Disease Information
General Information of the Disease (ID: DIS00096)
Name |
Prostate cancer
|
---|---|
ICD |
ICD-11: 2C82
|
Resistance Map |
Type(s) of Resistant Mechanism of This Disease
ADTT: Aberration of the Drug's Therapeutic Target
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)
25 drug(s) in total
Abiraterone
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Androgen receptor (AR) | [1] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Structural variation | Copy number gain |
||
Resistant Drug | Abiraterone | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Whole genome sequencing assay; Exome sequencing assay | |||
Mechanism Description | Accordingly, AR amplification was detected in circulating cell-free DNA and was shown to be associated with enzalutamide and abiraterone treatment resistance in a cohort of 62 CRPC patients. | |||
Key Molecule: Androgen receptor (AR) | [1] | |||
Resistant Disease | Primary prostate cancer [ICD-11: 2C82.Z] | |||
Molecule Alteration | Structural variation | Copy number gain |
||
Resistant Drug | Abiraterone | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Whole genome sequencing assay; Exome sequencing assay | |||
Mechanism Description | Accordingly, AR amplification was detected in circulating cell-free DNA and was shown to be associated with enzalutamide and abiraterone treatment resistance in a cohort of 62 CRPC patients. |
Apalutamide
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: Androgen receptor (AR) | [2] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Missense mutation | p.F877L (c.2629T>C) |
||
Resistant Drug | Apalutamide | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
In Vivo Model | SHO male mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Chromatin immunoprecipitation assay | |||
Mechanism Description | The missense mutation p.F877L (c.2629T>C) in gene AR cause the resistance of Apalutamide by aberration of the drug's therapeutic target | |||
Key Molecule: Androgen receptor (AR) | [2] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Missense mutation | p.F877L (c.2629T>C) |
||
Resistant Drug | Apalutamide | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
In Vivo Model | SHO male mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Chromatin immunoprecipitation assay | |||
Mechanism Description | The missense mutation p.F877L (c.2629T>C) in gene AR cause the resistance of Apalutamide by aberration of the drug's therapeutic target | |||
Key Molecule: Androgen receptor (AR) | [2] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Missense mutation | p.F877L (. |
||
Resistant Drug | Apalutamide | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
In Vivo Model | SHO male mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Chromatin immunoprecipitation assay | |||
Mechanism Description | The missense mutation p.F877L (. in gene AR cause the resistance of Apalutamide by aberration of the drug's therapeutic target |
Bicalutamide
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: Androgen receptor (AR) | [3] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Missense mutation | p.W742L (c.2225G>T) |
||
Resistant Drug | Bicalutamide | |||
Experimental Note | Identified from the Human Clinical Data | |||
Key Molecule: Androgen receptor (AR) | [3] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Missense mutation | p.W742C (c.2226G>T) |
||
Resistant Drug | Bicalutamide | |||
Experimental Note | Identified from the Human Clinical Data | |||
Regulation by the Disease Microenvironment (RTDM) | ||||
Key Molecule: Protocadherin beta-9 (PCDHB9) | [4] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Bicalutamide | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | LN229 cells | Brain | Homo sapiens (Human) | CVCL_0393 |
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Bicalutamide has been widely used as a first-line treatment for PCa. Although patients initially show a favorable response to bicalutamide treatment, PCa eventually acquires bicalutamide resistance. Several factors have been shown to be involved in bicalutamide resistance. However, the mechanism of bicalutamide resistance is not fully understood. In this study, the knockdown of protocadherin B9 reduced nuclear AR translocation and bicalutamide resistance in androgen-dependent LNCaP cells in the presence of DHT. The overexpression of protocadherin B9 had no effect on bicalutamide resistance in androgen-independent DU145 cells. These results further indicate that protocadherin B9 is involved in bicalutamide resistance through the modulation of AR signaling. Taken together, our findings suggest that protocadherin B9 targeted therapy could be more effective therapy than bicalutamide alone for patients with PCa. |
Cabazitaxel
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Phosphatase and tensin homolog (PTEN) | [5] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Cabazitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | CAL27 cells | Oral | Homo sapiens (Human) | CVCL_1107 |
LOVO cells | Colon | Homo sapiens (Human) | CVCL_0399 | |
BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 | |
C4-2 cells | Prostate | Homo sapiens (Human) | CVCL_4782 | |
HuTu80 cells | Small intestine | Homo sapiens (Human) | CVCL_1301 | |
DU145-DR cells | Brain | Homo sapiens (Human) | CVCL_4Y36 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | TUBB3 expression was upregulated in DTX-resistant and CBZ-resistant cells. TUBB3 knockdown re-sensitized DTX-resistant cells to DTX and CBZ-resistant cells to CBZ. Additionally, TUBB3 knockdown re-sensitized DTX-resistant cell lines to CBZ, indicating that TUBB3 mediates cross-resistance between DTX and CBZ. Knockdown of TUBB3 enhanced PTEN expression, and PTEN knockout enhanced TUBB3 expression. | |||
Key Molecule: Tubulin beta-3 chain (TUBB3) | [5] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Cabazitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | CAL27 cells | Oral | Homo sapiens (Human) | CVCL_1107 |
LOVO cells | Colon | Homo sapiens (Human) | CVCL_0399 | |
BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 | |
C4-2 cells | Prostate | Homo sapiens (Human) | CVCL_4782 | |
HuTu80 cells | Small intestine | Homo sapiens (Human) | CVCL_1301 | |
DU145-DR cells | Brain | Homo sapiens (Human) | CVCL_4Y36 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | TUBB3 expression was upregulated in DTX-resistant and CBZ-resistant cells. TUBB3 knockdown re-sensitized DTX-resistant cells to DTX and CBZ-resistant cells to CBZ. Additionally, TUBB3 knockdown re-sensitized DTX-resistant cell lines to CBZ, indicating that TUBB3 mediates cross-resistance between DTX and CBZ. Knockdown of TUBB3 enhanced PTEN expression, and PTEN knockout enhanced TUBB3 expression. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Phosphatase and tensin homolog (PTEN) | [5] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Cabazitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | CAL27 cells | Oral | Homo sapiens (Human) | CVCL_1107 |
LOVO cells | Colon | Homo sapiens (Human) | CVCL_0399 | |
BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 | |
C4-2 cells | Prostate | Homo sapiens (Human) | CVCL_4782 | |
HuTu80 cells | Small intestine | Homo sapiens (Human) | CVCL_1301 | |
DU145-DR cells | Brain | Homo sapiens (Human) | CVCL_4Y36 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | TUBB3 expression was upregulated in DTX-resistant and CBZ-resistant cells. TUBB3 knockdown re-sensitized DTX-resistant cells to DTX and CBZ-resistant cells to CBZ. Additionally, TUBB3 knockdown re-sensitized DTX-resistant cell lines to CBZ, indicating that TUBB3 mediates cross-resistance between DTX and CBZ. Knockdown of TUBB3 enhanced PTEN expression, and PTEN knockout enhanced TUBB3 expression. | |||
Key Molecule: Tubulin beta-3 chain (TUBB3) | [5] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Cabazitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | CAL27 cells | Oral | Homo sapiens (Human) | CVCL_1107 |
LOVO cells | Colon | Homo sapiens (Human) | CVCL_0399 | |
BxPC-3 cells | Pancreas | Homo sapiens (Human) | CVCL_0186 | |
C4-2 cells | Prostate | Homo sapiens (Human) | CVCL_4782 | |
HuTu80 cells | Small intestine | Homo sapiens (Human) | CVCL_1301 | |
DU145-DR cells | Brain | Homo sapiens (Human) | CVCL_4Y36 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | TUBB3 expression was upregulated in DTX-resistant and CBZ-resistant cells. TUBB3 knockdown re-sensitized DTX-resistant cells to DTX and CBZ-resistant cells to CBZ. Additionally, TUBB3 knockdown re-sensitized DTX-resistant cell lines to CBZ, indicating that TUBB3 mediates cross-resistance between DTX and CBZ. Knockdown of TUBB3 enhanced PTEN expression, and PTEN knockout enhanced TUBB3 expression. |
Cisplatin
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-mir-218 | [6] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Activation | hsa05200 | |
Cell migration | Activation | hsa04670 | ||
Cell viability | Activation | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometric analysis | |||
Mechanism Description | Overexpression of miR218 inhibited cell viability, migration, and invasion in PC3 and DU145 cells. Overexpression of BCAT1 decreased the chemosensitivity to CDDP treatment of PC3 and DU145 cells. The tumor suppressive role of miR218 was mediated by negatively regulating BCAT1 protein expression. | |||
Key Molecule: hsa-miR-17-92 | [7] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | AKT/ERK signaling pathway | Activation | hsa04010 | |
Cell apoptosis | Inhibition | hsa04210 | ||
Cell invasion | Activation | hsa05200 | ||
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
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-17-92 cluster plays a crucial role in cell growth of the DU145 prostate cancer cells due to regulation of cellular apoptosis-related and proliferation-related proteins, and causes chemo-resistance to cisplatin via activating AkT signaling together with upregulating ERCC1 also contributed to development of cisplatin-resistance. | |||
Key Molecule: hsa-mir-205 | [8] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
VCaP cells | Prostate | Homo sapiens (Human) | CVCL_2235 | |
WPE1-NA22 cells | Prostate | Homo sapiens (Human) | CVCL_3810 | |
WPE1-NB11 cells | Prostate | Homo sapiens (Human) | CVCL_3811 | |
WPE1-NB14 cells | Prostate | Homo sapiens (Human) | CVCL_3812 | |
WPE1-NB26 cells | Prostate | Homo sapiens (Human) | CVCL_3813 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-205 and miR-31 regulate apoptosis in prostate cancer cells by targeting antiapoptotic proteins Bcl-w and E2F6. | |||
Key Molecule: hsa-mir-31 | [8] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
VCaP cells | Prostate | Homo sapiens (Human) | CVCL_2235 | |
WPE1-NA22 cells | Prostate | Homo sapiens (Human) | CVCL_3810 | |
WPE1-NB11 cells | Prostate | Homo sapiens (Human) | CVCL_3811 | |
WPE1-NB14 cells | Prostate | Homo sapiens (Human) | CVCL_3812 | |
WPE1-NB26 cells | Prostate | Homo sapiens (Human) | CVCL_3813 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-205 and miR-31 regulate apoptosis in prostate cancer cells by targeting antiapoptotic proteins Bcl-w and E2F6. | |||
Irregularity in Drug Uptake and Drug Efflux (IDUE) | ||||
Key Molecule: ATP-binding cassette sub-family C2 (ABCC2) | [9] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | However, higher concentrations of probenecid (500 uM) failed to demonstrate a chemosensitizing effect. Consistent with this lower chemosensitizing efficacy in higher-concentration probenecid treatment, we observed that the expression of ABCG2, a drug-efflux transporter, increased in a dose-dependent manner following probenecid treatment. Thus, probenecid could enhance the chemosensitivity of 3D-cultured prostate cancer cells, but not at higher concentr. | |||
Key Molecule: Multidrug resistance-associated protein 1 (MRP1) | [9] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | However, higher concentrations of probenecid (500 uM) failed to demonstrate a chemosensitizing effect. Consistent with this lower chemosensitizing efficacy in higher-concentration probenecid treatment, we observed that the expression of ABCG2, a drug-efflux transporter, increased in a dose-dependent manner following probenecid treatment. Thus, probenecid could enhance the chemosensitivity of 3D-cultured prostate cancer cells, but not at higher concentr. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Branched-chain-amino-acid aminotransferase (BCAT1) | [6] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Cisplatin | |||
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 | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometric analysis | |||
