Disease Information
General Information of the Disease (ID: DIS00093)
Name |
Endometrial cancer
|
---|---|
ICD |
ICD-11: 2C76
|
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)
8 drug(s) in total
Cisplatin
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-mir-200b | [1] | |||
Resistant Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HEC-1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 |
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
Clonogenic assay | |||
Mechanism Description | The transcription factor AP-2alpha functions as a tumor suppressor by regulating various genes that are involved in cell proliferation and apoptosis. Chemotherapeutic drugs including cisplatin induce post-transcriptionally endogenous AP-2alpha, which contributes to chemosensitivity by enhancing therapy-induced apoptosis. miR-200b/200c/429 family recognized the MRE in the 3' UTR of AP-2alpha gene and negatively regulated the expression of endogenous AP-2alpha proteins. | |||
Key Molecule: hsa-mir-200c | [1] | |||
Resistant Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HEC-1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 |
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
Clonogenic assay | |||
Mechanism Description | The transcription factor AP-2alpha functions as a tumor suppressor by regulating various genes that are involved in cell proliferation and apoptosis. Chemotherapeutic drugs including cisplatin induce post-transcriptionally endogenous AP-2alpha, which contributes to chemosensitivity by enhancing therapy-induced apoptosis. miR-200b/200c/429 family recognized the MRE in the 3' UTR of AP-2alpha gene and negatively regulated the expression of endogenous AP-2alpha proteins. | |||
Key Molecule: hsa-miR-429 | [1] | |||
Resistant Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HEC-1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 |
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
Clonogenic assay | |||
Mechanism Description | The transcription factor AP-2alpha functions as a tumor suppressor by regulating various genes that are involved in cell proliferation and apoptosis. Chemotherapeutic drugs including cisplatin induce post-transcriptionally endogenous AP-2alpha, which contributes to chemosensitivity by enhancing therapy-induced apoptosis. miR-200b/200c/429 family recognized the MRE in the 3' UTR of AP-2alpha gene and negatively regulated the expression of endogenous AP-2alpha proteins. | |||
Irregularity in Drug Uptake and Drug Efflux (IDUE) | ||||
Key Molecule: Multidrug resistance protein 1 (ABCB1) | [2] | |||
Resistant Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell autophagy | Inhibition | hsa04140 | |
In Vitro Model | Ishikawa cells | Endometrium | Homo sapiens (Human) | CVCL_2529 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Dual-color autophagy reporter assay; CCK8 assay; Flow cytometric analysis | |||
Mechanism Description | HOTAIR can regulate the cisplatin-resistance ability of human endometrial cancer cells through the regulation of autophagy by increasing Beclin-1, MDR, and P-gp expression. | |||
Key Molecule: Multidrug resistance protein 3 (ABCB4) | [2] | |||
Resistant Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell autophagy | Inhibition | hsa04140 | |
In Vitro Model | Ishikawa cells | Endometrium | Homo sapiens (Human) | CVCL_2529 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Dual-color autophagy reporter assay; CCK8 assay; Flow cytometric analysis | |||
Mechanism Description | HOTAIR can regulate the cisplatin-resistance ability of human endometrial cancer cells through the regulation of autophagy by increasing Beclin-1, MDR, and P-gp expression. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Beclin-1 (BECN1) | [2] | |||
Resistant Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell autophagy | Inhibition | hsa04140 | |
In Vitro Model | Ishikawa cells | Endometrium | Homo sapiens (Human) | CVCL_2529 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Dual-color autophagy reporter assay; CCK8 assay; Flow cytometric analysis | |||
Mechanism Description | HOTAIR can regulate the cisplatin-resistance ability of human endometrial cancer cells through the regulation of autophagy by increasing Beclin-1, MDR, and P-gp expression. | |||
Key Molecule: Transcription factor AP2 alpha (TFAP2A) | [1] | |||
Resistant Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HEC-1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Clonogenic assay | |||
Mechanism Description | The transcription factor AP-2alpha functions as a tumor suppressor by regulating various genes that are involved in cell proliferation and apoptosis. Chemotherapeutic drugs including cisplatin induce post-transcriptionally endogenous AP-2alpha, which contributes to chemosensitivity by enhancing therapy-induced apoptosis. miR-200b/200c/429 family recognized the MRE in the 3' UTR of AP-2alpha gene and negatively regulated the expression of endogenous AP-2alpha proteins. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: HOX transcript antisense RNA (HOTAIR) | [2] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell autophagy | Activation | hsa04140 | |
In Vitro Model | Ishikawa cells | Endometrium | Homo sapiens (Human) | CVCL_2529 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
Dual-color autophagy reporter assay; CCK8 assay; Flow cytometric analysis | |||
Mechanism Description | HOTAIR can regulate the cisplatin-resistance ability of human endometrial cancer cells through the regulation of autophagy by increasing Beclin-1, MDR, and P-gp expression. | |||
Key Molecule: hsa-mir-23b | [3] | |||
