General Information of the Disease (ID: DIS00046)
Name
Brain cancer
ICD
ICD-11: 2A00
Resistance Map
Type(s) of Resistant Mechanism of This Disease
  ADTT: Aberration of the Drug's Therapeutic Target
  DISM: Drug Inactivation by Structure Modification
  EADR: Epigenetic Alteration of DNA, RNA or Protein
  IDUE: Irregularity in Drug Uptake and Drug Efflux
  RTDM: Regulation by the Disease Microenvironment
  UAPP: Unusual Activation of Pro-survival Pathway
Drug Resistance Data Categorized by Drug
Approved Drug(s)
40 drug(s) in total
Click to Show/Hide the Full List of Drugs
Alectinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: ALK tyrosine kinase receptor (ALK) [1]
Resistant Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Missense mutation
p.F1174L
Resistant Drug Alectinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model NBLW cells Brain Homo sapiens (Human) CVCL_VJ90
NBLW-R cells Brain Homo sapiens (Human) CVCL_VJ91
Experiment for
Molecule Alteration
Sangersequencing assay; Targeted deep sequencing assay
Experiment for
Drug Resistance
Array CGH assay
Mechanism Description Analysis of the sensitivity of NBLW and NBLW-R cells to a panel of ALk inhibitors (TAE-684, Crizotinib, Alectinib and Lorlatinib) revealed differences between the paired cell lines, and overall NBLW-R cells with the F1174L mutation were more resistant to ALk inhibitor induced apoptosis compared with NBLW cells.
Anagrelide
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: L1 cell adhesion molecule (L1CAM) [2]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Anagrelide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Activation hsa04670
In Vitro Model MDCK cells Kidney Canis lupus familiaris (Dog) (Canis familiaris) CVCL_0422
Experiment for
Molecule Alteration
Puromycin selection and monitored regularly for the maintenance of L1 silencing assay
Experiment for
Drug Resistance
Migration assay
Mechanism Description With OVCAR3 cells treated with anagrelide, 2-hydroxy-5-fluoropyrimidine and mestranol , the gap width closure was seen from 48 h onward at all concentrations tested. Similar results were obtained with U251 cells, and L1's metastatic potential is further evidenced by its promotion of epithelial-mesenchymal transition, endothelial cell transcytosis and resistance to chemo- and radiotherapy.
Azacitidine
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Oxalosuccinate decarboxylase (IDH1) [3]
Sensitive Disease FGFR-tacc positive glioblastoma [ICD-11: 2A00.01]
Molecule Alteration Missense mutation
p.R132H (c.395G>A)
Sensitive Drug Azacitidine
Experimental Note Identified from the Human Clinical Data
In Vitro Model Brain .
In Vivo Model Female athymic nude mouse (NCI-Frederick) model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Tumor volume measurement assay
Bevacizumab
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Oxalosuccinate decarboxylase (IDH1) [4]
Sensitive Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration Missense mutation
p.R132S (c.394C>A)
Sensitive Drug Bevacizumab
Experimental Note Identified from the Human Clinical Data
In Vitro Model Brain .
Mechanism Description The missense mutation p.R132S (c.394C>A) in gene IDH1 cause the sensitivity of Bevacizumab by aberration of the drug's therapeutic target
Key Molecule: Oxalosuccinate decarboxylase (IDH1) [4]
Sensitive Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration Missense mutation
p.R132C (c.394C>T)
Sensitive Drug Bevacizumab
Experimental Note Identified from the Human Clinical Data
In Vitro Model Brain .
Mechanism Description The missense mutation p.R132C (c.394C>T) in gene IDH1 cause the sensitivity of Bevacizumab by aberration of the drug's therapeutic target
Key Molecule: Oxalosuccinate decarboxylase (IDH1) [4]
Sensitive Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration Missense mutation
p.R132L (c.395G>T)
Sensitive Drug Bevacizumab
Experimental Note Identified from the Human Clinical Data
In Vitro Model Brain .
Mechanism Description The missense mutation p.R132L (c.395G>T) in gene IDH1 cause the sensitivity of Bevacizumab by aberration of the drug's therapeutic target
Brigatinib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: ALK tyrosine kinase receptor (ALK) [5]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Missense mutation
p.F1174V (c.3520T>G)
Sensitive Drug Brigatinib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model IMR-32 cells Abdomen Homo sapiens (Human) CVCL_0346
PC12 cells Adrenal gland Rattus norvegicus (Rat) CVCL_0481
CLB-PE cells Brain Homo sapiens (Human) CVCL_9534
CLB-GE cells Bone marrow Homo sapiens (Human) CVCL_9530
CLB-BAR cells Brain Homo sapiens (Human) CVCL_9519
In Vivo Model Female Balbc/nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
Resazurin disc test assay
Mechanism Description Anaplastic lymphoma kinase (ALK) is a tyrosine kinase receptor which has been implicated in numerous solid and hematologic cancers. Brigatinib is an effective inhibitor of ALK kinase activity in ALK addicted neuroblastoma
Carmustine
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Drug Inactivation by Structure Modification (DISM) Click to Show/Hide
Key Molecule: Glutathione S-transferase P (GSTP1) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Carmustine
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
Key Molecule: Glutathione S-transferase P (GSTP1) [6]
Resistant Disease Anaplastic astrocytoma [ICD-11: 2A00.04]
Molecule Alteration Expression
Up-regulation
Resistant Drug Carmustine
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
Oncotech EDR assay
Mechanism Description GSTP1 is the first major mechanism of resistance alkylator agents encounter after entering the cancer cell cytoplasm. GSTP1 acts to enzymatically conjugate glutathione to the reactive metabolites of BCNU. The mechanisms by which GSTP1 may be up-regulated in gliomas are under investigation. Constitutive expression is thought to be influenced by the proximal promoter factor Sp1, whereas increased expression levels may result from stabilization of GSTP1 mRNA. GSTP1 expression has been reported to be induced by drug exposure, indicating that it may play a role in acquired drug resistance.
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-21 [7]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Resistant Drug Carmustine
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SWOZ2 cells Brain Homo sapiens (Human) N.A.
SWOZ2-BCNU cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR21 enhanced glioma cells resistance to carmustine via decreasing Spry2 expression.
Key Molecule: hsa-mir-221 [8]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Carmustine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
PI3-k/PTEN/AKT signaling axis Activation hsa04151
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-221 regulated cell proliferation and BCNU resistance in glioma cells. Overexpression of miR-221 led to cell survival and BCNU resistance and reduced cell apoptosis induced by BCNU, whereas knockdown of miR-221 inhibited cell proliferation and prompted BCNU sensitivity and cell apoptosis. Further investigation revealed that miR-221 down-regulated PTEN and activated Akt, which resulted in cell survival and BCNU resistance. Overexpression of PTEN lacking 3'UTR or PI3-k/Akt specific inhibitor wortmannin attenuated miR-221-mediated BCNU resistance and prompted cell apoptosis. We propose that miR-221 regulated cell proliferation and BCNU resistance in glioma cells by targeting PI3-k/PTEN/Akt signaling axis.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Carmustine
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Protein sprouty homolog 2 (SPRY2) [7]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Resistant Drug Carmustine
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SWOZ2 cells Brain Homo sapiens (Human) N.A.
SWOZ2-BCNU cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR21 enhanced glioma cells resistance to carmustine via decreasing Spry2 expression.
Key Molecule: Phosphatase and tensin homolog (PTEN) [8]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Carmustine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
PI3K/PTEN/AKT signaling axis Activation hsa04151
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-221 regulated cell proliferation and BCNU resistance in glioma cells. Overexpression of miR-221 led to cell survival and BCNU resistance and reduced cell apoptosis induced by BCNU, whereas knockdown of miR-221 inhibited cell proliferation and prompted BCNU sensitivity and cell apoptosis. Further investigation revealed that miR-221 down-regulated PTEN and activated Akt, which resulted in cell survival and BCNU resistance. Overexpression of PTEN lacking 3'UTR or PI3-k/Akt specific inhibitor wortmannin attenuated miR-221-mediated BCNU resistance and prompted cell apoptosis. We propose that miR-221 regulated cell proliferation and BCNU resistance in glioma cells by targeting PI3-k/PTEN/Akt signaling axis.
Key Molecule: Methylated-DNA--protein-cysteine methyltransferase (MGMT) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Carmustine
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
Key Molecule: Methylated-DNA--protein-cysteine methyltransferase (MGMT) [6]
Resistant Disease Anaplastic astrocytoma [ICD-11: 2A00.04]
Molecule Alteration Expression
Up-regulation
Resistant Drug Carmustine
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
Oncotech EDR assay
Mechanism Description For drugs that have evaded cytosolic mechanisms of drug resistance, the nucleus is equipped with the capacity to remove BCNU or temozolomide alkyl groups from the O6-position of guanine via a reaction catalyzed by MGMT. Repair occurs before cross-link formation and involves an irreversible stoichiometric covalent transfer of the O6-alkyl DNA adduct to a cysteine within the active site of MGMT, resulting in the inactivation and subsequent depletion of enzyme activity. MGMT-mediated repair is rapid, with a half-life of 35 hours. MGMT enzyme recovery occurs via de novo synthesis. In malignant glioma patients, MGMT overexpression has been associated with resistance to BCNU and similar alkylating agents and was an independent predictor of poor survival. MGMT is also thought to contribute to temozolomide resistance, which we did not detect in our study. This may be related to the in vitro pharmacokinetic differences between BCNU and temozolomide.
Celecoxib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Prostaglandin G/H synthase 2 (PTGS2) [9]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Celecoxib
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
Cell apoptosis Activation hsa04210
Cell autophagy Activation hsa04140
In Vitro Model MDA-175 cells Pleural effusion Homo sapiens (Human) CVCL_1400
MMQ cells Pituitary gland Rattus norvegicus (Rat) CVCL_2117
Experiment for
Molecule Alteration
Western blot analysis; Fluorescence microscopy assay
Experiment for
Drug Resistance
MTS assay; Crystal violet staining assay; Fluorescence-activated cell sorting (FACS) assay; Flow cytometry
Mechanism Description Celecoxib reverses the glioblastoma chemo-resistance to temozolomide through mitochondrial metabolism.
Cisplatin
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Drug Inactivation by Structure Modification (DISM) Click to Show/Hide
Key Molecule: Glutathione S-transferase P (GSTP1) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Cisplatin
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-204 [10]
Resistant Disease Glioma [ICD-11: 2A00.1]
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 proliferation Activation hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
SNB19 cells Brain Homo sapiens (Human) CVCL_0535
U373 cells Brain Homo sapiens (Human) CVCL_2219
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Colony formation assays
Mechanism Description Knockdown of LncRNA HOXD-AS1 suppresses proliferation, migration and invasion and enhances cisplatin sensitivity of glioma cells by sponging miR-20.
Key Molecule: HOXD antisense growth-associated long non-coding RNA (HAGLR) [10]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-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 proliferation Activation hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
SNB19 cells Brain Homo sapiens (Human) CVCL_0535
U373 cells Brain Homo sapiens (Human) CVCL_2219
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Colony formation assays
Mechanism Description Knockdown of LncRNA HOXD-AS1 suppresses proliferation, migration and invasion and enhances cisplatin sensitivity of glioma cells by sponging miR-20.
Key Molecule: hsa-miR-214-3p [11]
Resistant Disease Pediatric intracranial nongerminomatous malignant germ cell tumors [ICD-11: 2A00.07]
Molecule Alteration Expression
Up-regulation
Resistant Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT Assay
Mechanism Description miR214-3p overexpression enhanced cisplatin resistance by downregulating the expression of its target, the apoptotic protein BCL2-like 11 (BCL2L11/BIM).
Key Molecule: hsa-mir-520f [12]
Resistant Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Expression
Down-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
In Vitro Model Sk-N-AS cells Adrenal Homo sapiens (Human) CVCL_1700
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Acid phosphatase assay
Mechanism Description Significant overexpression of NAIP mRNA and protein was documented, while experimental modulation of NAIP levels in both Sk-N-AsCis24 and in parental Sk-N-AS cells confirmed that NAIP was responsible for the drug resistant phenotype by apoptosis inhibition. Furthermore, a decrease in the NAIP targeting microRNA, miR-520f, was also demonstrated to be partially responsible for increased NAIP levels in Sk-N-AsCis24. Interestingly, miR-520f levels were determined to be significantly lower in postchemotherapy treatment tumours relative to matched prechemotherapy samples, consistent with a role for this miRNA in the acquisition of drug resistance in vivo, potentially through decreased NAIP targeting.
Key Molecule: hsa-let-7f-1 [13]
Resistant Disease Medulloblastoma [ICD-11: 2A00.10]
Molecule Alteration Expression
Down-regulation
Resistant Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model D425 cells Brain Homo sapiens (Human) CVCL_1275
UW228 cells Brain Homo sapiens (Human) CVCL_8585
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTS assay; TUNEL assay
Mechanism Description High-Mobility Group Box 1 (HMGB1) is a direct target of miR-let-7f-1. HMGB1 is a highly conserved nuclear protein that functions as a chromatin-binding factor that bends DNA and promotes access to transcriptional protein assemblies on specific DNA targets. Overexpression of HMGB1 in cells treated with pSP and cisplatin blocked SPARC-induced cisplatin resistance indicating that overexpression of miR-let-7f-1 and a reduction in HMGB1 protein levels result in cellular resistance to cisplatin in SPARC over expressed cells. Earlier studies demonstrated that HMGB1 functions as a regulator of the balance between autophagy and apoptosis.
Key Molecule: hsa-let-7b [14]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell cycle Inhibition hsa04110
Cell viability Activation hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Cisplatin treatment leads to Let-7b suppression, which in turn up-regulates cyclin D1 expression, resulting in resistance to cisplatin.
Key Molecule: hsa-mir-21 [15]
Resistant Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Expression
Up-regulation
Resistant Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SH-SY5Y cells Abdomen Homo sapiens (Human) CVCL_0019
BE(2) -M17 cells Brain Homo sapiens (Human) CVCL_0167
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Increased miR-21 expression might suppress the PTEN expression and eventually induce chemoresistance to cisplatin and increase cell proliferation.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Cisplatin
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
Key Molecule: Multidrug resistance protein 1 (ABCB1) [6]
Resistant Disease Anaplastic astrocytoma [ICD-11: 2A00.04]
Molecule Alteration Expression
Up-regulation
Resistant Drug Cisplatin
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
Oncotech EDR assay
Mechanism Description Cisplatin and etoposide are both substrates for membrane-bound efflux pumps, such as MRP and MDR1, which prevent their entry into the extracellular space of the central nervous system. The low levels of in vitro drug resistance noted for cisplatin and etoposide may be explained in part by the absence of such a barrier in our in vitro assay system.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Bcl-2-like protein 11 (BCL2L11) [11]
Resistant Disease Pediatric intracranial nongerminomatous malignant germ cell tumors [ICD-11: 2A00.07]
Molecule Alteration Expression
Down-regulation
Resistant Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model 293T cells Breast Homo sapiens (Human) CVCL_0063
Experiment for
Molecule Alteration
Immunoblotting assay; Immunohistochemistry
Experiment for
Drug Resistance
MTT Assay
Mechanism Description miR214-3p overexpression enhanced cisplatin resistance by downregulating the expression of its target, the apoptotic protein BCL2-like 11 (BCL2L11/BIM).
Key Molecule: Baculoviral IAP repeat-containing protein 1 (BIRC1) [12]
Resistant Disease Neuroblastoma [ICD-11: 2A00.11]
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
In Vitro Model Sk-N-AS cells Adrenal Homo sapiens (Human) CVCL_1700
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Acid phosphatase assay
Mechanism Description Significant overexpression of NAIP mRNA and protein was documented, while experimental modulation of NAIP levels in both Sk-N-AsCis24 and in parental Sk-N-AS cells confirmed that NAIP was responsible for the drug resistant phenotype by apoptosis inhibition. Furthermore, a decrease in the NAIP targeting microRNA, miR-520f, was also demonstrated to be partially responsible for increased NAIP levels in Sk-N-AsCis24. Interestingly, miR-520f levels were determined to be significantly lower in postchemotherapy treatment tumours relative to matched prechemotherapy samples, consistent with a role for this miRNA in the acquisition of drug resistance in vivo, potentially through decreased NAIP targeting.
Key Molecule: High mobility group protein B1 (HMGB1) [13]
Resistant Disease Medulloblastoma [ICD-11: 2A00.10]
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 proliferation Activation hsa05200
In Vitro Model D425 cells Brain Homo sapiens (Human) CVCL_1275
UW228 cells Brain Homo sapiens (Human) CVCL_8585
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay; TUNEL assay
Mechanism Description High-Mobility Group Box 1 (HMGB1) is a direct target of miR-let-7f-1. HMGB1 is a highly conserved nuclear protein that functions as a chromatin-binding factor that bends DNA and promotes access to transcriptional protein assemblies on specific DNA targets. Overexpression of HMGB1 in cells treated with pSP and cisplatin blocked SPARC-induced cisplatin resistance indicating that overexpression of miR-let-7f-1 and a reduction in HMGB1 protein levels result in cellular resistance to cisplatin in SPARC over expressed cells. Earlier studies demonstrated that HMGB1 functions as a regulator of the balance between autophagy and apoptosis.
Key Molecule: G1/S-specific cyclin-D1 (CCND1) [14]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
Cell viability Activation hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
Experiment for
Molecule Alteration
Luciferase reporter assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description Cisplatin treatment leads to Let-7b suppression, which in turn up-regulates cyclin D1 expression, resulting in resistance to cisplatin.
Key Molecule: Phosphatase and tensin homolog (PTEN) [15]
Resistant Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Expression
Down-regulation
Resistant Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SH-SY5Y cells Abdomen Homo sapiens (Human) CVCL_0019
BE(2) -M17 cells Brain Homo sapiens (Human) CVCL_0167
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Increased miR-21 expression might suppress the PTEN expression and eventually induce chemoresistance to cisplatin and increase cell proliferation.
Key Molecule: Methylated-DNA--protein-cysteine methyltransferase (MGMT) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Cisplatin
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-204 [10]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
SNB19 cells Brain Homo sapiens (Human) CVCL_0535
U373 cells Brain Homo sapiens (Human) CVCL_2219
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Colony formation assays
Mechanism Description miR-204 overexpression suppressed proliferation, migration and invasion and enhanced the DDP sensitivity in glioma cells.
Key Molecule: Maternally expressed 3 (MEG3) [16]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell autophagy Inhibition hsa04140
In Vitro Model U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Long non coding RNA MEG3 contributes to cisplatin induced apoptosis via inhibition of autophagy in human glioma cells.
Key Molecule: Long non-protein coding RNA (AC023115.3) [17]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation miR26a/GSk3Beta/Mcl1 signaling pathway Regulation hsa05206
In Vitro Model U251-MG cells Brain Homo sapiens (Human) CVCL_0021
U87MG cells Brain Homo sapiens (Human) CVCL_GP63
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Annexin V-FITC staining assay; Flow cytometry assay
Mechanism Description AC023115.3 sensitized glioma cell to cisplatin-induced apoptosis through regulation of the miR26a-GSk3beta-Mcl1 signalling. AC023115.3 acted as a miR26a sponge and inhibited its activity, thus increased the expression of GSk3beta.
Key Molecule: hsa-mir-26a [17]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell autophagy Inhibition hsa04140
miR26a/GSk3Beta/Mcl1 signaling pathway Regulation hsa05206
In Vitro Model U251-MG cells Brain Homo sapiens (Human) CVCL_0021
U87MG cells Brain Homo sapiens (Human) CVCL_GP63
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Annexin V-FITC staining assay; Flow cytometry assay
Mechanism Description Long non-coding RNA AC023115.3 suppresses chemoresistance of glioblastoma by reducing autophagy. AC023115.3 acts as a competing endogenous RNA for miR26a and attenuates the inhibitory effect of miR26a on GSk3beta, leading to an increase in GSk3beta and a decrease in autophagy.
Key Molecule: hsa-mir-186 [18]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Inhibition hsa05200
In Vitro Model LN229 cells Brain Homo sapiens (Human) CVCL_0393
U87MG cells Brain Homo sapiens (Human) CVCL_GP63
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR 186 reverses cisplatin resistance and inhibits the formation of the GIC phenotype by degrading YY1 in glioblastoma.
Key Molecule: hsa-miR-501-3p [19]
Sensitive Disease Glioma [ICD-11: 2A00.1]
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 viability Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-501-3p sensitizes glioma cells to cisplatin via reducing protein levels of MYCN.
Key Molecule: hsa-mir-141 [20]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model IMR-32 cells Abdomen Homo sapiens (Human) CVCL_0346
Sk-N-AS cells Adrenal Homo sapiens (Human) CVCL_1700
SH-SY5Y cells Abdomen Homo sapiens (Human) CVCL_0019
In Vivo Model Mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description In the IMR-32 and SH-SY5Y cells, lentivirus-induced miR-141 upregulation inhibited cancer proliferation, cell cycle progression, migration and increased cisplatin chemosensitivity in vitro. In addition, miR-141 upregulation reduced the in vivo growth of IMR-32 tumor explants. FUS was found to be inversely regulated by miR-141 in NB. Small interfering RNA (siRNA)-induced FUS downregulation had similar tumor-suppressive effects as miR-141 upregulation on NB cell proliferation, cell cycle progression, migration and cisplatin chemosensitivity.
Key Molecule: hsa-mir-873 [21]
Sensitive Disease Glioma [ICD-11: 2A00.1]
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 proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Bcl-2 was a direct target of miR 873, and miR 873 decreased the level of the Bcl-2 protein in cisplatin-resistant glioma cells. Notably, re-expression of Bcl-2 attenuated the function of miR 873 in cisplatin-resistant glioma cells and the sensitivity of the cells to cisplatin.
Key Molecule: hsa-mir-30d [22]
Sensitive Disease Glioma [ICD-11: 2A00.1]
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 proliferation Inhibition hsa05200
In Vitro Model T98G cells Brain Homo sapiens (Human) CVCL_0556
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR; qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The effect of miR-30d on cisplatin sensitivity is mediated through the beclin 1-regulated autophagy.
Key Molecule: hsa-mir-204 [23]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Cisplatin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model Kelly cells Adrenal Homo sapiens (Human) CVCL_2092
Sk-N-AS cells Adrenal Homo sapiens (Human) CVCL_1700
SH-SY5Y cells Abdomen Homo sapiens (Human) CVCL_0019
In Vivo Model Orthotopic xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-204 direct targeting of the 3' UTR of BCL2 and NTRk2 (TrkB). BCL2 has a critical role in ensuring the survival of early developing cell types, NTRk2 is also a well-established pro-survival oncogene in neuroblastoma, signalling the activation of the PI3k/AkT pathway, a significant mechanism of drug resistance in neuroblastoma. Ectopic miR-204 expression significantly increased sensitivity to cisplatin and etoposide in vitro.
Key Molecule: hsa-mir-34 [24]
Sensitive Disease Medulloblastoma [ICD-11: 2A00.10]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Cisplatin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
p53 signaling pathway Activation hsa04115
In Vitro Model UW228 cells Brain Homo sapiens (Human) CVCL_8585
R262 cells Bone marrow Homo sapiens (Human) CVCL_VU83
R300 cells Bone marrow Homo sapiens (Human) CVCL_VU84
UW426 cells Bone marrow Homo sapiens (Human) CVCL_DH82
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description The repression of MAGE-A by miR-34a results in increased expression of p53 thus lead to resistance.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Glycogen synthase kinase-3 beta (GSK3B) [17]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell autophagy Inhibition hsa04140
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
miR26a/GSk3Beta/Mcl1 signaling pathway Regulation hsa05206
In Vitro Model U251-MG cells Brain Homo sapiens (Human) CVCL_0021
U87MG cells Brain Homo sapiens (Human) CVCL_GP63
Experiment for
Molecule Alteration
Dual luciferase reporter assay; Western blot analysis
Experiment for
Drug Resistance
MTT assay; Annexin V-FITC staining assay; Flow cytometry assay
Mechanism Description Long non-coding RNA AC023115.3 suppresses chemoresistance of glioblastoma by reducing autophagy. AC023115.3 acts as a competing endogenous RNA for miR26a and attenuates the inhibitory effect of miR26a on GSk3beta, leading to an increase in GSk3beta and a decrease in autophagy.
Key Molecule: Transcriptional repressor protein YY1 (TYY1) [18]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Inhibition hsa05200
In Vitro Model LN229 cells Brain Homo sapiens (Human) CVCL_0393
U87MG cells Brain Homo sapiens (Human) CVCL_GP63
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR 186 reverses cisplatin resistance and inhibits the formation of the GIC phenotype by degrading YY1 in glioblastoma.
Key Molecule: N-myc proto-oncogene protein (MYCN) [19]
Sensitive Disease Glioma [ICD-11: 2A00.1]
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 viability Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-501-3p sensitizes glioma cells to cisplatin via reducing protein levels of MYCN.
Key Molecule: RNA-binding protein FUS (FUS) [20]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Cisplatin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model IMR-32 cells Abdomen Homo sapiens (Human) CVCL_0346
Sk-N-AS cells Adrenal Homo sapiens (Human) CVCL_1700
SH-SY5Y cells Abdomen Homo sapiens (Human) CVCL_0019
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description In the IMR-32 and SH-SY5Y cells, lentivirus-induced miR-141 upregulation inhibited cancer proliferation, cell cycle progression, migration and increased cisplatin chemosensitivity in vitro. In addition, miR-141 upregulation reduced the in vivo growth of IMR-32 tumor explants. FUS was found to be inversely regulated by miR-141 in NB. Small interfering RNA (siRNA)-induced FUS downregulation had similar tumor-suppressive effects as miR-141 upregulation on NB cell proliferation, cell cycle progression, migration and cisplatin chemosensitivity.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [21]
Sensitive Disease Glioma [ICD-11: 2A00.1]
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 proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Bcl-2 was a direct target of miR 873, and miR 873 decreased the level of the Bcl-2 protein in cisplatin-resistant glioma cells. Notably, re-expression of Bcl-2 attenuated the function of miR 873 in cisplatin-resistant glioma cells and the sensitivity of the cells to cisplatin.
Key Molecule: Beclin-1 (BECN1) [22]
Sensitive Disease Glioma [ICD-11: 2A00.1]
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 proliferation Inhibition hsa05200
In Vitro Model T98G cells Brain Homo sapiens (Human) CVCL_0556
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description The effect of miR-30d on cisplatin sensitivity is mediated through the beclin 1-regulated autophagy.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [23]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Cisplatin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model Kelly cells Adrenal Homo sapiens (Human) CVCL_2092
Sk-N-AS cells Adrenal Homo sapiens (Human) CVCL_1700
SH-SY5Y cells Abdomen Homo sapiens (Human) CVCL_0019
In Vivo Model Orthotopic xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-204 direct targeting of the 3' UTR of BCL2 and NTRk2 (TrkB). BCL2 has a critical role in ensuring the survival of early developing cell types, NTRk2 is also a well-established pro-survival oncogene in neuroblastoma, signalling the activation of the PI3k/AkT pathway, a significant mechanism of drug resistance in neuroblastoma. Ectopic miR-204 expression significantly increased sensitivity to cisplatin and etoposide in vitro.