Mechanism Description | Overexpression of miR218 inhibited cell viability, migration, and invasion in PC3 and DU145 cells. Overexpression of BCAT1 decreased the chemosensitivity to CDDP treatment of PC3 and DU145 cells. The tumor suppressive role of miR218 was mediated by negatively regulating BCAT1 protein expression. | |||
Key Molecule: Bcl-2-associated agonist of cell death (BAD) | [7] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | AKT/ERK signaling pathway | Activation | hsa04010 | |
Cell apoptosis | Inhibition | hsa04210 | ||
Cell invasion | Activation | hsa05200 | ||
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-17-92 cluster plays a crucial role in cell growth of the DU145 prostate cancer cells due to regulation of cellular apoptosis-related and proliferation-related proteins, and causes chemo-resistance to cisplatin via activating AkT signaling together with upregulating ERCC1 also contributed to development of cisplatin-resistance. | |||
Key Molecule: BH3-interacting domain death agonist (BID) | [7] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | AKT/ERK signaling pathway | Activation | hsa04010 | |
Cell apoptosis | Inhibition | hsa04210 | ||
Cell invasion | Activation | hsa05200 | ||
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-17-92 cluster plays a crucial role in cell growth of the DU145 prostate cancer cells due to regulation of cellular apoptosis-related and proliferation-related proteins, and causes chemo-resistance to cisplatin via activating AkT signaling together with upregulating ERCC1 also contributed to development of cisplatin-resistance. | |||
Key Molecule: Bcl-2-interacting killer (BIK) | [7] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | AKT/ERK signaling pathway | Activation | hsa04010 | |
Cell apoptosis | Inhibition | hsa04210 | ||
Cell invasion | Activation | hsa05200 | ||
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-17-92 cluster plays a crucial role in cell growth of the DU145 prostate cancer cells due to regulation of cellular apoptosis-related and proliferation-related proteins, and causes chemo-resistance to cisplatin via activating AkT signaling together with upregulating ERCC1 also contributed to development of cisplatin-resistance. | |||
Key Molecule: Bcl-2-like protein 11 (BCL2L11) | [7] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | AKT/ERK signaling pathway | Activation | hsa04010 | |
Cell apoptosis | Inhibition | hsa04210 | ||
Cell invasion | Activation | hsa05200 | ||
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
In Vivo Model | BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-17-92 cluster plays a crucial role in cell growth of the DU145 prostate cancer cells due to regulation of cellular apoptosis-related and proliferation-related proteins, and causes chemo-resistance to cisplatin via activating AkT signaling together with upregulating ERCC1 also contributed to development of cisplatin-resistance. | |||
Key Molecule: Bcl-2-like protein 2 (BCL2L2) | [8] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
VCaP cells | Prostate | Homo sapiens (Human) | CVCL_2235 | |
WPE1-NA22 cells | Prostate | Homo sapiens (Human) | CVCL_3810 | |
WPE1-NB11 cells | Prostate | Homo sapiens (Human) | CVCL_3811 | |
WPE1-NB14 cells | Prostate | Homo sapiens (Human) | CVCL_3812 | |
WPE1-NB26 cells | Prostate | Homo sapiens (Human) | CVCL_3813 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-205 and miR-31 regulate apoptosis in prostate cancer cells by targeting antiapoptotic proteins Bcl-w and E2F6. | |||
Key Molecule: Transcription factor E2F6 (E2F6) | [8] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
VCaP cells | Prostate | Homo sapiens (Human) | CVCL_2235 | |
WPE1-NA22 cells | Prostate | Homo sapiens (Human) | CVCL_3810 | |
WPE1-NB11 cells | Prostate | Homo sapiens (Human) | CVCL_3811 | |
WPE1-NB14 cells | Prostate | Homo sapiens (Human) | CVCL_3812 | |
WPE1-NB26 cells | Prostate | Homo sapiens (Human) | CVCL_3813 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-205 and miR-31 regulate apoptosis in prostate cancer cells by targeting antiapoptotic proteins Bcl-w and E2F6. | |||
Key Molecule: G1/S-specific cyclin-D1 (CCND1) | [10] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
ATP cell viability assay | |||
Mechanism Description | CCND1 may induce cisplatin resistance both through cell cycle control and inhibition of cellular apoptosis pathways, which have been previously observed37 and supported by our CCND1 knockdown study. The role of CCND1 in cell cycle control is well documented. CCND1 accumulates in cells at middle and late G1 phase and stimulate G1 progression to S phase. The proportion of parental cells in G1/0 correlated with the cisplatin sensitivity, with 833K cells having the highest G1/0 population cells and lowest EC50 value and GCT27 the lowest G1/0 population but highest EC50 score. | |||
Key Molecule: Bcl-2-associated agonist of cell death (BAD) | [11] | |||
Resistant Disease | Prostatic intraepithelial neoplasia [ICD-11: 2C82.2] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
In Vitro Model | LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 |
PC-3 cells | Bone | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
qRT-PCR; Western blotting assay | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | Forced expression of the PCPH protein or, in particular, of the mt-PCPH oncoprotein increased the levels of phosphorylated PKCalpha concurrently with those of Ser70-phosphorylated and total Bcl-2 protein, thus promoting cisplatin resistance. | |||
Key Molecule: Ectonucleoside triphosphate diphosphohydrolase 5 (ENTPD5) | [11] | |||
Resistant Disease | Prostatic intraepithelial neoplasia [ICD-11: 2C82.2] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
In Vitro Model | LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 |
PC-3 cells | Bone | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
qRT-PCR; Western blotting assay | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | Forced expression of the PCPH protein or, in particular, of the mt-PCPH oncoprotein increased the levels of phosphorylated PKCalpha concurrently with those of Ser70-phosphorylated and total Bcl-2 protein, thus promoting cisplatin resistance. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-mir-205 | [12] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Cisplatin | |||
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 | ||
ERK signaling pathway | Inhibition | hsa04210 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
VCaP cells | Prostate | Homo sapiens (Human) | CVCL_2235 | |
Experiment for Molecule Alteration |
RT-qPCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | UTMD mediated miR 205 transfection increased the expression of caspase 9, cleaved caspase 9, cytochrome c and E cadherin, and decreased the expression of MMP 9 and p ERk,inhibiting PCa cell proliferation, migration and invasion, and promoted apoptosis modulated by cisplatin. | |||
Key Molecule: hsa-mir-128a | [13] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Cisplatin | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
HEK293T cells | Kidney | Homo sapiens (Human) | CVCL_0063 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-128 binded to the 3'UTR of ZEB1 and inhibited its expression. And ZEB1 (+) PCa chemoresistance and invasion, while miR-128 could reverse that by down-regulated ZEB1. These indicated that miR-128-mediated sensitizing chemoresistance and inhibiting invasion of PCa cells by directly targeting ZEB1. | |||
Key Molecule: hsa-mir-205 | [14] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
In Vivo Model | SCID nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | Lysosome disturbance caused by miR-205-mediated down-regulation of RAB27A and LAMP3 constraints the completion of the autophagic flux by compromising the maturation step and, consequently, interferes with the detoxifying capabilities by which PCa cells may become resistant to CDDP. | |||
Regulation by the Disease Microenvironment (RTDM) | ||||
Key Molecule: Matrix metalloproteinase-9 (MMP9) | [12] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Cisplatin | |||
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 | ||
ERK signaling pathway | Inhibition | hsa04210 | ||
Epithelial mesenchymal transition signaling pathway | Inhibition | hsa01521 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
VCaP cells | Prostate | Homo sapiens (Human) | CVCL_2235 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | UTMD mediated miR 205 transfection increased the expression of caspase 9, cleaved caspase 9, cytochrome c and E cadherin, and decreased the expression of MMP 9 and p ERk,inhibiting PCa cell proliferation, migration and invasion, and promoted apoptosis modulated by cisplatin. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Zinc finger E-box-binding homeobox 1 (ZEB1) | [13] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Cisplatin | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
HEK293T cells | Kidney | Homo sapiens (Human) | CVCL_0063 | |
Experiment for Molecule Alteration |
RT-PCR; Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-128 binded to the 3'UTR of ZEB1 and inhibited its expression. And ZEB1 (+) PCa chemoresistance and invasion, while miR-128 could reverse that by down-regulated ZEB1. These indicated that miR-128-mediated sensitizing chemoresistance and inhibiting invasion of PCa cells by directly targeting ZEB1. | |||
Key Molecule: Lysosome-associated membrane glycoprotein 3 (LAMP3) | [14] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
In Vivo Model | SCID nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | Lysosome disturbance caused by miR-205-mediated down-regulation of RAB27A and LAMP3 constraints the completion of the autophagic flux by compromising the maturation step and, consequently, interferes with the detoxifying capabilities by which PCa cells may become resistant to CDDP. | |||
Key Molecule: Ras-related protein Rab-27A (RAP27A) | [14] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
In Vivo Model | SCID nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | Lysosome disturbance caused by miR-205-mediated down-regulation of RAB27A and LAMP3 constraints the completion of the autophagic flux by compromising the maturation step and, consequently, interferes with the detoxifying capabilities by which PCa cells may become resistant to CDDP. |
Docetaxel
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-mir-323 | [15] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometric analysis | |||
Mechanism Description | microRNA-323 upregulation promotes prostate cancer growth and docetaxel resistance by repressing p73. | |||
Key Molecule: hsa-mir-181a | [16] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | C4-2B cells | Prostate | Homo sapiens (Human) | CVCL_4784 |
TaxR cells | Prostate | Homo sapiens (Human) | CVCL_4V97 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
Beckman Coulter method; Rhodamine Assay; Cell Death ELISA | |||
Mechanism Description | Overexpression of miR181a in prostate cancer cells contributes to their resistance to docetaxel, this is due, in part, to modulation of p53 phosphorylation and apoptosis. | |||
Key Molecule: hsa-miR-125a-3p | [17] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | MTA1 signaling pathway | Activation | hsa05206 | |
In Vitro Model | LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
ELISA; MTT assay | |||
Mechanism Description | Regulation of docetaxel sensitivity in prostate cancer cells by hsa-miR125a-3p via modulation of metastasis-associated protein 1 signaling, MTA1 is a direct target of hsa-mir125a-3p in pca cells. | |||
Key Molecule: hsa-mir-375 | [18] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Annexin V-PE Apoptosis assay | |||
Mechanism Description | miR375 induces docetaxel resistance in prostate cancer by targeting SEC23A and YAP1. | |||
Key Molecule: hsa-mir-195 | [19] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell colony | Activation | hsa05200 | ||
Cell viability | Activation | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | miR-195 improved the sensitivity of resistant PC cells to DOC by suppressing CLU. | |||
Key Molecule: Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) | [20] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell invasion | Activation | hsa05200 | ||
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
MTT assay; Scratch Wound healing assay; Transwell Invasion assay; Flow cytometry assay | |||
