||
Experiment for Drug Resistance |
MTS assay | |||
Mechanism Description | miR3656 expression enhances the chemosensitivity of pancreatic cancer to gemcitabine through modulation of the RHOF/EMT axis. miR3656 could target RHOF, a member of the Rho subfamily of small GTPases, and regulate the EMT process, enforced EMT progression via TWIST1 overexpression compromised the chemotherapy-enhancing effects of miR3656. Reduced miR3656 expression levels activated the EMT pathway through upregulation of RHOF, eventually causing drug resistance. | |||
Key Molecule: hsa-miR-1243 | [63] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
Epithelial mesenchymal transition signaling pathway | Inhibition | hsa01521 | ||
TGF-beta signaling pathway | Inhibition | hsa04350 | ||
In Vitro Model | MDA-MB-231 cells | Breast | Homo sapiens (Human) | CVCL_0062 |
BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 | |
Su.86.86 cells | Pancreas | Homo sapiens (Human) | CVCL_3881 | |
CFPAC1 cells | Pancreas | Homo sapiens (Human) | CVCL_1119 | |
KMP3 cells | Pancreas | Homo sapiens (Human) | CVCL_8491 | |
KP4-4 cells | Pancreas | Homo sapiens (Human) | CVCL_Y142 | |
Panc1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
WST-8 assay; Crystal violet staining assay | |||
Mechanism Description | Overexpression of miR509-5p and miR1243 increased the expression of E-cadherin through the suppression of EMT-related gene expression and that drug sensitivity increased with a combination of each of these miRNAs and gemcitabine. miR1243 directly regulated SMAD2 and SMAD4, which regulate the TGF-beta signaling pathway, resulting in an induction of the MET phenotype. Suppressing SMADs reduced the effect of TGF-beta. | |||
Key Molecule: hsa-miR-509-5p | [63] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
Epithelial mesenchymal transition signaling pathway | Inhibition | hsa01521 | ||
TGF-beta signaling pathway | Inhibition | hsa04350 | ||
In Vitro Model | MDA-MB-231 cells | Breast | Homo sapiens (Human) | CVCL_0062 |
BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 | |
Su.86.86 cells | Pancreas | Homo sapiens (Human) | CVCL_3881 | |
CFPAC1 cells | Pancreas | Homo sapiens (Human) | CVCL_1119 | |
KMP3 cells | Pancreas | Homo sapiens (Human) | CVCL_8491 | |
KP4-4 cells | Pancreas | Homo sapiens (Human) | CVCL_Y142 | |
Panc1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
WST-8 assay; Crystal violet staining assay | |||
Mechanism Description | miR509-5p and miR1243 increase the sensitivity to gemcitabine by inhibiting epithelial-mesenchymal transition in pancreatic cancer miR509-5p induced an MET phenotype by directly regulating VIM and HMGA2. | |||
Key Molecule: Mothers against decapentaplegic homolog 2 (SMAD2) | [63] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
Epithelial mesenchymal transition signaling pathway | Inhibition | hsa01521 | ||
TGF-beta signaling pathway | Inhibition | hsa04350 | ||
In Vitro Model | MDA-MB-231 cells | Breast | Homo sapiens (Human) | CVCL_0062 |
BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 | |
Su.86.86 cells | Pancreas | Homo sapiens (Human) | CVCL_3881 | |
CFPAC1 cells | Pancreas | Homo sapiens (Human) | CVCL_1119 | |
KMP3 cells | Pancreas | Homo sapiens (Human) | CVCL_8491 | |
KP4-4 cells | Pancreas | Homo sapiens (Human) | CVCL_Y142 | |
Panc1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
WST-8 assay; Crystal violet staining assay | |||
Mechanism Description | Overexpression of miR509-5p and miR1243 increased the expression of E-cadherin through the suppression of EMT-related gene expression and that drug sensitivity increased with a combination of each of these miRNAs and gemcitabine. miR1243 directly regulated SMAD2 and SMAD4, which regulate the TGF-beta signaling pathway, resulting in an induction of the MET phenotype. Suppressing SMADs reduced the effect of TGF-beta. | |||
Key Molecule: Mothers against decapentaplegic homolog 4 (SMAD4) | [63] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
Epithelial mesenchymal transition signaling pathway | Inhibition | hsa01521 | ||
TGF-beta signaling pathway | Inhibition | hsa04350 | ||
In Vitro Model | MDA-MB-231 cells | Breast | Homo sapiens (Human) | CVCL_0062 |
BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 | |