Sensitive Disease | Endometrial carcinoma [ICD-11: 2C76.2] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Cisplatin | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | HEC1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 |
Human normal endometrial epithelial cell line | Uterus | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
RNA pull-down assay; qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometric analysis | |||
Mechanism Description | Long non-coding RNA TUSC7 acted as a potential tumor suppressor gene to inhibit cell growth as well as advance the chemotherapy sensitivity through targeted silencing of miR23b. | |||
Key Molecule: Tumor suppressor candidate 7 (TUSC7) | [3] | |||
Sensitive Disease | Endometrial carcinoma [ICD-11: 2C76.2] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Cisplatin | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | HEC1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 |
Human normal endometrial epithelial cell line | Uterus | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometric analysis | |||
Mechanism Description | Long non-coding RNA TUSC7 acted as a potential tumor suppressor gene to inhibit cell growth as well as advance the chemotherapy sensitivity through targeted silencing of miR23b. | |||
Irregularity in Drug Uptake and Drug Efflux (IDUE) | ||||
Key Molecule: Multidrug resistance protein (MDR) | [2] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell autophagy | Activation | hsa04140 | |
In Vitro Model | Ishikawa cells | Endometrium | Homo sapiens (Human) | CVCL_2529 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Dual-color autophagy reporter assay; CCK8 assay; Flow cytometric analysis | |||
Mechanism Description | HOTAIR can regulate the cisplatin-resistance ability of human endometrial cancer cells through the regulation of autophagy by increasing Beclin-1, MDR, and P-gp expression. | |||
Key Molecule: ATP-binding cassette sub-family B5 (ABCB5) | [2] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell autophagy | Activation | hsa04140 | |
In Vitro Model | Ishikawa cells | Endometrium | Homo sapiens (Human) | CVCL_2529 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Dual-color autophagy reporter assay; CCK8 assay; Flow cytometric analysis | |||
Mechanism Description | HOTAIR can regulate the cisplatin-resistance ability of human endometrial cancer cells through the regulation of autophagy by increasing Beclin-1, MDR, and P-gp expression. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Beclin-1 (BECN1) | [2] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Cisplatin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell autophagy | Activation | hsa04140 | |
In Vitro Model | Ishikawa cells | Endometrium | Homo sapiens (Human) | CVCL_2529 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Dual-color autophagy reporter assay; CCK8 assay; Flow cytometric analysis | |||
Mechanism Description | HOTAIR can regulate the cisplatin-resistance ability of human endometrial cancer cells through the regulation of autophagy by increasing Beclin-1, MDR, and P-gp expression. |
Docetaxel
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Polycomb complex protein BMI-1 (BMI1) | [4] | |||
Resistant Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Mutation | . |
||
Resistant Drug | Docetaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | PI3K/AKT signaling pathway | Activation | hsa04151 | |
Experiment for Molecule Alteration |
Low throughput experiment assay | |||
Experiment for Drug Resistance |
Disease-free survival analysis | |||
Mechanism Description | Recently, Dong et al. demonstrated that loss of BMI1 in endometrial cancer cells reduces expression of drug resistance gene MRP1, suggesting that BMI1 is required for the drug resistance. Overexpression of BMI1 rescues tumor cells from the apoptosis induced by Okadaic acid and Epigallocatechin-3-gallate, well-known apoptotic agents. |
Epothilone B
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Regulation by the Disease Microenvironment (RTDM) | ||||
Key Molecule: hsa-mir-200c | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Epothilone B | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Key Molecule: BDNF/NT-3 growth factors receptor (NTRK2) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Epothilone B | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Key Molecule: Protein quaking (QKI) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Epothilone B | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Key Molecule: Zinc finger E-box-binding homeobox 1 (ZEB1) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Epothilone B | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Key Molecule: Zinc finger E-box-binding homeobox 2 (ZEB2) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Epothilone B | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Fibronectin (FN1) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Epothilone B | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Key Molecule: Tubulin beta-3 chain (TUBB3) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Epothilone B | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. |
Medroxyprogesterone
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: Progesterone receptor (PGR) | [6] | |||
Resistant Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Medroxyprogesterone | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | SR786 cells | Pleural effusion | Homo sapiens (Human) | CVCL_1711 |
IshikawaPR cells | Endometrium | Homo sapiens (Human) | CVCL_2529 | |
Experiment for Molecule Alteration |
RTPCR | |||