Mechanism Description | Knockdown of MALAT1 in DTX-resistant PCa cells up-regulated miR-145-5p as well as suppressed AkAP12 expression, further inhibited cell viability and induced apoptosis. | |||
Key Molecule: SOCS2 antisense RNA 1 (SOCS2-AS1) | [21] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 |
VCaP cells | Prostate | Homo sapiens (Human) | CVCL_2235 | |
LTAD cells | Prostate | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
TUNEL assay; MTS assay | |||
Mechanism Description | Androgen-induced Long Noncoding RNA (LncRNA) SOCS2-AS1 Promotes Cell Growth and Inhibits Apoptosis in Prostate Cancer Cells.suppressor of cytokine signaling 2-antisense transcript 1 (SOCS2-AS1), the expression of which was higher in castration-resistant prostate cancer model cells.SOCS2-AS1 promoted castration-resistant and androgen-dependent cell growth. We found that SOCS2-AS1 knockdown up-regulated genes related to the apoptosis pathway, including tumor necrosis factor superfamily 10 (TNFSF10), and sensitized prostate cancer cells to docetaxel treatment. Moreover, we also demonstrated that SOCS2-AS1 promotes androgen signaling by modulating the epigenetic control for AR target genes including TNFSF10 These findings suggest that SOCS2-AS1 plays an important role in the development of castration-resistant prostate cancer by repressing apoptosis. | |||
Key Molecule: hsa-mir-34 | [22] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
Acid phosphatase assay | |||
Mechanism Description | miR-34a regulates BCL-2 and may, in part, regulate response to docetaxel. miR-34a was significantly decreased in prostate cancer versus normal tissues; in biochemical recurrence versus non-recurrence tissue and in metastatic versus primary disease prostate tissue. We confirmed BCL-2 as a target of miR-34a, by manipulating miR-34a expression in our parent and docetaxel resistant cell lines and subsequently assessing BCL-2 levels. Specifically, upon inhibition of miR-34a in sensitive parent cells (PC3 and 22Rv1) we observed an increase in BCL-2 expression, whereas mimicking miR-34a expression in docetaxel-resistant cells (PC3RD and 22Rv1RD) resulted in decreased BCL-2 expression. | |||
Key Molecule: hsa-mir-205 | [8], [23] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
VCaP cells | Prostate | Homo sapiens (Human) | CVCL_2235 | |
WPE1-NA22 cells | Prostate | Homo sapiens (Human) | CVCL_3810 | |
WPE1-NB11 cells | Prostate | Homo sapiens (Human) | CVCL_3811 | |
WPE1-NB14 cells | Prostate | Homo sapiens (Human) | CVCL_3812 | |
WPE1-NB26 cells | Prostate | Homo sapiens (Human) | CVCL_3813 | |
Experiment for Molecule Alteration |
RT-qPCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay | |||
Mechanism Description | Docetaxel-resistant cells showed a reduced E-cadherin and an increased vimentin expression accompanied by induced expression of stem cell markers compared with parental cells. Decreased Expression of miR-200c and miR-205 Is Responsible for E-Cadherin Loss in Chemotherapy-Resistant Cells. And miR-205 and miR-31 regulate apoptosis in prostate cancer cells by targeting antiapoptotic proteins Bcl-w and E2F6. | |||
Key Molecule: Growth arrest specific 5 (GAS5) | [24] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
Fluorescence microscopy test apoptosis assay | |||
Mechanism Description | Transient expression of GAS5 enhances apoptosis and decreases the survival of 22Rv1 cells, forced variation of GAS5 gene expression can modulate cellular responses to various apoptotic stimuli, including a range of chemotherapeutic drugs. | |||
Key Molecule: hsa-mir-200c | [23] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
RT-qPCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Docetaxel-resistant cells showed a reduced E-cadherin and an increased vimentin expression accompanied by induced expression of stem cell markers compared with parental cells. Decreased Expression of miR-200c and miR-205 Is Responsible for E-Cadherin Loss in Chemotherapy-Resistant Cells. | |||
Key Molecule: hsa-mir-31 | [8] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
VCaP cells | Prostate | Homo sapiens (Human) | CVCL_2235 | |
WPE1-NA22 cells | Prostate | Homo sapiens (Human) | CVCL_3810 | |
WPE1-NB11 cells | Prostate | Homo sapiens (Human) | CVCL_3811 | |
WPE1-NB14 cells | Prostate | Homo sapiens (Human) | CVCL_3812 | |
WPE1-NB26 cells | Prostate | Homo sapiens (Human) | CVCL_3813 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-205 and miR-31 regulate apoptosis in prostate cancer cells by targeting antiapoptotic proteins Bcl-w and E2F6. | |||
Key Molecule: hsa-mir-21 | [25] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Activation | hsa05200 | |
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | Programmed cell death 4 (PDCD4), is a novel suppressor of tumorigenesis, tumor progression and invasion. miR-21 can directly down-regulate the expression of PDCD4 by targeting its 3'UTR in PC3 cells. PDCD4, a direct target gene of miR-21, could mediate chemoresistance to docetaxel in PC3 cells. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Tumor protein p73 (TP73) | [15] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Luciferase reporter assay; Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometric analysis | |||
Mechanism Description | microRNA-323 upregulation promotes prostate cancer growth and docetaxel resistance by repressing p73. | |||
Key Molecule: Metastasis-associated protein MTA1 (MTA1) | [17] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | MTA1 signaling pathway | Activation | hsa05206 | |
In Vitro Model | LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
Promoter reporter assay; Western blot analysis | |||
Experiment for Drug Resistance |
ELISA; MTT assay | |||
Mechanism Description | Regulation of docetaxel sensitivity in prostate cancer cells by hsa-miR125a-3p via modulation of metastasis-associated protein 1 signaling, MTA1 is a direct target of hsa-mir125a-3p in pca cells. | |||
Key Molecule: Protein transport protein Sec23A (SEC23A) | [18] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Annexin V-PE Apoptosis assay | |||
Mechanism Description | miR375 induces docetaxel resistance in prostate cancer by targeting SEC23A and YAP1. | |||
Key Molecule: Transcriptional coactivator YAP1 (YAP1) | [18] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Annexin V-PE Apoptosis assay | |||
Mechanism Description | miR375 induces docetaxel resistance in prostate cancer by targeting SEC23A and YAP1. | |||
Key Molecule: Clusterin (CLU) | [19] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell colony | Activation | hsa05200 | ||
Cell viability | Activation | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | miR-195 improved the sensitivity of resistant PC cells to DOC by suppressing CLU. | |||
Key Molecule: A-kinase anchor protein 12 (AKAP12) | [20] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell invasion | Activation | hsa05200 | ||
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; Scratch Wound healing assay; Transwell Invasion assay; Flow cytometry assay | |||
Mechanism Description | Knockdown of MALAT1 in DTX-resistant PCa cells up-regulated miR-145-5p as well as suppressed AkAP12 expression, further inhibited cell viability and induced apoptosis. | |||
Key Molecule: Tumor necrosis factor ligand superfamily member 10 (TNFSF10) | [21] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 |
VCaP cells | Prostate | Homo sapiens (Human) | CVCL_2235 | |
LTAD cells | Prostate | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
Western blot analysis; Immunohistochemistry assay | |||
Experiment for Drug Resistance |
TUNEL assay; MTS assay | |||
Mechanism Description | Androgen-induced Long Noncoding RNA (LncRNA) SOCS2-AS1 Promotes Cell Growth and Inhibits Apoptosis in Prostate Cancer Cells.suppressor of cytokine signaling 2-antisense transcript 1 (SOCS2-AS1), the expression of which was higher in castration-resistant prostate cancer model cells.SOCS2-AS1 promoted castration-resistant and androgen-dependent cell growth. We found that SOCS2-AS1 knockdown up-regulated genes related to the apoptosis pathway, including tumor necrosis factor superfamily 10 (TNFSF10), and sensitized prostate cancer cells to docetaxel treatment. Moreover, we also demonstrated that SOCS2-AS1 promotes androgen signaling by modulating the epigenetic control for AR target genes including TNFSF10 These findings suggest that SOCS2-AS1 plays an important role in the development of castration-resistant prostate cancer by repressing apoptosis. | |||
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) | [22] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Acid phosphatase assay | |||
Mechanism Description | miR-34a regulates BCL-2 and may, in part, regulate response to docetaxel. miR-34a was significantly decreased in prostate cancer versus normal tissues; in biochemical recurrence versus non-recurrence tissue and in metastatic versus primary disease prostate tissue. We confirmed BCL-2 as a target of miR-34a, by manipulating miR-34a expression in our parent and docetaxel resistant cell lines and subsequently assessing BCL-2 levels. Specifically, upon inhibition of miR-34a in sensitive parent cells (PC3 and 22Rv1) we observed an increase in BCL-2 expression, whereas mimicking miR-34a expression in docetaxel-resistant cells (PC3RD and 22Rv1RD) resulted in decreased BCL-2 expression. | |||
Key Molecule: Bcl-2-like protein 2 (BCL2L2) | [8] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
VCaP cells | Prostate | Homo sapiens (Human) | CVCL_2235 | |
WPE1-NA22 cells | Prostate | Homo sapiens (Human) | CVCL_3810 | |
WPE1-NB11 cells | Prostate | Homo sapiens (Human) | CVCL_3811 | |
WPE1-NB14 cells | Prostate | Homo sapiens (Human) | CVCL_3812 | |
WPE1-NB26 cells | Prostate | Homo sapiens (Human) | CVCL_3813 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-205 and miR-31 regulate apoptosis in prostate cancer cells by targeting antiapoptotic proteins Bcl-w and E2F6. | |||
Key Molecule: Transcription factor E2F6 (E2F6) | [8] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
VCaP cells | Prostate | Homo sapiens (Human) | CVCL_2235 | |
WPE1-NA22 cells | Prostate | Homo sapiens (Human) | CVCL_3810 | |
WPE1-NB11 cells | Prostate | Homo sapiens (Human) | CVCL_3811 | |
WPE1-NB14 cells | Prostate | Homo sapiens (Human) | CVCL_3812 | |
WPE1-NB26 cells | Prostate | Homo sapiens (Human) | CVCL_3813 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-205 and miR-31 regulate apoptosis in prostate cancer cells by targeting antiapoptotic proteins Bcl-w and E2F6. | |||
Key Molecule: Programmed cell death protein 4 (PDCD4) | [25] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Activation | hsa05200 | |
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | Programmed cell death 4 (PDCD4), is a novel suppressor of tumorigenesis, tumor progression and invasion. miR-21 can directly down-regulate the expression of PDCD4 by targeting its 3'UTR in PC3 cells. PDCD4, a direct target gene of miR-21, could mediate chemoresistance to docetaxel in PC3 cells. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: Cancer susceptibility 2 (CASC2) | [26] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Docetaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | ERK signaling pathway | Regulation | hsa04210 | |
RTK signaling pathway | Inhibition | hsa04015 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
C4-2 cells | Prostate | Homo sapiens (Human) | CVCL_4782 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometer assay | |||
Mechanism Description | CASC2 directly targets miR183 to inhibit its expression. SPRY2 is regarded as a negative regulator RTk signaling pathway, antagonizing cell migration and/or cellular differentiation occurring through the ERk signaling. CASC2 competes with SPRY2 for miR183 binding to rescue the expression of SPRY2 in PC cells, thus suppressing the cell proliferation and promoting the apoptosis of PC cells, finally enhancing PC cells chemo-sensitivity to docetaxel through SPRY2 downstream ERk signaling pathway. | |||
Key Molecule: hsa-mir-183 | [26] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Docetaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | ERK signaling pathway | Regulation | hsa04210 | |
RTK signaling pathway | Inhibition | hsa04015 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
C4-2 cells | Prostate | Homo sapiens (Human) | CVCL_4782 | |
Experiment for Molecule Alteration |
Immunoblotting assay | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometer assay | |||