Su.86.86 cells | Pancreas | Homo sapiens (Human) | CVCL_3881 | |
CFPAC1 cells | Pancreas | Homo sapiens (Human) | CVCL_1119 | |
KMP3 cells | Pancreas | Homo sapiens (Human) | CVCL_8491 | |
KP4-4 cells | Pancreas | Homo sapiens (Human) | CVCL_Y142 | |
Panc1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
WST-8 assay; Crystal violet staining assay | |||
Mechanism Description | Overexpression of miR509-5p and miR1243 increased the expression of E-cadherin through the suppression of EMT-related gene expression and that drug sensitivity increased with a combination of each of these miRNAs and gemcitabine. miR1243 directly regulated SMAD2 and SMAD4, which regulate the TGF-beta signaling pathway, resulting in an induction of the MET phenotype. Suppressing SMADs reduced the effect of TGF-beta. | |||
Key Molecule: hsa-mir-155 | [64] | |||
Sensitive Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Panc1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
PSN1 cells | Pancreas | Homo sapiens (Human) | CVCL_1644 | |
In Vivo Model | Mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | The increase of miR155 induced two different functions; exosome secretion and chemoresistance ability via facilitating the anti-apoptotic activity. | |||
Key Molecule: Tyrosine-protein kinase Fyn (FYN) | [65] | |||
Sensitive Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
Cell viability | Inhibition | hsa05200 | ||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
PATU8988T cells | Pancreatic | Homo sapiens (Human) | CVCL_1847 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay; Transwell assay | |||
Mechanism Description | miR-125a-3p is responsible for chemosensitivity in PDAC by inhibiting epithelial-mesenchymal transition via Fyn. | |||
Key Molecule: hsa-miR-125a-3p | [65] | |||
Sensitive Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell invasion | Inhibition | hsa05200 | ||
Cell migration | Inhibition | hsa04670 | ||
Cell viability | Inhibition | hsa05200 | ||
Epithelial mesenchymal transition signaling pathway | Inhibition | hsa01521 | ||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
PATU8988T cells | Pancreatic | Homo sapiens (Human) | CVCL_1847 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay; Transwell assay | |||
Mechanism Description | miR-125a-3p is responsible for chemosensitivity in PDAC by inhibiting epithelial-mesenchymal transition via Fyn. | |||
Key Molecule: hsa-mir-223 | [66] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay; Transwell migration and invasion assay | |||
Mechanism Description | Down-regulation of miR-223 reverses epithelial-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells due to down-regulation of its target Fbw7 and subsequent upregulation of Notch-1, which enhances GR cells to gemcitabine sensitivity. | |||
Key Molecule: hsa-let-7b | [67] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
WST-1 assay | |||
Mechanism Description | The expression of miR-200b, miR-200c, let-7b, let-7c, let-7d, and let-7e was significantly down-regulated in gemcitabine-resistant cells that showed EMT characteristics such as elongated fibroblastoid morphology, lower expression of epithelial marker E-cadherin, and higher expression of mesenchymal markers such as vimentin and ZEB1. | |||
Key Molecule: hsa-let-7c | [67] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
WST-1 assay | |||
Mechanism Description | The expression of miR-200b, miR-200c, let-7b, let-7c, let-7d, and let-7e was significantly down-regulated in gemcitabine-resistant cells that showed EMT characteristics such as elongated fibroblastoid morphology, lower expression of epithelial marker E-cadherin, and higher expression of mesenchymal markers such as vimentin and ZEB1. | |||
Key Molecule: hsa-let-7d | [67] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
WST-1 assay | |||
Mechanism Description | The expression of miR-200b, miR-200c, let-7b, let-7c, let-7d, and let-7e was significantly down-regulated in gemcitabine-resistant cells that showed EMT characteristics such as elongated fibroblastoid morphology, lower expression of epithelial marker E-cadherin, and higher expression of mesenchymal markers such as vimentin and ZEB1. | |||
Key Molecule: hsa-let-7e | [67] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
WST-1 assay | |||