Key Molecule: BDNF/NT-3 growth factors receptor (NTRK2) [23]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Cisplatin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model Kelly cells Adrenal Homo sapiens (Human) CVCL_2092
Sk-N-AS cells Adrenal Homo sapiens (Human) CVCL_1700
SH-SY5Y cells Abdomen Homo sapiens (Human) CVCL_0019
In Vivo Model Orthotopic xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-204 direct targeting of the 3' UTR of BCL2 and NTRk2 (TrkB). BCL2 has a critical role in ensuring the survival of early developing cell types, NTRk2 is also a well-established pro-survival oncogene in neuroblastoma, signalling the activation of the PI3k/AkT pathway, a significant mechanism of drug resistance in neuroblastoma. Ectopic miR-204 expression significantly increased sensitivity to cisplatin and etoposide in vitro.
Key Molecule: Melanoma-associated antigen 12 (MAGEA12) [24]
Sensitive Disease Medulloblastoma [ICD-11: 2A00.10]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Cisplatin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
p53 signaling pathway Activation hsa04115
In Vitro Model UW228 cells Brain Homo sapiens (Human) CVCL_8585
R262 cells Bone marrow Homo sapiens (Human) CVCL_VU83
R300 cells Bone marrow Homo sapiens (Human) CVCL_VU84
UW426 cells Bone marrow Homo sapiens (Human) CVCL_DH82
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description The repression of MAGE-A by miR-34a results in increased expression of p53 thus lead to resistance.
Key Molecule: Melanoma-associated antigen 2 (MAGEA2) [24]
Sensitive Disease Medulloblastoma [ICD-11: 2A00.10]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Cisplatin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
p53 signaling pathway Activation hsa04115
In Vitro Model UW228 cells Brain Homo sapiens (Human) CVCL_8585
R262 cells Bone marrow Homo sapiens (Human) CVCL_VU83
R300 cells Bone marrow Homo sapiens (Human) CVCL_VU84
UW426 cells Bone marrow Homo sapiens (Human) CVCL_DH82
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description The repression of MAGE-A by miR-34a results in increased expression of p53 thus lead to resistance.
Key Molecule: Melanoma-associated antigen 3 (MAGEA3) [24]
Sensitive Disease Medulloblastoma [ICD-11: 2A00.10]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Cisplatin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
p53 signaling pathway Activation hsa04115
In Vitro Model UW228 cells Brain Homo sapiens (Human) CVCL_8585
R262 cells Bone marrow Homo sapiens (Human) CVCL_VU83
R300 cells Bone marrow Homo sapiens (Human) CVCL_VU84
UW426 cells Bone marrow Homo sapiens (Human) CVCL_DH82
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description The repression of MAGE-A by miR-34a results in increased expression of p53 thus lead to resistance.
Key Molecule: Melanoma-associated antigen 6 (MAGEA6) [24]
Sensitive Disease Medulloblastoma [ICD-11: 2A00.10]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Cisplatin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
p53 signaling pathway Activation hsa04115
In Vitro Model UW228 cells Brain Homo sapiens (Human) CVCL_8585
R262 cells Bone marrow Homo sapiens (Human) CVCL_VU83
R300 cells Bone marrow Homo sapiens (Human) CVCL_VU84
UW426 cells Bone marrow Homo sapiens (Human) CVCL_DH82
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description The repression of MAGE-A by miR-34a results in increased expression of p53 thus lead to resistance.
Crizotinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: ALK tyrosine kinase receptor (ALK) [1]
Resistant Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Missense mutation
p.F1174L
Resistant Drug Crizotinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model NBLW cells Brain Homo sapiens (Human) CVCL_VJ90
NBLW-R cells Brain Homo sapiens (Human) CVCL_VJ91
Experiment for
Molecule Alteration
Sangersequencing assay; Targeted deep sequencing assay
Experiment for
Drug Resistance
Array CGH assay
Mechanism Description Analysis of the sensitivity of NBLW and NBLW-R cells to a panel of ALk inhibitors (TAE-684, Crizotinib, Alectinib and Lorlatinib) revealed differences between the paired cell lines, and overall NBLW-R cells with the F1174L mutation were more resistant to ALk inhibitor induced apoptosis compared with NBLW cells.
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: ALK tyrosine kinase receptor (ALK) [25]
Resistant Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Missense mutation
p.F1174L
Resistant Drug Crizotinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation ALK signaling pathway Activation hsa05200
Cell apoptosis Inhibition hsa04210
Cell invasion Activation hsa05200
In Vitro Model NCI-H3122 cells Lung Homo sapiens (Human) CVCL_5160
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Direct sequencing assay
Experiment for
Drug Resistance
MTS assay
Mechanism Description There is a C to G mutation (asterix) in codon 3522 in exon 23 resulting in the F1174L mutation. When present in cis with an ALk translocation, this mutation (also detected in neuroblastomas) causes an increase in ALk phosphorylation, cell growth and downstream signaling. Furthermore, the F1174L mutation inhibits crizotinib mediated downregulation of ALk signaling and blocks apoptosis in RANBP2-ALk Ba/F3 cells.
Cyclophosphamide
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-129 [26]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Cyclophosphamide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell cycle Inhibition hsa04110
In Vitro Model IMR-32 cells Abdomen Homo sapiens (Human) CVCL_0346
BE-M17 cells Adrenal Homo sapiens (Human) N.A.
Kelly cells Adrenal Homo sapiens (Human) CVCL_2092
NB-1643 cells Adrenal Homo sapiens (Human) CVCL_5627
NB1 cells Adrenal Homo sapiens (Human) CVCL_1440
NBSD cells Adrenal Homo sapiens (Human) CVCL_LF68
Neuro-2a cells Adrenal Homo sapiens (Human) CVCL_0470
Sk-N-AS cells Adrenal Homo sapiens (Human) CVCL_1700
Sk-N-SH cells Adrenal Homo sapiens (Human) CVCL_0531
Sk-SY-5Y cells Adrenal Homo sapiens (Human) N.A.
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-129 suppressed cell growth and potentiated chemosensitivity by inhibiting MYO10.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Unconventional myosin-X (MYO10) [26]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Cyclophosphamide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell colony Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model IMR-32 cells Abdomen Homo sapiens (Human) CVCL_0346
BE-M17 cells Adrenal Homo sapiens (Human) N.A.
Kelly cells Adrenal Homo sapiens (Human) CVCL_2092
NB-1643 cells Adrenal Homo sapiens (Human) CVCL_5627
NB1 cells Adrenal Homo sapiens (Human) CVCL_1440
NBSD cells Adrenal Homo sapiens (Human) CVCL_LF68
Neuro-2a cells Adrenal Homo sapiens (Human) CVCL_0470
Sk-N-AS cells Adrenal Homo sapiens (Human) CVCL_1700
Sk-N-SH cells Adrenal Homo sapiens (Human) CVCL_0531
Sk-SY-5Y cells Adrenal Homo sapiens (Human) N.A.
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; RIP assay; Luciferase reporter assay
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-129 suppressed cell growth and potentiated chemosensitivity by inhibiting MYO10.
Dabrafenib/Trametinib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [27]
Sensitive Disease FGFR-tacc positive glioblastoma [ICD-11: 2A00.01]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Dabrafenib/Trametinib
Experimental Note Identified from the Human Clinical Data
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [28]
Sensitive Disease FGFR-tacc positive glioblastoma [ICD-11: 2A00.01]
Molecule Alteration Missense mutation
p.V600D (c.1799_1800delTGinsAC)
Sensitive Drug Dabrafenib/Trametinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation BRAF/MEK/MAPK signaling pathway Inhibition hsa04010
In Vitro Model Brain .
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [27]
Sensitive Disease Pleomorphic xanthoastrocytoma [ICD-11: 2A00.0Y]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Dabrafenib/Trametinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation ERK signaling pathway Inhibition hsa04210
In Vitro Model Brain .
Dacarbazine
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Drug Inactivation by Structure Modification (DISM) Click to Show/Hide
Key Molecule: Glutathione S-transferase P (GSTP1) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Dacarbazine
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Dacarbazine
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Methylated-DNA--protein-cysteine methyltransferase (MGMT) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Dacarbazine
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
Docetaxel
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Drug Inactivation by Structure Modification (DISM) Click to Show/Hide
Key Molecule: Glutathione S-transferase P (GSTP1) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Docetaxel
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [29]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Resistant Drug Docetaxel
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model U87-MG cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Athymic nu/nu female mice xenograft model Mus musculus
Experiment for
Drug Resistance
MTS assay
Mechanism Description In a cell line expressing a high level of P-glycoprotein, the IC50 of TTI-237 increased 25-fold whereas those of paclitaxel and vincristine increased 806-fold and 925-fold.
Key Molecule: Multidrug resistance protein 1 (ABCB1) [6]
Resistant Disease Anaplastic astrocytoma [ICD-11: 2A00.04]
Molecule Alteration Expression
Up-regulation
Resistant Drug Docetaxel
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Protein kinase C signaling pathways Inhibition hsa04310
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
Oncotech EDR assay
Mechanism Description On the other hand, the frequency of LDR that we noted for paclitaxel (20%) and vincristine (20%) was similar to the clinical response rates for these compounds. These data suggest that although MDR1 expression by glial tumors may not be the dominant direct cellular process responsible for tumor resistance to natural products, other mechanisms are present that diminish their activity. The clinical mechanisms of natural product resistance may be a multifactorial function of endothelial expression of MDR1 at the blood-brain barrier in conjunction with glial tumor cell expression of alternative efflux pumps, such as MRP, altered tubulin with lower affinity binding sites, and/or protein kinase C signaling pathways that suppress apoptosis.
Key Molecule: Multidrug resistance protein 1 (ABCB1) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Docetaxel
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Methylated-DNA--protein-cysteine methyltransferase (MGMT) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Docetaxel
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
Doxorubicin
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: piR-hsa-39980 [30]
Resistant Disease Neuroblastoma [ICD-11: 2A00.11]
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 metastasis Activation hsa05205
Cell proliferation Activation hsa05200
In Vitro Model IMR-32 cells Abdomen Homo sapiens (Human) CVCL_0346
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description piR-39980 is an oncogenic piRNA overexpressed in NB cells which induces the cancer cell growth, enhance metastasis, and inhibit the cellular senescence by targeting JAk3 as well as desensitizes the chemotherapeutic drug. And piR-39980 was found to desensitize the effect of doxorubicin and inhibit drug-induced apoptosis.
Key Molecule: hsa-mir-125b [31]
Resistant Disease Primitive neuroectodermal tumor [ICD-11: 2A00.08]
Molecule Alteration Expression
Up-regulation
Resistant Drug Doxorubicin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
miR125b-p53/BAKT signaling pathway Activation hsa05206
In Vitro Model RD-ES cells Bones Homo sapiens (Human) CVCL_2169
Sk-ES cells Bones Homo sapiens (Human) CVCL_0627
Sk-N-MC cells Bones Homo sapiens (Human) CVCL_0530
TC-71 cells Bones Homo sapiens (Human) CVCL_2213
VH-64 cells Bones Homo sapiens (Human) CVCL_9672
WE-68 cells Bones Homo sapiens (Human) CVCL_9717
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Celltiter-glo luminescent cell viability assay
Mechanism Description miR-125b led to the development of chemoresistance by suppressing the expression of p53 and Bak, and repression of miR-125b sensitized EWS cells to apoptosis induced by treatment with various cytotoxic drugs.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Tyrosine-protein kinase JAK3 (JAK3) [30]
Resistant Disease Neuroblastoma [ICD-11: 2A00.11]
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 metastasis Activation hsa05205
Cell proliferation Activation hsa05200
In Vitro Model IMR-32 cells Abdomen Homo sapiens (Human) CVCL_0346
Experiment for
Molecule Alteration
Dual-luciferase reporter assay
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description piR-39980 is an oncogenic piRNA overexpressed in NB cells which induces the cancer cell growth, enhance metastasis, and inhibit the cellular senescence by targeting JAk3 as well as desensitizes the chemotherapeutic drug. And piR-39980 was found to desensitize the effect of doxorubicin and inhibit drug-induced apoptosis.
Key Molecule: Bcl-2 homologous antagonist/killer (BAK1) [31]
Resistant Disease Primitive neuroectodermal tumor [ICD-11: 2A00.08]
Molecule Alteration Expression
Down-regulation
Resistant Drug Doxorubicin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
miR125b-p53/BAKT signaling pathway Activation hsa05206
In Vitro Model RD-ES cells Bones Homo sapiens (Human) CVCL_2169
Sk-ES cells Bones Homo sapiens (Human) CVCL_0627
Sk-N-MC cells Bones Homo sapiens (Human) CVCL_0530
TC-71 cells Bones Homo sapiens (Human) CVCL_2213
VH-64 cells Bones Homo sapiens (Human) CVCL_9672
WE-68 cells Bones Homo sapiens (Human) CVCL_9717
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Celltiter-glo luminescent cell viability assay
Mechanism Description miR-125b led to the development of chemoresistance by suppressing the expression of p53 and Bak, and repression of miR-125b sensitized EWS cells to apoptosis induced by treatment with various cytotoxic drugs.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-127 [32]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Doxorubicin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Inhibition hsa04151
Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87-MG cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay; Flow cytometry assay
Mechanism Description microRNA-127 silencing significantly affects cell growth and increases the sensitivity to adriamycin. microRNA-127 silencing arrests the cell cycle, potentiates adriamycin-induced apoptosis, and increases cellular Rh-123 uptake. microRNA-127 silencing down-regulates MDR1, MRP1, Runx2, Bcl-2, Survivin and ErbB4 expression while up-regulates p53 expression. microRNA-127 silencing inhibits AkT phosphorylation.
Key Molecule: hsa-mir-21 [33]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Doxorubicin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model A172 cells Brain Homo sapiens (Human) CVCL_0131
T98G cells Brain Homo sapiens (Human) CVCL_0556
U87MG cells Brain Homo sapiens (Human) CVCL_GP63
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; TUNEL assay
Mechanism Description To validate the possible association of miR-21 with drug resistance of T98G cells, we transfected anti-miR-21 inhibitor into the cells. The expression level of miR-21 was significantly lower in T98G transfected cells (than in the parental control cells). Transfected cells showed a high apoptotic rate compared to control after Dox treatment by TUNEL assay, suggesting that combined Dox and miR-21 inhibitor therapy can sensitize GBM resistant cells to anthracyclines by enhancing apoptosis.
Key Molecule: hsa-mir-137 [34]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Doxorubicin
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 UkF-NB3 cells Bone marrow Homo sapiens (Human) CVCL_9904
In Vivo Model Immunodeficient NCr nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Cell titer glo assay assay
Mechanism Description Hypermethylation of the miR-137 promoter and negative regulation of miR-137 by CAR contribute in part to reduced miR-137 expression and increased CAR and MDR1 expression in doxorubicin-resistant neuroblastoma cells.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [32]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Doxorubicin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Inhibition hsa04151
Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87-MG cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay; Flow cytometry assay
Mechanism Description microRNA-127 silencing significantly affects cell growth and increases the sensitivity to adriamycin. microRNA-127 silencing arrests the cell cycle, potentiates adriamycin-induced apoptosis, and increases cellular Rh-123 uptake. microRNA-127 silencing down-regulates MDR1, MRP1, Runx2, Bcl-2, Survivin and ErbB4 expression while up-regulates p53 expression. microRNA-127 silencing inhibits AkT phosphorylation.
Key Molecule: Multidrug resistance-associated protein 1 (MRP1) [32]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Doxorubicin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Inhibition hsa04151
Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87-MG cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay; Flow cytometry assay
Mechanism Description microRNA-127 silencing significantly affects cell growth and increases the sensitivity to adriamycin. microRNA-127 silencing arrests the cell cycle, potentiates adriamycin-induced apoptosis, and increases cellular Rh-123 uptake. microRNA-127 silencing down-regulates MDR1, MRP1, Runx2, Bcl-2, Survivin and ErbB4 expression while up-regulates p53 expression. microRNA-127 silencing inhibits AkT phosphorylation.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [32]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Doxorubicin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Inhibition hsa04151
Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87-MG cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay; Flow cytometry assay
Mechanism Description microRNA-127 silencing significantly affects cell growth and increases the sensitivity to adriamycin. microRNA-127 silencing arrests the cell cycle, potentiates adriamycin-induced apoptosis, and increases cellular Rh-123 uptake. microRNA-127 silencing down-regulates MDR1, MRP1, Runx2, Bcl-2, Survivin and ErbB4 expression while up-regulates p53 expression. microRNA-127 silencing inhibits AkT phosphorylation.
Key Molecule: Receptor tyrosine-protein kinase erbB-4 (ERBB4) [32]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Doxorubicin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Inhibition hsa04151
Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87-MG cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay; Flow cytometry assay
Mechanism Description microRNA-127 silencing significantly affects cell growth and increases the sensitivity to adriamycin. microRNA-127 silencing arrests the cell cycle, potentiates adriamycin-induced apoptosis, and increases cellular Rh-123 uptake. microRNA-127 silencing down-regulates MDR1, MRP1, Runx2, Bcl-2, Survivin and ErbB4 expression while up-regulates p53 expression. microRNA-127 silencing inhibits AkT phosphorylation.
Key Molecule: Cellular tumor antigen p53 (TP53) [32]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Doxorubicin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Inhibition hsa04151
Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87-MG cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay; Flow cytometry assay
Mechanism Description microRNA-127 silencing significantly affects cell growth and increases the sensitivity to adriamycin. microRNA-127 silencing arrests the cell cycle, potentiates adriamycin-induced apoptosis, and increases cellular Rh-123 uptake. microRNA-127 silencing down-regulates MDR1, MRP1, Runx2, Bcl-2, Survivin and ErbB4 expression while up-regulates p53 expression. microRNA-127 silencing inhibits AkT phosphorylation.
Key Molecule: Runt-related transcription factor 2 (RUNX2) [32]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Doxorubicin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Inhibition hsa04151
Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87-MG cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay; Flow cytometry assay
Mechanism Description microRNA-127 silencing significantly affects cell growth and increases the sensitivity to adriamycin. microRNA-127 silencing arrests the cell cycle, potentiates adriamycin-induced apoptosis, and increases cellular Rh-123 uptake. microRNA-127 silencing down-regulates MDR1, MRP1, Runx2, Bcl-2, Survivin and ErbB4 expression while up-regulates p53 expression. microRNA-127 silencing inhibits AkT phosphorylation.
Key Molecule: Baculoviral IAP repeat-containing protein 5 (BIRC5) [32]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Doxorubicin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Inhibition hsa04151
Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87-MG cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay; Flow cytometry assay
Mechanism Description microRNA-127 silencing significantly affects cell growth and increases the sensitivity to adriamycin. microRNA-127 silencing arrests the cell cycle, potentiates adriamycin-induced apoptosis, and increases cellular Rh-123 uptake. microRNA-127 silencing down-regulates MDR1, MRP1, Runx2, Bcl-2, Survivin and ErbB4 expression while up-regulates p53 expression. microRNA-127 silencing inhibits AkT phosphorylation.
Key Molecule: Nuclear receptor subfamily 1 group I3 (NR1I3) [34]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Doxorubicin
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 UkF-NB3 cells Bone marrow Homo sapiens (Human) CVCL_9904
In Vivo Model Immunodeficient NCr nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Chromatin immunoprecipitation assay
Experiment for
Drug Resistance
Cell titer glo assay assay
Mechanism Description Hypermethylation of the miR-137 promoter and negative regulation of miR-137 by CAR contribute in part to reduced miR-137 expression and increased CAR and MDR1 expression in doxorubicin-resistant neuroblastoma cells.
Key Molecule: Forkhead box protein M1 (FOXM1) [35]
Sensitive Disease Rhabdoid tumor [ICD-11: 2A00.0Y]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Doxorubicin
Experimental Note Identified from the Human Clinical Data
In Vitro Model Tm87-16 cells Kidney Homo sapiens (Human) CVCL_8001
TTC549 cells Liver Homo sapiens (Human) CVCL_8005
STM91-01 cells Lung Homo sapiens (Human) CVCL_8000
Experiment for
Molecule Alteration
qRT-PCR; Western blotting assay
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description FOXM1 siRNA and FOXM1 inhibitor (thiostrepton) successfully downregulated the mRNA and protein expression of FOXM1 in vitro and the downregulation of FOXM1 inhibited cell proliferation, drug resistance to doxorubicin, migration, invasion, and caused the cell cycle arrest and apoptosis of MRT cell lines.
Entrectinib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: ALK tyrosine kinase receptor (ALK) [36]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Missense mutation
p.F1245V (c.3733T>G)
Sensitive Drug Entrectinib
Experimental Note Identified from the Human Clinical Data
Etoposide
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-125b [31]
Resistant Disease Primitive neuroectodermal tumor [ICD-11: 2A00.08]
Molecule Alteration Expression
Up-regulation
Resistant Drug Etoposide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
miR125b-p53/BAKT signaling pathway Activation hsa05206
In Vitro Model RD-ES cells Bones Homo sapiens (Human) CVCL_2169
Sk-ES cells Bones Homo sapiens (Human) CVCL_0627
Sk-N-MC cells Bones Homo sapiens (Human) CVCL_0530
TC-71 cells Bones Homo sapiens (Human) CVCL_2213
VH-64 cells Bones Homo sapiens (Human) CVCL_9672
WE-68 cells Bones Homo sapiens (Human) CVCL_9717
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Celltiter-glo luminescent cell viability assay
Mechanism Description miR-125b led to the development of chemoresistance by suppressing the expression of p53 and Bak, and repression of miR-125b sensitized EWS cells to apoptosis induced by treatment with various cytotoxic drugs.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [37]
Resistant Disease Ependymoma [ICD-11: 2A00.05]
Molecule Alteration Expression
Up-regulation
Resistant Drug Etoposide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell invasion Activation hsa05200
In Vitro Model BXD-1425EPN cells Embryo Homo sapiens (Human) CVCL_Y105
EPN1 cells Embryo Homo sapiens (Human) N.A.
EPN7 cells Embryo Homo sapiens (Human) N.A.
EPN7R cells Embryo Homo sapiens (Human) N.A.
DKFZ-EP1 cells Embryo Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description ABCB1 gene expression was observed in 4 out of 5 paediatric ependymoma cell lines and increased in stem cell enriched neurospheres. Functional inhibition of ABCB1 using vardenafil or verapamil significantly (p < 0.05-0.001) potentiated the response to three chemotherapeutic drugs (vincristine, etoposide and methotrexate). Both inhibitors were also able to significantly reduce migration (p < 0.001) and invasion (p < 0.001).
Key Molecule: Multidrug resistance protein 1 (ABCB1) [6]
Resistant Disease Anaplastic astrocytoma [ICD-11: 2A00.04]
Molecule Alteration Expression
Up-regulation
Resistant Drug Etoposide
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
Oncotech EDR assay
Mechanism Description Cisplatin and etoposide are both substrates for membrane-bound efflux pumps, such as MRP and MDR1, which prevent their entry into the extracellular space of the central nervous system. The low levels of in vitro drug resistance noted for cisplatin and etoposide may be explained in part by the absence of such a barrier in our in vitro assay system.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Bcl-2 homologous antagonist/killer (BAK1) [31]
Resistant Disease Primitive neuroectodermal tumor [ICD-11: 2A00.08]
Molecule Alteration Expression
Down-regulation
Resistant Drug Etoposide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
miR125b-p53/BAKT signaling pathway Activation hsa05206
In Vitro Model RD-ES cells Bones Homo sapiens (Human) CVCL_2169
Sk-ES cells Bones Homo sapiens (Human) CVCL_0627
Sk-N-MC cells Bones Homo sapiens (Human) CVCL_0530
TC-71 cells Bones Homo sapiens (Human) CVCL_2213
VH-64 cells Bones Homo sapiens (Human) CVCL_9672
WE-68 cells Bones Homo sapiens (Human) CVCL_9717
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Celltiter-glo luminescent cell viability assay
Mechanism Description miR-125b led to the development of chemoresistance by suppressing the expression of p53 and Bak, and repression of miR-125b sensitized EWS cells to apoptosis induced by treatment with various cytotoxic drugs.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-296-3p [38]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Etoposide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U251AR cells Brain Homo sapiens (Human) CVCL_1G29
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description EAG1 channel might be involved in cell-cycle progression of tumour cells because a significant reduction in the proliferation of tumour cell lines could be achieved by inhibiting EAG1 expression using antisense oligonucleotides. Ectopic expression of miR-296-3p reduced EAG1 expression and suppressed cell proliferation drug resistance.
Key Molecule: hsa-mir-204 [23]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Etoposide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model Kelly cells Adrenal Homo sapiens (Human) CVCL_2092
Sk-N-AS cells Adrenal Homo sapiens (Human) CVCL_1700
SH-SY5Y cells Abdomen Homo sapiens (Human) CVCL_0019
In Vivo Model Orthotopic xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-204 direct targeting of the 3' UTR of BCL2 and NTRk2 (TrkB). BCL2 has a critical role in ensuring the survival of early developing cell types, NTRk2 is also a well-established pro-survival oncogene in neuroblastoma, signalling the activation of the PI3k/AkT pathway, a significant mechanism of drug resistance in neuroblastoma. Ectopic miR-204 expression significantly increased sensitivity to cisplatin and etoposide in vitro.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [37]
Sensitive Disease Ependymoma [ICD-11: 2A00.05]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Etoposide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell invasion Activation hsa05200
In Vitro Model BXD-1425EPN cells Embryo Homo sapiens (Human) CVCL_Y105
EPN1 cells Embryo Homo sapiens (Human) N.A.
EPN7 cells Embryo Homo sapiens (Human) N.A.
EPN7R cells Embryo Homo sapiens (Human) N.A.
DKFZ-EP1 cells Embryo Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description ABCB1 gene expression was observed in 4 out of 5 paediatric ependymoma cell lines and increased in stem cell enriched neurospheres. Functional inhibition of ABCB1 using vardenafil or verapamil significantly (p < 0.05-0.001) potentiated the response to three chemotherapeutic drugs (vincristine, etoposide and methotrexate). Both inhibitors were also able to significantly reduce migration (p < 0.001) and invasion (p < 0.001).
Key Molecule: Multidrug resistance protein 1 (ABCB1) [37]
Sensitive Disease Ependymoma [ICD-11: 2A00.05]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Etoposide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell invasion Activation hsa05200
In Vitro Model BXD-1425EPN cells Embryo Homo sapiens (Human) CVCL_Y105
EPN1 cells Embryo Homo sapiens (Human) N.A.
EPN7 cells Embryo Homo sapiens (Human) N.A.
EPN7R cells Embryo Homo sapiens (Human) N.A.