Mechanism Description | CASC2 directly targets miR183 to inhibit its expression. SPRY2 is regarded as a negative regulator RTk signaling pathway, antagonizing cell migration and/or cellular differentiation occurring through the ERk signaling. CASC2 competes with SPRY2 for miR183 binding to rescue the expression of SPRY2 in PC cells, thus suppressing the cell proliferation and promoting the apoptosis of PC cells, finally enhancing PC cells chemo-sensitivity to docetaxel through SPRY2 downstream ERk signaling pathway. | |||
Key Molecule: Protein sprouty homolog 2 (SPRY2) | [26] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Docetaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | ERK signaling pathway | Regulation | hsa04210 | |
RTK signaling pathway | Inhibition | hsa04015 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
C4-2 cells | Prostate | Homo sapiens (Human) | CVCL_4782 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometer assay | |||
Mechanism Description | SPRY2 is a direct downstream target of miR183 and can be negatively regulated by miR183 and is regarded as a negative regulator RTk signaling pathway, antagonizing cell migration and/or cellular differentiation occurring through the ERk signaling. CASC2 competes with SPRY2 for miR183 binding to rescue the expression of SPRY2 in PC cells, thus suppressing the cell proliferation and promoting the apoptosis of PC cells, finally enhancing PC cells chemo-sensitivity to docetaxel through SPRY2 downstream ERk signaling pathway. | |||
Key Molecule: hsa-miR-193a-5p | [27] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Docetaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | miR193a-5p/Bach2/HO1 signaling pathway | Inhibition | hsa05206 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
T24 cells | Bladder | Homo sapiens (Human) | CVCL_0554 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
UM-UC-3 cells | Bladder | Homo sapiens (Human) | CVCL_1783 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
TUNEL assays | |||
Mechanism Description | Silencing of miR193a-5p or blockade of the miR193a-5p-Bach2-HO-1 pathway enhances sensitization of PC3 cells to docetaxel-induced apoptosis. Docetaxel-induced miR193a-5p upregulation, which in turn inhibits Bach2 expression and thus relieves Bach2 repression of HO-1 expression, partly counteracted docetaxel-induced apoptosis. | |||
Key Molecule: hsa-mir-204 | [28] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | UCA1/miR204/Sirt1 signaling pathway | Activation | hsa05206 | |
In Vitro Model | LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
PNT2 cells | Prostate | Homo sapiens (Human) | CVCL_2164 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Annexin V-FITC Apoptosis assay; Flow cytometer | |||
Mechanism Description | The UCA1/miR204/Sirt1 axis modulates docetaxel sensitivity of prostate cancer cells. UCA1 upregulation directly resulted in decreased miR204 expression. | |||
Key Molecule: Urothelial cancer associated 1 (UCA1) | [28] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | UCA1/miR204/Sirt1 signaling pathway | Activation | hsa05206 | |
In Vitro Model | LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
PNT2 cells | Prostate | Homo sapiens (Human) | CVCL_2164 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Annexin V-FITC Apoptosis assay; Flow cytometer | |||
Mechanism Description | The UCA1/miR204/Sirt1 axis modulates docetaxel sensitivity of prostate cancer cells. UCA1 upregulation directly resulted in decreased miR204 expression. | |||
Key Molecule: hsa-miR-223-3p | [29] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell viability | Inhibition | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
C4-2 cells | Prostate | Homo sapiens (Human) | CVCL_4782 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qrt-PCR | |||
Experiment for Drug Resistance |
MTT assay; TUNEL assay; Flow cytometry assay | |||
Mechanism Description | miR-223-3p inhibitor sensitized prostatic cancer mouse model to docetaxel by increasing the expression of FOXO3. | |||
Key Molecule: hsa-mir-200b | [30] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | Bmi-1 is expressed at a high level in PCa. miR-200b plays a pivotal role in PCa at least in part via downregulation of the oncogene Bmi-1, inhibition of Bmi-1 enhanced the antitumor activity of docetaxel in PCa cells. | |||
Key Molecule: hsa-mir-143 | [31] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Docetaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
Cell proliferation | Inhibition | hsa05200 | ||
EGFR/RAS/MAPK signaling pathway | Inhibition | hsa01521 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-143 plays an important role in prostate cancer proliferation, migration and chemosensitivity by suppressing kRAS and subsequent inactivation of MAPk pathway. | |||
Regulation by the Disease Microenvironment (RTDM) | ||||
Key Molecule: Zinc finger E-box-binding homeobox 1 (ZEB1) | [32], [33] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Docetaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell invasion | Inhibition | hsa05200 | ||
Cell migration | Inhibition | hsa04670 | ||
Cell viability | Inhibition | hsa05200 | ||
ZEB1 signaling pathway | Inhibition | hsa05215 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
PrEC cells | Prostate | Homo sapiens (Human) | CVCL_0061 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis; Luciferase reporter assay | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometric analysis | |||
Mechanism Description | miR27b and miR34a enhance docetaxel sensitivity of prostate cancer cells through inhibiting epithelial-to-mesenchymal transition by targeting ZEB1. And microRNA-204 modulates chemosensitivity and apoptosis of prostate cancer cells by targeting ZEB1. Suppression of ZEB1 could effectively improve miR204 deficiency-triggered chemoresistance in PC cells. | |||
Key Molecule: hsa-mir-27b | [33] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Docetaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
PrEC cells | Prostate | Homo sapiens (Human) | CVCL_0061 | |
HEK293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR27b and miR34a enhance docetaxel sensitivity of prostate cancer cells through inhibiting epithelial-to-mesenchymal transition by targeting ZEB1. | |||
Key Molecule: hsa-mir-34 | [33] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Docetaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
PrEC cells | Prostate | Homo sapiens (Human) | CVCL_0061 | |
HEK293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR27b and miR34a enhance docetaxel sensitivity of prostate cancer cells through inhibiting epithelial-to-mesenchymal transition by targeting ZEB1. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Transcription regulator protein BACH2 (BACH2) | [27] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Docetaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | miR193a-5p/Bach2/HO1 signaling pathway | Inhibition | hsa05206 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
T24 cells | Bladder | Homo sapiens (Human) | CVCL_0554 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
UM-UC-3 cells | Bladder | Homo sapiens (Human) | CVCL_1783 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis; Luciferase reporter assay; Immunofluorescence staining assay | |||
Experiment for Drug Resistance |
TUNEL assays | |||
Mechanism Description | Silencing of miR193a-5p or blockade of the miR193a-5p-Bach2-HO-1 pathway enhances sensitization of PC3 cells to docetaxel-induced apoptosis. Docetaxel-induced miR193a-5p upregulation, which in turn inhibits Bach2 expression and thus relieves Bach2 repression of HO-1 expression, partly counteracted docetaxel-induced apoptosis. | |||
Key Molecule: NAD-dependent protein deacetylase sirtuin-1 (SIRT1) | [28] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | UCA1/miR204/Sirt1 signaling pathway | Activation | hsa05206 | |
In Vitro Model | LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
PNT2 cells | Prostate | Homo sapiens (Human) | CVCL_2164 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Annexin V-FITC Apoptosis assay; Flow cytometer | |||
Mechanism Description | The UCA1/miR204/Sirt1 axis modulates docetaxel sensitivity of prostate cancer cells. miR204 negatively modulated Sirt1 expression in prostate cancer cells. | |||
Key Molecule: Forkhead box protein O3 (FOXO3) | [29] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell viability | Inhibition | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
C4-2 cells | Prostate | Homo sapiens (Human) | CVCL_4782 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; TUNEL assay; Flow cytometry assay | |||
Mechanism Description | miR-223-3p inhibitor sensitized prostatic cancer mouse model to docetaxel by increasing the expression of FOXO3. | |||
Key Molecule: Polycomb complex protein BMI-1 (BMI1) | [30] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Docetaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | Bmi-1 is expressed at a high level in PCa. miR-200b plays a pivotal role in PCa at least in part via downregulation of the oncogene Bmi-1, inhibition of Bmi-1 enhanced the antitumor activity of docetaxel in PCa cells. | |||
Key Molecule: GTPase KRas (KRAS) | [31] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Docetaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
EGFR/RAS/MAPK signaling pathway | Inhibition | hsa01521 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-143 plays an important role in prostate cancer proliferation, migration and chemosensitivity by suppressing kRAS and subsequent inactivation of MAPk pathway. |
Doxorubicin
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: LOXL1 antisense RNA 1 (LOXL1-AS1) | [34] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Doxorubicin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | LncRNA LOXL1-AS1/miR-let-7a-5p/EGFR-related pathway regulates the doxorubicin resistance of prostate cancer DU-145 cells. | |||
Key Molecule: hsa-let-7a-5p | [34] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Doxorubicin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | LncRNA LOXL1-AS1/miR-let-7a-5p/EGFR-related pathway regulates the doxorubicin resistance of prostate cancer DU-145 cells. | |||
Key Molecule: PCGEM1 prostate-specific transcript (PCGEM1) | [35] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Doxorubicin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 |
Experiment for Molecule Alteration |
Northern blotting analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | PCGEM1overexpression in LNCaP cell culturemodel results in the inhibition of apoptosis induced by doxorubicin (DOX). Induction of p53 and p21Waf1/Cip1by DOX were delayed in LNCaP cells stably overexpressing PCGEM1(LNCaP-PCGEM1cells) compared tocontrol LNCaP cells. | |||
Irregularity in Drug Uptake and Drug Efflux (IDUE) | ||||
Key Molecule: ATP-binding cassette sub-family C2 (ABCC2) | [9] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Doxorubicin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | However, higher concentrations of probenecid (500 uM) failed to demonstrate a chemosensitizing effect. Consistent with this lower chemosensitizing efficacy in higher-concentration probenecid treatment, we observed that the expression of ABCG2, a drug-efflux transporter, increased in a dose-dependent manner following probenecid treatment. Thus, probenecid could enhance the chemosensitivity of 3D-cultured prostate cancer cells, but not at higher concentr. | |||
Key Molecule: Multidrug resistance-associated protein 1 (MRP1) | [9] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Doxorubicin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | However, higher concentrations of probenecid (500 uM) failed to demonstrate a chemosensitizing effect. Consistent with this lower chemosensitizing efficacy in higher-concentration probenecid treatment, we observed that the expression of ABCG2, a drug-efflux transporter, increased in a dose-dependent manner following probenecid treatment. Thus, probenecid could enhance the chemosensitivity of 3D-cultured prostate cancer cells, but not at higher concentr. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Epidermal growth factor receptor (EGFR) | [34] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Doxorubicin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell migration | Activation | hsa04670 | ||
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
Experiment for Molecule Alteration |
Western blot analysis; RT-qPCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | LncRNA LOXL1-AS1/miR-let-7a-5p/EGFR-related pathway regulates the doxorubicin resistance of prostate cancer DU-145 cells. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Irregularity in Drug Uptake and Drug Efflux (IDUE) | ||||