Mechanism Description | The expression of miR-200b, miR-200c, let-7b, let-7c, let-7d, and let-7e was significantly down-regulated in gemcitabine-resistant cells that showed EMT characteristics such as elongated fibroblastoid morphology, lower expression of epithelial marker E-cadherin, and higher expression of mesenchymal markers such as vimentin and ZEB1. | |||
Key Molecule: hsa-mir-200b | [67] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
WST-1 assay | |||
Mechanism Description | Re-expression of miR-200 in gemcitabine-resistant cells showed partial reversal of EMT characteristics as documented by increased expression of E-cadherin and decreased expression of vimentin, ZEB1, and slug. These results suggest that miR-200 family regulates the expression of ZEB1, slug, E-cadherin, and vimentin and that the re-expression of miR-200 could be useful for the reversal of EMT phenotype to mesenchymal-epithelial transition (MET). re-expression of miR-200 by transfection studies or treatment of gemcitabine-resistant cells with either DIM or isoflavone resulted in the down-regulation of ZEB1, slug, and vimentin, which was consistent with morphological reversal of EMT phenotype leading to epithelial morphology. | |||
Key Molecule: hsa-mir-200c | [67] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
WST-1 assay | |||
Mechanism Description | Re-expression of miR-200 in gemcitabine-resistant cells showed partial reversal of EMT characteristics as documented by increased expression of E-cadherin and decreased expression of vimentin, ZEB1, and slug. These results suggest that miR-200 family regulates the expression of ZEB1, slug, E-cadherin, and vimentin and that the re-expression of miR-200 could be useful for the reversal of EMT phenotype to mesenchymal-epithelial transition (MET). re-expression of miR-200 by transfection studies or treatment of gemcitabine-resistant cells with either DIM or isoflavone resulted in the down-regulation of ZEB1, slug, and vimentin, which was consistent with morphological reversal of EMT phenotype leading to epithelial morphology. | |||
Key Molecule: Zinc finger E-box-binding homeobox 1 (ZEB1) | [67] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
WST-1 assay | |||
Mechanism Description | Re-expression of miR-200 in gemcitabine-resistant cells showed partial reversal of EMT characteristics as documented by increased expression of E-cadherin and decreased expression of vimentin, ZEB1, and slug. These results suggest that miR-200 family regulates the expression of ZEB1, slug, E-cadherin, and vimentin and that the re-expression of miR-200 could be useful for the reversal of EMT phenotype to mesenchymal-epithelial transition (MET). re-expression of miR-200 by transfection studies or treatment of gemcitabine-resistant cells with either DIM or isoflavone resulted in the down-regulation of ZEB1, slug, and vimentin, which was consistent with morphological reversal of EMT phenotype leading to epithelial morphology. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Cadherin-1 (CDH1) | [18] | |||
Sensitive Disease | Pancreatic carcinoma [ICD-11: 2C10.2] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell invasion | Inhibition | hsa05200 | ||
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Capan-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0026 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Colorimetric methylene blue assay; Flow cytometry assay | |||
Mechanism Description | Forced expression of miR-200b induces CDH1 expression and promotes gemcitabine sensitivity in Capan-2 and Panc-1 cells. | |||
Key Molecule: Transforming protein RhoA (RHOA) | [38] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
Su.86.86 cells | Pancreas | Homo sapiens (Human) | CVCL_3881 | |
In Vivo Model | Engrafted tumor mouse model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis; qRT-PCR; IHC analyses | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | RhoA inhibition leads to improved efficacy of gemcitabine in PC cells. | |||
Key Molecule: Proto-oncogene tyrosine-protein kinase Src (SRC) | [38] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
Su.86.86 cells | Pancreas | Homo sapiens (Human) | CVCL_3881 | |