DKFZ-EP1 cells Embryo Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description ABCB1 gene expression was observed in 4 out of 5 paediatric ependymoma cell lines and increased in stem cell enriched neurospheres. Functional inhibition of ABCB1 using vardenafil or verapamil significantly (p < 0.05-0.001) potentiated the response to three chemotherapeutic drugs (vincristine, etoposide and methotrexate). Both inhibitors were also able to significantly reduce migration (p < 0.001) and invasion (p < 0.001).
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Potassium voltage-gated channel subfamily H member 1 (KCNH1) [38]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Etoposide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U251AR cells Brain Homo sapiens (Human) CVCL_1G29
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description EAG1 channel might be involved in cell-cycle progression of tumour cells because a significant reduction in the proliferation of tumour cell lines could be achieved by inhibiting EAG1 expression using antisense oligonucleotides. Ectopic expression of miR-296-3p reduced EAG1 expression and suppressed cell proliferation drug resistance.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [23]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Etoposide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model Kelly cells Adrenal Homo sapiens (Human) CVCL_2092
Sk-N-AS cells Adrenal Homo sapiens (Human) CVCL_1700
SH-SY5Y cells Abdomen Homo sapiens (Human) CVCL_0019
In Vivo Model Orthotopic xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-204 direct targeting of the 3' UTR of BCL2 and NTRk2 (TrkB). BCL2 has a critical role in ensuring the survival of early developing cell types, NTRk2 is also a well-established pro-survival oncogene in neuroblastoma, signalling the activation of the PI3k/AkT pathway, a significant mechanism of drug resistance in neuroblastoma. Ectopic miR-204 expression significantly increased sensitivity to cisplatin and etoposide in vitro.
Key Molecule: BDNF/NT-3 growth factors receptor (NTRK2) [23]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Etoposide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model Kelly cells Adrenal Homo sapiens (Human) CVCL_2092
Sk-N-AS cells Adrenal Homo sapiens (Human) CVCL_1700
SH-SY5Y cells Abdomen Homo sapiens (Human) CVCL_0019
In Vivo Model Orthotopic xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-204 direct targeting of the 3' UTR of BCL2 and NTRk2 (TrkB). BCL2 has a critical role in ensuring the survival of early developing cell types, NTRk2 is also a well-established pro-survival oncogene in neuroblastoma, signalling the activation of the PI3k/AkT pathway, a significant mechanism of drug resistance in neuroblastoma. Ectopic miR-204 expression significantly increased sensitivity to cisplatin and etoposide in vitro.
Imatinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: hsa-mir-203 [39]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Imatinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell migration Activation hsa04670
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description SNAI2 is a direct target of miR-203 and that miR-203-mediated inhibition of SNAI2 is dependent on a conversed motif in the 3'-UTR of SNAI2. Recent independent studies have shown that overexpression of SNAI2 alters cell invasion, motility, chemoresistance, metastasis and poor prognosis in several human cancers. As a member of the snail family of transcription factors, SNAI2 can repress E-cadherin transcription and induce EMT directly. Therefore, SNAI2 overexpression due to reduction of miR-203 may result in EMT and chemoresistance in GBM via these pathways. Additionally, miR-203 may relieve E-cadherin from transcriptional repression by targeting SNAI2 signaling. Nevertheless, because one single miRNA might have multiple targets, judicious considerations are essential for identi cation of the main functional targets.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Zinc finger protein SNAI2 (SNAI2) [39]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Imatinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell migration Activation hsa04670
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description SNAI2 is a direct target of miR-203 and that miR-203-mediated inhibition of SNAI2 is dependent on a conversed motif in the 3'-UTR of SNAI2. Recent independent studies have shown that overexpression of SNAI2 alters cell invasion, motility, chemoresistance, metastasis and poor prognosis in several human cancers. As a member of the snail family of transcription factors, SNAI2 can repress E-cadherin transcription and induce EMT directly. Therefore, SNAI2 overexpression due to reduction of miR-203 may result in EMT and chemoresistance in GBM via these pathways. Additionally, miR-203 may relieve E-cadherin from transcriptional repression by targeting SNAI2 signaling. Nevertheless, because one single miRNA might have multiple targets, judicious considerations are essential for identi cation of the main functional targets.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-296-3p [38]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Imatinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U251AR cells Brain Homo sapiens (Human) CVCL_1G29
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description EAG1 channel might be involved in cell-cycle progression of tumour cells because a significant reduction in the proliferation of tumour cell lines could be achieved by inhibiting EAG1 expression using antisense oligonucleotides. Ectopic expression of miR-296-3p reduced EAG1 expression and suppressed cell proliferation drug resistance.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Potassium voltage-gated channel subfamily H member 1 (KCNH1) [38]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Imatinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U251AR cells Brain Homo sapiens (Human) CVCL_1G29
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description EAG1 channel might be involved in cell-cycle progression of tumour cells because a significant reduction in the proliferation of tumour cell lines could be achieved by inhibiting EAG1 expression using antisense oligonucleotides. Ectopic expression of miR-296-3p reduced EAG1 expression and suppressed cell proliferation drug resistance.
Irinotecan
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Drug Inactivation by Structure Modification (DISM) Click to Show/Hide
Key Molecule: Glutathione S-transferase P (GSTP1) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Irinotecan
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Irinotecan
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Methylated-DNA--protein-cysteine methyltransferase (MGMT) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Irinotecan
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
Lorlatinib
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: ALK tyrosine kinase receptor (ALK) [1]
Resistant Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Missense mutation
p.F1174L
Resistant Drug Lorlatinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model NBLW cells Brain Homo sapiens (Human) CVCL_VJ90
NBLW-R cells Brain Homo sapiens (Human) CVCL_VJ91
Experiment for
Molecule Alteration
Sangersequencing assay; Targeted deep sequencing assay
Experiment for
Drug Resistance
Array CGH assay
Mechanism Description Analysis of the sensitivity of NBLW and NBLW-R cells to a panel of ALk inhibitors (TAE-684, Crizotinib, Alectinib and Lorlatinib) revealed differences between the paired cell lines, and overall NBLW-R cells with the F1174L mutation were more resistant to ALk inhibitor induced apoptosis compared with NBLW cells.
Maraviroc
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: C-C motif chemokine receptor 5 (CCR5) [40]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Function
Inhibition
Sensitive Drug Maraviroc
Experimental Note Discovered Using In-vivo Testing Model
Cell Pathway Regulation CCL5-CCR5 signaling pathway Inhibition has05163
In Vivo Model Intracranial GBM patient-derived xenograft model Mus musculus
Experiment for
Molecule Alteration
Neutral comet assay; ELISA assay; Immunofluorescence staining analysis; Immunohistochemistry staining analysi; Immunoblot assay
Experiment for
Drug Resistance
CCK-8 assay
Mechanism Description The authors uncovered that pericytes potentiate DNA damage repair (DDR) in GBM cells residing in the perivascular niche, which induces temozolomide (TMZ) chemoresistance. Disrupting CCL5-CCR5 paracrine signaling through the brain-penetrable CCR5 antagonist maraviroc (MVC) potently inhibits pericyte-promoted DDR and effectively improves the chemotherapeutic efficacy of TMZ.
Matrine
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa_circ_104075 [41]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Resistant Drug Matrine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell growth Activation hsa05200
Wnt/Beta-catenin/PI3K/AKT signaling pathway Inhibition hsa04310
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Matrine inhibits Bcl-9 expression through down-regulating circRNA-104075 expression in U251 cells.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: B-cell CLL/lymphoma 9 protein (BCL9) [41]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Resistant Drug Matrine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell autophagy Inhibition hsa04140
Wnt/Beta-catenin/PI3K/AKT signaling pathway Inhibition hsa04310
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Matrine inhibits Bcl-9 expression through down-regulating circRNA-104075 expression in U251 cells.
Meclizine
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase mTOR (mTOR) [42]
Sensitive Disease Glioblastoma multiforme [ICD-11: 2A00.03]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Meclizine
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SH-1-V4 cells Esophagus Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Mechanism Description GBM stem cells (GBMSC) resist the standard-of-care therapy, temozolomide, and are considered a major contributor to tumor resistance. GBMSCs are resistant to the standard-of-care temozolomide therapy, but temozolomide supplemented with tight-binding piperazine meclizine and flunarizine greatly enhanced GBMSC death over temozolomide alone.
Mestranol
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: L1 cell adhesion molecule (L1CAM) [2]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Mestranol
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Activation hsa04670
In Vitro Model MDCK cells Kidney Canis lupus familiaris (Dog) (Canis familiaris) CVCL_0422
Experiment for
Molecule Alteration
Puromycin selection and monitored regularly for the maintenance of L1 silencing assay
Experiment for
Drug Resistance
Migration assay
Mechanism Description With OVCAR3 cells treated with anagrelide, 2-hydroxy-5-fluoropyrimidine and mestranol , the gap width closure was seen from 48 h onward at all concentrations tested. Similar results were obtained with U251 cells, and L1's metastatic potential is further evidenced by its promotion of epithelial-mesenchymal transition, endothelial cell transcytosis and resistance to chemo- and radiotherapy.
Methotrexate
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [37]
Resistant Disease Ependymoma [ICD-11: 2A00.05]
Molecule Alteration Expression
Up-regulation
Resistant Drug Methotrexate
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell invasion Activation hsa05200
In Vitro Model BXD-1425EPN cells Embryo Homo sapiens (Human) CVCL_Y105
EPN1 cells Embryo Homo sapiens (Human) N.A.
EPN7 cells Embryo Homo sapiens (Human) N.A.
EPN7R cells Embryo Homo sapiens (Human) N.A.
DKFZ-EP1 cells Embryo Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description ABCB1 gene expression was observed in 4 out of 5 paediatric ependymoma cell lines and increased in stem cell enriched neurospheres. Functional inhibition of ABCB1 using vardenafil or verapamil significantly (p < 0.05-0.001) potentiated the response to three chemotherapeutic drugs (vincristine, etoposide and methotrexate). Both inhibitors were also able to significantly reduce migration (p < 0.001) and invasion (p < 0.001).
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [37]
Sensitive Disease Ependymoma [ICD-11: 2A00.05]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Methotrexate
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell invasion Activation hsa05200
In Vitro Model BXD-1425EPN cells Embryo Homo sapiens (Human) CVCL_Y105
EPN1 cells Embryo Homo sapiens (Human) N.A.
EPN7 cells Embryo Homo sapiens (Human) N.A.
EPN7R cells Embryo Homo sapiens (Human) N.A.
DKFZ-EP1 cells Embryo Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description ABCB1 gene expression was observed in 4 out of 5 paediatric ependymoma cell lines and increased in stem cell enriched neurospheres. Functional inhibition of ABCB1 using vardenafil or verapamil significantly (p < 0.05-0.001) potentiated the response to three chemotherapeutic drugs (vincristine, etoposide and methotrexate). Both inhibitors were also able to significantly reduce migration (p < 0.001) and invasion (p < 0.001).
Key Molecule: Multidrug resistance protein 1 (ABCB1) [37]
Sensitive Disease Ependymoma [ICD-11: 2A00.05]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Methotrexate
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell invasion Activation hsa05200
In Vitro Model BXD-1425EPN cells Embryo Homo sapiens (Human) CVCL_Y105
EPN1 cells Embryo Homo sapiens (Human) N.A.
EPN7 cells Embryo Homo sapiens (Human) N.A.
EPN7R cells Embryo Homo sapiens (Human) N.A.
DKFZ-EP1 cells Embryo Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description ABCB1 gene expression was observed in 4 out of 5 paediatric ependymoma cell lines and increased in stem cell enriched neurospheres. Functional inhibition of ABCB1 using vardenafil or verapamil significantly (p < 0.05-0.001) potentiated the response to three chemotherapeutic drugs (vincristine, etoposide and methotrexate). Both inhibitors were also able to significantly reduce migration (p < 0.001) and invasion (p < 0.001).
Mitomycin
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-34 [24]
Sensitive Disease Medulloblastoma [ICD-11: 2A00.10]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Mitomycin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
p53 signaling pathway Activation hsa04115
In Vitro Model UW228 cells Brain Homo sapiens (Human) CVCL_8585
R262 cells Bone marrow Homo sapiens (Human) CVCL_VU83
R300 cells Bone marrow Homo sapiens (Human) CVCL_VU84
UW426 cells Bone marrow Homo sapiens (Human) CVCL_DH82
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description The repression of MAGE-A by miR-34a results in increased expression of p53 thus lead to resistance.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Melanoma-associated antigen 12 (MAGEA12) [24]
Sensitive Disease Medulloblastoma [ICD-11: 2A00.10]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Mitomycin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
p53 signaling pathway Activation hsa04115
In Vitro Model UW228 cells Brain Homo sapiens (Human) CVCL_8585
R262 cells Bone marrow Homo sapiens (Human) CVCL_VU83
R300 cells Bone marrow Homo sapiens (Human) CVCL_VU84
UW426 cells Bone marrow Homo sapiens (Human) CVCL_DH82
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description The repression of MAGE-A by miR-34a results in increased expression of p53 thus lead to resistance.
Key Molecule: Melanoma-associated antigen 2 (MAGEA2) [24]
Sensitive Disease Medulloblastoma [ICD-11: 2A00.10]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Mitomycin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
p53 signaling pathway Activation hsa04115
In Vitro Model UW228 cells Brain Homo sapiens (Human) CVCL_8585
R262 cells Bone marrow Homo sapiens (Human) CVCL_VU83
R300 cells Bone marrow Homo sapiens (Human) CVCL_VU84
UW426 cells Bone marrow Homo sapiens (Human) CVCL_DH82
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description The repression of MAGE-A by miR-34a results in increased expression of p53 thus lead to resistance.
Key Molecule: Melanoma-associated antigen 3 (MAGEA3) [24]
Sensitive Disease Medulloblastoma [ICD-11: 2A00.10]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Mitomycin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
p53 signaling pathway Activation hsa04115
In Vitro Model UW228 cells Brain Homo sapiens (Human) CVCL_8585
R262 cells Bone marrow Homo sapiens (Human) CVCL_VU83
R300 cells Bone marrow Homo sapiens (Human) CVCL_VU84
UW426 cells Bone marrow Homo sapiens (Human) CVCL_DH82
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description The repression of MAGE-A by miR-34a results in increased expression of p53 thus lead to resistance.
Key Molecule: Melanoma-associated antigen 6 (MAGEA6) [24]
Sensitive Disease Medulloblastoma [ICD-11: 2A00.10]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Mitomycin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
p53 signaling pathway Activation hsa04115
In Vitro Model UW228 cells Brain Homo sapiens (Human) CVCL_8585
R262 cells Bone marrow Homo sapiens (Human) CVCL_VU83
R300 cells Bone marrow Homo sapiens (Human) CVCL_VU84
UW426 cells Bone marrow Homo sapiens (Human) CVCL_DH82
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description The repression of MAGE-A by miR-34a results in increased expression of p53 thus lead to resistance.
Nivolumab
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Mismatch repair endonuclease PMS2 (PMS2) [43]
Sensitive Disease FGFR-tacc positive glioblastoma [ICD-11: 2A00.01]
Molecule Alteration FS-deletion
p.K706fs*19 (c.2118delG)
Sensitive Drug Nivolumab
Experimental Note Identified from the Human Clinical Data
Paclitaxel
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-34 [44]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Paclitaxel
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation miR34a/PD-L1 signaling pathway Regulation hsa05206
In Vitro Model U87 cells Brain Homo sapiens (Human) CVCL_0022
U87-P cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Annexin V/PI apoptosis assay; Cell cycle assay; MTT assay
Mechanism Description miR34a attenuates glioma cells progression and chemoresistance via targeting PD-L1.
Key Molecule: hsa-mir-21 [45]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
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 migration Inhibition hsa04670
EGFR/STAT3 signaling pathway Inhibition hsa01521
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description The miR-21 inhibitor could enhance the chemo-sensitivity of human glioblastoma cells to taxol. A combination of miR-21 inhibitor and taxol could be an effective therapeutic strategy for controlling the growth of GBM by inhibiting STAT3 expression and phosphorylation.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Programmed cell death 1 ligand 1 (PD-L1) [44]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Paclitaxel
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation miR34a/PD-L1 signaling pathway Regulation hsa05206
In Vitro Model U87 cells Brain Homo sapiens (Human) CVCL_0022
U87-P cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis; Luciferase reporter assay
Experiment for
Drug Resistance
Annexin V/PI apoptosis assay; Cell cycle assay; MTT assay
Mechanism Description miR34a attenuates glioma cells progression and chemoresistance via targeting PD-L1.
Panobinostat
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Quinolinate phosphoribosyltransferase (QPRT) [46]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Resistant Drug Panobinostat
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model ES-2 cells Ovary Homo sapiens (Human) CVCL_3509
MG-63 cells Bone Homo sapiens (Human) CVCL_0426
MMQ cells Pituitary gland Rattus norvegicus (Rat) CVCL_2117
MOLM-13 cells Peripheral blood Homo sapiens (Human) CVCL_2119
MOLM-14 cells Peripheral blood Homo sapiens (Human) CVCL_7916
SH-1-V8 cells Esophagus Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis; RNA-sequencing analysis
Experiment for
Drug Resistance
Flow cytometry
Mechanism Description RNA-sequencing identifies quinolinic acid phosphoribosyltransferase (QPRT) as a highly expressed gene in bortezomib-panobinostat resistant U87 cells. QPRT, an enzyme catalyzing the rate-determining conversion of quinolinic acid (QA) to nicotinic acid mononucleotide (NAMN) a precursor for de novo NAD+ biosynthesis from tryptophan.
Perphenazine
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [47]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Perphenazine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
Cell invasion Inhibition hsa05200
In Vitro Model SHI-1 cells Bone marrow Homo sapiens (Human) CVCL_2191
Experiment for
Molecule Alteration
Western blotting analysis
Mechanism Description The present study explored the effects of perphenazine and prochlorperazine on the levels of ABCB1, ABCG2, E-cadherin, alpha-tubulin and integrins (alpha3, alpha5, and beta1), as well as on the migratory and invasive ability of U87-MG cells. The results suggested that perphenazine and prochlorperazine may modulate the expression levels of multidrug resistance proteins (they decreased ABCB1 and increased ABCG2 expression), E-cadherin, alpha-tubulin and integrins, and could impair the migration and invasion of U-87 MG cells.
Key Molecule: ATP-binding cassette sub-family G2 (ABCG2) [47]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Perphenazine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
Cell invasion Inhibition hsa05200
In Vitro Model SHI-1 cells Bone marrow Homo sapiens (Human) CVCL_2191
Experiment for
Molecule Alteration
Western blotting analysis
Mechanism Description The present study explored the effects of perphenazine and prochlorperazine on the levels of ABCB1, ABCG2, E-cadherin, alpha-tubulin and integrins (alpha3, alpha5, and beta1), as well as on the migratory and invasive ability of U87-MG cells. The results suggested that perphenazine and prochlorperazine may modulate the expression levels of multidrug resistance proteins (they decreased ABCB1 and increased ABCG2 expression), E-cadherin, alpha-tubulin and integrins, and could impair the migration and invasion of U-87 MG cells.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Integrin alpha-3 (ITA3) [47]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Perphenazine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
Cell invasion Inhibition hsa05200
In Vitro Model SHI-1 cells Bone marrow Homo sapiens (Human) CVCL_2191
Experiment for
Molecule Alteration
Western blotting analysis
Mechanism Description The present study explored the effects of perphenazine and prochlorperazine on the levels of ABCB1, ABCG2, E-cadherin, alpha-tubulin and integrins (alpha3, alpha5, and beta1), as well as on the migratory and invasive ability of U87-MG cells. The results suggested that perphenazine and prochlorperazine may modulate the expression levels of multidrug resistance proteins (they decreased ABCB1 and increased ABCG2 expression), E-cadherin, alpha-tubulin and integrins, and could impair the migration and invasion of U-87 MG cells.
Key Molecule: Integrin beta-1 (ITGB1) [47]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Perphenazine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
Cell invasion Inhibition hsa05200
In Vitro Model SHI-1 cells Bone marrow Homo sapiens (Human) CVCL_2191
Experiment for
Molecule Alteration
Western blotting analysis
Mechanism Description The present study explored the effects of perphenazine and prochlorperazine on the levels of ABCB1, ABCG2, E-cadherin, alpha-tubulin and integrins (alpha3, alpha5, and beta1), as well as on the migratory and invasive ability of U87-MG cells. The results suggested that perphenazine and prochlorperazine may modulate the expression levels of multidrug resistance proteins (they decreased ABCB1 and increased ABCG2 expression), E-cadherin, alpha-tubulin and integrins, and could impair the migration and invasion of U-87 MG cells.
Picrotoxin
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Gamma-aminobutyric acid receptor subunit alpha-1 (GABAARs) [48]
Resistant Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Function
Inhibition
Resistant Drug Picrotoxin
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SH-1-V7 cells Esophagus Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Whole-cell patch-clamp recording assay
Experiment for
Drug Resistance
Slice whole-cell recording
Mechanism Description Receptors containing Delta T6'Y had marginally weaker sensitivity to positive allosteric modulators, likely a secondary consequence of differing GABA sensitivity. Overexpression of DeltaT6'Y in neurons resulted in robust PTX-insensitive IPSCs, suggesting that Delta-containing receptors are readily recruited by synaptically released GABA.
Prochlorperazine
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [47]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Prochlorperazine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model SHI-1 cells Bone marrow Homo sapiens (Human) CVCL_2191
Experiment for
Molecule Alteration
Western blotting analysis; RNA-sequencing analysis
Experiment for
Drug Resistance
Wound healing assay;Transwell assay
Mechanism Description Prochlorperazine may modulate the expression levels of multidrug resistance proteins (they decreased ABCB1 and increased ABCG2 expression), E-cadherin, alpha-tubulin and integrins, and could impair the migration and invasion of U-87 MG cells.
Key Molecule: ATP-binding cassette sub-family G2 (ABCG2) [47]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Prochlorperazine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model SHI-1 cells Bone marrow Homo sapiens (Human) CVCL_2191
Experiment for
Molecule Alteration
Western blotting analysis; RNA-sequencing analysis
Experiment for
Drug Resistance
Wound healing assay;Transwell assay
Mechanism Description Prochlorperazine may modulate the expression levels of multidrug resistance proteins (they decreased ABCB1 and increased ABCG2 expression), E-cadherin, alpha-tubulin and integrins, and could impair the migration and invasion of U-87 MG cells.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Integrin alpha-3 (ITA3) [47]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Prochlorperazine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model SHI-1 cells Bone marrow Homo sapiens (Human) CVCL_2191
Experiment for
Molecule Alteration
Western blotting analysis; RNA-sequencing analysis
Experiment for
Drug Resistance
Wound healing assay;Transwell assay
Mechanism Description Prochlorperazine may modulate the expression levels of multidrug resistance proteins (they decreased ABCB1 and increased ABCG2 expression), E-cadherin, alpha-tubulin and integrins, and could impair the migration and invasion of U-87 MG cells.
Key Molecule: Integrin beta-1 (ITGB1) [47]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Prochlorperazine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model SHI-1 cells Bone marrow Homo sapiens (Human) CVCL_2191
Experiment for
Molecule Alteration
Western blotting analysis; RNA-sequencing analysis
Experiment for
Drug Resistance
Wound healing assay;Transwell assay
Mechanism Description Prochlorperazine may modulate the expression levels of multidrug resistance proteins (they decreased ABCB1 and increased ABCG2 expression), E-cadherin, alpha-tubulin and integrins, and could impair the migration and invasion of U-87 MG cells.
Rabeprazole
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: Vimentin (VIM) [49]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Rabeprazole
Experimental Note Discovered Using In-vivo Testing Model
Cell Pathway Regulation AKT/GSK3beta signaling pathway Inhibition hsa04931
NF-KappaB signaling pathway Inhibition hsa04064
In Vitro Model MDA-231 cells Pleural effusion Homo sapiens (Human) CVCL_0062
MJ cells Peripheral blood Homo sapiens (Human) CVCL_1414
MMQ cells Pituitary gland Rattus norvegicus (Rat) CVCL_2117
MOLM-13 cells Peripheral blood Homo sapiens (Human) CVCL_2119
In Vivo Model Male Wistar rats-Stereotaxic glioma model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis; Gene expression analysis
Experiment for
Drug Resistance
MTT assay; Scratch wound healing migration assay; Transwell invasion assay
Mechanism Description Epithelial to mesenchymal transition (EMT) is pivotal in embryonic development and wound healing, whereas in cancer it inflicts malignancy and drug resistance. Rabeprazole has efficacy per se and reduces resistance to temozolomide in glioma via EMT inhibition. Rabeprazole suppressed EMT by impeding AKT/GSK3beta phosphorylation and/or NF-kappaB signaling and sensitized temozolomide resistance.
Key Molecule: Dishevelled binding antagonist of beta catenin 1 (DACT1) [49]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Rabeprazole
Experimental Note Discovered Using In-vivo Testing Model
Cell Pathway Regulation AKT/GSK3beta signaling pathway Inhibition hsa04931
NF-KappaB signaling pathway Inhibition hsa04064
In Vitro Model MDA-231 cells Pleural effusion Homo sapiens (Human) CVCL_0062
MJ cells Peripheral blood Homo sapiens (Human) CVCL_1414
MMQ cells Pituitary gland Rattus norvegicus (Rat) CVCL_2117
MOLM-13 cells Peripheral blood Homo sapiens (Human) CVCL_2119
In Vivo Model Male Wistar rats-Stereotaxic glioma model Rattus norvegicus
Experiment for
Molecule Alteration
Western blotting analysis; Gene expression analysis
Experiment for
Drug Resistance
MTT assay; Scratch wound healing migration assay; Transwell invasion assay
Mechanism Description Epithelial to mesenchymal transition (EMT) is pivotal in embryonic development and wound healing, whereas in cancer it inflicts malignancy and drug resistance. Rabeprazole has efficacy per se and reduces resistance to temozolomide in glioma via EMT inhibition. Rabeprazole suppressed EMT by impeding AKT/GSK3beta phosphorylation and/or NF-kappaB signaling and sensitized temozolomide resistance.
Key Molecule: Glial fibrillary acidic protein (GFAP) [49]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Rabeprazole
Experimental Note Discovered Using In-vivo Testing Model
Cell Pathway Regulation AKT/GSK3beta signaling pathway Inhibition hsa04931
NF-KappaB signaling pathway Inhibition hsa04064
In Vitro Model MDA-231 cells Pleural effusion Homo sapiens (Human) CVCL_0062
MJ cells Peripheral blood Homo sapiens (Human) CVCL_1414
MMQ cells Pituitary gland Rattus norvegicus (Rat) CVCL_2117
MOLM-13 cells Peripheral blood Homo sapiens (Human) CVCL_2119
In Vivo Model Male Wistar rats-Stereotaxic glioma model Rattus norvegicus
Experiment for
Molecule Alteration
Western blotting analysis; Gene expression analysis
Experiment for
Drug Resistance
MTT assay; Scratch wound healing migration assay; Transwell invasion assay
Mechanism Description Epithelial to mesenchymal transition (EMT) is pivotal in embryonic development and wound healing, whereas in cancer it inflicts malignancy and drug resistance. Rabeprazole has efficacy per se and reduces resistance to temozolomide in glioma via EMT inhibition. Rabeprazole suppressed EMT by impeding AKT/GSK3beta phosphorylation and/or NF-kappaB signaling and sensitized temozolomide resistance.