Key Molecule: ATP-binding cassette sub-family B5 (ABCB5) | [36] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Doxorubicin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
DU-145Nox1 cells | Prostate | Homo sapiens (Human) | CVCL_0105 | |
Experiment for Molecule Alteration |
Immunohistochemistry assay | |||
Experiment for Drug Resistance |
Annexin V staining assay | |||
Mechanism Description | In DU-145Nox1 tumor spheroids, expression of HIF-1alpha as well as P-gp was significantly decreased as compared to DU-145 spheroids, which resulted in an increased retention of the anticancer agent doxorubicin. Pretreatment with the free radical scavengers vitamin E and vitamin C increased the expression of P-gp as well as HIF-1alpha in Nox-1-overexpressing cells, whereas no effect of free radical scavengers was observed on mdr-1 mRNA expression. |
Enzalutamide
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: Androgen receptor (AR) | [1] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Structural variation | Copy number gain |
||
Resistant Drug | Enzalutamide | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Whole genome sequencing assay; Exome sequencing assay | |||
Mechanism Description | Accordingly, AR amplification was detected in circulating cell-free DNA and was shown to be associated with enzalutamide and abiraterone treatment resistance in a cohort of 62 CRPC patients. | |||
Key Molecule: Androgen receptor (AR) | [1] | |||
Resistant Disease | Primary prostate cancer [ICD-11: 2C82.Z] | |||
Molecule Alteration | Structural variation | Copy number gain |
||
Resistant Drug | Enzalutamide | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Whole genome sequencing assay; Exome sequencing assay | |||
Mechanism Description | Accordingly, AR amplification was detected in circulating cell-free DNA and was shown to be associated with enzalutamide and abiraterone treatment resistance in a cohort of 62 CRPC patients. |
Everolimus
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: Growth arrest specific 5 (GAS5) | [37] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Everolimus | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
PNT2C2 cells | Prostate | Homo sapiens (Human) | CVCL_4889 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
GAS5 assay; MTS assay | |||
Mechanism Description | First generation mTORC1, combined mTORC1/mTORC2 and dual PI3k/mTOR inhibitors all increased cellular GAS5 levels and inhibited culture growth in androgen-dependent (LNCaP) and androgen-sensitive (22Rv1) cell lines, but not in androgen-independent (PC-3 and DU 145) cell lines. The latter exhibited low endogenous GAS5 expression, and GAS5 silencing in LNCaP and 22Rv1 cells decreased the sensitivity to mTOR inhibitors, whereas transfection of GAS5 LncRNA sensitized PC-3 and DU 145 cells to these agents. |
Flutamide
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: Androgen receptor (AR) | [38] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Missense mutation | p.T878A (c.2632A>G) |
||
Resistant Drug | Flutamide | |||
Experimental Note | Identified from the Human Clinical Data | |||
Key Molecule: Androgen receptor (AR) | [38] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Missense mutation | p.T877A |
||
Resistant Drug | Flutamide | |||
Experimental Note | Identified from the Human Clinical Data |
Hydroxyflutamide
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: Androgen receptor (AR) | [39] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Missense mutation | p.T877A |
||
Resistant Drug | Hydroxyflutamide | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Energy decomposition assay | |||
Mechanism Description | However, a drug resistance problem appears after about one year's treatment. AR T877A is the first mutation that was found to cause a resistance problem. Then W741C_T877A and F876L_T877A mutations were also reported to cause resistance to HF, while W741C and F876L single mutations cannot. | |||
Key Molecule: Androgen receptor (AR) | [39] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Missense mutation+Missense mutation | p.W741C+T877 |
||
Resistant Drug | Hydroxyflutamide | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Energy decomposition assay | |||
Mechanism Description | However, a drug resistance problem appears after about one year's treatment. AR T877A is the first mutation that was found to cause a resistance problem. Then W741C_T877A and F876L_T877A mutations were also reported to cause resistance to HF, while W741C and F876L single mutations cannot. | |||
Key Molecule: Androgen receptor (AR) | [39] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Missense mutation+Missense mutation | p.F876L+T877A |
||
Resistant Drug | Hydroxyflutamide | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Energy decomposition assay | |||
Mechanism Description | However, a drug resistance problem appears after about one year's treatment. AR T877A is the first mutation that was found to cause a resistance problem. Then W741C_T877A and F876L_T877A mutations were also reported to cause resistance to HF, while W741C and F876L single mutations cannot. |
Ibrutinib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-mir-214 | [40] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Ibrutinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell invasion | Inhibition | hsa05200 | ||
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
RT-qPCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay | |||
Mechanism Description | microRNA-214 targets PTk6 to inhibit tumorigenic potential and increase drug sensitivity of prostate cancer cells. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Protein-tyrosine kinase 6 (PTK6) | [40] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Ibrutinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell invasion | Inhibition | hsa05200 | ||
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay | |||
Mechanism Description | microRNA-214 targets PTk6 to inhibit tumorigenic potential and increase drug sensitivity of prostate cancer cells. |
Levofloxacin
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: DNA topoisomerase (ATP-hydrolyzing) (PARC) | [41] | |||
Resistant Disease | Mycoplasma hominis prostate cancer [ICD-11: 2C82.Y] | |||
Molecule Alteration | Missense mutation | p.K134R |
||
Resistant Drug | Levofloxacin | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Mycoplasma hominis ATCC 23114(PG21) | 347256 | ||
Mycoplasma hominis isolate | 2098 | |||
Experiment for Molecule Alteration |
Whole genome sequence assay | |||
Mechanism Description | The single amino acid mutation in ParC of MH may relate to the resistance to OFX and LVX and the high-level resistance to fluoroquinolones for MH is associated with mutations in both DNA gyrase and the ParC subunit of topoisomerase IV. |
Mitoxantrone
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: Growth arrest specific 5 (GAS5) | [24] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Mitoxantrone | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
Fluorescence microscopy test apoptosis assay | |||
Mechanism Description | Transient expression of GAS5 enhances apoptosis and decreases the survival of 22Rv1 cells, forced variation of GAS5 gene expression can modulate cellular responses to various apoptotic stimuli, including a range of chemotherapeutic drugs. |
Moclobemide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: Monoamine oxidase A (MAOA) | [42] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Function | Inhibition |
||
Sensitive Drug | Moclobemide | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | Monoamine oxidase A (MAOA) may promote tumor burden and drug/castration resistance in PCa. A positive association will pave the way for MAOA inhibitors such as moclobemide for PCa therapy. Association of key molecules of oncogenesis and metastasis with MAOA suggests that MAOA inhibitors such as moclobemide might be effective in the management of PCa. |
Ofloxacin
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: DNA topoisomerase (ATP-hydrolyzing) (PARC) | [41] | |||
Resistant Disease | Mycoplasma hominis prostate cancer [ICD-11: 2C82.Y] | |||
Molecule Alteration | Missense mutation | p.K134R |
||
Resistant Drug | Ofloxacin | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Mycoplasma hominis ATCC 23114(PG21) | 347256 | ||
Mycoplasma hominis isolate | 2098 | |||
Experiment for Molecule Alteration |
Whole genome sequence assay | |||
Mechanism Description | The single amino acid mutation in ParC of MH may relate to the resistance to OFX and LVX and the high-level resistance to fluoroquinolones for MH is associated with mutations in both DNA gyrase and the ParC subunit of topoisomerase IV. |
Olaparib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: Prostate cancer associated transcript 1 (PCAT1) | [43] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Olaparib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE cells | Prostate | Homo sapiens (Human) | CVCL_1736 | |
In Vivo Model | SCID nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
WST assay | |||
Mechanism Description | PCAT-1 expressing cells exhibit a BRCA-like phenotype, resulting in cell sensitization to PARP1 inhibitors. In human prostate cancer tissues, high PCAT-1 expression predicts for low BRCA2 expression, supporting our observations in model systems. |
Paclitaxel
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-miR-216b-5p | [44] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Long non-coding RNA Linc00518 Can enhance GATA6 expression by suppressing miR-216b-5p expression to promotes paclitaxel resistance in the human prostate cancer. | |||
Key Molecule: Long non-protein coding RNA 518 (LINC00518) | [44] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Long non-coding RNA Linc00518 Can enhance GATA6 expression by suppressing miR-216b-5p expression to promotes paclitaxel resistance in the human prostate cancer. | |||
Key Molecule: hsa-mir-199a | [45] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Paclitaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | PC3/TXR cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay; Annexin V-FITC and PI Flow cytometry assay | |||
Mechanism Description | Overexpression of miR199a inhibited PTX resistance. YES1 was a target of miR199a, and overexpression of YES1 reversed the effect of miR199a in suppressing PTX resistance. In vivo, miR199a increased tumor PTX sensitivity. | |||
Key Molecule: hsa-mir-130a | [46] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Caspase-3 signaling pathway | Activation | hsa04210 | |
Cell apoptosis | Inhibition | hsa04210 | ||
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CellTiter-Glo luminescent cell viability assay | |||
Mechanism Description | Restoration of miR-130a activated caspase-8 and increased the drug sensitivity in taxane-resistant prostate cancer cells, suggesting that miR-130a may become a potential target for therapy of taxane-resistant CRPC. Since the mechanism of the action of miR-130a was different from that of paclitaxel, a combination therapy of paclitaxel and miR-130a mimic may be effective in treatment of CRPC. Furthermore, it was reported that miR-130a expression was decreased in prostate cancer tissues. It is therefore possible that the restoration of miR-130a could be an effective approach for treating not only taxane-resistant prostate cancer but also prostate cancer with reduced expression of miR-130a. | |||
Key Molecule: hsa-mir-135a | [47] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | A549 cells | Lung | Homo sapiens (Human) | CVCL_0023 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) is a protein that binds to HIF-1alpha and inhibits its transcriptional activity. HIF1AN is a potential miR-135a target listed in both the TargetScan and PicTar databases. miR-135a-mediated paclitaxel resistance is in part mediated by downregulation of APC. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Transcription factor GATA6 (GATA6) | [44] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
Experiment for Molecule Alteration |
Western blot analysis; RT-qPCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Long non-coding RNA Linc00518 Can enhance GATA6 expression by suppressing miR-216b-5p expression to promotes paclitaxel resistance in the human prostate cancer. | |||
Key Molecule: Tyrosine-protein kinase Yes (YES1) | [45] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Paclitaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | PC3/TXR cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis; Luciferase reporter assay | |||