Mechanism Description | The presence of the progesterone receptor (PR) is the precondition for progesterone response and PR is a predictive marker for response of progesterone. Progesterone binds to its receptor PR-A and PR-B, subsequently inhibiting tumor growth and promoting tumor apoptosis by regulating downstream genes. Constant stimulation of progesterone reduced the expression of PGR and promoted the development of drug resistance. Thus, downregulation of PR especially PRB must be involved in progesterone resistance. However, the molecular mechanism of PGR dysfunction remains unclear. |
Medroxyprogesterone acetate
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: Long non-protein coding RNA (LNC-CETP-3) | [7] | |||
Resistant Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Up-regulation | Expression |
||
Resistant Drug | Medroxyprogesterone acetate | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | Ishikawa cells | Endometrium | Homo sapiens (Human) | CVCL_2529 |
KLE cells | Ovary | Homo sapiens (Human) | CVCL_1329 | |
Experiment for Molecule Alteration |
Microarray assay; qRT-PCR | |||
Mechanism Description | Medroxyprogesterone acetate causes the alterations of endoplasmic reticulum related mRNAs and LncRNAs in endometrial cancer cells. |
Paclitaxel
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: Long non-protein coding RNA 672 (LINC00672) | [8] | |||
Resistant Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | 293T cells | Breast | Homo sapiens (Human) | CVCL_0063 |
Ishikawa cells | Endometrium | Homo sapiens (Human) | CVCL_2529 | |
HEC-1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Transwell migration assay; Matrigel invasion assay; Flow cytometry assay; TUNEL assay; Wound healing assay; Colony formation assay | |||
Mechanism Description | LINC00672 can down-regulate LASP1 expression as a locus-restricted cofactor for p53-mediated gene suppression, thus impacting EC malig.ncies and chemosensitivity to paclitaxel. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: LIM and SH3 domain protein 1 (LASP1) | [8] | |||
Resistant Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | 293T cells | Breast | Homo sapiens (Human) | CVCL_0063 |
Ishikawa cells | Endometrium | Homo sapiens (Human) | CVCL_2529 | |
HEC-1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Transwell migration assay; Matrigel invasion assay; Flow cytometry assay; TUNEL assay; Wound healing assay; Colony formation assay | |||
Mechanism Description | LINC00672 can down-regulate LASP1 expression as a locus-restricted cofactor for p53-mediated gene suppression, thus impacting EC malig.ncies and chemosensitivity to paclitaxel. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: hsa-mir-23b | [3] | |||
Sensitive Disease | Endometrial carcinoma [ICD-11: 2C76.2] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | HEC1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 |
Human normal endometrial epithelial cell line | Uterus | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
RNA pull-down assay; qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometric analysis | |||
Mechanism Description | Long non-coding RNA TUSC7 acted as a potential tumor suppressor gene to inhibit cell growth as well as advance the chemotherapy sensitivity through targeted silencing of miR23b. | |||
Key Molecule: Tumor suppressor candidate 7 (TUSC7) | [3] | |||
Sensitive Disease | Endometrial carcinoma [ICD-11: 2C76.2] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | HEC1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 |
Human normal endometrial epithelial cell line | Uterus | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometric analysis | |||
Mechanism Description | Long non-coding RNA TUSC7 acted as a potential tumor suppressor gene to inhibit cell growth as well as advance the chemotherapy sensitivity through targeted silencing of miR23b. | |||
Key Molecule: hsa-miR-29c-3p | [9] | |||
Sensitive Disease | Endometrial carcinoma [ICD-11: 2C76.2] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell colony | Inhibition | hsa05200 | |
Cell invasion | Inhibition | hsa05200 | ||
Cell viability | Inhibition | hsa05200 | ||
miR125a-5p/BCL2/MRP4 signaling pathway | Regulation | hsa05206 | ||
In Vitro Model | Ishikawa cells | Endometrium | Homo sapiens (Human) | CVCL_2529 |
HEC-1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 | |
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | The up-regulation of miR-29c-3p using exogenous mimic molecules markedly increased PTX sensitivity in both cell lines and reduced expression of kDM5B while the inhibitor of miR-29-3p resulted in the opposite effects. | |||
Key Molecule: hsa-mir-24 | [10] | |||
Sensitive Disease | Endometrial carcinoma [ICD-11: 2C76.2] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | HEC-1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 |
In Vivo Model | Crl:NU-Foxn1nu nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-24, which is under-expressed in EC, functions as a tumor-suppressing gene to inhibit malignant proliferation and advance chemotherapy sensitivity to paclitaxel in EC by targeted silencing of S100A8. | |||
Regulation by the Disease Microenvironment (RTDM) | ||||
Key Molecule: hsa-mir-200c | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Key Molecule: BDNF/NT-3 growth factors receptor (NTRK2) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Key Molecule: Protein quaking (QKI) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Key Molecule: Zinc finger E-box-binding homeobox 1 (ZEB1) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Key Molecule: Zinc finger E-box-binding homeobox 2 (ZEB2) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Lysine-specific demethylase 5B (KDM5B) | [9] | |||
Sensitive Disease | Endometrial carcinoma [ICD-11: 2C76.2] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell colony | Inhibition | hsa05200 | |