In Vivo Model | Engrafted tumor mouse model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis; qRT-PCR; IHC analyses | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | SRC inhibition leads to improved efficacy of gemcitabine in PC cells. | |||
Key Molecule: High mobility group protein HMGI-C (HMGA2) | [63], [42] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | CXCR4/let-7a/HMGA2 pathway | Regulation | hsa05206 | |
In Vitro Model | HPDE6-C7 cells | Pancreas | Homo sapiens (Human) | CVCL_0P38 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR; Western blot analysis; Luciferase reporter assay | |||
Experiment for Drug Resistance |
MTT assay; Transwell assay; Flow cytometric analysis | |||
Mechanism Description | CXCR4/Let-7a axis regulates metastasis and chemoresistance of pancreatic cancer cells through targeting HMGA2. overexpression of HMGA2 restores cell proliferation, metastasis and chemosensitivity of gem inhibited by let-7a. | |||
Key Molecule: Bcl-2-binding component 3 (BBC3) | [39] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Slug/PUMA signaling pathway | Activation | hsa04390 | |
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Experiment for Molecule Alteration |
RT-PCR; Western blot analysis | |||
Experiment for Drug Resistance |
Flow Cytometric Analysis, MTT assay; TUNEL staining | |||
Mechanism Description | miR34 induces Slug-mediated upregulation of PUMA expression. miR34 sensitizes to gemcitabine-mediated apoptosis by PUMA upregulation. | |||
Key Molecule: Zinc finger protein SNAI2 (SNAI2) | [39] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Slug/PUMA signaling pathway | Regulation | hsa04390 | |
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Experiment for Molecule Alteration |
RT-PCR; Western blot analysis | |||
Experiment for Drug Resistance |
Flow Cytometric Analysis, MTT assay; TUNEL staining | |||
Mechanism Description | miR34 increases in vitro PANC-1 cell sensitivity to gemcitabine via targeting Slug/PUMA. miR34 enhances sensitization against gemcitabine-mediated apoptosis through the down-regulation of Slug expression, and up-regulation of Slug-dependent PUMA expression. | |||
Key Molecule: Programmed cell death protein 4 (PDCD4) | [40] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR429 sensitized gemcitabine response in GZ-resistant pancreatic cancer cells via its direct upregulation of PDCD4 expression. | |||
Key Molecule: Zinc finger protein SNAI1 (SNAI1) | [45] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0026 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
CFPAC1 cells | Pancreas | Homo sapiens (Human) | CVCL_1119 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTS assay; Annexin-V/PI Apoptosis assay; TUNEL assay | |||
Mechanism Description | miR153 enhanced gemcitabine sensitivity by targeting Snail in pancreatic cancer. | |||
Key Molecule: DNA-dependent catalytic protein kinase (PRKDC) | [46] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; Annexin V apoptosis assay; Caspase-3 activity assay | |||
Mechanism Description | microRNA-101 silences RNA-Pkcs and sensitizes pancreatic cancer cells to gemcitabine. AntagomiR101 expression causes RNA-Pkcs upregulation and gemcitabine resistance. miR101 expression inhibits Akt activation in PANC-1 cells. | |||
Key Molecule: Ribosomal protein S6 kinase beta-1 (RPS6KB1) | [48] | |||
Sensitive Disease | Pancreatic adenocarcinoma [ICD-11: 2C10.4] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Transwell migration assay | |||
Mechanism Description | miRNA-145 increases therapeutic sensibility to gemcitabine treatment of pancreatic adenocarcinoma cells, miR145 negatively regulated p70S6k1 expression at the posttranscriptional level in colon cancer. | |||
Key Molecule: Ribonucleoside-diphosphate reductase subunit M2 (RRM2) | [49] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell viability | Inhibition | hsa05200 | ||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay | |||
Mechanism Description | miR-20a-5p inhibits protein expression of RRM2 and reverses gemcitabine resistance. | |||
Key Molecule: Insulin receptor substrate 1 (IRS1) | [50] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell colony | Inhibition | hsa05200 | |
Cell proliferation | Inhibition | hsa05200 | ||
SNAI1/IRS1/AKT signaling pathway | Regulation | hsa04151 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