Talazoparib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: Oxalosuccinate decarboxylase (IDH1) [50]
Sensitive Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration Missense mutation
p.R132C (c.394C>T)
Sensitive Drug Talazoparib
Experimental Note Identified from the Human Clinical Data
In Vitro Model IDH2 cells N.A. Homo sapiens (Human) N.A.
IDH1 cells N.A. Homo sapiens (Human) N.A.
In Vivo Model Female athymic nu/nu mouse PDX model Mus musculus
Experiment for
Drug Resistance
Promega assay
Mechanism Description The oncometabolite, 2-hydroxyglutarate, renders IDH1/2 mutant cancer cells deficient in homologous recombination and confers vulnerability to synthetic lethal targeting with PARP inhibitors.
Key Molecule: Oxalosuccinate decarboxylase (IDH1) [50]
Sensitive Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration Missense mutation
p.R132H (c.395G>A)
Sensitive Drug Talazoparib
Experimental Note Identified from the Human Clinical Data
In Vitro Model IDH2 cells N.A. Homo sapiens (Human) N.A.
IDH1 cells N.A. Homo sapiens (Human) N.A.
In Vivo Model Female athymic nu/nu mouse PDX model Mus musculus
Experiment for
Drug Resistance
Promega assay
Mechanism Description The oncometabolite, 2-hydroxyglutarate, renders IDH1/2 mutant cancer cells deficient in homologous recombination and confers vulnerability to synthetic lethal targeting with PARP inhibitors.
Temozolomide
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Drug Inactivation by Structure Modification (DISM) Click to Show/Hide
Key Molecule: Glutathione S-transferase P (GSTP1) [6]
Resistant Disease Anaplastic astrocytoma [ICD-11: 2A00.04]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
Oncotech EDR assay
Mechanism Description GSTP1 is the first major mechanism of resistance alkylator agents encounter after entering the cancer cell cytoplasm. GSTP1 acts to enzymatically conjugate glutathione to the reactive metabolites of BCNU. The mechanisms by which GSTP1 may be up-regulated in gliomas are under investigation. Constitutive expression is thought to be influenced by the proximal promoter factor Sp1, whereas increased expression levels may result from stabilization of GSTP1 mRNA. GSTP1 expression has been reported to be induced by drug exposure, indicating that it may play a role in acquired drug resistance.
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-20b-3p [51]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
c-Met signaling signaling pathway Inhibition hsa01521
In Vitro Model HG7 cells Brain Homo sapiens (Human) N.A.
LN229 cells Brain Homo sapiens (Human) CVCL_0393
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay; Flow cytometry assay
Mechanism Description Lnc-TALC promotes O6-methylguanine-DNA methyltransferase expression via regulating the c-Met pathway by competitively binding with miR-20b-3p.
Key Molecule: LncRNA regulator of Akt signaling associated with HCC and RCC (LNCARSR) [51]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
c-Met signaling signaling pathway Inhibition hsa01521
In Vitro Model HG7 cells Brain Homo sapiens (Human) N.A.
LN229 cells Brain Homo sapiens (Human) CVCL_0393
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay; Flow cytometry assay
Mechanism Description Lnc-TALC promotes O6-methylguanine-DNA methyltransferase expression via regulating the c-Met pathway by competitively binding with miR-20b-3p.
Key Molecule: hsa-miR-20b-3p [51]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
c-Met signaling signaling pathway Inhibition hsa01521
In Vitro Model HG7 cells Brain Homo sapiens (Human) N.A.
LN229 cells Brain Homo sapiens (Human) CVCL_0393
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay; Flow cytometry assay
Mechanism Description Lnc-TALC promotes O6-methylguanine-DNA methyltransferase expression via regulating the c-Met pathway by competitively binding with miR-20b-3p.
Key Molecule: Cancer susceptibility 2 (CASC2) [52], [53]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation DNA damage repair signaling pathway Activation hsa03410
In Vitro Model U87 cells Brain Homo sapiens (Human) CVCL_0022
U257 cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
Flow cytometry assay; MTT assay; Transwell assay
Mechanism Description Upregulation of CASC2 sensitized glioma to temozolomide cytotoxicity through autophagy inhibition by sponging miR193a-5p and regulating mTOR expression. CASC2 is downregulated in gliomas, resulting in increased miR193a-5p level and a decrease in mTOR expression, which further induces protective autophagy, leading to TMZ resistance.
Key Molecule: Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) [54], [55]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation miR203-TS signaling pathway Regulation hsa05206
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometric analysis
Mechanism Description LncRNA MALAT1 inhibition re-sensitized TMZ resistant cells through up-regulating miR203 and down-regulating TS expression. MALAT1 decreased the sensitivity of resistant glioma cell lines to TMZ by upregulating ZEB1.
Key Molecule: hsa-mir-132 [56]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model U87MG cells Brain Homo sapiens (Human) CVCL_GP63
U87MG-res cells Brain Homo sapiens (Human) CVCL_GP63
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Soft agar assay; MTT assay; Sphere formation assay
Mechanism Description microRNA-132 induces temozolomide resistance and promotes the formation of cancer stem cell phenotypes by targeting tumor suppressor candidate 3 in glioblastoma.
Key Molecule: hsa-mir-29c [57]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation DNA damage repair signaling pathway Activation hsa03410
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
U373 cells Brain Homo sapiens (Human) CVCL_2219
U118 cells Brain Homo sapiens (Human) CVCL_0633
NHA Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qRT-PCR; RIP assay; Dual luciferase reporter assay
Experiment for
Drug Resistance
MTT assay; BrdU incorporation assay
Mechanism Description XIST can amplify the chemoresistance of glioma cell lines to TMZ through directly targetting miR29c via SP1 and MGMT. XIST expression was up-regulated by miR29c inhibition while down-regulated by ectopic miR29, and XIST directly binds to miR29c to inhibit its expression, XIST and miR29c neatively regulates each other.
Key Molecule: X inactive specific transcript (XIST) [57]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation DNA damage repair signaling pathway Activation hsa03410
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
U373 cells Brain Homo sapiens (Human) CVCL_2219
U118 cells Brain Homo sapiens (Human) CVCL_0633
NHA Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay; BrdU incorporation assay
Mechanism Description XIST was inversely correlated with miR29c, positively correlated with PS1, positively related with MGMT. XIST can inhibit miR29c expression by directly binding to miR29c and subsequently up-regulate the expression of SP1 and MGMT to promote the chemoresistance of glioma cells to TMZ.
Key Molecule: hsa-mir-223 [58]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Transwell assay; Transwell matrix penetration assay; MTT assay; BrdU incorporation assay
Mechanism Description miR223/PAX6 axis regulates glioblastoma stem cell proliferation and the chemo resistance to TMZ via inhibition of PI3k/Akt pathway.
Key Molecule: hsa-mir-181a [52]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell growth Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
SNB19 cells Brain Homo sapiens (Human) CVCL_0535
U373 cells Brain Homo sapiens (Human) CVCL_2219
U118 cells Brain Homo sapiens (Human) CVCL_0633
NHA cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; BrdU incorporation assay
Mechanism Description LncRNA CASC2 interacts with miR181a to modulate glioma growth and resistance to TMZ through PTEN pathway.
Key Molecule: hsa-mir-497 [59]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation IGF1R/IRS1 signaling pathway Activation hsa04212
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
U138 cells Brain Homo sapiens (Human) CVCL_0020
HEK293 cells Kidney Homo sapiens (Human) CVCL_0045
NHA cells Brain Homo sapiens (Human) N.A.
LN382 cells Brain Homo sapiens (Human) CVCL_3956
SF295 cells Brain Homo sapiens (Human) CVCL_1690
SHG-44 cells Brain Homo sapiens (Human) CVCL_6728
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Up-regulation of miR-497 confers resistance to temozolomide in human glioma cells by targeting mTOR/Bcl-2. The silencing of miR-497 decreased the protein levels of IGF1R/IRS1 pathway-related proteins, that is, IGF1R, IRS1, mTOR, and Bcl-2.
Key Molecule: hsa-mir-195 [60]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model U251-MG cells Brain Homo sapiens (Human) CVCL_0021
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Hsa-miR-195 could negatively regulate the expression of CCNE1 in glioma and microRNA-195 reverses the resistance to temozolomide through targeting cyclin E1 in glioma cells.
Key Molecule: hsa-mir-151a [61]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell colony Activation hsa05200
Cell viability Activation hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
A172 cells Brain Homo sapiens (Human) CVCL_0131
U87 cells Brain Homo sapiens (Human) CVCL_0022
T98G cells Brain Homo sapiens (Human) CVCL_0556
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 Inhibiting miR-151a leads to increased XRCC4 levels, resulting in activated DNA repair and subsequent resistance to TMZ.
Key Molecule: hsa-mir-101 [62]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expressiom
Down-regulation
Resistant Drug Temozolomide
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 U251 cells Brain Homo sapiens (Human) CVCL_0021
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; TUNEL assay; Flow cytometry assay
Mechanism Description The endogenous protein level of GSk3beta and MGMT was significantly suppressed by combination of MALAT1 knockdown and miR-101 overexpression and the promoter methylation of MGMT was largely promoted by the combination of MALAT1 knockdown and miR-101 overexpression.
Key Molecule: Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) [62]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
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 U251 cells Brain Homo sapiens (Human) CVCL_0021
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay; TUNEL assay; Flow cytometry assay
Mechanism Description The endogenous protein level of GSk3beta and MGMT was significantly suppressed by combination of MALAT1 knockdown and miR-101 overexpression and the promoter methylation of MGMT was largely promoted by the combination of MALAT1 knockdown and miR-101 overexpression.
Key Molecule: hsa-miR-634 [63]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell colony Inhibition hsa05200
Cell invasion Inhibition hsa05200
Cell viability Inhibition hsa05200
RAF/ERK signaling pathway Activation hsa04010
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Overexpression of CYR61 increased the survival rate of U251/TMZ and U87/TMZ cells after TMZ treatment, while induction of miR-634 significantly suppressed the survival of U251/TMZ and U87/TMZ cells after TMZ treatment.
Key Molecule: hsa-mir-10a [64]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Upregulation of TUSC7,which acted by directly targeting and silencing expression of miR-10a gene, suppressed both TMZ resistance and expression of multidrug resistance protein 1 (MDR1) in U87TR cells,, and miR-10a mediated TUSC7-induced inhibition on TMZ resistance in U87TR cells.
Key Molecule: Tumor suppressor candidate 7 (TUSC7) [64]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Upregulation of TUSC7,which acted by directly targeting and silencing expression of miR-10a gene, suppressed both TMZ resistance and expression of multidrug resistance protein 1 (MDR1) in U87TR cells,, and miR-10a mediated TUSC7-induced inhibition on TMZ resistance in U87TR cells.
Key Molecule: hsa-miR-423-5p [65]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT/ERK signaling pathway Activation hsa04010
Cell invasion Activation hsa05200
Cell proliferation Activation hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
N3 GBM cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
RT-PCR; qRT-PCR
Experiment for
Drug Resistance
Cell-cycle assay
Mechanism Description miR-423-5p contributes to a malignant phenotype and temozolomide chemoresistance in glioblastomas.
Key Molecule: H19, imprinted maternally expressed transcript (H19) [66]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Knockdown of long noncoding RNA H19 sensitizes human glioma cells to temozolomide therapy.the expression level of H19 transcripts was increased compared to wild-type or nonresistant clones.Furthermore, the reduced expression of H19 altered major drug resistance genes, such as ABCB1 (MDR1), ABCC (MRP), and ABCG2 (BCRP), both at the mRNA and protein levels. Taken together, these findings suggest that H19 plays an important role in the development of TMZ resistance, and may represent a novel therapeutic target for TMZ-resistant gliomas.Our results suggested that knockdown of H19 significantly downregulated the expression of these drug-resistant genes, both at the mRNA (P<0.001 vs respective control siRNA) and protein levels. These data confirm that the H19-induced TMZ resistance is in part mediated by MDR, MRP, and ABCG2.
Key Molecule: hsa-mir-138 [67]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
miR138/BIM signaling pathway Regulation hsa05206
In Vitro Model LN229 cells Brain Homo sapiens (Human) CVCL_0393
A172 cells Brain Homo sapiens (Human) CVCL_0131
LN-18 cells Brain Homo sapiens (Human) CVCL_0392
T98G cells Brain Homo sapiens (Human) CVCL_0556
U87MG cells Brain Homo sapiens (Human) CVCL_GP63
LN308 cells Brain Homo sapiens (Human) CVCL_0394
D247MG cells Brain Homo sapiens (Human) CVCL_1153
LN-319 cells Brain Homo sapiens (Human) CVCL_3958
LN-428 cells Brain Homo sapiens (Human) CVCL_3959
In Vivo Model BALB/c nu/nu nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Trypan blue dye exclusion assay
Mechanism Description Transient transfection of miR-138 mimics in glioma cells with low basal miR-138 expression increased glioma cell proliferation. Moreover, miR-138 overexpression increased TMZ resistance in long-term glioblastoma cell lines and glioma initiating cell cultures. The apoptosis regulator BIM was identified as a direct target of miR-138, and its silencing mediated the induced TMZ resistance phenotype.
Key Molecule: hsa-mir-16 [68]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model A172 cells Brain Homo sapiens (Human) CVCL_0131
U138-MG cells Brain Homo sapiens (Human) CVCL_0020
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description The mechanism responsible for resistance of glioma cells to temozolomide was associated with miR-16-mediated downregulation of Bcl-2. miR-16 may function as an important modifier of the response of glioma cells to temozolomide.
Key Molecule: hsa-mir-497 [69]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Ectopic overexpression of miR-497 promotes chemotherapy resistance in glioma cells by targeting PDCD4, a tumor suppressor that is involved in apoptosis. In contrast, the inhibition of miR-497 enhances apoptosis and increases the sensitivity of glioma cells to TMZ.
Key Molecule: hsa-mir-125b-2 [70]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Mitochondrial apoptotic signaling pathway Inhibition hsa04210
In Vitro Model Human glioblastoma tissues and PRGMTTT samples Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-125b-2 is overexpressed in glioblastoma multiforme tissues and the corresponding stem cells (GBMSC); downregulation of miR-125b-2 expression in GBMSC could allow TMZ to induce GBMSC apoptosis. Additionally, the expression of the anti-apop-totic protein Bcl-2 was decreased after the TMZ+miR-125b-2 inhibitor treatment, while the expression of the proapoptotic protein Bax was increased. he induction of apoptosis in GBMSC is also associated with increased cytochrome c release from mitochondria, induction of Apaf-1, activation of caspase-3 and poly-ADP-ribose polymerase (PARP). miR-125b-2 overexpression might confer glioblastoma stem cells resistance to TMZ.
Key Molecule: hsa-mir-21 [71]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
In Vitro Model U87-MG cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Trypan blue dye exclusion assay
Mechanism Description miR-21 could inhibit TMZ-induced apoptosis in U87MG cells, at least in part, by decreasing Bax/Bcl-2 ratio and caspase-3 activity.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [64]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Upregulation of TUSC7,which acted by directly targeting and silencing expression of miR-10a gene, suppressed both TMZ resistance and expression of multidrug resistance protein 1 (MDR1) in U87TR cells,, and miR-10a mediated TUSC7-induced inhibition on TMZ resistance in U87TR cells.
Key Molecule: Multidrug resistance protein 1 (ABCB1) [66]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Knockdown of long noncoding RNA H19 sensitizes human glioma cells to temozolomide therapy.the expression level of H19 transcripts was increased compared to wild-type or nonresistant clones.Furthermore, the reduced expression of H19 altered major drug resistance genes, such as ABCB1 (MDR1), ABCC (MRP), and ABCG2 (BCRP), both at the mRNA and protein levels. Taken together, these findings suggest that H19 plays an important role in the development of TMZ resistance, and may represent a novel therapeutic target for TMZ-resistant gliomas.Our results suggested that knockdown of H19 significantly downregulated the expression of these drug-resistant genes, both at the mRNA (P<0.001 vs respective control siRNA) and protein levels. These data confirm that the H19-induced TMZ resistance is in part mediated by MDR, MRP, and ABCG2.
Key Molecule: Multidrug resistance-associated protein 1 (MRP1) [66]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Knockdown of long noncoding RNA H19 sensitizes human glioma cells to temozolomide therapy.the expression level of H19 transcripts was increased compared to wild-type or nonresistant clones.Furthermore, the reduced expression of H19 altered major drug resistance genes, such as ABCB1 (MDR1), ABCC (MRP), and ABCG2 (BCRP), both at the mRNA and protein levels. Taken together, these findings suggest that H19 plays an important role in the development of TMZ resistance, and may represent a novel therapeutic target for TMZ-resistant gliomas.Our results suggested that knockdown of H19 significantly downregulated the expression of these drug-resistant genes, both at the mRNA (P<0.001 vs respective control siRNA) and protein levels. These data confirm that the H19-induced TMZ resistance is in part mediated by MDR, MRP, and ABCG2.
Key Molecule: ATP-binding cassette sub-family G2 (ABCG2) [66]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Knockdown of long noncoding RNA H19 sensitizes human glioma cells to temozolomide therapy.the expression level of H19 transcripts was increased compared to wild-type or nonresistant clones.Furthermore, the reduced expression of H19 altered major drug resistance genes, such as ABCB1 (MDR1), ABCC (MRP), and ABCG2 (BCRP), both at the mRNA and protein levels. Taken together, these findings suggest that H19 plays an important role in the development of TMZ resistance, and may represent a novel therapeutic target for TMZ-resistant gliomas.Our results suggested that knockdown of H19 significantly downregulated the expression of these drug-resistant genes, both at the mRNA (P<0.001 vs respective control siRNA) and protein levels. These data confirm that the H19-induced TMZ resistance is in part mediated by MDR, MRP, and ABCG2.
Key Molecule: Multidrug resistance protein 1 (ABCB1) [72]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model Glioblastoma tissue .
Experiment for
Molecule Alteration
Real-time PCR
Experiment for
Drug Resistance
Patient survival time
Mechanism Description In the chemosensitive MDR1-negative parental cell line k562 10 ug/ml temozolomide resulted in pronounced cell death with only 47.1% surviving 48 h compared with the control. In contrast, in the highly MDR1-expressing resistant subline k562-VP16, cell death was significantly lower after exposure to temozolomide with 73.4% surviving 48 h (P = 0.002). Addition of a nontoxic dose of the MDR1-modulator cyclosporine A (1 uM) to temozolomide resulted in a trend towards restoration of chemosensitivity in the resistant MDR1-expressing cell line.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: SBF2 antisense RNA 1 (SBF2-AS1) [73]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation NF-kB/XIAP signaling pathway Activation hsa04218
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
A172 cells Brain Homo sapiens (Human) CVCL_0131
T98 cells Brain Homo sapiens (Human) CVCL_B368
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Subcutaneous and orthotopic xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Exosomal SBF2-AS1 functions as a ceRNA for miR-151a-3p, leading to the disinhibition of its endogenous target, X-ray repair cross complementing 4 (XRCC4), which enhances DSB repair in GBM cells. Exosomes selected from temozolomide-resistant GBM cells had high levels of SBF2-AS1 and spread TMZ resistance to chemoresponsive GBM cells.
Key Molecule: hsa-miR-151a-3p [73]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation DNA damage repair signaling pathway Activation hsa03410
miR151a-3p/XRCC4 signaling pathway Regulation hsa05206
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
A172 cells Brain Homo sapiens (Human) CVCL_0131
T98 cells Brain Homo sapiens (Human) CVCL_B368
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Subcutaneous and orthotopic xenograft model Mus musculus
Experiment for
Molecule Alteration
RIP experiments; qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Exosomal SBF2-AS1 functions as a ceRNA for miR-151a-3p, leading to the disinhibition of its endogenous target, X-ray repair cross complementing 4 (XRCC4), which enhances DSB repair in GBM cells. Exosomes selected from temozolomide-resistant GBM cells had high levels of SBF2-AS1 and spread TMZ resistance to chemoresponsive GBM cells.
Key Molecule: H19, imprinted maternally expressed transcript (H19) [74]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell colony Activation hsa05200
Cell viability Activation hsa05200
Wnt/beta-catenin signaling pathway Activation hsa04310
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
M059J cells Brain Homo sapiens (Human) CVCL_0400
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Silencing of H19 decreases chemoresistance through suppressing EMT via the Wnt/beta-Catenin pathway.
Key Molecule: Vimentin (VIM) [74]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
Wnt/beta-catenin signaling pathway Activation hsa04310
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
M059J cells Brain Homo sapiens (Human) CVCL_0400
Experiment for
Molecule Alteration
Western blot analysis; RNAi assay
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Silencing of H19 decreases chemoresistance through suppressing EMT via the Wnt/beta-Catenin pathway.
Key Molecule: Zinc finger E-box-binding homeobox 1 (ZEB1) [74]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
Wnt/beta-catenin signaling pathway Activation hsa04310
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
M059J cells Brain Homo sapiens (Human) CVCL_0400
Experiment for
Molecule Alteration
Western blot analysis; RNAi assay
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Silencing of H19 decreases chemoresistance through suppressing EMT via the Wnt/beta-Catenin pathway.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Hepatocyte growth factor receptor (MET) [51]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
c-Met signaling signaling pathway Inhibition hsa01521
In Vitro Model HG7 cells Brain Homo sapiens (Human) N.A.
LN229 cells Brain Homo sapiens (Human) CVCL_0393
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay; Flow cytometry assay
Mechanism Description Lnc-TALC promotes O6-methylguanine-DNA methyltransferase expression via regulating the c-Met pathway by competitively binding with miR-20b-3p.
Key Molecule: Hepatocyte growth factor receptor (MET) [51]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
c-Met signaling signaling pathway Inhibition hsa01521
In Vitro Model HG7 cells Brain Homo sapiens (Human) N.A.
LN229 cells Brain Homo sapiens (Human) CVCL_0393
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay; Flow cytometry assay
Mechanism Description Lnc-TALC promotes O6-methylguanine-DNA methyltransferase expression via regulating the c-Met pathway by competitively binding with miR-20b-3p.
Key Molecule: Serine/threonine-protein kinase mTOR (mTOR) [53]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell autophagy Inhibition hsa04140
Cell cytotoxicity Activation hsa04650
In Vitro Model U87 cells Brain Homo sapiens (Human) CVCL_0022
U257 cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Luciferase reporter assay; Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay; MTT assay; Transwell assay
Mechanism Description Upregulation of CASC2 sensitized glioma to temozolomide cytotoxicity through autophagy inhibition by sponging miR193a-5p and regulating mTOR expression. CASC2 is downregulated in gliomas, resulting in increased miR193a-5p level and a decrease in mTOR expression, which further induces protective autophagy, leading to TMZ resistance.
Key Molecule: Tumor suppressor candidate 3 (TUSC3) [56]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model U87MG cells Brain Homo sapiens (Human) CVCL_GP63
U87MG-res cells Brain Homo sapiens (Human) CVCL_GP63
Experiment for
Molecule Alteration
Immunofluorescence staining; Western blot analysis
Experiment for
Drug Resistance
Soft agar assay; MTT assay; Sphere formation assay
Mechanism Description microRNA-132 induces temozolomide resistance and promotes the formation of cancer stem cell phenotypes by targeting tumor suppressor candidate 3 in glioblastoma.
Key Molecule: Methylated-DNA--protein-cysteine methyltransferase (MGMT) [57]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Epithelial mesenchymal transition signaling pathway Inhibition hsa01521
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
U373 cells Brain Homo sapiens (Human) CVCL_2219
U118 cells Brain Homo sapiens (Human) CVCL_0633
NHA Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; BrdU incorporation assay
Mechanism Description XIST was inversely correlated with miR29c, positively correlated with PS1, positively related with MGMT. XIST can inhibit miR29c expression by directly binding to miR29c and subsequently up-regulate the expression of SP1 and MGMT to promote the chemoresistance of glioma cells to TMZ.
Key Molecule: Transcription factor Sp1 (SP1) [57]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation DNA damage repair signaling pathway Activation hsa03410
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
U373 cells Brain Homo sapiens (Human) CVCL_2219
U118 cells Brain Homo sapiens (Human) CVCL_0633
NHA Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; BrdU incorporation assay
Mechanism Description XIST was inversely correlated with miR29c, positively correlated with PS1, positively related with MGMT. XIST can inhibit miR29c expression by directly binding to miR29c and subsequently up-regulate the expression of SP1 and MGMT to promote the chemoresistance of glioma cells to TMZ.
Key Molecule: Zinc finger E-box-binding homeobox 1 (ZEB1) [55]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description MALAT1 decreased the sensitivity of resistant glioma cell lines to TMZ by upregulating ZEB1.
Key Molecule: Paired box protein Pax-6 (PAX6) [58]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Transwell assay; Transwell matrix penetration assay; MTT assay; BrdU incorporation assay
Mechanism Description miR223/PAX6 axis regulates glioblastoma stem cell proliferation and the chemo resistance to TMZ via inhibition of PI3k/Akt pathway.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [59]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation IGF1R/IRS1 signaling pathway Activation hsa04212
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
U138 cells Brain Homo sapiens (Human) CVCL_0020
HEK293 cells Kidney Homo sapiens (Human) CVCL_0045
NHA cells Brain Homo sapiens (Human) N.A.
LN382 cells Brain Homo sapiens (Human) CVCL_3956
SF295 cells Brain Homo sapiens (Human) CVCL_1690
SHG-44 cells Brain Homo sapiens (Human) CVCL_6728
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Up-regulation of miR-497 confers resistance to temozolomide in human glioma cells by targeting mTOR/Bcl-2. The silencing of miR-497 decreased the protein levels of IGF1R/IRS1 pathway-related proteins, that is, IGF1R, IRS1, mTOR, and Bcl-2.
Key Molecule: Serine/threonine-protein kinase mTOR (mTOR) [59]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation IGF1R/IRS1 signaling pathway Activation hsa04212
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
U138 cells Brain Homo sapiens (Human) CVCL_0020
HEK293 cells Kidney Homo sapiens (Human) CVCL_0045
NHA cells Brain Homo sapiens (Human) N.A.
LN382 cells Brain Homo sapiens (Human) CVCL_3956
SF295 cells Brain Homo sapiens (Human) CVCL_1690
SHG-44 cells Brain Homo sapiens (Human) CVCL_6728
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Up-regulation of miR-497 confers resistance to temozolomide in human glioma cells by targeting mTOR/Bcl-2. The silencing of miR-497 decreased the protein levels of IGF1R/IRS1 pathway-related proteins, that is, IGF1R, IRS1, mTOR, and Bcl-2.