Cell invasion | Inhibition | hsa05200 | ||
Cell viability | Inhibition | hsa05200 | ||
miR125a-5p/BCL2/MRP4 signaling pathway | Regulation | hsa05206 | ||
In Vitro Model | Ishikawa cells | Endometrium | Homo sapiens (Human) | CVCL_2529 |
HEC-1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | The up-regulation of miR-29c-3p using exogenous mimic molecules markedly increased PTX sensitivity in both cell lines and reduced expression of kDM5B while the inhibitor of miR-29-3p resulted in the opposite effects. | |||
Key Molecule: Protein S100-A8 (S100A8) | [10] | |||
Sensitive Disease | Endometrial carcinoma [ICD-11: 2C76.2] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | HEC-1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-24, which is under-expressed in EC, functions as a tumor-suppressing gene to inhibit malignant proliferation and advance chemotherapy sensitivity to paclitaxel in EC by targeted silencing of S100A8. | |||
Key Molecule: Fibronectin (FN1) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Key Molecule: Tubulin beta-3 chain (TUBB3) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Paclitaxel | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. |
Progesterone
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Epigenetic Alteration of DNA, RNA or Protein (EADR) | ||||
Key Molecule: Nuclear paraspeckle assembly transcript 1 (NEAT1) | [11] | |||
Resistant Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Progesterone | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell cycle arrest | Activation | hsa04110 | ||
In Vitro Model | 293T cells | Breast | Homo sapiens (Human) | CVCL_0063 |
Ishikawa cells | Endometrium | Homo sapiens (Human) | CVCL_2529 | |
Experiment for Molecule Alteration |
Microarray | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | Progesterone Repressed LncRNA NEAT1,LEF1, c-myc, and MMP9 in Wnt/beta-catenin Signaling Pathway via Inhibition of NEAT1/miRNA-146b-5p Axis in Endometrial Cancer. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Lymphoid enhancer-binding factor 1 (LEF1) | [11] | |||
Resistant Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Progesterone | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell colony | Activation | hsa05200 | ||
Cell cycle | Activation | hsa04110 | ||
In Vitro Model | 293T cells | Breast | Homo sapiens (Human) | CVCL_0063 |
Ishikawa cells | Endometrium | Homo sapiens (Human) | CVCL_2529 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | Progesterone Repressed LncRNA NEAT1,LEF1, c-myc, and MMP9 in Wnt/beta-catenin Signaling Pathway via Inhibition of NEAT1/miRNA-146b-5p Axis in Endometrial Cancer. |
Vincristine
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Regulation by the Disease Microenvironment (RTDM) | ||||
Key Molecule: hsa-mir-200c | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Vincristine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Key Molecule: BDNF/NT-3 growth factors receptor (NTRK2) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Vincristine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Key Molecule: Protein quaking (QKI) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Vincristine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Key Molecule: Zinc finger E-box-binding homeobox 1 (ZEB1) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Vincristine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Key Molecule: Zinc finger E-box-binding homeobox 2 (ZEB2) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Vincristine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Fibronectin (FN1) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Vincristine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. | |||
Key Molecule: Tubulin beta-3 chain (TUBB3) | [5] | |||
Sensitive Disease | Endometrial cancer [ICD-11: 2C76.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Vincristine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell migration | Inhibition | hsa04670 | |
In Vitro Model | Hec50 cells | Endometrium | Homo sapiens (Human) | CVCL_2929 |
Experiment for Molecule Alteration |
Immunoblotting analysis | |||
Experiment for Drug Resistance |
ELISA assay | |||
Mechanism Description | Low or absent miR-200c results in aberrant expression of ZEB1 and consequent repression of E-cadherin. Reinstatement of miR-200c to such cells restores E-cadherin and dramatically reduces migration and invasion. One such gene, class IIIbeta-tubulin (TUBB3), which encodes a tubulin isotype normally found only in neuronal cells, is a direct target of miR-200c. Restoration of miR-200c increases sensitivity to microtubule-targeting agents by up to 85%. Since expression of TUBB3 is a common mechanism of resistance to microtubule-binding chemotherapeutic agents in many types of solid tumors, the ability of miR-200c to restore chemosensitivity to such agents may be explained by its ability to reduce TUBB3. |
Clinical Trial Drug(s)
12 drug(s) in total
Brivanib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Fibroblast growth factor receptor 2 (FGFR2) | [12] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.S252W (c.755C>G) |
||
Sensitive Drug | Brivanib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | SUP-M2 cells | Colon | Homo sapiens (Human) | CVCL_2209 |
KARPAS-299 cells | Peripheral blood | Homo sapiens (Human) | CVCL_1324 | |
In Vivo Model | mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTS assay | |||
Mechanism Description | The missense mutation p.S252W (c.755C>G) in gene FGFR2 cause the sensitivity of Brivanib by unusual activation of pro-survival pathway |