In Vivo Model | BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTS assay | |||
Mechanism Description | miR-30a overexpression suppresses cell proliferation, and sensitizes pancreatic cancer cells to gemcitabine and miR-30a overexpression reduced IRS1 and SNAI1 protein level. | |||
Key Molecule: Zinc finger protein SNAI1 (SNAI1) | [50] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell colony | Inhibition | hsa05200 | |
Cell proliferation | Inhibition | hsa05200 | ||
SNAI1/IRS1/AKT signaling pathway | Regulation | hsa04151 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
In Vivo Model | BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTS assay | |||
Mechanism Description | miR-30a overexpression suppresses cell proliferation, and sensitizes pancreatic cancer cells to gemcitabine and miR-30a overexpression reduced IRS1 and SNAI1 protein level. | |||
Key Molecule: G1/S-specific cyclin-D2 (CCND2) | [51] | |||
Sensitive Disease | Pancreatic carcinoma [ICD-11: 2C10.2] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell invasion | Inhibition | hsa05200 | ||
Cell migration | Inhibition | hsa04670 | ||
Cell viability | Inhibition | hsa05200 | ||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay | |||
Mechanism Description | miR-373-3p enhances the chemosensitivity of gemcitabine through cell cycle pathway by downregulating CCND2 in pancreatic carcinoma cells. | |||
Key Molecule: Ubiquitin carboxyl-terminal hydrolase 22 (USP22) | [52] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell colony | Inhibition | hsa05200 | ||
Cell viability | Inhibition | hsa05200 | ||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis; RIP assay; Luciferase reporter assay | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay | |||
Mechanism Description | miR-29c targets USP22 and suppresses autophagy-mediated chemoresistance in a xenograft tumor model in vivo. | |||
Key Molecule: Ribonucleoside-diphosphate reductase large subunit (RRM1) | [53] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTS assay | |||
Mechanism Description | Long-term treatment of PDA cells with gemcitabine induced pronounced therapy resistance. The RRM1 gene is a major mediator of resistance and its expression is regulated by direct binding of miR-101-3p to two binding sites in the RRM1 3'UTR. The overexpression of miR-101-3p mimics inhibited the expression of RRM1 and partially reversed gemcitabine-resistance. | |||
Key Molecule: Ribonucleoside-diphosphate reductase subunit M2 (RRM2) | [54] | |||
Sensitive Disease | KRAS mutant pancreatic ductal adenocarcinoma [ICD-11: 2C10.5] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
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) | [54] | |||
Sensitive Disease | KRAS mutant pancreatic ductal adenocarcinoma [ICD-11: 2C10.5] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell invasion | Inhibition | hsa05200 | ||
Cell migration | Inhibition | hsa04670 | ||
MEK/ERK /PI3K/AKT signaling pathway | Inhibition | hsa04151 | ||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | Let-7b repletion selectively sensitized kRAS mutant tumor cells to the cytotoxicity of paclitaxel and gemcitabine. Transfection of let-7b mimic downregulated the expression of mutant but not wild-type kRAS. Combination of let-7b mimic with paclitaxel or gemcitabine diminished MEk/ERk and PI3k/AkT signaling concurrently, triggered the onset of apoptosis, and reverted the epithelial-mesenchymal transition in kRAS mutant tumor cells. In addition, let-7b repletion downregulated the expression of beta-tubulin III and ribonucleotide reductase subunit M2, two proteins known to mediate tumor resistance to paclitaxel and gemcitabine, respectively. Let-7 may represent a new class of chemosensitizer for the treatment of kRAS mutant tumors. | |||
Key Molecule: PI3-kinase regulatory subunit alpha (PIK3R1) | [55] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | PI3K/AKT signaling pathway | Inhibition | hsa04151 | |
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Hs-578T cells | Breast | Homo sapiens (Human) | CVCL_0332 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Increased p85alpha expression in PDAC TCs results in decreased PI3k-AkT signaling and increased gemcitabine sensitivity. Expression of p85alpha inversely correlates with miR-21 levels in human PDAC. Overexpression of miR-21 results in decreased levels of p85alpha and increased PI3k-AkT activation. | |||