Experiment for Drug Resistance |
MTT assay; Annexin V-FITC and PI Flow cytometry assay | |||
Mechanism Description | Overexpression of miR199a inhibited PTX resistance. YES1 was a target of miR199a, and overexpression of YES1 reversed the effect of miR199a in suppressing PTX resistance. In vivo, miR199a increased tumor PTX sensitivity. | |||
Key Molecule: SLAIN motif-containing protein 1 (SLAIN1) | [46] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Caspase-3 signaling pathway | Activation | hsa04210 | |
Cell apoptosis | Inhibition | hsa04210 | ||
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CellTiter-Glo luminescent cell viability assay | |||
Mechanism Description | Restoration of miR-130a activated caspase-8 and increased the drug sensitivity in taxane-resistant prostate cancer cells, suggesting that miR-130a may become a potential target for therapy of taxane-resistant CRPC. Since the mechanism of the action of miR-130a was different from that of paclitaxel, a combination therapy of paclitaxel and miR-130a mimic may be effective in treatment of CRPC. Furthermore, it was reported that miR-130a expression was decreased in prostate cancer tissues. It is therefore possible that the restoration of miR-130a could be an effective approach for treating not only taxane-resistant prostate cancer but also prostate cancer with reduced expression of miR-130a. | |||
Key Molecule: Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) | [47] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | A549 cells | Lung | Homo sapiens (Human) | CVCL_0023 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) is a protein that binds to HIF-1alpha and inhibits its transcriptional activity. HIF1AN is a potential miR-135a target listed in both the TargetScan and PicTar databases. miR-135a-mediated paclitaxel resistance is in part mediated by downregulation of APC. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-mir-148a | [48] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
MSK1 signaling pathway | Inhibition | hsa04010 | ||
In Vitro Model | PC3PR cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Cell Growth Assay | |||
Mechanism Description | MSk1 is a novel target gene of miR-148a in both PC3 and PC3PR cells and miR-148 attenuates paclitaxel-resistance of PC3PR cells by modulating MSk1 expression. | |||
Key Molecule: hsa-mir-34 | [49] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell proliferation | Inhibition | hsa05200 | ||
Notch1 signaling pathway | Inhibition | hsa04330 | ||
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | microRNA-34a Attenuates Paclitaxel Resistance in Prostate Cancer Cells via Direct Suppression of JAG1/Notch1 Axis. | |||
Key Molecule: hsa-mir-34 | [50] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell viability | Inhibition | hsa05200 | |
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
Trypan blue dye exclusion assay | |||
Mechanism Description | SIRT1 plays crucial roles in various cellular processes including cell survival under genotoxic and oxidative stresses. Bcl2I is an anti-apoptotic factor. In PC3PR cells, reduced expression of miR-34a confers paclitaxel resistance via up-regulating SIRT1 and Bcl2 expression. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Ribosomal protein S6 kinase alpha-5 (RPS6KA5) | [48] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell invasion | Inhibition | hsa05200 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
MSK1 signaling pathway | Inhibition | hsa04010 | ||
In Vitro Model | PC3PR cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Cell Growth Assay | |||
Mechanism Description | MSk1 is a novel target gene of miR-148a in both PC3 and PC3PR cells and miR-148 attenuates paclitaxel-resistance of PC3PR cells by modulating MSk1 expression. | |||
Key Molecule: Protein jagged-1 (JAG1) | [49] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
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 colony | Inhibition | hsa05200 | ||
Cell proliferation | Inhibition | hsa05200 | ||
Notch1 signaling pathway | Inhibition | hsa04330 | ||
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | microRNA-34a Attenuates Paclitaxel Resistance in Prostate Cancer Cells via Direct Suppression of JAG1/Notch1 Axis. | |||
Key Molecule: Neurogenic locus notch homolog protein 1 (NOTCH1) | [49] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
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 colony | Inhibition | hsa05200 | ||
Cell proliferation | Inhibition | hsa05200 | ||
Notch1 signaling pathway | Inhibition | hsa04330 | ||
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | microRNA-34a Attenuates Paclitaxel Resistance in Prostate Cancer Cells via Direct Suppression of JAG1/Notch1 Axis. | |||
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) | [50] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell viability | Inhibition | hsa05200 | |
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Trypan blue dye exclusion assay | |||
Mechanism Description | SIRT1 plays crucial roles in various cellular processes including cell survival under genotoxic and oxidative stresses. Bcl2I is an anti-apoptotic factor. In PC3PR cells, reduced expression of miR-34a confers paclitaxel resistance via up-regulating SIRT1 and Bcl2 expression. | |||
Key Molecule: NAD-dependent protein deacetylase sirtuin-1 (SIRT1) | [50] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell viability | Inhibition | hsa05200 | |
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Trypan blue dye exclusion assay | |||
Mechanism Description | SIRT1 plays crucial roles in various cellular processes including cell survival under genotoxic and oxidative stresses. Bcl2I is an anti-apoptotic factor. In PC3PR cells, reduced expression of miR-34a confers paclitaxel resistance via up-regulating SIRT1 and Bcl2 expression. |
Probenecid
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Irregularity in Drug Uptake and Drug Efflux (IDUE) | ||||
Key Molecule: ATP-binding cassette sub-family C2 (ABCC2) | [9] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Probenecid | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell autophagy | Inhibition | hsa04140 | |
Cell cytotoxicity | Activation | hsa04650 | ||
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | However, probenecid only weakly inhibits ABCG2. Thus, probenecid enhanced the efficacy of anticancer drugs against 22Rv1 spheroids by inhibiting drug resistance-related transporters such as MRP; at high probenecid concentrations, the chemosensitization effect may be reduced owing to promotion of alternate drug excretion pathways via upregulated ABCG2 expression. | |||
Key Molecule: Multidrug resistance-associated protein 1 (MRP1) | [9] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Probenecid | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell colony | Inhibition | hsa05200 | |
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | However, probenecid only weakly inhibits ABCG2. Thus, probenecid enhanced the efficacy of anticancer drugs against 22Rv1 spheroids by inhibiting drug resistance-related transporters such as MRP; at high probenecid concentrations, the chemosensitization effect may be reduced owing to promotion of alternate drug excretion pathways via upregulated ABCG2 expression. |
Quercetin
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) | [51] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Down-regulation | Expression |
||
Resistant Drug | Quercetin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | PC-3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
In Vivo Model | Male BALB/c nude mice xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Overexpression assay; qRT-PCR; Western bloting analysis; Immunohistochemistry analysis | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | MALAT1 Overexpression in PC cells resulted in the resistance against quercetin treatment. |
Sirolimus
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: Growth arrest specific 5 (GAS5) | [37] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Sirolimus | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
PNT2C2 cells | Prostate | Homo sapiens (Human) | CVCL_4889 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
GAS5 assay; MTS assay | |||
Mechanism Description | First generation mTORC1, combined mTORC1/mTORC2 and dual PI3k/mTOR inhibitors all increased cellular GAS5 levels and inhibited culture growth in androgen-dependent (LNCaP) and androgen-sensitive (22Rv1) cell lines, but not in androgen-independent (PC-3 and DU 145) cell lines. The latter exhibited low endogenous GAS5 expression, and GAS5 silencing in LNCaP and 22Rv1 cells decreased the sensitivity to mTOR inhibitors, whereas transfection of GAS5 LncRNA sensitized PC-3 and DU 145 cells to these agents. |
Sparfloxacin
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: DNA topoisomerase (ATP-hydrolyzing) (PARC) | [41] | |||
Resistant Disease | Mycoplasma hominis prostate cancer [ICD-11: 2C82.Y] | |||
Molecule Alteration | Missense mutation | p.K134R |
||
Resistant Drug | Sparfloxacin | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Mycoplasma hominis ATCC 23114(PG21) | 347256 | ||
Mycoplasma hominis isolate | 2098 | |||
Experiment for Molecule Alteration |
Whole genome sequence assay | |||
Mechanism Description | The single amino acid mutation in ParC of MH may relate to the resistance to OFX and LVX and the high-level resistance to fluoroquinolones for MH is associated with mutations in both DNA gyrase and the ParC subunit of topoisomerase IV. |
Temsirolimus
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: Growth arrest specific 5 (GAS5) | [37] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Temsirolimus | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
PNT2C2 cells | Prostate | Homo sapiens (Human) | CVCL_4889 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
GAS5 assay; MTS assay | |||
Mechanism Description | First generation mTORC1, combined mTORC1/mTORC2 and dual PI3k/mTOR inhibitors all increased cellular GAS5 levels and inhibited culture growth in androgen-dependent (LNCaP) and androgen-sensitive (22Rv1) cell lines, but not in androgen-independent (PC-3 and DU 145) cell lines. The latter exhibited low endogenous GAS5 expression, and GAS5 silencing in LNCaP and 22Rv1 cells decreased the sensitivity to mTOR inhibitors, whereas transfection of GAS5 LncRNA sensitized PC-3 and DU 145 cells to these agents. |
Verteporfin
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Myb-related protein B (MYBL2) | [52] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Verteporfin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HEp-2 cells | Skin | Homo sapiens (Human) | CVCL_1906 |
U251 cells | Brain | Homo sapiens (Human) | CVCL_0021 | |
BT474 cells | Breast | Homo sapiens (Human) | CVCL_0179 | |
A172 cells | Brain | Homo sapiens (Human) | CVCL_0131 | |
U87 cells | Brain | Homo sapiens (Human) | CVCL_0022 | |
H1299 cells | Lung | Homo sapiens (Human) | CVCL_0060 | |
Calu-3 cells | Lung | Homo sapiens (Human) | CVCL_0609 | |
HuTu80 cells | Small intestine | Homo sapiens (Human) | CVCL_1301 | |
In Vivo Model | Male BALB/c nude mouse model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis; qRT-PCR | |||
Experiment for Drug Resistance |
WST-1 assay; Colony formation assay; Annexin V-FITC/PI Apoptosis assay | |||
Mechanism Description | MYBL2 expression was significantly upregulated in CRPC tissues and cell lines. Overexpression of MYBL2 could facilitate castration-resistant growth and metastatic capacity in androgen-dependent PCa cells by promoting YAP1 transcriptional activity via modulating the activity of the Rho GTPases RhoA and LATS1 kinase. Importantly, targeting MYBL2, or treatment with either the YAP/TAZ inhibitor Verteporfin or the RhoA inhibitor Simvastatin, reversed the resistance to ADT and blocked bone metastasis in CRPC cells. | |||
Key Molecule: Transcriptional coactivator YAP1 (YAP1) | [52] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Verteporfin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HEp-2 cells | Skin | Homo sapiens (Human) | CVCL_1906 |
U251 cells | Brain | Homo sapiens (Human) | CVCL_0021 | |
BT474 cells | Breast | Homo sapiens (Human) | CVCL_0179 | |
A172 cells | Brain | Homo sapiens (Human) | CVCL_0131 | |
U87 cells | Brain | Homo sapiens (Human) | CVCL_0022 | |
H1299 cells | Lung | Homo sapiens (Human) | CVCL_0060 | |
Calu-3 cells | Lung | Homo sapiens (Human) | CVCL_0609 | |
HuTu80 cells | Small intestine | Homo sapiens (Human) | CVCL_1301 | |
In Vivo Model | Male BALB/c nude mouse model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis; qRT-PCR | |||