Capivasertib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: RAC-alpha serine/threonine-protein kinase (AKT1) | [13] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.E17K (c.49G>A) |
||
Sensitive Drug | Capivasertib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Capivasertib by aberration of the drug's therapeutic target | |||
Key Molecule: RAC-alpha serine/threonine-protein kinase (AKT1) | [14] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.E17K (c.49G>A) |
||
Sensitive Drug | Capivasertib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Breast | . | ||
Mechanism Description | The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Capivasertib by aberration of the drug's therapeutic target |
Cediranib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Fibroblast growth factor receptor 2 (FGFR2) | [12] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.N549K (c.1647T>G) |
||
Sensitive Drug | Cediranib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | SUP-M2 cells | Colon | Homo sapiens (Human) | CVCL_2209 |
KARPAS-299 cells | Peripheral blood | Homo sapiens (Human) | CVCL_1324 | |
In Vivo Model | mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTS assay | |||
Mechanism Description | The missense mutation p.N549K (c.1647T>G) in gene FGFR2 cause the sensitivity of Cediranib by unusual activation of pro-survival pathway | |||
Key Molecule: Fibroblast growth factor receptor 2 (FGFR2) | [12] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.S252W (c.755C>G) |
||
Sensitive Drug | Cediranib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | SUP-M2 cells | Colon | Homo sapiens (Human) | CVCL_2209 |
KARPAS-299 cells | Peripheral blood | Homo sapiens (Human) | CVCL_1324 | |
In Vivo Model | mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTS assay | |||
Mechanism Description | The missense mutation p.S252W (c.755C>G) in gene FGFR2 cause the sensitivity of Cediranib by unusual activation of pro-survival pathway |
Derazantinib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Fibroblast growth factor receptor 2 (FGFR2) | [15] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.S252W (c.755C>G) |
||
Sensitive Drug | Derazantinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | FGF/FGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | A2780 cells | Ovary | Homo sapiens (Human) | CVCL_0134 |
KG-1 cells | Bone marrow | Homo sapiens (Human) | CVCL_0374 | |
J82 cells | Bladder | Homo sapiens (Human) | CVCL_0359 | |
RT4 cells | Bladder | Homo sapiens (Human) | CVCL_0036 | |
NCI-H716 cells | Colon | Homo sapiens (Human) | CVCL_1581 | |
SNU-16 cells | Gastric | Homo sapiens (Human) | CVCL_0076 | |
AN3CA cells | Ovary | Homo sapiens (Human) | CVCL_0028 | |
SkOV3 cells | Ovary | Homo sapiens (Human) | CVCL_0532 | |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
SW780 cells | Bladder | Homo sapiens (Human) | CVCL_1728 | |
KATO-3 cells | Gastric | Homo sapiens (Human) | CVCL_0371 | |
RT-112 cells | Urinary bladder | Homo sapiens (Human) | CVCL_1670 | |
MFM-223 cells | Pleural effusion | Homo sapiens (Human) | CVCL_1408 | |
MFE296 cells | Endometrium | Homo sapiens (Human) | CVCL_1406 | |
MFE280 cells | Endometrium | Homo sapiens (Human) | CVCL_1405 | |
COS-1 cells | Kidney | Chlorocebus aethiops (Green monkey) | CVCL_0223 | |
In Vivo Model | SCID beige mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTS assay | |||
Mechanism Description | In cells, inhibition of FGFR2 auto-phosphorylation and other proteins downstream in the FGFR pathway (FRS2alpha, AKT, ERK) was evident by the response to ARQ 087 treatment. Cell proliferation studies demonstrated ARQ 087 has anti-proliferative activity in cell lines driven by FGFR dysregulation, including amplifications, fusions, and mutations. Cell cycle studies in cell lines with high levels of FGFR2 protein showed a positive relationship between ARQ 087 induced G1 cell cycle arrest and subsequent induction of apoptosis. In addition, ARQ 087 was effective at inhibiting tumor growth in vivo in FGFR2 altered, SNU-16 and NCI-H716, xenograft tumor models with gene amplifications and fusions. | |||
Key Molecule: Fibroblast growth factor receptor 2 (FGFR2) | [15] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.N549K (c.1647T>G) |
||
Sensitive Drug | Derazantinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | FGF/FGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | A2780 cells | Ovary | Homo sapiens (Human) | CVCL_0134 |
KG-1 cells | Bone marrow | Homo sapiens (Human) | CVCL_0374 | |
J82 cells | Bladder | Homo sapiens (Human) | CVCL_0359 | |
RT4 cells | Bladder | Homo sapiens (Human) | CVCL_0036 | |
NCI-H716 cells | Colon | Homo sapiens (Human) | CVCL_1581 | |
SNU-16 cells | Gastric | Homo sapiens (Human) | CVCL_0076 | |
AN3CA cells | Ovary | Homo sapiens (Human) | CVCL_0028 | |
SkOV3 cells | Ovary | Homo sapiens (Human) | CVCL_0532 | |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
SW780 cells | Bladder | Homo sapiens (Human) | CVCL_1728 | |
KATO-3 cells | Gastric | Homo sapiens (Human) | CVCL_0371 | |
RT-112 cells | Urinary bladder | Homo sapiens (Human) | CVCL_1670 | |
MFM-223 cells | Pleural effusion | Homo sapiens (Human) | CVCL_1408 | |
MFE296 cells | Endometrium | Homo sapiens (Human) | CVCL_1406 | |
MFE280 cells | Endometrium | Homo sapiens (Human) | CVCL_1405 | |
COS-1 cells | Kidney | Chlorocebus aethiops (Green monkey) | CVCL_0223 | |
In Vivo Model | SCID beige mouse PDX model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTS assay | |||