Key Molecule: F-box/WD repeat-containing protein 7 (FBXW7) | [66] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; Transwell migration and invasion assay | |||
Mechanism Description | Down-regulation of miR-223 reverses epithelial-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells due to down-regulation of its target Fbw7 and subsequent upregulation of Notch-1, which enhances GR cells to gemcitabine sensitivity. | |||
Key Molecule: Fibroblast growth factor 2 (FGF1) | [5] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
FGF/FGFR signaling pathway | Inhibition | hsa01521 | ||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | miR-497 suppressed cells proliferation, decreased the percentage of S phase cells, re-sensitized cells to gemcitabine and erlotinib, and attenuated migration and invasion capacities. Furthermore, fibroblast growth factor 2 and fibroblast growth factor receptor 1 were confirmed as miR-497 targets. Overexpression of miR-497 inhibited tumor growth in vivo. Additionally, miR-497 expression was significantly downregulated in pancreatic cancer tissues compared with tumor-adjacent samples. Low expression of miR-497 was an independent adverse prognostic factor of pancreatic cancer. miR-497 plays a role in modulating the malignant phenotype and chemosensitivity of pancreatic cancer cells by directly inhibition of FGF2 and FGFR1 expression. | |||
Key Molecule: Fibroblast growth factor receptor 1 (FGFR1) | [5] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
FGF/FGFR signaling pathway | Inhibition | hsa01521 | ||
In Vitro Model | MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 |
SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | miR-497 suppressed cells proliferation, decreased the percentage of S phase cells, re-sensitized cells to gemcitabine and erlotinib, and attenuated migration and invasion capacities. Furthermore, fibroblast growth factor 2 and fibroblast growth factor receptor 1 were confirmed as miR-497 targets. Overexpression of miR-497 inhibited tumor growth in vivo. Additionally, miR-497 expression was significantly downregulated in pancreatic cancer tissues compared with tumor-adjacent samples. Low expression of miR-497 was an independent adverse prognostic factor of pancreatic cancer. miR-497 plays a role in modulating the malignant phenotype and chemosensitivity of pancreatic cancer cells by directly inhibition of FGF2 and FGFR1 expression. | |||
Key Molecule: Ribonucleoside-diphosphate reductase subunit M2 (RRM2) | [56] | |||
Sensitive Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
In Vitro Model | Suit2 cells | Pancreas | Homo sapiens (Human) | CVCL_3172 |
SUIT2-007 cells | Pancreas | Homo sapiens (Human) | CVCL_B279 | |
SUIT2-028 cells | Pancreas | Homo sapiens (Human) | CVCL_B282 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Transwell assay | |||
Mechanism Description | The induction of the miR-211 expression in the cells increased the sensitivity to gemcitabine and reduced the expression of its target ribonucleotide reductase subunit 2 (RRM2). | |||
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) | [58] | |||
Sensitive Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Gemcitabine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
In Vitro Model | SW1990 cells | Pancreas | Homo sapiens (Human) | CVCL_1723 |
CFPAC1 cells | Pancreas | Homo sapiens (Human) | CVCL_1119 | |
In Vivo Model | BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | BCL-2 facilitates cell survival against chemotherapy via the blockage of Bax/Bak-induced apoptosis, miRNA-181b sensitizes PDAC cells to gemcitabine by targeting BCL-2. |
Paclitaxel
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-mir-181c | [12] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Paclitaxel | |||
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) | [12] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Paclitaxel | |||
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) | [12] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Paclitaxel | |||
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) | [12] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Paclitaxel | |||