Key Molecule: G1/S-specific cyclin-E1 (CCNE1) [60]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model U251-MG cells Brain Homo sapiens (Human) CVCL_0021
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Hsa-miR-195 could negatively regulate the expression of CCNE1 in glioma and microRNA-195 reverses the resistance to temozolomide through targeting cyclin E1 in glioma cells.
Key Molecule: DNA repair protein XRCC4 (XRCC4) [61]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell colony Activation hsa05200
Cell viability Activation hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
A172 cells Brain Homo sapiens (Human) CVCL_0131
U87 cells Brain Homo sapiens (Human) CVCL_0022
T98G cells Brain Homo sapiens (Human) CVCL_0556
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 Inhibiting miR-151a leads to increased XRCC4 levels, resulting in activated DNA repair and subsequent resistance to TMZ.
Key Molecule: CCN family member 1 (CYR61) [63]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell colony Activation hsa05200
Cell viability Activation hsa05200
RAF/ERK signaling pathway Activation hsa04010
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Overexpression of CYR61 increased the survival rate of U251/TMZ and U87/TMZ cells after TMZ treatment, while induction of miR-634 significantly suppressed the survival of U251/TMZ and U87/TMZ cells after TMZ treatment.
Key Molecule: Growth protein 4 inhibitor (ING4) [65]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT/ERK signaling pathway Activation hsa04010
Cell invasion Activation hsa05200
Cell proliferation Activation hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
N3 GBM cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Cell-cycle assay
Mechanism Description miR-423-5p contributes to a malignant phenotype and temozolomide chemoresistance in glioblastomas.
Key Molecule: Bcl-2-like protein 11 (BCL2L11) [67]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
miR138/BIM signaling pathway Regulation hsa05206
In Vitro Model LN229 cells Brain Homo sapiens (Human) CVCL_0393
A172 cells Brain Homo sapiens (Human) CVCL_0131
LN-18 cells Brain Homo sapiens (Human) CVCL_0392
T98G cells Brain Homo sapiens (Human) CVCL_0556
U87MG cells Brain Homo sapiens (Human) CVCL_GP63
LN308 cells Brain Homo sapiens (Human) CVCL_0394
D247MG cells Brain Homo sapiens (Human) CVCL_1153
LN-319 cells Brain Homo sapiens (Human) CVCL_3958
LN-428 cells Brain Homo sapiens (Human) CVCL_3959
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Trypan blue dye exclusion assay
Mechanism Description Transient transfection of miR-138 mimics in glioma cells with low basal miR-138 expression increased glioma cell proliferation. Moreover, miR-138 overexpression increased TMZ resistance in long-term glioblastoma cell lines and glioma initiating cell cultures. The apoptosis regulator BIM was identified as a direct target of miR-138, and its silencing mediated the induced TMZ resistance phenotype.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [68]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model A172 cells Brain Homo sapiens (Human) CVCL_0131
U138-MG cells Brain Homo sapiens (Human) CVCL_0020
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description The mechanism responsible for resistance of glioma cells to temozolomide was associated with miR-16-mediated downregulation of Bcl-2. miR-16 may function as an important modifier of the response of glioma cells to temozolomide.
Key Molecule: Programmed cell death protein 4 (PDCD4) [69]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Resistant Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Ectopic overexpression of miR-497 promotes chemotherapy resistance in glioma cells by targeting PDCD4, a tumor suppressor that is involved in apoptosis. In contrast, the inhibition of miR-497 enhances apoptosis and increases the sensitivity of glioma cells to TMZ.
Key Molecule: DNA mismatch repair protein Msh6 (MSH6) [75]
Resistant Disease FGFR-tacc positive glioblastoma [ICD-11: 2A00.01]
Molecule Alteration Mutation
.
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
SSCP assay; Direct sequencing assay
Mechanism Description Prominent example of therapy-induced molecular alterations in gliomas which themselves ensue therapeutic consequences are MSH6 mutations in glioblastomas which arise during temozolomide chemotherapy and which are able to convey temozolomide resistance in affected tumors.
Key Molecule: Endoplasmic reticulum chaperone BiP (HSPA5) [76]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation UPR signaling pathway Activation hsa0414)
In Vitro Model U87MG cells Brain Homo sapiens (Human) CVCL_GP63
In Vivo Model BALB/c nu/nu athymic mice xenografts model Mus musculus
Experiment for
Molecule Alteration
Northern blot analysis
Experiment for
Drug Resistance
Clonogenic assay
Mechanism Description Transcripts for the ER chaperones GRP94 and GRP78 were upregulated in the U87MG and U87+EGFR gliomas, relative to normal mouse brain from healthy animals. Elevated levels of UPR transcription factors and ER chaperones correlated with poor patient prognosis; western blots of high grade gliomas and tissue microarray immunohistochemistry verified high expression of UPR players, especially GRP94, in high grade gliomas. Activation of the UPR signaling pathways is a prominent feature of glioma biology that leads to metabolic shifts and enhances chemoresistant features of gliomas.
Key Molecule: Endoplasmin (HSP90B1) [76]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation UPR signaling pathway Activation hsa0414)
In Vitro Model U87MG cells Brain Homo sapiens (Human) CVCL_GP63
In Vivo Model BALB/c nu/nu athymic mice xenografts model Mus musculus
Experiment for
Molecule Alteration
Northern blot analysis
Experiment for
Drug Resistance
Clonogenic assay
Mechanism Description Transcripts for the ER chaperones GRP94 and GRP78 were upregulated in the U87MG and U87+EGFR gliomas, relative to normal mouse brain from healthy animals. Elevated levels of UPR transcription factors and ER chaperones correlated with poor patient prognosis; western blots of high grade gliomas and tissue microarray immunohistochemistry verified high expression of UPR players, especially GRP94, in high grade gliomas. Activation of the UPR signaling pathways is a prominent feature of glioma biology that leads to metabolic shifts and enhances chemoresistant features of gliomas.
Key Molecule: Methylated-DNA--protein-cysteine methyltransferase (MGMT) [6]
Resistant Disease Anaplastic astrocytoma [ICD-11: 2A00.04]
Molecule Alteration Expression
Up-regulation
Resistant Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
Oncotech EDR assay
Mechanism Description For drugs that have evaded cytosolic mechanisms of drug resistance, the nucleus is equipped with the capacity to remove BCNU or temozolomide alkyl groups from the O6-position of guanine via a reaction catalyzed by MGMT. Repair occurs before cross-link formation and involves an irreversible stoichiometric covalent transfer of the O6-alkyl DNA adduct to a cysteine within the active site of MGMT, resulting in the inactivation and subsequent depletion of enzyme activity. MGMT-mediated repair is rapid, with a half-life of 35 hours. MGMT enzyme recovery occurs via de novo synthesis. In malignant glioma patients, MGMT overexpression has been associated with resistance to BCNU and similar alkylating agents and was an independent predictor of poor survival. MGMT is also thought to contribute to temozolomide resistance, which we did not detect in our study. This may be related to the in vitro pharmacokinetic differences between BCNU and temozolomide.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-126-3p [77]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell autophagy Inhibition hsa04140
Wnt/Beta-catenin signaling pathway Activation hsa04310
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay; Flow cytometry assay
Mechanism Description miR-126-3p sensitizes glioblastoma cells to temozolomide by inactivating Wnt/beta-catenin signaling via targeting SOX2.
Key Molecule: hsa-mir-23b [78]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
In Vitro Model U87 GSCs Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-23b overexpression sensitized U87 glioma stem cells to TMZ-induced growth inhibition. And miR-23b had a synergistically suppressive effect on the expression of HMGA2 with TMZ in U87 GSCs.
Key Molecule: Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) [79]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
LN-18 cells Brain Homo sapiens (Human) CVCL_0392
T98G cells Brain Homo sapiens (Human) CVCL_0556
U87-luc2 Brain Homo sapiens (Human) CVCL_5J12
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
XTT assay; CellTiter-Glo Luminescent Cell Viability Assay
Mechanism Description Targeted nanocomplex carrying siRNA against MALAT1 sensitizes glioblastoma to temozolomide.
Key Molecule: Cancer susceptibility 2 (CASC2) [53]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
In Vitro Model U87 cells Brain Homo sapiens (Human) CVCL_0022
U257 cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
Flow cytometry assay; MTT assay; Transwell assay
Mechanism Description CASC2 negatively regulates miR193a-5p expression by direct interaction in glioma cells. Overexpression of CASC2 or inhibition of miR193a-5p reduced TMZ-induced autophagy via mTOR upregulation, which makes the glioma cells become sensitive to TMZ cytotoxicity.
Key Molecule: hsa-miR-193a-5p [53]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
In Vitro Model U87 cells Brain Homo sapiens (Human) CVCL_0022
U257 cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
Flow cytometry assay; MTT assay; Transwell assay
Mechanism Description Upregulation of CASC2 sensitized glioma to temozolomide cytotoxicity through autophagy inhibition by sponging miR193a-5p and regulating mTOR expression. mTOR or CASC2 overexpression or miR193a-5p inhibition remarkably reduced autophagy-related proteins expression.
Key Molecule: hsa-miR-146b-5p [80]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AKT/NF-kappaB signaling pathway Inhibition hsa05135
In Vitro Model U251-MG cells Brain Homo sapiens (Human) CVCL_0021
U87-MG cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR 146b 5p suppresses glioblastoma cell resistance to temozolomide through targeting TRAF6. Overexpression of miR 146b 5p or TRAF6 knockdown significantly decreased the level of p AkT and p p65.
Key Molecule: hsa-mir-29c [57]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation DNA mismatch repair pathway Regulation hsa03430
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
U373 cells Brain Homo sapiens (Human) CVCL_2219
U118 cells Brain Homo sapiens (Human) CVCL_0633
NHA Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qRT-PCR; RIP assay; Dual luciferase reporter assay
Experiment for
Drug Resistance
MTT assay; BrdU incorporation assay
Mechanism Description XIST can amplify the chemoresistance of glioma cell lines to TMZ through directly targetting miR29c via SP1 and MGMT. XIST/miR29c axis regulated glioma cell chemoresistance to TMZ through RNA mismatch repair pathway. XIST expression was up-regulated by miR29c inhibition while down-regulated by ectopic miR29, and XIST directly binds to miR29c to inhibit its expression, XIST and miR29c neatively regulates each other.
Key Molecule: X inactive specific transcript (XIST) [57]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation DNA mismatch repair pathway Regulation hsa03430
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
U373 cells Brain Homo sapiens (Human) CVCL_2219
U118 cells Brain Homo sapiens (Human) CVCL_0633
NHA Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay; BrdU incorporation assay
Mechanism Description XIST can amplify the chemoresistance of glioma cell lines to TMZ through directly targetting miR29c via SP1 and MGMT. XIST/miR29c axis regulated glioma cell chemoresistance to TMZ through RNA mismatch repair pathway. XIST expression was up-regulated by miR29c inhibition while down-regulated by ectopic miR29, and XIST directly binds to miR29c to inhibit its expression, XIST and miR29c neatively regulates each other.
Key Molecule: hsa-mir-181 [81]
Sensitive Disease Glioblastoma multiforme [ICD-11: 2A00.03]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation EGFR signaling pathway Inhibition hsa01521
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometric analysis
Mechanism Description miR181b modulates chemosensitivity of glioblastoma multiforme cells to temozolomide by targeting the epidermal growth factor receptor.
Key Molecule: hsa-miR-198 [82]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
A172 cells Brain Homo sapiens (Human) CVCL_0131
T98 cells Brain Homo sapiens (Human) CVCL_B368
U87 cells Brain Homo sapiens (Human) CVCL_0022
U118 cells Brain Homo sapiens (Human) CVCL_0633
U138 cells Brain Homo sapiens (Human) CVCL_0020
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay
Mechanism Description miR-198 enhances temozolomide sensitivity in glioblastoma by targeting MGMT.
Key Molecule: hsa-mir-124 [83]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model LN229 cells Brain Homo sapiens (Human) CVCL_0393
A172 cells Brain Homo sapiens (Human) CVCL_0131
T98G cells Brain Homo sapiens (Human) CVCL_0556
M059J cells Brain Homo sapiens (Human) CVCL_0400
M059k cells Brain Homo sapiens (Human) CVCL_0401
U-87 MG cells Brain Homo sapiens (Human) CVCL_0022
U118 MG cells Brain Homo sapiens (Human) CVCL_0633
U138-MG cells Brain Homo sapiens (Human) CVCL_0020
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR124 suppresses glioblastoma growth and potentiates chemosensitivity by inhibiting AURkA. Re-expression of AURkA rescued miR124-mediated growth suppression.
Key Molecule: hsa-mir-203 [54]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation miR203-TS signaling pathway Regulation hsa05206
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometric analysis
Mechanism Description miR203 re-sensitizes TMZ resistant cells through directly targeting TS.
Key Molecule: Cancer susceptibility 2 (CASC2) [52]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PTEN signaling pathway Activation hsa05235
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
SNB19 cells Brain Homo sapiens (Human) CVCL_0535
U373 cells Brain Homo sapiens (Human) CVCL_2219
U118 cells Brain Homo sapiens (Human) CVCL_0633
NHA cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; BrdU incorporation assay
Mechanism Description LncRNA CASC2 interacts with miR181a to modulate glioma growth and resistance to TMZ through PTEN pathway.
Key Molecule: Cancer susceptibility 2 (CASC2) [52]
Sensitive Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell growth Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
SNB19 cells Brain Homo sapiens (Human) CVCL_0535
U373 cells Brain Homo sapiens (Human) CVCL_2219
U118 cells Brain Homo sapiens (Human) CVCL_0633
NHA cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTT assay; BrdU incorporation assay
Mechanism Description CASC2 up-regulated PTEN protein and down-regulated p-AkT protein through regulating miR181a, and the effect of CASC2 on PTEN and p-AkT could be partially restored by miR181a.
Key Molecule: hsa-mir-181a [52]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PTEN signaling pathway Activation hsa05235
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
SNB19 cells Brain Homo sapiens (Human) CVCL_0535
U373 cells Brain Homo sapiens (Human) CVCL_2219
U118 cells Brain Homo sapiens (Human) CVCL_0633
NHA cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; BrdU incorporation assay
Mechanism Description CASC2 up-regulated PTEN protein and down-regulated p-AkT protein through regulating miR181a, and the effect of CASC2 on PTEN and p-AkT could be partially restored by miR181a.
Key Molecule: hsa-miR-433-3p [84]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
U87 cells Brain Homo sapiens (Human) CVCL_0022
SNB19 cells Brain Homo sapiens (Human) CVCL_0535
LN308 cells Brain Homo sapiens (Human) CVCL_0394
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay; Transwell migration assay; Annexin V/fluorescein isothiocyanate (FITC) apoptosis assay
Mechanism Description miR433-3p suppresses cell growth and enhances chemosensitivity by targeting CREB in human glioma, the overexpression of CREB can rescue the phenotype changes induced by miR433-3p overexpression.
Key Molecule: hsa-mir-101 [85]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A172 cells Brain Homo sapiens (Human) CVCL_0131
T98G cells Brain Homo sapiens (Human) CVCL_0556
U251-MG cells Brain Homo sapiens (Human) CVCL_0021
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay
Mechanism Description microRNA-101 reverses temozolomide resistance by inhibition of GSk3beta in glioblastoma.
Key Molecule: hsa-miR-370-3p [86]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
U87 cells Brain Homo sapiens (Human) CVCL_0022
U373 cells Brain Homo sapiens (Human) CVCL_2219
LN-18 cells Brain Homo sapiens (Human) CVCL_0392
T98G cells Brain Homo sapiens (Human) CVCL_0556
SHG-44 cells Brain Homo sapiens (Human) CVCL_6728
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
TMZ cytotoxicity assay; Colony formation assay; gamma -H2AX foci formation assay
Mechanism Description Up-regulation of miR370-3p restores glioblastoma multiforme sensitivity to temozolomide by influencing MGMT expression. MGMT was found to be inversely correlated with miR370-3p expression.
Key Molecule: hsa-mir-196b [87]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell colony Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Down-regulation of miR-196b increased glioma cell sensitivity to TMZ and E2F1 decreased following transfection with miR-196b inhibitors.
Key Molecule: hsa-miR-7-5p [88]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell colony Inhibition hsa05200
Cell viability Inhibition hsa05200
In Vitro Model LN229 cells Brain Homo sapiens (Human) CVCL_0393
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-7-5p suppresses stemness and enhances temozolomide sensitivity of drug-resistant glioblastoma cells by targeting Yin Yang 1.
Key Molecule: hsa-miR-181b-5p [89]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
Cell viability Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87MG cells Brain Homo sapiens (Human) CVCL_GP63
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Transwell assay
Mechanism Description Upregulation of miR-181b-5p targets Bcl-2 directly and may function as an important modifier to sensitize glioma cells to TMZ.
Key Molecule: hsa-mir-1271 [90]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model A172 cells Brain Homo sapiens (Human) CVCL_0131
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description The chemoresistant cell survival mediated with Bcl-2 was inhibited by overexpression of miR-1271 and was enhanced by depletion of miR-1271.
Key Molecule: hsa-miR-299-5p [91]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell growth Inhibition hsa05200
MAPK/ERK signaling pathway Inhibition hsa04010
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
A172 cells Brain Homo sapiens (Human) CVCL_0131
SNB19 cells Brain Homo sapiens (Human) CVCL_0535
T98G cells Brain Homo sapiens (Human) CVCL_0556
LN308 cells Brain Homo sapiens (Human) CVCL_0394
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description Inhibition of microRNA-299-5p sensitizes glioblastoma cells to temozolomide via upregulating GOLPH3 and inactivating the MAPk/ERk signaling pathway.
Key Molecule: hsa-miR-1268a [92]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Inhibition hsa05200
Hedgehog signaling pathway Inhibition hsa04340
MAPK signaling pathway Inhibition hsa04010
p53 signaling pathway Inhibition hsa04115
In Vitro Model LN229 cells Brain Homo sapiens (Human) CVCL_0393
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Overexpression of miR-1268a inhibited protein translation of ABCC1, which enhanced sensitivity of GBM cells to TMZ.
Key Molecule: Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) [62]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell colony Inhibition hsa05200
Cell viability Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; TUNEL assay; Flow cytometry assay
Mechanism Description The endogenous protein level of GSk3beta and MGMT was significantly suppressed by combination of MALAT1 knockdown and miR-101 overexpression and the promoter methylation of MGMT was largely promoted by the combination of MALAT1 knockdown and miR-101 overexpression.
Key Molecule: hsa-mir-101 [62]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell colony Inhibition hsa05200
Cell viability Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; TUNEL assay; Flow cytometry assay
Mechanism Description The endogenous protein level of GSk3beta and MGMT was significantly suppressed by combination of MALAT1 knockdown and miR-101 overexpression and the promoter methylation of MGMT was largely promoted by the combination of MALAT1 knockdown and miR-101 overexpression.
Key Molecule: hsa-mir-29c [93]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
DNA mismatch repair pathway Regulation hsa03430
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Ectopic expression of miR-29c increased TMZ sensitivity by inhibiting cell growth and promoting apoptosis in U251/TR cells.
Key Molecule: hsa-mir-10a [94]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
TGF-beta signaling pathway Regulation hsa04350
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description LncRNA RP11-838N2.4 (+) TMZ sensitivity in GBM by serving as a ceRNA, sequestering with miR-10a on an epigenetic level.
Key Molecule: Long non-protein coding RNA (RP11-838N2.4) [94]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
TGF-beta signaling pathway Regulation hsa04350
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description LncRNA RP11-838N2.4 (+) TMZ sensitivity in GBM by serving as a ceRNA, sequestering with miR-10a on an epigenetic level.
Key Molecule: hsa-mir-203 [95]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
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 A172 cells Brain Homo sapiens (Human) CVCL_0131
U251-MG cells Brain Homo sapiens (Human) CVCL_0021
U87MG cells Brain Homo sapiens (Human) CVCL_GP63
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-203 was reversely associated with migration and invasion, and positively associated with chemosensitivity in glioma cells. E2F3 was shown to be a novel target of miR-203 and E2F3 knockdown exerted a similar effect to that of miR-203 overexpression. These results indicate that miR-203 may act as a tumor suppressor by targeting E2F3 in glioma cells and that miR-203/E2F3 may be a novel candidate for developing rational therapeutic strategies in glioma treatment.
Key Molecule: hsa-mir-128a [96]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Expression of Rap1B is negatively regulated by miR-128 and miR-149. TMZ inhibits Rap1B expression by upregulating miR-128 and miR-149. miR-128 and miR-149 suppress cell proliferation and invasion, and alter cytoskeletal remodeling by affecting Rap1B-associated small GTPase. miR-128 and miR-149 increase the chemosensitivity of TMZ in glioblastoma cells.
Key Molecule: hsa-mir-149 [96]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Expression of Rap1B is negatively regulated by miR-128 and miR-149. TMZ inhibits Rap1B expression by upregulating miR-128 and miR-149. miR-128 and miR-149 suppress cell proliferation and invasion, and alter cytoskeletal remodeling by affecting Rap1B-associated small GTPase. miR-128 and miR-149 increase the chemosensitivity of TMZ in glioblastoma cells.
Key Molecule: hsa-mir-125b [97]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
NF-kappaB signaling pathway Inhibition hsa04064
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN-18 cells Brain Homo sapiens (Human) CVCL_0392
T98G cells Brain Homo sapiens (Human) CVCL_0556
U87-MG cells Brain Homo sapiens (Human) CVCL_0022
HS683 cells Brain Homo sapiens (Human) CVCL_0844
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Promega assay
Mechanism Description A novel mechanism independent of TP53 and MGMT by which oncogenic miR-125b confers TMZ resistance by targeting TNFAIP3 and NkIRAS2. GBM cells overexpressing miR-125b showed increased NF-kB activity and upregulation of anti-apoptotic and cell cycle genes. This was significantly associated with resistance of GBM cells to TNFalpha- and TNF-related inducing ligand-induced apoptosis as well as resistance to TMZ. Conversely, overexpression of anti-miR-125b resulted in cell cycle arrest, increased apoptosis and increased sensitivity to TMZ, indicating that endogenous miR-125b is sufficient to control these processes. GBM cells overexpressing TNFAIP3 and NkIRAS2 were refractory to miR-125b-induced apoptosis resistance as well as TMZ resistance, indicating that both genes are relevant targets of miR-125b.
Key Molecule: hsa-mir-181a [98]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Ras-associated protein 1 (Rap1), a growth regulatory protein, belongs to a member of RAS-like small GTP-binding protein superfamily. Rap1 regulates several basic cellular functions: migration, adhesion and growth. TMZ can inhibit the Rap1B expression to exert its cell killing by upregulating miR-181a/b/c/d subunits; conversely, each miR-181a/b/c/d subunit enhanced the chemosensitivity of TMZ in glioblastoma.
Key Molecule: hsa-mir-181 [98]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Ras-associated protein 1 (Rap1), a growth regulatory protein, belongs to a member of RAS-like small GTP-binding protein superfamily. Rap1 regulates several basic cellular functions: migration, adhesion and growth. TMZ can inhibit the Rap1B expression to exert its cell killing by upregulating miR-181a/b/c/d subunits; conversely, each miR-181a/b/c/d subunit enhanced the chemosensitivity of TMZ in glioblastoma.
Key Molecule: hsa-mir-181c [98]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Ras-associated protein 1 (Rap1), a growth regulatory protein, belongs to a member of RAS-like small GTP-binding protein superfamily. Rap1 regulates several basic cellular functions: migration, adhesion and growth. TMZ can inhibit the Rap1B expression to exert its cell killing by upregulating miR-181a/b/c/d subunits; conversely, each miR-181a/b/c/d subunit enhanced the chemosensitivity of TMZ in glioblastoma.
Key Molecule: hsa-mir-181d [98]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description Ras-associated protein 1 (Rap1), a growth regulatory protein, belongs to a member of RAS-like small GTP-binding protein superfamily. Rap1 regulates several basic cellular functions: migration, adhesion and growth. TMZ can inhibit the Rap1B expression to exert its cell killing by upregulating miR-181a/b/c/d subunits; conversely, each miR-181a/b/c/d subunit enhanced the chemosensitivity of TMZ in glioblastoma.
Key Molecule: hsa-mir-125b [99]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
In Vitro Model GSCs cells Brain Homo sapiens (Human) N.A.
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
PCR
Experiment for
Drug Resistance
Transwell invasion assay
Mechanism Description Inhibition of miR-125b expression may enhance sensitivity of GSCs to temozolomide by targeting PIAS3 on cell invasion.
Key Molecule: hsa-mir-17 [100]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell autophagy Activation hsa04140
In Vitro Model T98G cells Brain Homo sapiens (Human) CVCL_0556
U373-MG Brain Homo sapiens (Human) CVCL_2219
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
Celltiter 96 aqueous one solution cell proliferation assay
Mechanism Description ATG7 is a potential target for miR-17, and this miRNA could negatively regulate ATG7 expression, resulting in a modulation of the autophagic status in T98G glioblastoma cells, the autophagy activation by anti-miR-17 resulted in a decrease of the threshold resistance at temozolomide doses in T98G cells.
Key Molecule: hsa-mir-181 [101]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
MAPK signaling pathway Inhibition hsa04010
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-181b independently predicted chemoresponse to temozolomide and enhanced temozolomide sensitivity via MEk1 downregulation.
Key Molecule: hsa-miR-296-3p [38]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U251AR cells Brain Homo sapiens (Human) CVCL_1G29
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description EAG1 channel might be involved in cell-cycle progression of tumour cells because a significant reduction in the proliferation of tumour cell lines could be achieved by inhibiting EAG1 expression using antisense oligonucleotides. Ectopic expression of miR-296-3p reduced EAG1 expression and suppressed cell proliferation drug resistance.
Key Molecule: hsa-mir-21 [102]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model D54MG cells Brain Homo sapiens (Human) CVCL_5735
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
TUNEL Analysis
Mechanism Description miR-21 is anti-apoptotic, and may promote glioma invasion and proliferation.
Key Molecule: hsa-mir-200c [103]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell growth Inhibition hsa05200
Cell metastasis Inhibition hsa05205
In Vitro Model U87MG cells Brain Homo sapiens (Human) CVCL_GP63
DBTRG-05MG cells Brain Homo sapiens (Human) CVCL_1169
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Flow cytometry assay
Mechanism Description miR-200c overexpression in DBTRG cells was able to downregulate both PDHA1 and TIGAR, which are involved in OXPHOS and glycolysis regulation. PDHA1 was described as the major provider of carbon for the TCA in GB. miR-200c overexpression was able to decrease DBTRG cell mobility. The observed effect of miR-200c on the mobility of DBTRG cells could be attributed to miR-200c modulation of E-cadherin levels, and consequent repression of epithelial-mesenchymal transition (EMT).