Mechanism Description | In cells, inhibition of FGFR2 auto-phosphorylation and other proteins downstream in the FGFR pathway (FRS2alpha, AKT, ERK) was evident by the response to ARQ 087 treatment. Cell proliferation studies demonstrated ARQ 087 has anti-proliferative activity in cell lines driven by FGFR dysregulation, including amplifications, fusions, and mutations. Cell cycle studies in cell lines with high levels of FGFR2 protein showed a positive relationship between ARQ 087 induced G1 cell cycle arrest and subsequent induction of apoptosis. In addition, ARQ 087 was effective at inhibiting tumor growth in vivo in FGFR2 altered, SNU-16 and NCI-H716, xenograft tumor models with gene amplifications and fusions. |
Selumetinib
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Fibroblast growth factor receptor 2 (FGFR2) | [16] | |||
Resistant Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.S252W (c.755C>G) |
||
Resistant Drug | Selumetinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HCT116 cells | Colon | Homo sapiens (Human) | CVCL_0291 |
NCI-N87 cells | Gastric | Homo sapiens (Human) | CVCL_1603 | |
MkN-45 cells | Gastric | Homo sapiens (Human) | CVCL_0434 | |
NCI-H716 cells | Colon | Homo sapiens (Human) | CVCL_1581 | |
SNU-16 cells | Gastric | Homo sapiens (Human) | CVCL_0076 | |
NCI-H520 cells | Lung | Homo sapiens (Human) | CVCL_1566 | |
RT-4 cells | Urinary bladder | Homo sapiens (Human) | CVCL_0036 | |
ZR75-1 cells | Breast | Homo sapiens (Human) | CVCL_0588 | |
NCI-N87 cells | Gastric | Homo sapiens (Human) | CVCL_1603 | |
NCI-H716 cells | Colon | Homo sapiens (Human) | CVCL_1581 | |
KATO-3 cells | Gastric | Homo sapiens (Human) | CVCL_0371 | |
SNU-16 cells | Gastric | Homo sapiens (Human) | CVCL_0076 | |
NCI-H520 cells | Lung | Homo sapiens (Human) | CVCL_1566 | |
RT-4 cells | Urinary bladder | Homo sapiens (Human) | CVCL_0036 | |
UM-UC-14 cells | Kidney | Homo sapiens (Human) | CVCL_2747 | |
SUM-52PE cells | Pleural effusion | Homo sapiens (Human) | CVCL_3425 | |
NCI-H1581 cells | Lung | Homo sapiens (Human) | CVCL_1479 | |
MFE296 cells | Endometrium | Homo sapiens (Human) | CVCL_1406 | |
MFE280 cells | Endometrium | Homo sapiens (Human) | CVCL_1405 | |
KMS-11 cells | Pleural effusion | Homo sapiens (Human) | CVCL_2989 | |
HSC-39 cells | Ascites | Homo sapiens (Human) | CVCL_A385 | |
DMS-114 cells | Lung | Homo sapiens (Human) | CVCL_1174 | |
AN3 CA cells | Endometrium | Homo sapiens (Human) | CVCL_0028 | |
UM-UC-14 cells | Kidney | Homo sapiens (Human) | CVCL_2747 | |
KATO-III cells | Pleural effusion | Homo sapiens (Human) | CVCL_0371 | |
AN3 CA cells | Endometrium | Homo sapiens (Human) | CVCL_0028 | |
Experiment for Molecule Alteration |
Microarray assay; Western blotting analysis | |||
Experiment for Drug Resistance |
CCK-8 assay |
AZD-4547
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Fibroblast growth factor receptor 2 (FGFR2) | [17] | |||
Resistant Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.S252W (c.755C>G) |
||
Resistant Drug | AZD-4547 | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HEK293T cells | Kidney | Homo sapiens (Human) | CVCL_0063 |
U2OS cells | Bone | Homo sapiens (Human) | CVCL_0042 | |
Ishikawa cells | Endometrium | Homo sapiens (Human) | CVCL_2529 | |
HEC1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 | |
MV4-11 cells | Peripheral blood | Homo sapiens (Human) | CVCL_0064 | |
TT cells | Thyroid gland | Homo sapiens (Human) | CVCL_1774 | |
MOLM-1 cells | Bone marrow | Homo sapiens (Human) | CVCL_2188 | |
MFE296 cells | Endometrium | Homo sapiens (Human) | CVCL_1406 | |
MFE296 cells | Endometrium | Homo sapiens (Human) | CVCL_1406 | |
MFE280 cells | Endometrium | Homo sapiens (Human) | CVCL_1405 | |
MFE280 cells | Endometrium | Homo sapiens (Human) | CVCL_1405 | |
In Vivo Model | Female balb/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Phospho-kinase array analysis; Reporter gene assay; Microarray analysis; RT-PCR; Gene set enrichment analysis | |||
Experiment for Drug Resistance |
MTT assay; Soft-agar colony assay | |||
Key Molecule: Fibroblast growth factor receptor 2 (FGFR2) | [17] | |||
Resistant Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.N550K (c.1650T>G) |
||
Resistant Drug | AZD-4547 | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HEK293T cells | Kidney | Homo sapiens (Human) | CVCL_0063 |
U2OS cells | Bone | Homo sapiens (Human) | CVCL_0042 | |
Ishikawa cells | Endometrium | Homo sapiens (Human) | CVCL_2529 | |
HEC1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 | |
MV4-11 cells | Peripheral blood | Homo sapiens (Human) | CVCL_0064 | |
TT cells | Thyroid gland | Homo sapiens (Human) | CVCL_1774 | |
MOLM-1 cells | Bone marrow | Homo sapiens (Human) | CVCL_2188 | |
MFE296 cells | Endometrium | Homo sapiens (Human) | CVCL_1406 | |
MFE296 cells | Endometrium | Homo sapiens (Human) | CVCL_1406 | |
MFE280 cells | Endometrium | Homo sapiens (Human) | CVCL_1405 | |
MFE280 cells | Endometrium | Homo sapiens (Human) | CVCL_1405 | |
In Vivo Model | Female balb/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Phospho-kinase array analysis; Reporter gene assay; Microarray analysis; RT-PCR; Gene set enrichment analysis | |||
Experiment for Drug Resistance |
MTT assay; Soft-agar colony assay |
DEBIO-1347
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: Fibroblast growth factor receptor 2 (FGFR2) | [18] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.S252W (c.755C>G) |
||
Sensitive Drug | DEBIO-1347 | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | 327 cells | N.A. | . | N.A. |
In Vivo Model | Female BALB-nu/nu mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CCK-8 assay | |||
Key Molecule: Fibroblast growth factor receptor 2 (FGFR2) | [18] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.N549K (c.1647T>G) |
||
Sensitive Drug | DEBIO-1347 | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | 327 cells | N.A. | . | N.A. |