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) | [12] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Paclitaxel | |||
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 | [54] | |||
Sensitive Disease | KRAS mutant pancreatic ductal adenocarcinoma [ICD-11: 2C10.5] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Paclitaxel | |||
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) | [54] | |||
Sensitive Disease | KRAS mutant pancreatic ductal adenocarcinoma [ICD-11: 2C10.5] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Paclitaxel | |||
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) | [54] | |||
Sensitive Disease | KRAS mutant pancreatic ductal adenocarcinoma [ICD-11: 2C10.5] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Paclitaxel | |||
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. |
Penfluridol
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Autophagy-related protein LC3 B (MAP1LC3B) | [68] | |||
Sensitive Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Penfluridol | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
In Vitro Model | A549/Taxol cells | Lung | Homo sapiens (Human) | CVCL_W218 |
SW403 cells | Colon | Homo sapiens (Human) | CVCL_0545 | |
In Vivo Model | Athymic nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
SRB assay | |||
Mechanism Description | One of the hallmarks of autophagy is the accumulation of LC3B and its localization in vesicular structures. We observed that penfluridol treatment enhanced the expression of LC3B and hence induced autophagy in pancreatic cancer cells. |
Trametinib
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) | [69] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Missense mutation | p.K601E (c.1801A>G) |
||
Resistant Drug | Trametinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | SkBR3 cells | Breast | Homo sapiens (Human) | CVCL_0033 |
A375 cells | Skin | Homo sapiens (Human) | CVCL_0132 | |
NIH-3T3 cells | Embryo | Mus musculus (Mouse) | CVCL_0594 | |
NT-3 cells | Lymph node | Homo sapiens (Human) | CVCL_VG81 | |
Experiment for Drug Resistance |
BioRad TC20 automated cell counter assay |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) | [70] | |||
Sensitive Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Complex-indel | p.V487_P492delinsA (c.1460_1474del15) |
||
Sensitive Drug | Trametinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HCT116 cells | Colon | Homo sapiens (Human) | CVCL_0291 |
A375 cells | Skin | Homo sapiens (Human) | CVCL_0132 | |
BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 | |
NIH 3T3 cells | Colon | Homo sapiens (Human) | CVCL_0594 | |
HEK 293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
OV-90 cells | Ascites | Homo sapiens (Human) | CVCL_3768 | |
H2405 cells | Lung | Homo sapiens (Human) | CVCL_1551 | |
In Vivo Model | NIH nude rat xenograft model | Rattus norvegicus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CellTiter-Glo assay; Colony transformation assay; Cell-cycle analysis; BrdUrd incorporation assay | |||
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) | [71] | |||
Sensitive Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | IF-deletion | p.N486_P490delNVTAP (c.1457_1471del15) |
||
Sensitive Drug | Trametinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Blood sample | . |
Clinical Trial Drug(s)
4 drug(s) in total
TRAIL
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: HOX transcript antisense RNA (HOTAIR) | [72] | |||
Resistant Disease | Pancreatic cancer [ICD-11: 2C10.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | TRAIL | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Suit2 cells | Pancreas | Homo sapiens (Human) | CVCL_3172 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Flow cytometric analysis | |||
Mechanism Description | The long non-coding RNA HOTAIR enhances pancreatic cancer resistance to TNF-related apoptosis-inducing ligand. |
MK-2206
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: GTPase KRas (KRAS) | [73] | |||
Sensitive Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Missense mutation | p.G12D (c.35G>A) |
||
Sensitive Drug | MK-2206 | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | The missense mutation p.G12D (c.35G>A) in gene KRAS cause the sensitivity of MK-2206 by unusual activation of pro-survival pathway |