Key Molecule: hsa-mir-144 [104]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
In Vitro Model U87MG cells Brain Homo sapiens (Human) CVCL_GP63
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Colorimetric SRB assay
Mechanism Description The increase of miR-144 levels, shown to be downregulated in U87 and DBTRG human GB cell lines, as well as in GB tumor samples, promoted the downregulation of mRNA of enzymes involved in bioenergetic pathways, with consequent alterations in cell metabolism, impairment of migratory capacity, and sensitization of DBTRG cells to a chemotherapeutic drug, the dichloroacetate (DCA).
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance-associated protein 1 (MRP1) [92]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell viability Inhibition hsa05200
Hedgehog signaling pathway Inhibition hsa04340
MAPK signaling pathway Inhibition hsa04010
p53 signaling pathway Inhibition hsa04115
In Vitro Model LN229 cells Brain Homo sapiens (Human) CVCL_0393
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; RIP assay; Luciferase reporter assay
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description Overexpression of miR-1268a inhibited protein translation of ABCC1, which enhanced sensitivity of GBM cells to TMZ.
Key Molecule: Multidrug resistance protein 1 (ABCB1) [72]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model Glioblastoma tissue .
Experiment for
Molecule Alteration
Real-time PCR
Experiment for
Drug Resistance
Patient survival time
Mechanism Description In the chemosensitive MDR1-negative parental cell line k562 10 ug/ml temozolomide resulted in pronounced cell death with only 47.1% surviving 48 h compared with the control. In contrast, in the highly MDR1-expressing resistant subline k562-VP16, cell death was significantly lower after exposure to temozolomide with 73.4% surviving 48 h (P = 0.002). Addition of a nontoxic dose of the MDR1-modulator cyclosporine A (1 uM) to temozolomide resulted in a trend towards restoration of chemosensitivity in the resistant MDR1-expressing cell line.
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: hsa-mir-26b [105]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SNB19 cells Brain Homo sapiens (Human) CVCL_0535
T98G cells Brain Homo sapiens (Human) CVCL_0556
SNB19 TR cells Brain Homo sapiens (Human) CVCL_0535
T98G TR cells Brain Homo sapiens (Human) CVCL_0556
Experiment for
Molecule Alteration
RT-qPCR
Experiment for
Drug Resistance
MTT Assay; Wound healing assay; Transwell invasion assays
Mechanism Description miR26b reverses temozolomide resistance via targeting Wee1 in glioma cells. miR26b governed TR-mediate EMT partly due to governing its target Wee1.
Key Molecule: Wee1-like protein kinase (WEE1) [105]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SNB19 cells Brain Homo sapiens (Human) CVCL_0535
T98G cells Brain Homo sapiens (Human) CVCL_0556
SNB19 TR cells Brain Homo sapiens (Human) CVCL_0535
T98G TR cells Brain Homo sapiens (Human) CVCL_0556
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT Assay; Wound healing assay; Transwell invasion assays
Mechanism Description miR26b reverses temozolomide resistance via targeting Wee1 in glioma cells. miR26b governed TR-mediate EMT partly due to governing its target Wee1.
Key Molecule: Dynamin-3 (DNM3) [106]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87MG cells Brain Homo sapiens (Human) CVCL_GP63
SHG-44 cells Brain Homo sapiens (Human) CVCL_6728
HEB cells Brain Homo sapiens (Human) N.A.
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Transwell matrigel invasion assay; Scratch wound assay
Mechanism Description Exosomal miR221 targets DNM3 to induce tumor progression and temozolomide resistance in glioma. DNM3 is the target of miR221 and overexpression of DNM3 could reverse the miR221's tumour-promoting effect.
Key Molecule: hsa-mir-221 [106]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87MG cells Brain Homo sapiens (Human) CVCL_GP63
SHG-44 cells Brain Homo sapiens (Human) CVCL_6728
HEB cells Brain Homo sapiens (Human) N.A.
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Transwell matrigel invasion assay; Scratch wound assay
Mechanism Description Exosomal miR221 targets DNM3 to induce tumor progression and temozolomide resistance in glioma. DNM3 is the target of miR221 and overexpression of DNM3 could reverse the miR221's tumour-promoting effect.
Key Molecule: hsa-mir-143 [107]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
MAPK/ERK signaling pathway Inhibition hsa04010
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model BALB/c nude mice Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis; RT-PCR; qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Matrigel assay; Flow cytometry assay
Mechanism Description Overexpression of miR-143 decreased glioma cell migration, invasion, tube formation and slowed tumor growth and angiogenesis in a manner associated with N-RAS downregulation in vitro and in vivo. miR-143 also sensitizes glioma cells to temozolomide (TMZ),the first-line drug for glioma treatment.
Key Molecule: hsa-mir-143 [107]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PI3K/AKT signaling pathway Inhibition hsa04151
MAPK/ERK signaling pathway Inhibition hsa04010
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model BALB/c nude mice Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis; RT-PCR; qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Matrigel assay; Flow cytometry assay
Mechanism Description Overexpression of miR-143 decreased glioma cell migration, invasion, tube formation and slowed tumor growth and angiogenesis in a manner associated with N-RAS downregulation in vitro and in vivo. miR-143 also sensitizes glioma cells to temozolomide (TMZ),the first-line drug for glioma treatment.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Transcription factor SOX-2 (SOX2) [77]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell apoptosis Inhibition hsa04210
Wnt/Beta-catenin signaling pathway Activation hsa04310
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay; Flow cytometry assay
Mechanism Description miR-126-3p sensitizes glioblastoma cells to temozolomide by inactivating Wnt/beta-catenin signaling via targeting SOX2.
Key Molecule: High mobility group protein HMGI-C (HMGA2) [78]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
In Vitro Model U87 GSCs Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description miR-23b overexpression sensitized U87 glioma stem cells to TMZ-induced growth inhibition. And miR-23b had a synergistically suppressive effect on the expression of HMGA2 with TMZ in U87 GSCs.
Key Molecule: Methylated-DNA--protein-cysteine methyltransferase (MGMT) [82], [86]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
A172 cells Brain Homo sapiens (Human) CVCL_0131
T98 cells Brain Homo sapiens (Human) CVCL_B368
U87 cells Brain Homo sapiens (Human) CVCL_0022
U118 cells Brain Homo sapiens (Human) CVCL_0633
U138 cells Brain Homo sapiens (Human) CVCL_0020
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Luciferase reporter assay
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay; TMZ cytotoxicity assay; gamma -H2AX foci formation assay
Mechanism Description miR-198 enhances temozolomide sensitivity in glioblastoma by targeting MGMT.
Key Molecule: Serine/threonine-protein kinase mTOR (mTOR) [53]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
In Vitro Model U87 cells Brain Homo sapiens (Human) CVCL_0022
U257 cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Luciferase reporter assay; Western blot analysis
Experiment for
Drug Resistance
Flow cytometry assay; MTT assay; Transwell assay
Mechanism Description Upregulation of CASC2 sensitized glioma to temozolomide cytotoxicity through autophagy inhibition by sponging miR193a-5p and regulating mTOR expression. mTOR or CASC2 overexpression or miR193a-5p inhibition remarkably reduced autophagy-related proteins expression.
Key Molecule: TNF receptor-associated factor 6 (TRAF6) [80]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AKT/NF-kappaB signaling pathway Inhibition hsa05135
In Vitro Model U251-MG cells Brain Homo sapiens (Human) CVCL_0021
U87-MG cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR 146b 5p suppresses glioblastoma cell resistance to temozolomide through targeting TRAF6. Overexpression of miR 146b 5p or TRAF6 knockdown significantly decreased the level of p AkT and p p65.
Key Molecule: Epidermal growth factor receptor (EGFR) [81]
Sensitive Disease Glioblastoma multiforme [ICD-11: 2A00.03]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation EGFR signaling pathway Inhibition hsa01521
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; Luciferase reporter assay
Experiment for
Drug Resistance
CCK8 assay; Flow cytometric analysis
Mechanism Description miR181b modulates chemosensitivity of glioblastoma multiforme cells to temozolomide by targeting the epidermal growth factor receptor.
Key Molecule: Aurora kinase A (AURKA) [83]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model LN229 cells Brain Homo sapiens (Human) CVCL_0393
A172 cells Brain Homo sapiens (Human) CVCL_0131
T98G cells Brain Homo sapiens (Human) CVCL_0556
M059J cells Brain Homo sapiens (Human) CVCL_0400
M059k cells Brain Homo sapiens (Human) CVCL_0401
U-87 MG cells Brain Homo sapiens (Human) CVCL_0022
U118 MG cells Brain Homo sapiens (Human) CVCL_0633
U138-MG cells Brain Homo sapiens (Human) CVCL_0020
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Dual luciferase assay; Western blot analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR124 suppresses glioblastoma growth and potentiates chemosensitivity by inhibiting AURkA. Re-expression of AURkA rescued miR124-mediated growth suppression.
Key Molecule: Thymidylate synthase (TYMS) [54]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis; Immunofluorescence assay
Experiment for
Drug Resistance
CCK8 assay; Flow cytometric analysis
Mechanism Description LncRNA MALAT1 inhibition re-sensitized TMZ resistant cells through up-regulating miR203 and down-regulating TS expression.
Key Molecule: Phosphatase and tensin homolog (PTEN) [52]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation PTEN signaling pathway Activation hsa05235
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
SNB19 cells Brain Homo sapiens (Human) CVCL_0535
U373 cells Brain Homo sapiens (Human) CVCL_2219
U118 cells Brain Homo sapiens (Human) CVCL_0633
NHA cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; BrdU incorporation assay
Mechanism Description CASC2 up-regulated PTEN protein and down-regulated p-AkT protein through regulating miR181a, and the effect of CASC2 on PTEN and p-AkT could be partially restored by miR181a.
Key Molecule: Phosphatase and tensin homolog (PTEN) [52]
Sensitive Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell growth Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
SNB19 cells Brain Homo sapiens (Human) CVCL_0535
U373 cells Brain Homo sapiens (Human) CVCL_2219
U118 cells Brain Homo sapiens (Human) CVCL_0633
NHA cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; BrdU incorporation assay
Mechanism Description CASC2 up-regulated PTEN protein and down-regulated p-AkT protein through regulating miR181a, and the effect of CASC2 on PTEN and p-AkT could be partially restored by miR181a.
Key Molecule: Cyclic AMP-responsive element-binding protein 1 (CREB1) [84]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN229 cells Brain Homo sapiens (Human) CVCL_0393
U87 cells Brain Homo sapiens (Human) CVCL_0022
SNB19 cells Brain Homo sapiens (Human) CVCL_0535
LN308 cells Brain Homo sapiens (Human) CVCL_0394
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Transwell migration assay; Annexin V/fluorescein isothiocyanate (FITC) apoptosis assay
Mechanism Description miR433-3p suppresses cell growth and enhances chemosensitivity by targeting CREB in human glioma, the overexpression of CREB can rescue the phenotype changes induced by miR433-3p overexpression.
Key Molecule: Glycogen synthase kinase-3 beta (GSK3B) [85]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A172 cells Brain Homo sapiens (Human) CVCL_0131
T98G cells Brain Homo sapiens (Human) CVCL_0556
U251-MG cells Brain Homo sapiens (Human) CVCL_0021
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Colony formation assay
Mechanism Description microRNA-101 reverses temozolomide resistance by inhibition of GSk3beta in glioblastoma.
Key Molecule: Transcription factor E2F1 (E2F1) [87]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell colony Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Down-regulation of miR-196b increased glioma cell sensitivity to TMZ and E2F1 decreased following transfection with miR-196b inhibitors.
Key Molecule: Transcriptional repressor protein YY1 (TYY1) [88]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell colony Inhibition hsa05200
Cell viability Inhibition hsa05200
In Vitro Model LN229 cells Brain Homo sapiens (Human) CVCL_0393
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-7-5p suppresses stemness and enhances temozolomide sensitivity of drug-resistant glioblastoma cells by targeting Yin Yang 1.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [89]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
Cell viability Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87MG cells Brain Homo sapiens (Human) CVCL_GP63
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay; Transwell assay
Mechanism Description Upregulation of miR-181b-5p targets Bcl-2 directly and may function as an important modifier to sensitize glioma cells to TMZ.
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) [90]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
In Vitro Model A172 cells Brain Homo sapiens (Human) CVCL_0131
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description The chemoresistant cell survival mediated with Bcl-2 was inhibited by overexpression of miR-1271 and was enhanced by depletion of miR-1271.
Key Molecule: Golgi phosphoprotein 3 (GOLPH3) [91]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell formation Inhibition hsa05200
Cell invasion Inhibition hsa05200
MAPK/ERK signaling pathway Inhibition hsa04010
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
A172 cells Brain Homo sapiens (Human) CVCL_0131
SNB19 cells Brain Homo sapiens (Human) CVCL_0535
T98G cells Brain Homo sapiens (Human) CVCL_0556
LN308 cells Brain Homo sapiens (Human) CVCL_0394
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 Inhibition of microRNA-299-5p sensitizes glioblastoma cells to temozolomide via upregulating GOLPH3 and inactivating the MAPk/ERk signaling pathway.
Key Molecule: Methylated-DNA--protein-cysteine methyltransferase (MGMT) [62]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Methylation
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell colony Inhibition hsa05200
Cell viability Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; TUNEL assay; Flow cytometry assay
Mechanism Description The endogenous protein level of GSk3beta and MGMT was significantly suppressed by combination of MALAT1 knockdown and miR-101 overexpression and the promoter methylation of MGMT was largely promoted by the combination of MALAT1 knockdown and miR-101 overexpression.
Key Molecule: Transcription factor Sp1 (SP1) [93]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
DNA mismatch repair pathway Regulation hsa03430
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Ectopic expression of miR-29c increased TMZ sensitivity by inhibiting cell growth and promoting apoptosis in U251/TR cells.
Key Molecule: Transcription factor E2F3 (E2F3) [95]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
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 A172 cells Brain Homo sapiens (Human) CVCL_0131
U251-MG cells Brain Homo sapiens (Human) CVCL_0021
U87MG cells Brain Homo sapiens (Human) CVCL_GP63
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTS assay
Mechanism Description miR-203 was reversely associated with migration and invasion, and positively associated with chemosensitivity in glioma cells. E2F3 was shown to be a novel target of miR-203 and E2F3 knockdown exerted a similar effect to that of miR-203 overexpression. These results indicate that miR-203 may act as a tumor suppressor by targeting E2F3 in glioma cells and that miR-203/E2F3 may be a novel candidate for developing rational therapeutic strategies in glioma treatment.
Key Molecule: Ras-related protein Rap-1b (RAP1B) [96]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Expression of Rap1B is negatively regulated by miR-128 and miR-149. TMZ inhibits Rap1B expression by upregulating miR-128 and miR-149. miR-128 and miR-149 suppress cell proliferation and invasion, and alter cytoskeletal remodeling by affecting Rap1B-associated small GTPase. miR-128 and miR-149 increase the chemosensitivity of TMZ in glioblastoma cells.
Key Molecule: NF-kappa-B inhibitor-interacting Ras-like protein 2 (NKIRAS2) [97]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
NF-kappaB signaling pathway Inhibition hsa04064
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN-18 cells Brain Homo sapiens (Human) CVCL_0392
T98G cells Brain Homo sapiens (Human) CVCL_0556
U87-MG cells Brain Homo sapiens (Human) CVCL_0022
HS683 cells Brain Homo sapiens (Human) CVCL_0844
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Promega assay
Mechanism Description A novel mechanism independent of TP53 and MGMT by which oncogenic miR-125b confers TMZ resistance by targeting TNFAIP3 and NkIRAS2. GBM cells overexpressing miR-125b showed increased NF-kB activity and upregulation of anti-apoptotic and cell cycle genes. This was significantly associated with resistance of GBM cells to TNFalpha- and TNF-related inducing ligand-induced apoptosis as well as resistance to TMZ. Conversely, overexpression of anti-miR-125b resulted in cell cycle arrest, increased apoptosis and increased sensitivity to TMZ, indicating that endogenous miR-125b is sufficient to control these processes. GBM cells overexpressing TNFAIP3 and NkIRAS2 were refractory to miR-125b-induced apoptosis resistance as well as TMZ resistance, indicating that both genes are relevant targets of miR-125b.
Key Molecule: Tumor necrosis factor alpha-induced protein 3 (TNFAIP3) [97]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
NF-kappaB signaling pathway Inhibition hsa04064
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
LN-18 cells Brain Homo sapiens (Human) CVCL_0392
T98G cells Brain Homo sapiens (Human) CVCL_0556
U87-MG cells Brain Homo sapiens (Human) CVCL_0022
HS683 cells Brain Homo sapiens (Human) CVCL_0844
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Promega assay
Mechanism Description A novel mechanism independent of TP53 and MGMT by which oncogenic miR-125b confers TMZ resistance by targeting TNFAIP3 and NkIRAS2. GBM cells overexpressing miR-125b showed increased NF-kB activity and upregulation of anti-apoptotic and cell cycle genes. This was significantly associated with resistance of GBM cells to TNFalpha- and TNF-related inducing ligand-induced apoptosis as well as resistance to TMZ. Conversely, overexpression of anti-miR-125b resulted in cell cycle arrest, increased apoptosis and increased sensitivity to TMZ, indicating that endogenous miR-125b is sufficient to control these processes. GBM cells overexpressing TNFAIP3 and NkIRAS2 were refractory to miR-125b-induced apoptosis resistance as well as TMZ resistance, indicating that both genes are relevant targets of miR-125b.
Key Molecule: Ras-related protein Rap-1b (RAP1B) [98]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description Ras-associated protein 1 (Rap1), a growth regulatory protein, belongs to a member of RAS-like small GTP-binding protein superfamily. Rap1 regulates several basic cellular functions: migration, adhesion and growth. TMZ can inhibit the Rap1B expression to exert its cell killing by upregulating miR-181a/b/c/d subunits; conversely, each miR-181a/b/c/d subunit enhanced the chemosensitivity of TMZ in glioblastoma.
Key Molecule: E3 SUMO-protein ligase PIAS3 (PIAS3) [99]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell migration Inhibition hsa04670
In Vitro Model GSCs cells Brain Homo sapiens (Human) N.A.
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Transwell invasion assay
Mechanism Description Inhibition of miR-125b expression may enhance sensitivity of GSCs to temozolomide by targeting PIAS3 on cell invasion.
Key Molecule: Ubiquitin-like modifier-activating enzyme ATG7 (ATG7) [100]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Temozolomide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell autophagy Activation hsa04140
In Vitro Model T98G cells Brain Homo sapiens (Human) CVCL_0556
U373-MG Brain Homo sapiens (Human) CVCL_2219
Experiment for
Molecule Alteration
Immunoblotting analysis
Experiment for
Drug Resistance
Celltiter 96 aqueous one solution cell proliferation assay
Mechanism Description ATG7 is a potential target for miR-17, and this miRNA could negatively regulate ATG7 expression, resulting in a modulation of the autophagic status in T98G glioblastoma cells, the autophagy activation by anti-miR-17 resulted in a decrease of the threshold resistance at temozolomide doses in T98G cells.
Key Molecule: MAPK/ERK kinase 1 (MEK1) [101]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
MAPK signaling pathway Inhibition hsa04010
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-181b independently predicted chemoresponse to temozolomide and enhanced temozolomide sensitivity via MEk1 downregulation.
Key Molecule: Potassium voltage-gated channel subfamily H member 1 (KCNH1) [38]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Temozolomide
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U251AR cells Brain Homo sapiens (Human) CVCL_1G29
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description EAG1 channel might be involved in cell-cycle progression of tumour cells because a significant reduction in the proliferation of tumour cell lines could be achieved by inhibiting EAG1 expression using antisense oligonucleotides. Ectopic expression of miR-296-3p reduced EAG1 expression and suppressed cell proliferation drug resistance.
Teniposide
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-21 [108]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Teniposide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
NF-kappaB signaling pathway Activation hsa04064
In Vitro Model U373 MG cells Brain Homo sapiens (Human) CVCL_2219
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-21 likely contributes to VM-26 resistance through depression of the expression of LRRFIP1, leading to the reduction of the cytotoxicity of chemotherapy drugs through activation of the NF-kB pathway.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Leucine-rich repeat flightless-interacting protein 1 (LRRFIP1) [108]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Teniposide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Activation hsa05200
NF-kappaB signaling pathway Activation hsa04064
In Vitro Model U373 MG cells Brain Homo sapiens (Human) CVCL_2219
Experiment for
Molecule Alteration
Fluorescent reporter assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-21 likely contributes to VM-26 resistance through depression of the expression of LRRFIP1, leading to the reduction of the cytotoxicity of chemotherapy drugs through activation of the NF-kB pathway.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-181 [109]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Teniposide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description MDM2 is a candidate target of miR-181b. MDM2 knockdown mimicked the sensitization effect of miR-181b. Further study revealed that miR-181b binds to the 3'-UTR region of MDM2 leading to the decrease in MDM2 levels and subsequent increase in teniposide sensitivity. Partial restoration of MDM2 attenuated the sensitivity enhancement by miR-181b.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: E3 ubiquitin-protein ligase Mdm2 (MDM2) [109]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Teniposide
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
In Vitro Model U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description MDM2 is a candidate target of miR-181b. MDM2 knockdown mimicked the sensitization effect of miR-181b. Further study revealed that miR-181b binds to the 3'-UTR region of MDM2 leading to the decrease in MDM2 levels and subsequent increase in teniposide sensitivity. Partial restoration of MDM2 attenuated the sensitivity enhancement by miR-181b.
Tretinoin
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Neurofibromin (NF1) [110], [111]
Resistant Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Alteration
.
Resistant Drug Tretinoin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK/RAS signaling pathway Activation hsa04010
In Vitro Model Kelly cells Adrenal Homo sapiens (Human) CVCL_2092
Sk-N-AS cells Adrenal Homo sapiens (Human) CVCL_1700
IMR-5 cells Brain Homo sapiens (Human) CVCL_1306
NBL-S cells Brain Homo sapiens (Human) CVCL_2136
Experiment for
Molecule Alteration
Whole genome sequencing assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description NF1 i.ctivation has been reported in neuroblastoma and confers activation of RAS-MAPk signalling and resistance to retinoic acid.
Vinblastine
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [29]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Resistant Drug Vinblastine
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model U87-MG cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Athymic nu/nu female mice xenograft model Mus musculus
Experiment for
Drug Resistance
MTS assay
Mechanism Description In a cell line expressing a high level of P-glycoprotein, the IC50 of TTI-237 increased 25-fold whereas those of paclitaxel and vincristine increased 806-fold and 925-fold.
Vincristine
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Drug Inactivation by Structure Modification (DISM) Click to Show/Hide
Key Molecule: Glutathione S-transferase P (GSTP1) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Vincristine
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-125b [31]
Resistant Disease Primitive neuroectodermal tumor [ICD-11: 2A00.08]
Molecule Alteration Expression
Up-regulation
Resistant Drug Vincristine
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
miR125b-p53/BAKT signaling pathway Activation hsa05206
In Vitro Model RD-ES cells Bones Homo sapiens (Human) CVCL_2169
Sk-ES cells Bones Homo sapiens (Human) CVCL_0627
Sk-N-MC cells Bones Homo sapiens (Human) CVCL_0530
TC-71 cells Bones Homo sapiens (Human) CVCL_2213
VH-64 cells Bones Homo sapiens (Human) CVCL_9672
WE-68 cells Bones Homo sapiens (Human) CVCL_9717
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Celltiter-glo luminescent cell viability assay
Mechanism Description miR-125b led to the development of chemoresistance by suppressing the expression of p53 and Bak, and repression of miR-125b sensitized EWS cells to apoptosis induced by treatment with various cytotoxic drugs.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [37]
Resistant Disease Ependymoma [ICD-11: 2A00.05]
Molecule Alteration Expression
Up-regulation
Resistant Drug Vincristine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell invasion Activation hsa05200
In Vitro Model BXD-1425EPN cells Embryo Homo sapiens (Human) CVCL_Y105
EPN1 cells Embryo Homo sapiens (Human) N.A.
EPN7 cells Embryo Homo sapiens (Human) N.A.
EPN7R cells Embryo Homo sapiens (Human) N.A.
DKFZ-EP1 cells Embryo Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description ABCB1 gene expression was observed in 4 out of 5 paediatric ependymoma cell lines and increased in stem cell enriched neurospheres. Functional inhibition of ABCB1 using vardenafil or verapamil significantly (p < 0.05-0.001) potentiated the response to three chemotherapeutic drugs (vincristine, etoposide and methotrexate). Both inhibitors were also able to significantly reduce migration (p < 0.001) and invasion (p < 0.001).
Key Molecule: Multidrug resistance protein 1 (ABCB1) [6]
Resistant Disease Anaplastic astrocytoma [ICD-11: 2A00.04]
Molecule Alteration Expression
Up-regulation
Resistant Drug Vincristine
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Protein kinase C signaling pathways Inhibition hsa04310
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
Oncotech EDR assay
Mechanism Description On the other hand, the frequency of LDR that we noted for paclitaxel (20%) and vincristine (20%) was similar to the clinical response rates for these compounds. These data suggest that although MDR1 expression by glial tumors may not be the dominant direct cellular process responsible for tumor resistance to natural products, other mechanisms are present that diminish their activity. The clinical mechanisms of natural product resistance may be a multifactorial function of endothelial expression of MDR1 at the blood-brain barrier in conjunction with glial tumor cell expression of alternative efflux pumps, such as MRP, altered tubulin with lower affinity binding sites, and/or protein kinase C signaling pathways that suppress apoptosis.
Key Molecule: Multidrug resistance protein 1 (ABCB1) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Vincristine
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Bcl-2 homologous antagonist/killer (BAK1) [31]
Resistant Disease Primitive neuroectodermal tumor [ICD-11: 2A00.08]
Molecule Alteration Expression
Down-regulation
Resistant Drug Vincristine
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
miR125b-p53/BAKT signaling pathway Activation hsa05206
In Vitro Model RD-ES cells Bones Homo sapiens (Human) CVCL_2169
Sk-ES cells Bones Homo sapiens (Human) CVCL_0627
Sk-N-MC cells Bones Homo sapiens (Human) CVCL_0530
TC-71 cells Bones Homo sapiens (Human) CVCL_2213
VH-64 cells Bones Homo sapiens (Human) CVCL_9672
WE-68 cells Bones Homo sapiens (Human) CVCL_9717
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Celltiter-glo luminescent cell viability assay
Mechanism Description miR-125b led to the development of chemoresistance by suppressing the expression of p53 and Bak, and repression of miR-125b sensitized EWS cells to apoptosis induced by treatment with various cytotoxic drugs.
Key Molecule: Methylated-DNA--protein-cysteine methyltransferase (MGMT) [6]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug Vincristine
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue .
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [37]
Sensitive Disease Ependymoma [ICD-11: 2A00.05]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Vincristine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell invasion Activation hsa05200
In Vitro Model BXD-1425EPN cells Embryo Homo sapiens (Human) CVCL_Y105
EPN1 cells Embryo Homo sapiens (Human) N.A.
EPN7 cells Embryo Homo sapiens (Human) N.A.
EPN7R cells Embryo Homo sapiens (Human) N.A.
DKFZ-EP1 cells Embryo Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description ABCB1 gene expression was observed in 4 out of 5 paediatric ependymoma cell lines and increased in stem cell enriched neurospheres. Functional inhibition of ABCB1 using vardenafil or verapamil significantly (p < 0.05-0.001) potentiated the response to three chemotherapeutic drugs (vincristine, etoposide and methotrexate). Both inhibitors were also able to significantly reduce migration (p < 0.001) and invasion (p < 0.001).
Key Molecule: Multidrug resistance protein 1 (ABCB1) [37]
Sensitive Disease Ependymoma [ICD-11: 2A00.05]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Vincristine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell invasion Activation hsa05200
In Vitro Model BXD-1425EPN cells Embryo Homo sapiens (Human) CVCL_Y105
EPN1 cells Embryo Homo sapiens (Human) N.A.
EPN7 cells Embryo Homo sapiens (Human) N.A.
EPN7R cells Embryo Homo sapiens (Human) N.A.
DKFZ-EP1 cells Embryo Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description ABCB1 gene expression was observed in 4 out of 5 paediatric ependymoma cell lines and increased in stem cell enriched neurospheres. Functional inhibition of ABCB1 using vardenafil or verapamil significantly (p < 0.05-0.001) potentiated the response to three chemotherapeutic drugs (vincristine, etoposide and methotrexate). Both inhibitors were also able to significantly reduce migration (p < 0.001) and invasion (p < 0.001).
Curcumin
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-326 [112]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Curcumin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation SHH/GLI1 signaling pathway Inhibition hsa05217
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR326 exerts a tumor inhibition effect by decreasing the activity of the SHH/GLI1 pathway. miR326 could target the SMO oncogene to inhibit the biological behaviors and stemness of glioma cells.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Smoothened homolog (SMO) [112]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Curcumin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation SHH/GLI1 signaling pathway Inhibition hsa05217
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR326 exerts a tumor inhibition effect by decreasing the activity of the SHH/GLI1 pathway. miR326 could target the SMO oncogene to inhibit the biological behaviors and stemness of glioma cells.
Dichloroacetate
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-144 [104]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Dichloroacetate
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model DBTRG cells Brain Homo sapiens (Human) CVCL_1169
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
Colorimetric SRB assay
Mechanism Description The potential of miR-144 overexpression to reduce GB cell malignancy, both by decreasing Cell migration and invasion abilities and by sensitizing resistant tumor cells to chemotherapy, paving the way to a novel and more effective GB therapy.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Oxalosuccinate decarboxylase (IDH1) [104]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Dichloroacetate
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model DBTRG cells Brain Homo sapiens (Human) CVCL_1169
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
Colorimetric SRB assay
Mechanism Description The potential of miR-144 overexpression to reduce GB cell malignancy, both by decreasing Cell migration and invasion abilities and by sensitizing resistant tumor cells to chemotherapy, paving the way to a novel and more effective GB therapy.
Key Molecule: Isocitrate dehydrogenase NADP 2 (IDH2) [104]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Dichloroacetate
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model DBTRG cells Brain Homo sapiens (Human) CVCL_1169
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
Colorimetric SRB assay
Mechanism Description The potential of miR-144 overexpression to reduce GB cell malignancy, both by decreasing Cell migration and invasion abilities and by sensitizing resistant tumor cells to chemotherapy, paving the way to a novel and more effective GB therapy.
Key Molecule: phosphoinositide-3-dependent protein kinase 1 (PDPK1) [104]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Dichloroacetate
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model DBTRG cells Brain Homo sapiens (Human) CVCL_1169
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
Colorimetric SRB assay
Mechanism Description The potential of miR-144 overexpression to reduce GB cell malignancy, both by decreasing Cell migration and invasion abilities and by sensitizing resistant tumor cells to chemotherapy, paving the way to a novel and more effective GB therapy.
Key Molecule: Fructose-2,6-bisphosphatase TIGAR (TIGAR) [104]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Dichloroacetate
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model DBTRG cells Brain Homo sapiens (Human) CVCL_1169
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
Colorimetric SRB assay
Mechanism Description The potential of miR-144 overexpression to reduce GB cell malignancy, both by decreasing Cell migration and invasion abilities and by sensitizing resistant tumor cells to chemotherapy, paving the way to a novel and more effective GB therapy.
Clinical Trial Drug(s)
8 drug(s) in total
Click to Show/Hide the Full List of Drugs
Capivasertib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: RAC-alpha serine/threonine-protein kinase (AKT1) [113]
Sensitive Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Sensitive Drug Capivasertib
Experimental Note Identified from the Human Clinical Data
In Vitro Model Brain .
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
Carboxyamidotriazole orotate
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Epidermal growth factor receptor/Protein Mdm4 (EGFR/MDM4) [114]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Structural mutation
Structural variation
Resistant Drug Carboxyamidotriazole orotate
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PI3K signaling pathway Activation hsa04151
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
FISH assay; Whole-exome sequencing assay; Exome sequencing assay
Experiment for
Drug Resistance
Magnetic resonance imaging assay
Mechanism Description Indeed, EGFR and MDM4 FISH analysis of the patient-derived primary GBM cells from the second recurrence showed the presence of DMs, which are known to be resistant to targeted chemotherapies as previously reported.
Enasidenib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: Isocitrate dehydrogenase NADP 2 (IDH2) [115]
Sensitive Disease FGFR-tacc positive glioblastoma [ICD-11: 2A00.01]
Molecule Alteration Missense mutation
p.R172K (c.515G>A)
Sensitive Drug Enasidenib
Experimental Note Identified from the Human Clinical Data
In Vitro Model U87MG cells Brain Homo sapiens (Human) CVCL_GP63
TF-1 cells Bone marrow Homo sapiens (Human) CVCL_0559
TF-1a cells Bone marrow Homo sapiens (Human) CVCL_3608
IDH2 cells N.A. Homo sapiens (Human) N.A.
In Vivo Model NSG mouse PDX model Mus musculus
Mechanism Description Somatic gain-of-function mutations in isocitrate dehydrogenases (IDH) 1 and 2 are found in multiple hematologic and solid tumors, leading to accumulation of the oncometabolite (R)-2-hydroxyglutarate (2HG). 2HG competitively inhibits alpha-ketoglutarate-dependent dioxygenases, including histone demethylases and methylcytosine dioxygenases of the TET family, causing epigenetic dysregulation and a block in cellular differentiation.
Key Molecule: Isocitrate dehydrogenase NADP 2 (IDH2) [115]
Sensitive Disease FGFR-tacc positive glioblastoma [ICD-11: 2A00.01]
Molecule Alteration Missense mutation
p.R140Q (c.419G>A)
Sensitive Drug Enasidenib
Experimental Note Identified from the Human Clinical Data
In Vitro Model U87MG cells Brain Homo sapiens (Human) CVCL_GP63
TF-1 cells Bone marrow Homo sapiens (Human) CVCL_0559
TF-1a cells Bone marrow Homo sapiens (Human) CVCL_3608
IDH2 cells N.A. Homo sapiens (Human) N.A.
In Vivo Model NSG mouse PDX model Mus musculus
Mechanism Description Somatic gain-of-function mutations in isocitrate dehydrogenases (IDH) 1 and 2 are found in multiple hematologic and solid tumors, leading to accumulation of the oncometabolite (R)-2-hydroxyglutarate (2HG). 2HG competitively inhibits alpha-ketoglutarate-dependent dioxygenases, including histone demethylases and methylcytosine dioxygenases of the TET family, causing epigenetic dysregulation and a block in cellular differentiation.
Selumetinib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [116]
Sensitive Disease FGFR-tacc positive glioblastoma [ICD-11: 2A00.01]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Selumetinib
Experimental Note Identified from the Human Clinical Data
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [116]
Sensitive Disease FGFR-tacc positive glioblastoma [ICD-11: 2A00.01]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Selumetinib
Experimental Note Identified from the Human Clinical Data
Beta-lapachone
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-218-2 [117]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug Beta-lapachone
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Inhibition hsa04151
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
U118 cells Brain Homo sapiens (Human) CVCL_0633
HA-1800 cells Brain Homo sapiens (Human) N.A.
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay; Soft agar colony formation assay; Wound-healing analysis
Mechanism Description miR218-2 promotes glioblastomas growth, invasion and drug resistance by targeting CDC27, and the overexpression of CDC27 counteracted the function of miR218-2 in glioma cells. miR218-2 induces glioma malig.ncy by targeting CDC27, which leads to a decrease in the activation of the APC/C biquitin-proteosome pathway, probably downstream of the TGFbeta signaling pathways.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Cell division cycle protein 27 homolog (CDC27) [117]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug Beta-lapachone
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT signaling pathway Inhibition hsa04151
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
U118 cells Brain Homo sapiens (Human) CVCL_0633
HA-1800 cells Brain Homo sapiens (Human) N.A.
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Dual luciferase assay; Western blot analysis; Immunohistochemistry (IHC) assay
Experiment for
Drug Resistance
CCK8 assay; Soft agar colony formation assay; Wound-healing analysis
Mechanism Description miR218-2 promotes glioblastomas growth, invasion and drug resistance by targeting CDC27, and the overexpression of CDC27 counteracted the function of miR218-2 in glioma cells. miR218-2 induces glioma malig.ncy by targeting CDC27, which leads to a decrease in the activation of the APC/C biquitin-proteosome pathway, probably downstream of the TGFbeta signaling pathways.
Patidegib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Smoothened homolog (SMO) [118]
Sensitive Disease Medulloblastoma [ICD-11: 2A00.10]
Molecule Alteration Missense mutation
p.D473H (c.1417G>C)
Sensitive Drug Patidegib
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model Bone marrow .
In Vivo Model Mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
DNA sequencing assay
Mechanism Description The missense mutation p.D473H (c.1417G>C) in gene SMO cause the sensitivity of Patidegib by unusual activation of pro-survival pathway
Ulixertinib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [119]
Sensitive Disease FGFR-tacc positive glioblastoma [ICD-11: 2A00.01]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Ulixertinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation MAPK signaling pathway Inhibition hsa04010
PLX4720
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [120]
Resistant Disease FGFR-tacc positive glioblastoma [ICD-11: 2A00.01]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Resistant Drug PLX4720
Experimental Note Identified from the Human Clinical Data
In Vitro Model GBM cells Brain Homo sapiens (Human) N.A.
In Vivo Model Athymic mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
Alamar blue proliferation assay
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [121]
Sensitive Disease FGFR-tacc positive glioblastoma [ICD-11: 2A00.01]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug PLX4720
Experimental Note Identified from the Human Clinical Data
In Vitro Model LN229 cells Brain Homo sapiens (Human) CVCL_0393
A172 cells Brain Homo sapiens (Human) CVCL_0131
U87 cells Brain Homo sapiens (Human) CVCL_0022
SNB19 cells Brain Homo sapiens (Human) CVCL_0535
U138 cells Brain Homo sapiens (Human) CVCL_0020
T98G cells Brain Homo sapiens (Human) CVCL_0556
HS683 cells Brain Homo sapiens (Human) CVCL_0844
DBTRG-05MG cells Brain Homo sapiens (Human) CVCL_1169
NMC-G1 cells Brain Homo sapiens (Human) CVCL_1608
MO59J cells Brain Homo sapiens (Human) CVCL_0400
LN405 cells Brain Homo sapiens (Human) CVCL_1378
LN172 cells N.A. . N.A.
KG1c cells Brain Homo sapiens (Human) CVCL_2971
H4 cells Brain Homo sapiens (Human) CVCL_1239
GMS10 cells Brain Homo sapiens (Human) CVCL_1233
GAMG cells Brain Homo sapiens (Human) CVCL_1226
CCF-STTG1 cells Brain Homo sapiens (Human) CVCL_1118
AM-38 cells Brain Homo sapiens (Human) CVCL_1070
8MGBA cells Brain Homo sapiens (Human) CVCL_1052
42MGBA cells Brain Homo sapiens (Human) CVCL_1798
In Vivo Model BALB/c nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Mechanism Description PLX4720 suppresses MEK-ERK phosphorylation and cell proliferation in MA cells containing BRAFV600E mutation.
Discontinued Drug(s)
1 drug(s) in total
Click to Show/Hide the Full List of Drugs
Cevipabulin
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [29]
Resistant Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Up-regulation
Resistant Drug Cevipabulin
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model U87-MG cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Athymic nu/nu female mice xenograft model Mus musculus
Experiment for
Drug Resistance
MTS assay
Mechanism Description The compound was a weak substrate of multidrug resistance 1 (multidrug resistance transporter or P-glycoprotein). In a cell line expressing a high level of P-glycoprotein, the IC50 of TTI-237 increased 25-fold whereas those of paclitaxel and vincristine increased 806-fold and 925-fold, respectively.
Preclinical Drug(s)
11 drug(s) in total
Click to Show/Hide the Full List of Drugs
AGI-5198
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Oxalosuccinate decarboxylase (IDH1) [122]
Sensitive Disease FGFR-tacc positive glioblastoma [ICD-11: 2A00.01]
Molecule Alteration Missense mutation
p.R132H (c.395G>A)
Sensitive Drug AGI-5198
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model TS676 cells Brain Homo sapiens (Human) CVCL_A5HX
TS603 cells Brain Homo sapiens (Human) CVCL_A5HW
TS516 cells Brain Homo sapiens (Human) CVCL_A5HY
In Vivo Model SCID mouse xenograft model Mus musculus
Experiment for
Drug Resistance
Soft agar assay
Mechanism Description The missense mutation p.R132H (c.395G>A) in gene IDH1 cause the sensitivity of AGI-5198 by aberration of the drug's therapeutic target
Key Molecule: Oxalosuccinate decarboxylase (IDH1) [122]
Sensitive Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration Missense mutation
p.R132C (c.394C>T)
Sensitive Drug AGI-5198
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model TS676 cells Brain Homo sapiens (Human) CVCL_A5HX
TS603 cells Brain Homo sapiens (Human) CVCL_A5HW
TS516 cells Brain Homo sapiens (Human) CVCL_A5HY
In Vivo Model SCID mouse xenograft model Mus musculus
Experiment for
Drug Resistance
Soft agar assay
Mechanism Description The missense mutation p.R132C (c.394C>T) in gene IDH1 cause the sensitivity of AGI-5198 by aberration of the drug's therapeutic target
Key Molecule: Oxalosuccinate decarboxylase (IDH1) [122]
Sensitive Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration Missense mutation
p.R132H (c.395G>A)
Sensitive Drug AGI-5198
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model TS676 cells Brain Homo sapiens (Human) CVCL_A5HX
TS603 cells Brain Homo sapiens (Human) CVCL_A5HW
TS516 cells Brain Homo sapiens (Human) CVCL_A5HY
In Vivo Model SCID mouse xenograft model Mus musculus
Experiment for
Drug Resistance
Soft agar assay
Mechanism Description The missense mutation p.R132H (c.395G>A) in gene IDH1 cause the sensitivity of AGI-5198 by aberration of the drug's therapeutic target
AZD3463/Doxorubicin
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: ALK tyrosine kinase receptor (ALK) [123]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Missense mutation
p.D1091N (c.3271G>A)
Sensitive Drug AZD3463/Doxorubicin
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PI3K signaling pathway Inhibition hsa04151
In Vitro Model IMR-32 cells Abdomen Homo sapiens (Human) CVCL_0346
SH-SY5Y cells Abdomen Homo sapiens (Human) CVCL_0019
LA-N-6 cells Bone marrow Homo sapiens (Human) CVCL_1363
In Vivo Model Athymic NCR nude mouse PDX model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay; FACS assay; Propidium iodide staining assay; MTT assay
Mechanism Description The novel ALK inhibitor alectinib effectively suppressed cell proliferation and induces apoptosis in NB cell lines with either wild-type ALK or mutated ALK (F1174L and D1091N) by blocking ALK-mediated PI3K/Akt/mTOR signaling. In addition, alectinib enhanced doxorubicin-induced cytotoxicity and apoptosis in NB cells. Furthermore, alectinib induced apoptosis in an orthotopic xenograft NB mouse model. Also, in the TH-MYCN transgenic mouse model, alectinib resulted in decreased tumor growth and prolonged survival time.
BI-2536
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [120]
Sensitive Disease FGFR-tacc positive glioblastoma [ICD-11: 2A00.01]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug BI-2536
Experimental Note Identified from the Human Clinical Data
In Vitro Model GBM cells Brain Homo sapiens (Human) N.A.
In Vivo Model Athymic mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
Alamar blue proliferation assay
BPTES
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Oxalosuccinate decarboxylase (IDH1) [124]
Sensitive Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration Missense mutation
p.R132H (c.395G>A)
Sensitive Drug BPTES
Experimental Note Revealed Based on the Cell Line Data
Experiment for
Drug Resistance
3H-thymidine incorporation assay
Mechanism Description The missense mutation p.R132H (c.395G>A) in gene IDH1 cause the sensitivity of BPTES by unusual activation of pro-survival pathway
Everolimus/Binimetinib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: GTPase Nras (NRAS) [125]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Missense mutation
p.Q61K (c.181C>A)
Sensitive Drug Everolimus/Binimetinib
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PI3K/mTOR signaling pathway Inhibition hsa04151
In Vitro Model Sk-N-AS cells Adrenal Homo sapiens (Human) CVCL_1700
SH-SY5Y cells Abdomen Homo sapiens (Human) CVCL_0019
NGP cells Lung Homo sapiens (Human) CVCL_2141
CHP-212 cells Brain Homo sapiens (Human) CVCL_1125
CHP-134 cells Adrenal gland Homo sapiens (Human) CVCL_1124
Experiment for
Molecule Alteration
Western blotting analysis; PCR
Experiment for
Drug Resistance
Promega assay; FACS assay
Mechanism Description Combination of mTOR and MEK inhibitors synergistically inhibit downstream signaling and cell growth of NRAS mutant cell lines.
Everolimus/PLX4720
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [126]
Sensitive Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Everolimus/PLX4720
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation BRAF/MEK/MAPK signaling pathway Inhibition hsa04010
In Vitro Model DBTRG cells Brain Homo sapiens (Human) CVCL_1169
SF9427 cells N.A. Homo sapiens (Human) N.A.
SF8628 cells Brain Homo sapiens (Human) CVCL_IT46
SF188 cells Brain Homo sapiens (Human) CVCL_6948
BT40 cells N.A. Homo sapiens (Human) N.A.
AM-38 cells Brain Homo sapiens (Human) CVCL_1070
In Vivo Model NSG mouse PDX model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Promega assay
Mechanism Description Combination therapy in BRAFV600E-mutant human glioma cells enhances cell cycle arrest and apoptosis and reduces proliferation compared to monotherapy.
Everolimus/Selumetinib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [126]
Sensitive Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Everolimus/Selumetinib
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation BRAF/MEK/MAPK signaling pathway Inhibition hsa04010
In Vitro Model DBTRG cells Brain Homo sapiens (Human) CVCL_1169
SF9427 cells N.A. Homo sapiens (Human) N.A.
SF8628 cells Brain Homo sapiens (Human) CVCL_IT46
SF188 cells Brain Homo sapiens (Human) CVCL_6948
BT40 cells N.A. Homo sapiens (Human) N.A.
AM-38 cells Brain Homo sapiens (Human) CVCL_1070
In Vivo Model NSG mouse PDX model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Promega assay
Mechanism Description Combination therapy in BRAFV600E-mutant human glioma cells enhances cell cycle arrest and apoptosis and reduces proliferation compared to monotherapy.
hEGFR vIII-CD3 bi-scFv
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Epidermal growth factor receptor (EGFR) [127]
Sensitive Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration Complex-indel
(c.89_889del11867)
Sensitive Drug hEGFR vIII-CD3 bi-scFv
Experimental Note Identified from the Human Clinical Data
In Vitro Model U87-MG cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Drug Resistance
Chromium release assay; FACS assay
IDH1 inhibitors
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Oxalosuccinate decarboxylase (IDH1) [122]
Sensitive Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration Missense mutation
p.R132H (c.395G>A)
Sensitive Drug IDH1 inhibitors
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model TS676 cells Brain Homo sapiens (Human) CVCL_A5HX
TS603 cells Brain Homo sapiens (Human) CVCL_A5HW
TS516 cells Brain Homo sapiens (Human) CVCL_A5HY
In Vivo Model SCID mouse xenograft model Mus musculus
Experiment for
Drug Resistance
Soft agar assay
Mechanism Description The missense mutation p.R132H (c.395G>A) in gene IDH1 cause the sensitivity of IDH1 inhibitors by aberration of the drug's therapeutic target
SF1126
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: GTPase Hras (HRAS) [128]
Sensitive Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration Missense mutation
p.G12V (c.35G>T)
Sensitive Drug SF1126
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model U87MG glioma cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Athymic female (CD-1 Nu/Nu) mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Kinase-Glo luminescent assay
YM-024
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: PI3-kinase alpha (PIK3CA) [129]
Sensitive Disease FGFR-tacc positive glioblastoma [ICD-11: 2A00.01]
Molecule Alteration Missense mutation
p.H1047Y (c.3139C>T)
Sensitive Drug YM-024
Experimental Note Identified from the Human Clinical Data
In Vitro Model SK-MG-26 cell N.A. Homo sapiens (Human) CVCL_D701
SK-MG-17 cells N.A. Homo sapiens (Human) CVCL_8574
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CellTiter96 AQueous assay; Soft-agar colony formation assay
Investigative Drug(s)
15 drug(s) in total
Click to Show/Hide the Full List of Drugs
2-hydroxy-5-fluoropyrimidine
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Key Molecule: L1 cell adhesion molecule (L1CAM) [2]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Resistant Drug 2-hydroxy-5-fluoropyrimidine
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell migration Activation hsa04670
In Vitro Model MDCK cells Kidney Canis lupus familiaris (Dog) (Canis familiaris) CVCL_0422
Experiment for
Molecule Alteration
Puromycin selection and monitored regularly for the maintenance of L1 silencing assay
Experiment for
Drug Resistance
Migration assay
Mechanism Description With OVCAR3 cells treated with anagrelide, 2-hydroxy-5-fluoropyrimidine and mestranol , the gap width closure was seen from 48 h onward at all concentrations tested. Similar results were obtained with U251 cells, and L1's metastatic potential is further evidenced by its promotion of epithelial-mesenchymal transition, endothelial cell transcytosis and resistance to chemo- and radiotherapy.
Apigenin
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-miR-423-5p [130]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Apigenin
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Bax/BCL2/caspase-3 signaling pathway Activation hsa04933
Mitochondrial signaling pathway Regulation hsa04217
In Vitro Model CD133-positive cells Brain Homo sapiens (Human) CVCL_IR55
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
MTT assay; Annexin V/PI apoptosis assay; Caspase-3 activity assay
Mechanism Description Retracted-miR423-5p knockdown enhances the sensitivity of glioma stem cells to apigenin through the mitochondrial pathway.
AZD3463
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: ALK tyrosine kinase receptor (ALK) [131]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Missense mutation
p.F1174L (c.3520T>C)
Sensitive Drug AZD3463
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model IMR-32 cells Abdomen Homo sapiens (Human) CVCL_0346
Sk-N-AS cells Adrenal Homo sapiens (Human) CVCL_1700
SH-SY5Y cells Abdomen Homo sapiens (Human) CVCL_0019
NGP cells Lung Homo sapiens (Human) CVCL_2141
N b-19 cells N.A. Homo sapiens (Human) N.A.
In Vivo Model Orthotopic Mouse Model of NB Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay; Colony formation assay
Key Molecule: ALK tyrosine kinase receptor (ALK) [131]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Missense mutation
p.F1174L (c.3522C>A)
Sensitive Drug AZD3463
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model IMR-32 cells Abdomen Homo sapiens (Human) CVCL_0346
Sk-N-AS cells Adrenal Homo sapiens (Human) CVCL_1700
SH-SY5Y cells Abdomen Homo sapiens (Human) CVCL_0019
NGP cells Lung Homo sapiens (Human) CVCL_2141
N b-19 cells N.A. Homo sapiens (Human) N.A.
In Vivo Model Orthotopic Mouse Model of NB Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay; Colony formation assay
CEP-28122
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: ALK tyrosine kinase receptor (ALK) [132]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Missense mutation
p.F1174L (c.3520T>C)
Sensitive Drug CEP-28122
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.F1174L (c.3520T>C) in gene ALK cause the sensitivity of CEP-28122 by aberration of the drug's therapeutic target
Key Molecule: ALK tyrosine kinase receptor (ALK) [132]
Sensitive Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Missense mutation
p.R1275Q (c.3824G>A)
Sensitive Drug CEP-28122
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.R1275Q (c.3824G>A) in gene ALK cause the sensitivity of CEP-28122 by aberration of the drug's therapeutic target
Cisplatinum
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: HOXD antisense growth-associated long non-coding RNA (HAGLR) [10]
Resistant Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration Up-regulation
Interaction
Resistant Drug Cisplatinum
Experimental Note Identified from the Human Clinical Data
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
SNB19 cells Brain Homo sapiens (Human) CVCL_0535
U373 cells Brain Homo sapiens (Human) CVCL_2219
NHA cells Brain Homo sapiens (Human) N.A.
In Vivo Model BALB/c nude mice model Mus musculus
Experiment for
Molecule Alteration
Knockdown assay; Overexpression assay
Experiment for
Drug Resistance
MTT assay
Mechanism Description Knockdown of LncRNA HOXD-AS1 suppresses proliferation, migration and invasion and enhances cisplatin sensitivity of glioma cells by sponging miR-204.
Cobimetinib/Vemurafenib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Serine/threonine-protein kinase B-raf (BRAF) [133]
Sensitive Disease FGFR-tacc positive glioblastoma [ICD-11: 2A00.01]
Molecule Alteration Missense mutation
p.V600E (c.1799T>A)
Sensitive Drug Cobimetinib/Vemurafenib
Experimental Note Identified from the Human Clinical Data
EGFR TKIs
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Epidermal growth factor receptor (EGFR) [134]
Resistant Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration IF-deletion
p.V30_V336 (c.88_1008)
Resistant Drug EGFR TKIs
Experimental Note Identified from the Human Clinical Data
In Vitro Model Brain .
Mechanism Description The if-deletion p.V30_V336 (c.88_1008) in gene EGFR cause the resistance of EGFR TKIs by aberration of the drug's therapeutic target.
Key Molecule: Epidermal growth factor receptor (EGFR) [135]
Resistant Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration Copy number gain
.