Experiment for Drug Resistance |
WST-1 assay; Colony formation assay; Annexin V-FITC/PI Apoptosis assay | |||
Mechanism Description | MYBL2 expression was significantly upregulated in CRPC tissues and cell lines. Overexpression of MYBL2 could facilitate castration-resistant growth and metastatic capacity in androgen-dependent PCa cells by promoting YAP1 transcriptional activity via modulating the activity of the Rho GTPases RhoA and LATS1 kinase. Importantly, targeting MYBL2, or treatment with either the YAP/TAZ inhibitor Verteporfin or the RhoA inhibitor Simvastatin, reversed the resistance to ADT and blocked bone metastasis in CRPC cells. |
Sulforaphane
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: Glyceraldehyde-3-phosphate dehydrogenase 1 (GAPDH) | [53] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Sulforaphane | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Activation | hsa05200 | |
In Vitro Model | LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
Cell proliferation assay | |||
Mechanism Description | Knockdown of LINC01116 with siRNA decreased proliferation of prostate cancer cells, and significantly upregulated several genes including GAPDH (regulates glycolysis), MAP1LC3B2 (autophagy) and H2AFY (chromatin structure) and LncRNA LINC01116 is upregulated in a human prostate cancer cell line, is decreased by SFN treatment, and promotes cell proliferation in a human cancer cell line. | |||
Key Molecule: Core histone macro-H2A.1 (H2AFY) | [53] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Sulforaphane | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Activation | hsa05200 | |
In Vitro Model | LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
Cell proliferation assay | |||
Mechanism Description | Knockdown of LINC01116 with siRNA decreased proliferation of prostate cancer cells, and significantly upregulated several genes including GAPDH (regulates glycolysis), MAP1LC3B2 (autophagy) and H2AFY (chromatin structure) and LncRNA LINC01116 is upregulated in a human prostate cancer cell line, is decreased by SFN treatment, and promotes cell proliferation in a human cancer cell line. | |||
Key Molecule: Long non-protein coding RNA 1116 (LINC01116) | [53] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Sulforaphane | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Activation | hsa05200 | |
In Vitro Model | LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
Sequencing assay; qRT-PCR | |||
Experiment for Drug Resistance |
Cell proliferation assay | |||
Mechanism Description | Knockdown of LINC01116 with siRNA decreased proliferation of prostate cancer cells, and significantly upregulated several genes including GAPDH (regulates glycolysis), MAP1LC3B2 (autophagy) and H2AFY (chromatin structure) and LncRNA LINC01116 is upregulated in a human prostate cancer cell line, is decreased by SFN treatment, and promotes cell proliferation in a human cancer cell line. | |||
Key Molecule: Microtubule-associated proteins 1A/1B light chain 3 beta 2 (MAP1LC3B2) | [53] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Sulforaphane | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Activation | hsa05200 | |
In Vitro Model | LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
Cell proliferation assay | |||
Mechanism Description | Knockdown of LINC01116 with siRNA decreased proliferation of prostate cancer cells, and significantly upregulated several genes including GAPDH (regulates glycolysis), MAP1LC3B2 (autophagy) and H2AFY (chromatin structure) and LncRNA LINC01116 is upregulated in a human prostate cancer cell line, is decreased by SFN treatment, and promotes cell proliferation in a human cancer cell line. |
Clinical Trial Drug(s)
8 drug(s) in total
Camptothecin
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-mir-34 | [54] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Camptothecin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell growth | Inhibition | hsa05200 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
PrEC cells | Prostate | Homo sapiens (Human) | CVCL_0061 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
Trypan blue dye exclusion assay | |||
Mechanism Description | Inhibition of the SIRT1 activity or expression resulted in attenuation of cell proliferation and chemoresistance in PC3 and DU145 cells. Ectopic expression of miR-34a decreased the SIRT1 mRNA and protein levels as well as protein levels of known direct target genes. Ectopic miR-34a expression resulted in cell cycle arrest and growth inhibition and attenuated chemoresistance to anticancer drug camptothecin by inducing apoptosis. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: NAD-dependent protein deacetylase sirtuin-1 (SIRT1) | [54] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Camptothecin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell growth | Inhibition | hsa05200 | ||
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
PrEC cells | Prostate | Homo sapiens (Human) | CVCL_0061 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Trypan blue dye exclusion assay | |||
Mechanism Description | Inhibition of the SIRT1 activity or expression resulted in attenuation of cell proliferation and chemoresistance in PC3 and DU145 cells. Ectopic expression of miR-34a decreased the SIRT1 mRNA and protein levels as well as protein levels of known direct target genes. Ectopic miR-34a expression resulted in cell cycle arrest and growth inhibition and attenuated chemoresistance to anticancer drug camptothecin by inducing apoptosis. |
Dactolisib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: Growth arrest specific 5 (GAS5) | [37] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Dactolisib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
PNT2C2 cells | Prostate | Homo sapiens (Human) | CVCL_4889 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
GAS5 assay; MTS assay | |||
Mechanism Description | First generation mTORC1, combined mTORC1/mTORC2 and dual PI3k/mTOR inhibitors all increased cellular GAS5 levels and inhibited culture growth in androgen-dependent (LNCaP) and androgen-sensitive (22Rv1) cell lines, but not in androgen-independent (PC-3 and DU 145) cell lines. The latter exhibited low endogenous GAS5 expression, and GAS5 silencing in LNCaP and 22Rv1 cells decreased the sensitivity to mTOR inhibitors, whereas transfection of GAS5 LncRNA sensitized PC-3 and DU 145 cells to these agents. |
Genistein
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: HOX transcript antisense RNA (HOTAIR) | [55] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Genistein | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTS assay | |||
Mechanism Description | Knockdown (siRNA) of HOTAIR decreased PCa cell proliferation, migration and invasion and induced apoptosis and cell cycle arrest. miR-34a was also up-regulated by genistein and may directly target HOTAIR in both PC3 and DU145 PCa cells. | |||
Key Molecule: hsa-mir-34 | [55] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Genistein | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell migration | Inhibition | hsa04670 | ||
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Dual-luciferase reporter assays | |||
Experiment for Drug Resistance |
MTS assay | |||
Mechanism Description | Knockdown (siRNA) of HOTAIR decreased PCa cell proliferation, migration and invasion and induced apoptosis and cell cycle arrest. miR-34a was also up-regulated by genistein and may directly target HOTAIR in both PC3 and DU145 PCa cells. |
Veliparib dihydrochloride
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: Prostate cancer associated transcript 1 (PCAT1) | [43] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Veliparib dihydrochloride | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
RWPE cells | Prostate | Homo sapiens (Human) | CVCL_1736 | |
In Vivo Model | SCID nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
WST assay | |||
Mechanism Description | PCAT-1 expressing cells exhibit a BRCA-like phenotype, resulting in cell sensitization to PARP1 inhibitors. In human prostate cancer tissues, high PCAT-1 expression predicts for low BRCA2 expression, supporting our observations in model systems. |
Canertinib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-mir-203 | [56] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Canertinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Activation | hsa04670 | |
Cell proliferation | Activation | hsa05200 | ||
EGFR/RAS signaling pathway | Activation | hsa01521 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | The induction of bone metastasis and TkI resistance require miR-203 down-regulation, activation of the EGFR pathway via altered expression of EGFR ligands (EREG and TGFA) and anti-apoptotic proteins (API5, BIRC2, and TRIAP1). Importantly, a sufficient reconstitution of invasiveness and resistance to TkIs treatment was observed in cells transfected with anti-miR-203. In prostate cancer patients, miR-203 levels were inversely correlated with the expression of two EGFR ligands, EREG and TGFA, and an EGFR dependent gene signature. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Amphiregulin (AREG) | [56] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Canertinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Activation | hsa04670 | |
Cell proliferation | Activation | hsa05200 | ||
EGFR/RAS signaling pathway | Activation | hsa01521 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | The induction of bone metastasis and TkI resistance require miR-203 down-regulation, activation of the EGFR pathway via altered expression of EGFR ligands (EREG and TGFA) and anti-apoptotic proteins (API5, BIRC2, and TRIAP1). Importantly, a sufficient reconstitution of invasiveness and resistance to TkIs treatment was observed in cells transfected with anti-miR-203. In prostate cancer patients, miR-203 levels were inversely correlated with the expression of two EGFR ligands, EREG and TGFA, and an EGFR dependent gene signature. | |||
Key Molecule: Proepiregulin (EREG) | [56] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Canertinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Activation | hsa04670 | |
Cell proliferation | Activation | hsa05200 | ||
EGFR/RAS signaling pathway | Activation | hsa01521 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | The induction of bone metastasis and TkI resistance require miR-203 down-regulation, activation of the EGFR pathway via altered expression of EGFR ligands (EREG and TGFA) and anti-apoptotic proteins (API5, BIRC2, and TRIAP1). Importantly, a sufficient reconstitution of invasiveness and resistance to TkIs treatment was observed in cells transfected with anti-miR-203. In prostate cancer patients, miR-203 levels were inversely correlated with the expression of two EGFR ligands, EREG and TGFA, and an EGFR dependent gene signature. | |||
Key Molecule: Protransforming growth factor alpha (TGFA) | [56] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Canertinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Activation | hsa04670 | |
Cell proliferation | Activation | hsa05200 | ||
EGFR/RAS signaling pathway | Activation | hsa01521 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | The induction of bone metastasis and TkI resistance require miR-203 down-regulation, activation of the EGFR pathway via altered expression of EGFR ligands (EREG and TGFA) and anti-apoptotic proteins (API5, BIRC2, and TRIAP1). Importantly, a sufficient reconstitution of invasiveness and resistance to TkIs treatment was observed in cells transfected with anti-miR-203. In prostate cancer patients, miR-203 levels were inversely correlated with the expression of two EGFR ligands, EREG and TGFA, and an EGFR dependent gene signature. |
CH-5132799
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: PI3-kinase alpha (PIK3CA) | [57] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Missense mutation | p.Q546R (c.1637A>G) |
||
Sensitive Drug | CH-5132799 | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | KPL-4 cells | Breast | Homo sapiens (Human) | CVCL_5310 |
IGROV1 cells | Ovary | Homo sapiens (Human) | CVCL_1304 | |
GXF97 cells | N.A. | . | N.A. | |
In Vivo Model | Female BALB-nu/nu mouse xenograft model | Mus musculus | ||
Experiment for Drug Resistance |
CCK-8 assay | |||
Mechanism Description | The missense mutation p.Q546R (c.1637A>G) in gene PIK3CA cause the sensitivity of CH-5132799 by aberration of the drug's therapeutic target |
AZD-8055
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: Growth arrest specific 5 (GAS5) | [37] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | AZD-8055 | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