LY3009120
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) | [70] | |||
Sensitive Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Complex-indel | p.V487_P492delinsA (c.1460_1474del15) |
||
Sensitive Drug | LY3009120 | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HCT116 cells | Colon | Homo sapiens (Human) | CVCL_0291 |
A375 cells | Skin | Homo sapiens (Human) | CVCL_0132 | |
BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 | |
NIH 3T3 cells | Colon | Homo sapiens (Human) | CVCL_0594 | |
HEK 293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
OV-90 cells | Ascites | Homo sapiens (Human) | CVCL_3768 | |
H2405 cells | Lung | Homo sapiens (Human) | CVCL_1551 | |
In Vivo Model | NIH nude rat xenograft model | Rattus norvegicus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CellTiter-Glo assay; Colony transformation assay; Cell-cycle analysis; BrdUrd incorporation assay |
OSI-027
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-miR-663a | [37] | |||
Sensitive Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | OSI-027 | |||
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 | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
T3-M4 cells | Pancreas | Homo sapiens (Human) | CVCL_VQ95 | |
Experiment for Molecule Alteration |
RT-PCR, qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Colony formation assay; Flow cytometry assay | |||
Mechanism Description | Gemcitabine enhances OSI-027 cytotoxicity by upregulation of miR-663a in pancreatic ductal adenocarcinoma cells. |
Preclinical Drug(s)
1 drug(s) in total
Spliceostatin A
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: Splicing factor 3B subunit 1 (SF3B1) | [74] | |||
Sensitive Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Missense mutation | p.K700E (c.2098A>G) |
||
Sensitive Drug | Spliceostatin A | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
Capan-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0026 | |
HEC1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 | |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
Capan-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0026 | |
Pancreatic Panc 0504 cells | Pancreas | Homo sapiens (Human) | CVCL_1637 | |
MFE296 cells | Endometrium | Homo sapiens (Human) | CVCL_1406 | |
HEC59 cells | Endometrium | Homo sapiens (Human) | CVCL_2930 | |
ESS-1 cells | Endometrium | Homo sapiens (Human) | CVCL_1205 | |
DSMZ cells | N.A. | . | N.A. | |
ESS-1 cells | Endometrium | Homo sapiens (Human) | CVCL_1205 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CellTiter-Glo assay |
Investigative Drug(s)
2 drug(s) in total
Cisplatinum
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: MACC1 antisense RNA 1 (MACC1-AS1) | [75] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Up-regulation | Expression |
||
Resistant Drug | Cisplatinum | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | PAX8/NOTCH1 signaling pathway | Activation | hsa04330 | |
In Vitro Model | BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 |
MIA PaCa-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0428 | |
PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 | |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
AsPC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0152 | |
KP-2 cells | Pancreas | Homo sapiens (Human) | CVCL_3004 | |
In Vivo Model | Male BALB/c nude mice xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Microarray assay; qRT-PCR; Western bloting analysis; Luciferase assay; RIP experiments assay; RNA pull down assay | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | Long non-coding RNA MACC1-AS1 promoted pancreatic carcinoma progression through activation of PAX8/NOTCH1 signaling pathway. |
Gemcitabine/Trametinib
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: GTPase KRas (KRAS) | [76] | |||
Resistant Disease | Pancreatic ductal adenocarcinoma [ICD-11: 2C10.0] | |||
Molecule Alteration | Other | p.G12_G13 (c.34_39) |
||
Resistant Drug | Gemcitabine/Trametinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Pancreas | . | ||
Experiment for Drug Resistance |
Plasma analysis |
References
If you find any error in data or bug in web service, please kindly report it to Dr. Sun and Dr. Zhang.