In Vivo Model | Female BALB-nu/nu mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
CCK-8 assay |
Miransertib
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: RAC-alpha serine/threonine-protein kinase (AKT1) | [19] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.E17K (c.49G>A) |
||
Sensitive Drug | Miransertib | |||
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 | |
MDA-MB-231 cells | Breast | Homo sapiens (Human) | CVCL_0062 | |
Hela cells | Cervix uteri | Homo sapiens (Human) | CVCL_0030 | |
T47D cells | Breast | Homo sapiens (Human) | CVCL_0553 | |
NCI-H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 | |
MDA-MB-453 cells | Breast | Homo sapiens (Human) | CVCL_0418 | |
MDA-MB-468 cells | Breast | Homo sapiens (Human) | CVCL_0419 | |
HCC70 cells | Breast | Homo sapiens (Human) | CVCL_1270 | |
A2058 cells | Skin | Homo sapiens (Human) | CVCL_1059 | |
Caco-2 cells | Colon | Homo sapiens (Human) | CVCL_0025 | |
MCF-7 cells | Breast | Homo sapiens (Human) | CVCL_0031 | |
ZR75-1 cells | Breast | Homo sapiens (Human) | CVCL_0588 | |
NCI-60 cells | N.A. | Homo sapiens (Human) | N.A. | |
KU-19 cells | Blood | Bos taurus (Bovine) | CVCL_VN09 | |
EVSA-T cells | Ascites | Homo sapiens (Human) | CVCL_1207 | |
CAL-120 cells | Pleural effusion | Homo sapiens (Human) | CVCL_1104 | |
BT-549 cells | Breast | Homo sapiens (Human) | CVCL_1092 | |
BT-474 cells | Breast | Homo sapiens (Human) | CVCL_0179 | |
BT-20 cells | Mammary gland | Homo sapiens (Human) | CVCL_0178 | |
B16F10 cells | Skin | Mus musculus (Mouse) | CVCL_0159 | |
In Vivo Model | Female NMRI (nu/nu) mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Mechanism Description | The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Miransertib by aberration of the drug's therapeutic target |
RO-5126766 free base
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Fibroblast growth factor receptor 2 (FGFR2) | [16] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.S252W (c.755C>G) |
||
Sensitive Drug | RO-5126766 free base | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HCT116 cells | Colon | Homo sapiens (Human) | CVCL_0291 |
NCI-N87 cells | Gastric | Homo sapiens (Human) | CVCL_1603 | |
MkN-45 cells | Gastric | Homo sapiens (Human) | CVCL_0434 | |
NCI-H716 cells | Colon | Homo sapiens (Human) | CVCL_1581 | |
SNU-16 cells | Gastric | Homo sapiens (Human) | CVCL_0076 | |
NCI-H520 cells | Lung | Homo sapiens (Human) | CVCL_1566 | |
RT-4 cells | Urinary bladder | Homo sapiens (Human) | CVCL_0036 | |
ZR75-1 cells | Breast | Homo sapiens (Human) | CVCL_0588 | |
NCI-N87 cells | Gastric | Homo sapiens (Human) | CVCL_1603 | |
NCI-H716 cells | Colon | Homo sapiens (Human) | CVCL_1581 | |
KATO-3 cells | Gastric | Homo sapiens (Human) | CVCL_0371 | |
SNU-16 cells | Gastric | Homo sapiens (Human) | CVCL_0076 | |
NCI-H520 cells | Lung | Homo sapiens (Human) | CVCL_1566 | |
RT-4 cells | Urinary bladder | Homo sapiens (Human) | CVCL_0036 | |
UM-UC-14 cells | Kidney | Homo sapiens (Human) | CVCL_2747 | |
SUM-52PE cells | Pleural effusion | Homo sapiens (Human) | CVCL_3425 | |
NCI-H1581 cells | Lung | Homo sapiens (Human) | CVCL_1479 | |
MFE296 cells | Endometrium | Homo sapiens (Human) | CVCL_1406 | |
MFE280 cells | Endometrium | Homo sapiens (Human) | CVCL_1405 | |
KMS-11 cells | Pleural effusion | Homo sapiens (Human) | CVCL_2989 | |
HSC-39 cells | Ascites | Homo sapiens (Human) | CVCL_A385 | |
DMS-114 cells | Lung | Homo sapiens (Human) | CVCL_1174 | |
AN3 CA cells | Endometrium | Homo sapiens (Human) | CVCL_0028 | |
UM-UC-14 cells | Kidney | Homo sapiens (Human) | CVCL_2747 | |
KATO-III cells | Pleural effusion | Homo sapiens (Human) | CVCL_0371 | |
AN3 CA cells | Endometrium | Homo sapiens (Human) | CVCL_0028 | |
Experiment for Molecule Alteration |
Microarray assay; Western blotting analysis | |||
Experiment for Drug Resistance |
CCK-8 assay |
CH-5132799
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: PI3-kinase alpha (PIK3CA) | [20] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.H1047Y (c.3139C>T) |
||
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.H1047Y (c.3139C>T) in gene PIK3CA cause the sensitivity of CH-5132799 by aberration of the drug's therapeutic target |
MTOR inhibitors
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: PI3-kinase regulatory subunit beta (PIK3R2) | [21] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.N561D (c.1681A>G) |
||
Sensitive Drug | MTOR inhibitors | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
Experiment for Molecule Alteration |
Whole-gene resequencing assay | |||
Mechanism Description | The missense mutation p.N561D (c.1681A>G) in gene PIK3R2 cause the sensitivity of MTOR inhibitors by unusual activation of pro-survival pathway | |||
Key Molecule: PI3-kinase regulatory subunit beta (PIK3R2) | [21] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.A171V (c.512C>T) |
||
Sensitive Drug | MTOR inhibitors | |||
Experimental Note | Identified from the Human Clinical Data |
PRN1371
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Unusual Activation of Pro-survival Pathway (UAPP) | ||||
Key Molecule: Fibroblast growth factor receptor 2 (FGFR2) | [22] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.N549K (c.1647T>G) |
||
Sensitive Drug | PRN1371 | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | FGF/FGFR signaling pathway | Inhibition | hsa01521 | |
In Vitro Model | HCT116 cells | Colon | Homo sapiens (Human) | CVCL_0291 |
Hep3B cells | Liver | Homo sapiens (Human) | CVCL_0326 | |
RT4 cells | Bladder | Homo sapiens (Human) | CVCL_0036 | |
NCI-H716 cells | Colon | Homo sapiens (Human) | CVCL_1581 | |
RT112 cells | Bladder | Homo sapiens (Human) | CVCL_1670 | |
AN3CA cells | Ovary | Homo sapiens (Human) | CVCL_0028 | |
Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 | |
SNU878 cells | Liver | Homo sapiens (Human) | CVCL_5102 | |
SNU16 cells | Ascites | Homo sapiens (Human) | CVCL_0076 | |
OPM2 cells | Peripheral blood | Homo sapiens (Human) | CVCL_1625 | |
LI7 cells | Liver | Homo sapiens (Human) | CVCL_3840 | |
JHH7 cells | Liver | Homo sapiens (Human) | CVCL_2805 | |
In Vivo Model | Nude mouse PDX model | Mus musculus | ||
Experiment for Drug Resistance |
Promega assay | |||
Mechanism Description | PRN1371 exhibits potent and durable pathway inhibition, and robust antiproliferative activity. PRN1371 demonstrates prolonged FGFR inhibition in vivo. |
Preclinical Drug(s)
5 drug(s) in total
ARQ 751
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: RAC-alpha serine/threonine-protein kinase (AKT1) | [19] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.E17K (c.49G>A) |
||
Sensitive Drug | ARQ 751 | |||
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 | |
MDA-MB-231 cells | Breast | Homo sapiens (Human) | CVCL_0062 | |
Hela cells | Cervix uteri | Homo sapiens (Human) | CVCL_0030 | |
T47D cells | Breast | Homo sapiens (Human) | CVCL_0553 | |
NCI-H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 | |
MDA-MB-453 cells | Breast | Homo sapiens (Human) | CVCL_0418 | |
MDA-MB-468 cells | Breast | Homo sapiens (Human) | CVCL_0419 | |
HCC70 cells | Breast | Homo sapiens (Human) | CVCL_1270 | |
A2058 cells | Skin | Homo sapiens (Human) | CVCL_1059 | |
Caco-2 cells | Colon | Homo sapiens (Human) | CVCL_0025 | |
MCF-7 cells | Breast | Homo sapiens (Human) | CVCL_0031 | |
ZR75-1 cells | Breast | Homo sapiens (Human) | CVCL_0588 | |
NCI-60 cells | N.A. | Homo sapiens (Human) | N.A. | |
KU-19 cells | Blood | Bos taurus (Bovine) | CVCL_VN09 | |
EVSA-T cells | Ascites | Homo sapiens (Human) | CVCL_1207 | |
CAL-120 cells | Pleural effusion | Homo sapiens (Human) | CVCL_1104 | |
BT-549 cells | Breast | Homo sapiens (Human) | CVCL_1092 | |
BT-474 cells | Breast | Homo sapiens (Human) | CVCL_0179 | |
BT-20 cells | Mammary gland | Homo sapiens (Human) | CVCL_0178 | |
B16F10 cells | Skin | Mus musculus (Mouse) | CVCL_0159 | |
In Vivo Model | Female NMRI (nu/nu) mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Mechanism Description | The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of ARQ 751 by aberration of the drug's therapeutic target |
E7090
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: Fibroblast growth factor receptor 2 (FGFR2) | [23] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.S252W (c.755C>G) |
||
Sensitive Drug | E7090 | |||
Experimental Note | Identified from the Human Clinical Data |
FIIN-1
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: Fibroblast growth factor receptor 2 (FGFR2) | [24] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.N549K (c.1647T>G) |
||
Sensitive Drug | FIIN-1 | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | Ba/F3 cells | Colon | Homo sapiens (Human) | CVCL_0161 |
Experiment for Drug Resistance |
CellTiter-Glo Luminescent Cell Viability Assay | |||
Mechanism Description | The missense mutation p.N549K (c.1647T>G) in gene FGFR2 cause the sensitivity of FIIN-1 by aberration of the drug's therapeutic target |
GSK3052230
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: Fibroblast growth factor receptor 2 (FGFR2) | [25] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.S252W (c.755C>G) |
||
Sensitive Drug | GSK3052230 | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | Lung | . | ||
Experiment for Molecule Alteration |
QuantiGene Plex DNA assay | |||
Mechanism Description | The missense mutation p.S252W (c.755C>G) in gene FGFR2 cause the sensitivity of GSK3052230 by aberration of the drug's therapeutic target |
Spliceostatin A
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: Splicing factor 3B subunit 1 (SF3B1) | [26] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.K666N (c.1998G>C) |
||
Sensitive Drug | Spliceostatin A | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | PANC-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0480 |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
Capan-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0026 | |
HEC1A cells | Uterus | Homo sapiens (Human) | CVCL_0293 | |
Capan-1 cells | Pancreas | Homo sapiens (Human) | CVCL_0237 | |
Capan-2 cells | Pancreas | Homo sapiens (Human) | CVCL_0026 | |
Pancreatic Panc 0504 cells | Pancreas | Homo sapiens (Human) | CVCL_1637 | |
MFE296 cells | Endometrium | Homo sapiens (Human) | CVCL_1406 | |
HEC59 cells | Endometrium | Homo sapiens (Human) | CVCL_2930 | |
ESS-1 cells | Endometrium | Homo sapiens (Human) | CVCL_1205 | |
DSMZ cells | N.A. | . | N.A. | |
ESS-1 cells | Endometrium | Homo sapiens (Human) | CVCL_1205 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CellTiter-Glo assay |
Investigative Drug(s)
1 drug(s) in total
PD173074
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
Aberration of the Drug's Therapeutic Target (ADTT) | ||||
Key Molecule: Fibroblast growth factor receptor 2 (FGFR2) | [27] | |||
Sensitive Disease | Endometrial adenocarcinoma [ICD-11: 2C76.0] | |||
Molecule Alteration | Missense mutation | p.N550K (c.1650T>A) |
||
Sensitive Drug | PD173074 | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Uterus | . | ||
Experiment for Molecule Alteration |
DNA sequencing assay | |||
Experiment for Drug Resistance |
Sulforhodamine B assay | |||
Mechanism Description | The missense mutation p.N550K (c.1650T>A) in gene FGFR2 cause the sensitivity of PD173074 by aberration of the drug's therapeutic target |
References
If you find any error in data or bug in web service, please kindly report it to Dr. Sun and Dr. Zhang.