LNCaP cells | Prostate | Homo sapiens (Human) | CVCL_0395 | |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
PNT2C2 cells | Prostate | Homo sapiens (Human) | CVCL_4889 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
GAS5 assay; MTS assay | |||
Mechanism Description | First generation mTORC1, combined mTORC1/mTORC2 and dual PI3k/mTOR inhibitors all increased cellular GAS5 levels and inhibited culture growth in androgen-dependent (LNCaP) and androgen-sensitive (22Rv1) cell lines, but not in androgen-independent (PC-3 and DU 145) cell lines. The latter exhibited low endogenous GAS5 expression, and GAS5 silencing in LNCaP and 22Rv1 cells decreased the sensitivity to mTOR inhibitors, whereas transfection of GAS5 LncRNA sensitized PC-3 and DU 145 cells to these agents. |
Trichostatin A
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-mir-181a | [58] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Trichostatin A | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
In Vitro Model | C4-2B cells | Prostate | Homo sapiens (Human) | CVCL_4784 |
Experiment for Molecule Alteration |
RT-PCR; qRT-PCR | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | GRP78 up-regulation is a major contributor to tumorigenesis and therapeutic resistance, miR-30d, miR-181a and miR-199a-5p regulate GRP78 and that their decreased expression in tumor cells results in increased GRP78 levels, which in turn promotes tumorigenesis and therapeutic resistance. | |||
Key Molecule: hsa-mir-30d | [58] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Trichostatin A | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
In Vitro Model | HL60 cells | Peripheral blood | Homo sapiens (Human) | CVCL_0002 |
Experiment for Molecule Alteration |
RT-PCR; qRT-PCR | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | GRP78 up-regulation is a major contributor to tumorigenesis and therapeutic resistance, miR-30d, miR-181a and miR-199a-5p regulate GRP78 and that their decreased expression in tumor cells results in increased GRP78 levels, which in turn promotes tumorigenesis and therapeutic resistance. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Endoplasmic reticulum chaperone BiP (HSPA5) | [58] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Trichostatin A | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
In Vitro Model | C4-2B cells | Prostate | Homo sapiens (Human) | CVCL_4784 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | GRP78 up-regulation is a major contributor to tumorigenesis and therapeutic resistance, miR-30d, miR-181a and miR-199a-5p regulate GRP78 and that their decreased expression in tumor cells results in increased GRP78 levels, which in turn promotes tumorigenesis and therapeutic resistance. |
Discontinued Drug(s)
1 drug(s) in total
AZD-6482
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Phosphatase and tensin homolog (PTEN) | [59] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Missense mutation | p.A126G (c.377C>G) |
||
Sensitive Drug | AZD-6482 | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Presto blue assay; Clonogenic cell survival assay |
Investigative Drug(s)
8 drug(s) in total
Alpha-solanine
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: Growth arrest specific 5 (GAS5) | [60] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Down-regulation | Expression |
||
Resistant Drug | Alpha-solanine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | GAS5/miR-18a signaling pathway | Activation | hsa05206 | |
In Vitro Model | RWPE-1 cells | Prostate | Homo sapiens (Human) | CVCL_3791 |
DU145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 | |
Experiment for Molecule Alteration |
Overexpression assay | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | Long noncoding RNA GAS5 modulates alpha-Solanine-induced radiosensitivity by negatively regulating miR-18a in human prostate cancer cells. |
Bufalin
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: HOX transcript antisense RNA (HOTAIR) | [61] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Down-regulation | Interaction |
||
Resistant Drug | Bufalin | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | DU145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC-3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
Experiment for Molecule Alteration |
In situ hybridization assay; Immunohistochemical assay; Overexpression assay; Microarray assay | |||
Experiment for Drug Resistance |
IC50 assay | |||
Mechanism Description | Bufalin suppresses the migration and invasion of prostate cancer cells through HOTAIR, the sponge of miR-520b. |
Gardiquimod
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Irregularity in Drug Uptake and Drug Efflux (IDUE) | ||||
Key Molecule: Multidrug resistance protein 1 (ABCB1) | [62] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Gardiquimod | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | Hep3B cells | Liver | Homo sapiens (Human) | CVCL_0326 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Resazurin Cell Viability Assay | |||
Mechanism Description | Imidazoquinolines IMQ, RSQ, and GDQ are substrates for P-gp and begins to elucidate differences in their trafficking in cancer cells as a consequence of acquired drug resistance. We believe this work that begins to examine imidazoquinoline trafficking will prove useful in the future rational design of immunotherapeutics with enhanced susceptibility to P-gp efflux that enable increased bioavailability, in MDR cancers. |
Imiquimod
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Irregularity in Drug Uptake and Drug Efflux (IDUE) | ||||
Key Molecule: Multidrug resistance protein 1 (ABCB1) | [62] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imiquimod | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | Hep3B cells | Liver | Homo sapiens (Human) | CVCL_0326 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Resazurin Cell Viability Assay | |||
Mechanism Description | Imidazoquinolines IMQ, RSQ, and GDQ are substrates for P-gp and begins to elucidate differences in their trafficking in cancer cells as a consequence of acquired drug resistance. We believe this work that begins to examine imidazoquinoline trafficking will prove useful in the future rational design of immunotherapeutics with enhanced susceptibility to P-gp efflux that enable increased bioavailability, in MDR cancers. |
Nutlin-3
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: Growth arrest specific 5 (GAS5) | [24] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Nutlin-3 | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
In Vitro Model | PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 |
22RV1 cells | Prostate | Homo sapiens (Human) | CVCL_1045 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
Fluorescence microscopy test apoptosis assay | |||
Mechanism Description | Transient expression of GAS5 enhances apoptosis and decreases the survival of 22Rv1 cells, forced variation of GAS5 gene expression can modulate cellular responses to various apoptotic stimuli, including a range of chemotherapeutic drugs. |
Platinum
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: Schlafen family member 11 (SLFN11) | [63] | |||
Resistant Disease | Primary prostate cancer [ICD-11: 2C82.Z] | |||
Molecule Alteration | Alteration | Epigenetic silencing |
||
Resistant Drug | Platinum | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
PC3 cells | Prostate | Homo sapiens (Human) | CVCL_0035 | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Methyl-CpG binding domain protein-enriched genome sequencing assay;Methylated DNA Immunoprecipitation Sequencing assay | |||
Mechanism Description | Epigenetic silencing of SLFN11 has been associated with resistance to platinum-based chemotherapies in a number of cell lines including DU-145 and PC3. |
Resiquimod
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Irregularity in Drug Uptake and Drug Efflux (IDUE) | ||||
Key Molecule: Multidrug resistance protein 1 (ABCB1) | [62] | |||
Resistant Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Resiquimod | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | Hep3B cells | Liver | Homo sapiens (Human) | CVCL_0326 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Resazurin Cell Viability Assay | |||
Mechanism Description | Imidazoquinolines IMQ, RSQ, and GDQ are substrates for P-gp and begins to elucidate differences in their trafficking in cancer cells as a consequence of acquired drug resistance. We believe this work that begins to examine imidazoquinoline trafficking will prove useful in the future rational design of immunotherapeutics with enhanced susceptibility to P-gp efflux that enable increased bioavailability, in MDR cancers. |
Tyrphostin AG-1478
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-mir-203 | [56] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Tyrphostin AG-1478 | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Activation | hsa04670 | |
Cell proliferation | Activation | hsa05200 | ||
EGFR/RAS signaling pathway | Activation | hsa01521 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | The induction of bone metastasis and TkI resistance require miR-203 down-regulation, activation of the EGFR pathway via altered expression of EGFR ligands (EREG and TGFA) and anti-apoptotic proteins (API5, BIRC2, and TRIAP1). Importantly, a sufficient reconstitution of invasiveness and resistance to TkIs treatment was observed in cells transfected with anti-miR-203. In prostate cancer patients, miR-203 levels were inversely correlated with the expression of two EGFR ligands, EREG and TGFA, and an EGFR dependent gene signature. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Amphiregulin (AREG) | [56] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Tyrphostin AG-1478 | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Activation | hsa04670 | |
Cell proliferation | Activation | hsa05200 | ||
EGFR/RAS signaling pathway | Activation | hsa01521 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | The induction of bone metastasis and TkI resistance require miR-203 down-regulation, activation of the EGFR pathway via altered expression of EGFR ligands (EREG and TGFA) and anti-apoptotic proteins (API5, BIRC2, and TRIAP1). Importantly, a sufficient reconstitution of invasiveness and resistance to TkIs treatment was observed in cells transfected with anti-miR-203. In prostate cancer patients, miR-203 levels were inversely correlated with the expression of two EGFR ligands, EREG and TGFA, and an EGFR dependent gene signature. | |||
Key Molecule: Proepiregulin (EREG) | [56] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Tyrphostin AG-1478 | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Activation | hsa04670 | |
Cell proliferation | Activation | hsa05200 | ||
EGFR/RAS signaling pathway | Activation | hsa01521 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | The induction of bone metastasis and TkI resistance require miR-203 down-regulation, activation of the EGFR pathway via altered expression of EGFR ligands (EREG and TGFA) and anti-apoptotic proteins (API5, BIRC2, and TRIAP1). Importantly, a sufficient reconstitution of invasiveness and resistance to TkIs treatment was observed in cells transfected with anti-miR-203. In prostate cancer patients, miR-203 levels were inversely correlated with the expression of two EGFR ligands, EREG and TGFA, and an EGFR dependent gene signature. | |||
Key Molecule: Protransforming growth factor alpha (TGFA) | [56] | |||
Sensitive Disease | Prostate cancer [ICD-11: 2C82.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Tyrphostin AG-1478 | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Activation | hsa04670 | |
Cell proliferation | Activation | hsa05200 | ||
EGFR/RAS signaling pathway | Activation | hsa01521 | ||
In Vitro Model | DU-145 cells | Prostate | Homo sapiens (Human) | CVCL_0105 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | The induction of bone metastasis and TkI resistance require miR-203 down-regulation, activation of the EGFR pathway via altered expression of EGFR ligands (EREG and TGFA) and anti-apoptotic proteins (API5, BIRC2, and TRIAP1). Importantly, a sufficient reconstitution of invasiveness and resistance to TkIs treatment was observed in cells transfected with anti-miR-203. In prostate cancer patients, miR-203 levels were inversely correlated with the expression of two EGFR ligands, EREG and TGFA, and an EGFR dependent gene signature. |
References
If you find any error in data or bug in web service, please kindly report it to Dr. Sun and Dr. Zhang.