Resistant Drug EGFR TKIs
Experimental Note Identified from the Human Clinical Data
FGFR inhibitors
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: Fibroblast growth factor receptor (FGFR) [136]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Chromosomal translocations
FGFR-TACC gene fusions
Resistant Drug FGFR inhibitors
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation ERK/MAPKsignaling pathway Activation hsa04210
PI3K/AKT signaling pathway Activation hsa04151
STAT3 signaling pathway Activation hsa04550
Experiment for
Molecule Alteration
Sanger sequencing assay
Experiment for
Drug Resistance
Screening assay
Mechanism Description In particular, epidermal growth factor receptor (EGFR) activation has been identified as a mechanism of resistance in bladder cancer cells with FGFR3 mutations after treatment with FGFR inhibitors.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Hepatocyte growth factor receptor (MET) [136]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Mutation
.
Resistant Drug FGFR inhibitors
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation ERK/MAPKsignaling pathway Activation hsa04210
PI3K/AKT signaling pathway Activation hsa04151
STAT3 signaling pathway Activation hsa04550
Experiment for
Molecule Alteration
Sanger sequencing assay
Experiment for
Drug Resistance
Screening assay
Mechanism Description In particular, epidermal growth factor receptor (EGFR) activation has been identified as a mechanism of resistance in bladder cancer cells with FGFR3 mutations after treatment with FGFR inhibitors.
GDP-beta-L-galactose
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Smoothened homolog (SMO) [137]
Resistant Disease Medulloblastoma [ICD-11: 2A00.10]
Molecule Alteration Missense mutation
p.D473H
Resistant Drug GDP-beta-L-galactose
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Hedgehog signaling pathway Activation hsa04340
Experiment for
Molecule Alteration
Deep sequencing assay
Experiment for
Drug Resistance
Fluorescence-activated cell sorting (FACS) analysis
Mechanism Description Molecular profiling of the medulloblastoma patient's primary and metastatic tumor taken before treatment with GDC-0449 revealed an underlying somatic mutation in PTCH1 (PTCH1-W844C) as well as up-regulated expression of Hh pathway target genes, supporting the hypothesis that the tumor was driven by dysregulated Hh signaling. SMO-D473H transfection induced Hh pathway activity to levels comparable with that seen with SMO-WT, demonstrating that SMO-D473H is fully capable of activating Hh signaling.
I-BET151
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: HOX transcript antisense RNA (HOTAIR) [138]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug I-BET151
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model U87Luc cells Brain Homo sapiens (Human) CVCL_5J12
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
Colony forming assay; Flow cytometry assay
Mechanism Description The Bromodomain protein BRD4 controls HOTAIR, a long noncoding RNA essential for glioblastoma proliferation to promote I-bet151 resistance in Glioblastoma.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Bromodomain-containing protein 4 (BRD4) [138]
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Resistant Drug I-BET151
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
In Vitro Model U87Luc cells Brain Homo sapiens (Human) CVCL_5J12
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qPCR
Experiment for
Drug Resistance
Colony forming assay; Flow cytometry assay
Mechanism Description The Bromodomain protein BRD4 controls HOTAIR, a long noncoding RNA essential for glioblastoma proliferation to promote I-bet151 resistance in Glioblastoma.
Isoarnebin 4
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-143 [139]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Up-regulation
Sensitive Drug Isoarnebin 4
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
Mitochondrial apoptotic signaling pathway Activation hsa04210
In Vitro Model GBM cells Brain Homo sapiens (Human) N.A.
In Vivo Model Mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR-143 enhances the antitumor activity of shikonin by targeting BAG3 and reducing its expression in human glioblastoma stem cell.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: BAG family molecular chaperone regulator 3 (BAG3) [139]
Sensitive Disease Glioblastoma [ICD-11: 2A00.02]
Molecule Alteration Expression
Down-regulation
Sensitive Drug Isoarnebin 4
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
Mitochondrial apoptotic signaling pathway Activation hsa04210
In Vitro Model GBM cells Brain Homo sapiens (Human) N.A.
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR-143 enhances the antitumor activity of shikonin by targeting BAG3 and reducing its expression in human glioblastoma stem cell.
Isoliquiritigenin
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: Nuclear paraspeckle assembly transcript 1 (NEAT1) [140]
Resistant Disease Brain glioma [ICD-11: 2A00.0]
Molecule Alteration Down-regulation
Interaction
Resistant Drug Isoliquiritigenin
Experimental Note Discovered Using In-vivo Testing Model
In Vitro Model C6 cells Brain Rattus norvegicus (Rat) CVCL_0194
Experiment for
Molecule Alteration
qRT-PCR; Western bloting analysis; Immunofluorescence assay; ELISA assay; Luciferase assay; Overexpression assay
Mechanism Description LncRNA NEAT1 overexpression reversed ISL-mediated increase in miR-194-5p expression, and thereby attenuated FGF-2, TGF-beta and VEGF production.
NSC141562
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-155 [141]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug NSC141562
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Epithelial mesenchymal transition signaling pathway Activation hsa01521
Wnt/beta-catenin signaling pathway Activation hsa04310
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay; Wound healing assay; Transwell assay; MTT assay
Mechanism Description miR155HG Is a Mesenchymal Transition-Associated Long Noncoding RNA, miR155-5p and miR155-3p Are key Derivatives of MIR155HG. miR155-5p or miR155-3p Targets Protocadherin 9 or 7, Respectively, Protocadherin 9 and 7 Function as Tumor Suppressor Genes by Inhibiting the Wnt/ beta-catenin signaling pathway.
Key Molecule: MIR155 host gene (MIR155HG) [141]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Molecule Alteration Expression
Up-regulation
Resistant Drug NSC141562
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Epithelial mesenchymal transition signaling pathway Activation hsa01521
Wnt/beta-catenin signaling pathway Activation hsa04310
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qPCR; Microarray assay
Experiment for
Drug Resistance
CCK8 assay; Wound healing assay; Transwell assay; MTT assay
Mechanism Description miR155HG Is a Mesenchymal Transition-Associated Long Noncoding RNA, miR155-5p and miR155-3p Are key Derivatives of MIR155HG. miR155-5p or miR155-3p Targets Protocadherin 9 or 7, Respectively, Protocadherin 9 and 7 Function as Tumor Suppressor Genes by Inhibiting the Wnt/ beta-catenin signaling pathway.
NVP-TAE684
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Key Molecule: ALK tyrosine kinase receptor (ALK) [1]
Resistant Disease Neuroblastoma [ICD-11: 2A00.11]
Molecule Alteration Missense mutation
p.F1174L
Resistant Drug NVP-TAE684
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Activation hsa05200
In Vitro Model NBLW cells Brain Homo sapiens (Human) CVCL_VJ90
NBLW-R cells Brain Homo sapiens (Human) CVCL_VJ91
Experiment for
Molecule Alteration
Sangersequencing assay; Targeted deep sequencing assay
Experiment for
Drug Resistance
Array CGH assay
Mechanism Description Analysis of the sensitivity of NBLW and NBLW-R cells to a panel of ALk inhibitors (TAE-684, Crizotinib, Alectinib and Lorlatinib) revealed differences between the paired cell lines, and overall NBLW-R cells with the F1174L mutation were more resistant to ALk inhibitor induced apoptosis compared with NBLW cells.
Temozolomide/Vandetanib
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Oxalosuccinate decarboxylase (IDH1) [142]
Sensitive Disease FGFR-tacc positive glioblastoma [ICD-11: 2A00.01]
Molecule Alteration Missense mutation
p.R132H (c.395G>A)
Sensitive Drug Temozolomide/Vandetanib
Experimental Note Identified from the Human Clinical Data
In Vitro Model Brain .
Experiment for
Molecule Alteration
Multiplex array; Standard ELISA assay
Experiment for
Drug Resistance
Pharmacokinetics analysis
References
Ref 1 Identification of different ALK mutations in a pair of neuroblastoma cell lines established at diagnosis and relapse. Oncotarget. 2016 Dec 27;7(52):87301-87311. doi: 10.18632/oncotarget.13541.
Ref 2 Antagonistic L1 Adhesion Molecule Mimetic Compounds Inhibit Glioblastoma Cell Migration In Vitro .Biomolecules. 2022 Mar 12;12(3):439. doi: 10.3390/biom12030439. 10.3390/biom12030439
Ref 3 5-azacytidine reduces methylation, promotes differentiation and induces tumor regression in a patient-derived IDH1 mutant glioma xenograftOncotarget. 2013 Oct;4(10):1737-47. doi: 10.18632/oncotarget.1408.
Ref 4 Correlation between IDH1 gene mutation status and survival of patients treated for recurrent gliomaAnticancer Res. 2011 Dec;31(12):4457-63.
Ref 5 Brigatinib, an anaplastic lymphoma kinase inhibitor, abrogates activity and growth in ALK-positive neuroblastoma cells, Drosophila and miceOncotarget. 2016 May 17;7(20):29011-22. doi: 10.18632/oncotarget.8508.
Ref 6 In vitro drug response and molecular markers associated with drug resistance in malignant gliomas .Clin Cancer Res. 2006 Aug 1;12(15):4523-32. doi: 10.1158/1078-0432.CCR-05-1830. 10.1158/1078-0432.CCR-05-1830
Ref 7 MiR-21 enhanced glioma cells resistance to carmustine via decreasing Spry2 expression. Eur Rev Med Pharmacol Sci. 2017 Nov;21(22):5065-5071. doi: 10.26355/eurrev_201711_13819.
Ref 8 MicroRNA-221 targeting PI3-K/Akt signaling axis induces cell proliferation and BCNU resistance in human glioblastoma. Neuropathology. 2014 Oct;34(5):455-64. doi: 10.1111/neup.12129. Epub 2014 Apr 30.
Ref 9 Celecoxib reverses the glioblastoma chemo-resistance to temozolomide through mitochondrial metabolism .Aging (Albany NY). 2021 Sep 8;13(17):21268-21282. doi: 10.18632/aging.203443. Epub 2021 Sep 8. 10.18632/aging.203443
Ref 10 Knockdown of lncRNA HOXD-AS1 suppresses proliferation, migration and invasion and enhances cisplatin sensitivity of glioma cells by sponging miR-204. Biomed Pharmacother. 2019 Apr;112:108633. doi: 10.1016/j.biopha.2019.108633. Epub 2019 Feb 20.
Ref 11 Global DNA methylation analysis reveals miR-214-3p contributes to cisplatin resistance in pediatric intracranial nongerminomatous malignant germ cell tumors. Neuro Oncol. 2018 Mar 27;20(4):519-530. doi: 10.1093/neuonc/nox186.
Ref 12 Modulation of chemotherapeutic drug resistance in neuroblastoma SK-N-AS cells by the neural apoptosis inhibitory protein and miR-520f. Int J Cancer. 2015 Apr 1;136(7):1579-88. doi: 10.1002/ijc.29144. Epub 2014 Sep 2.
Ref 13 miR-let-7f-1 regulates SPARC mediated cisplatin resistance in medulloblastoma cells. Cell Signal. 2014 Oct;26(10):2193-201. doi: 10.1016/j.cellsig.2014.06.014. Epub 2014 Jul 8.
Ref 14 Let-7b expression determines response to chemotherapy through the regulation of cyclin D1 in glioblastoma. J Exp Clin Cancer Res. 2013 Jun 27;32(1):41. doi: 10.1186/1756-9966-32-41.
Ref 15 Micro-RNA-21 regulates the sensitivity to cisplatin in human neuroblastoma cells. J Pediatr Surg. 2012 Oct;47(10):1797-805. doi: 10.1016/j.jpedsurg.2012.05.013.
Ref 16 Long non coding RNA MEG3 contributes to cisplatin induced apoptosis via inhibition of autophagy in human glioma cells. Mol Med Rep. 2017 Sep;16(3):2946-2952. doi: 10.3892/mmr.2017.6897. Epub 2017 Jun 30.
Ref 17 Long non-coding RNA AC023115.3 suppresses chemoresistance of glioblastoma by reducing autophagy. Biochim Biophys Acta Mol Cell Res. 2017 Aug;1864(8):1393-1404. doi: 10.1016/j.bbamcr.2017.05.008. Epub 2017 May 9.
Ref 18 miR-186 reverses cisplatin resistance and inhibits the formation of the glioblastoma-initiating cell phenotype by degrading Yin Yang 1 in glioblastoma. Int J Mol Med. 2019 Jan;43(1):517-524. doi: 10.3892/ijmm.2018.3940. Epub 2018 Oct 18.
Ref 19 miR 501 3p sensitizes glioma cells to cisplatin by targeting MYCN. Mol Med Rep. 2018 Nov;18(5):4747-4752. doi: 10.3892/mmr.2018.9458. Epub 2018 Sep 5.
Ref 20 MicroRNA-141 and its associated gene FUS modulate proliferation, migration and cisplatin chemosensitivity in neuroblastoma cell lines. Oncol Rep. 2016 May;35(5):2943-51. doi: 10.3892/or.2016.4640. Epub 2016 Feb 26.
Ref 21 MiR-873 acts as a novel sensitizer of glioma cells to cisplatin by targeting Bcl-2. Int J Oncol. 2015 Oct;47(4):1603-11. doi: 10.3892/ijo.2015.3143. Epub 2015 Aug 31.
Ref 22 Regulation of autophagy by miR-30d impacts sensitivity of anaplastic thyroid carcinoma to cisplatin. Biochem Pharmacol. 2014 Feb 15;87(4):562-70. doi: 10.1016/j.bcp.2013.12.004. Epub 2013 Dec 15.
Ref 23 MicroRNA-204 increases sensitivity of neuroblastoma cells to cisplatin and is associated with a favourable clinical outcome. Br J Cancer. 2012 Sep 4;107(6):967-76. doi: 10.1038/bjc.2012.356. Epub 2012 Aug 14.
Ref 24 miR-34a confers chemosensitivity through modulation of MAGE-A and p53 in medulloblastoma. Neuro Oncol. 2011 Feb;13(2):165-75. doi: 10.1093/neuonc/noq179. Epub 2010 Dec 22.
Ref 25 The neuroblastoma-associated F1174L ALK mutation causes resistance to an ALK kinase inhibitor in ALK-translocated cancers. Cancer Res. 2010 Dec 15;70(24):10038-43. doi: 10.1158/0008-5472.CAN-10-2956. Epub 2010 Oct 28.
Ref 26 miR-129 inhibits tumor growth and potentiates chemosensitivity of neuroblastoma by targeting MYO10. Biomed Pharmacother. 2018 Jul;103:1312-1318. doi: 10.1016/j.biopha.2018.04.153. Epub 2018 May 7.
Ref 27 Concurrent BRAF/MEK Inhibitors in BRAF V600-Mutant High-Grade Primary Brain TumorsJ Natl Compr Canc Netw. 2018 Apr;16(4):343-347. doi: 10.6004/jnccn.2017.7052.
Ref 28 Targeted MAPK Pathway Inhibitors in Patients With Disseminated Pilocytic AstrocytomasJ Natl Compr Canc Netw. 2017 Aug;15(8):978-982. doi: 10.6004/jnccn.2017.0139.
Ref 29 TTI-237: a novel microtubule-active compound with in vivo antitumor activity. Cancer Res. 2008 Apr 1;68(7):2292-300. doi: 10.1158/0008-5472.CAN-07-1420.
Ref 30 PIWI-interacting RNA 39980 promotes tumor progression and reduces drug sensitivity in neuroblastoma cells. J Cell Physiol. 2020 Mar;235(3):2286-2299. doi: 10.1002/jcp.29136. Epub 2019 Sep 3.
Ref 31 miR-125b develops chemoresistance in Ewing sarcoma/primitive neuroectodermal tumor. Cancer Cell Int. 2013 Mar 4;13(1):21. doi: 10.1186/1475-2867-13-21.
Ref 32 Knockdown of microRNA-127 reverses adriamycin resistance via cell cycle arrest and apoptosis sensitization in adriamycin-resistant human glioma cells. Int J Clin Exp Pathol. 2015 Jun 1;8(6):6107-16. eCollection 2015.
Ref 33 Anti-miR21 oligonucleotide enhances chemosensitivity of T98G cell line to doxorubicin by inducing apoptosis. Am J Cancer Res. 2014 Dec 15;5(1):231-42. eCollection 2015.
Ref 34 miR-137 regulates the constitutive androstane receptor and modulates doxorubicin sensitivity in parental and doxorubicin-resistant neuroblastoma cells. Oncogene. 2014 Jul 10;33(28):3717-29. doi: 10.1038/onc.2013.330. Epub 2013 Aug 12.
Ref 35 The forkhead box M1 (FOXM1) expression and antitumor effect of FOXM1 inhibition in malignant rhabdoid tumor .J Cancer Res Clin Oncol. 2021 May;147(5):1499-1518. doi: 10.1007/s00432-020-03438-w. Epub 2020 Nov 21. 10.1007/s00432-020-03438-w
Ref 36 Safety and Antitumor Activity of the Multitargeted Pan-TRK, ROS1, and ALK Inhibitor Entrectinib: Combined Results from Two Phase I Trials (ALKA-372-001 and STARTRK-1)Cancer Discov. 2017 Apr;7(4):400-409. doi: 10.1158/2159-8290.CD-16-1237. Epub 2017 Feb 9.
Ref 37 A role for ABCB1 in prognosis, invasion and drug resistance in ependymoma .Sci Rep. 2019 Jul 16;9(1):10290. doi: 10.1038/s41598-019-46700-z. 10.1038/s41598-019-46700-z
Ref 38 MiR-296-3p regulates cell growth and multi-drug resistance of human glioblastoma by targeting ether-a-go-go (EAG1). Eur J Cancer. 2013 Feb;49(3):710-24. doi: 10.1016/j.ejca.2012.08.020. Epub 2012 Sep 18.
Ref 39 MiR-203 downregulation is responsible for chemoresistance in human glioblastoma by promoting epithelial-mesenchymal transition via SNAI2. Oncotarget. 2015 Apr 20;6(11):8914-28. doi: 10.18632/oncotarget.3563.
Ref 40 Pericytes augment glioblastoma cell resistance to temozolomide through CCL5-CCR5 paracrine signaling .Cell Res. 2021 Oct;31(10):1072-1087. doi: 10.1038/s41422-021-00528-3. Epub 2021 Jul 8. 10.1038/s41422-021-00528-3
Ref 41 Matrine induces apoptosis and autophagy of glioma cell line U251 by regulation of circRNA-104075/BCL-9. Chem Biol Interact. 2019 Aug 1;308:198-205. doi: 10.1016/j.cbi.2019.05.030. Epub 2019 May 18.
Ref 42 Novel mTORC1 Inhibitors Kill Glioblastoma Stem Cells .Pharmaceuticals (Basel). 2020 Nov 24;13(12):419. doi: 10.3390/ph13120419. 10.3390/ph13120419
Ref 43 Immune Checkpoint Inhibition for Hypermutant Glioblastoma Multiforme Resulting From Germline Biallelic Mismatch Repair DeficiencyJ Clin Oncol. 2016 Jul 1;34(19):2206-11. doi: 10.1200/JCO.2016.66.6552. Epub 2016 Mar 21.
Ref 44 miR-34a attenuates glioma cells progression and chemoresistance via targeting PD-L1. Biotechnol Lett. 2017 Oct;39(10):1485-1492. doi: 10.1007/s10529-017-2397-z. Epub 2017 Jul 18.
Ref 45 MicroRNA-21 inhibitor sensitizes human glioblastoma cells U251 (PTEN-mutant) and LN229 (PTEN-wild type) to taxol. BMC Cancer. 2010 Jan 31;10:27. doi: 10.1186/1471-2407-10-27.
Ref 46 Targeting NAD(+) Biosynthesis Overcomes Panobinostat and Bortezomib-Induced Malignant Glioma Resistance .Mol Cancer Res. 2020 Jul;18(7):1004-1017. doi: 10.1158/1541-7786.MCR-19-0669. Epub 2020 Apr 1. 10.1158/1541-7786.MCR-19-0669
Ref 47 Perphenazine and prochlorperazine decrease glioblastoma U-87 MG cell migration and invasion: Analysis of the ABCB1 and ABCG2 transporters, E-cadherin, Alpha-tubulin and integrins (Alpha3, Alpha5, and Beta1) levels .Oncol Lett. 2022 Jun;23(6):182. doi: 10.3892/ol.2022.13302. Epub 2022 Apr 15. 10.3892/ol.2022.13302
Ref 48 Pharmacological and Biophysical Characteristics of Picrotoxin-Resistant, Delta Subunit-Containing GABA(A) Receptors .Front Synaptic Neurosci. 2021 Nov 18;13:763411. doi: 10.3389/fnsyn.2021.763411. eCollection 2021. 10.3389/fnsyn.2021.763411
Ref 49 Rabeprazole has efficacy per se and reduces resistance to temozolomide in glioma via EMT inhibition .Cell Oncol (Dordr). 2021 Aug;44(4):889-905. doi: 10.1007/s13402-021-00609-w. Epub 2021 May 4. 10.1007/s13402-021-00609-w
Ref 50 2-Hydroxyglutarate produced by neomorphic IDH mutations suppresses homologous recombination and induces PARP inhibitor sensitivitySci Transl Med. 2017 Feb 1;9(375):eaal2463. doi: 10.1126/scitranslmed.aal2463.
Ref 51 Lnc-TALC promotes O(6)-methylguanine-DNA methyltransferase expression via regulating the c-Met pathway by competitively binding with miR-20b-3p. Nat Commun. 2019 May 3;10(1):2045. doi: 10.1038/s41467-019-10025-2.
Ref 52 LncRNA CASC2 Interacts With miR-181a to Modulate Glioma Growth and Resistance to TMZ Through PTEN Pathway. J Cell Biochem. 2017 Jul;118(7):1889-1899. doi: 10.1002/jcb.25910. Epub 2017 Feb 13.
Ref 53 Upregulation of CASC2 sensitized glioma to temozolomide cytotoxicity through autophagy inhibition by sponging miR-193a-5p and regulating mTOR expression. Biomed Pharmacother. 2018 Jan;97:844-850. doi: 10.1016/j.biopha.2017.10.146. Epub 2017 Nov 7.
Ref 54 MALAT1 is a prognostic factor in glioblastoma multiforme and induces chemoresistance to temozolomide through suppressing miR-203 and promoting thymidylate synthase expression. Oncotarget. 2017 Apr 4;8(14):22783-22799. doi: 10.18632/oncotarget.15199.
Ref 55 Long Non-Coding RNA MALAT1 Decreases the Sensitivity of Resistant Glioblastoma Cell Lines to Temozolomide. Cell Physiol Biochem. 2017;42(3):1192-1201. doi: 10.1159/000478917. Epub 2017 Jul 3.
Ref 56 MicroRNA-132 induces temozolomide resistance and promotes the formation of cancer stem cell phenotypes by targeting tumor suppressor candidate 3 in glioblastoma. Int J Mol Med. 2017 Nov;40(5):1307-1314. doi: 10.3892/ijmm.2017.3124. Epub 2017 Sep 7.
Ref 57 LncRNA-XIST interacts with miR-29c to modulate the chemoresistance of glioma cell to TMZ through DNA mismatch repair pathway. Biosci Rep. 2017 Sep 7;37(5):BSR20170696. doi: 10.1042/BSR20170696. Print 2017 Oct 31.
Ref 58 MiR-223/PAX6 Axis Regulates Glioblastoma Stem Cell Proliferation and the Chemo Resistance to TMZ via Regulating PI3K/Akt Pathway. J Cell Biochem. 2017 Oct;118(10):3452-3461. doi: 10.1002/jcb.26003. Epub 2017 Jun 6.
Ref 59 Up-regulation of miR-497 confers resistance to temozolomide in human glioma cells by targeting mTOR/Bcl-2. Cancer Med. 2017 Feb;6(2):452-462. doi: 10.1002/cam4.987. Epub 2017 Jan 8.
Ref 60 MicroRNA-195 reverses the resistance to temozolomide through targeting cyclin E1 in glioma cells. Anticancer Drugs. 2019 Jan;30(1):81-88. doi: 10.1097/CAD.0000000000000700.
Ref 61 Exosomal transfer of miR-151a enhances chemosensitivity to temozolomide in drug-resistant glioblastoma. Cancer Lett. 2018 Nov 1;436:10-21. doi: 10.1016/j.canlet.2018.08.004. Epub 2018 Aug 10.
Ref 62 Long noncoding RNA MALAT1 knockdown reverses chemoresistance to temozolomide via promoting microRNA-101 in glioblastoma. Cancer Med. 2018 Apr;7(4):1404-1415. doi: 10.1002/cam4.1384. Epub 2018 Feb 26.
Ref 63 MiR-634 sensitizes glioma cells to temozolomide by targeting CYR61 through Raf-ERK signaling pathway. Cancer Med. 2018 Mar;7(3):913-921. doi: 10.1002/cam4.1351. Epub 2018 Feb 23.
Ref 64 Long non-coding RNA TUSC7 inhibits temozolomide resistance by targeting miR-10a in glioblastoma. Cancer Chemother Pharmacol. 2018 Apr;81(4):671-678. doi: 10.1007/s00280-018-3522-y. Epub 2018 Feb 3.
Ref 65 miR-423-5p contributes to a malignant phenotype and temozolomide chemoresistance in glioblastomas. Neuro Oncol. 2017 Jan;19(1):55-65. doi: 10.1093/neuonc/now129. Epub 2016 Jul 28.
Ref 66 Knockdown of long noncoding RNA H19 sensitizes human glioma cells to temozolomide therapy. Onco Targets Ther. 2016 Jun 13;9:3501-9. doi: 10.2147/OTT.S96278. eCollection 2016.
Ref 67 MicroRNA-138 promotes acquired alkylator resistance in glioblastoma by targeting the Bcl-2-interacting mediator BIM. Oncotarget. 2016 Mar 15;7(11):12937-50. doi: 10.18632/oncotarget.7346.
Ref 68 MiR-16 modulate temozolomide resistance by regulating BCL-2 in human glioma cells. Int J Clin Exp Pathol. 2015 Oct 1;8(10):12698-707. eCollection 2015.
Ref 69 Hypoxia-induced miR-497 decreases glioma cell sensitivity to TMZ by inhibiting apoptosis. FEBS Lett. 2014 Sep 17;588(18):3333-9. doi: 10.1016/j.febslet.2014.07.021. Epub 2014 Jul 29.
Ref 70 MicroRNA-125b-2 confers human glioblastoma stem cells resistance to temozolomide through the mitochondrial pathway of apoptosis. Int J Oncol. 2012 Jan;40(1):119-29. doi: 10.3892/ijo.2011.1179. Epub 2011 Aug 29.
Ref 71 MiR-21 protected human glioblastoma U87MG cells from chemotherapeutic drug temozolomide induced apoptosis by decreasing Bax/Bcl-2 ratio and caspase-3 activity. Brain Res. 2010 Sep 17;1352:255-64. doi: 10.1016/j.brainres.2010.07.009. Epub 2010 Jul 13.
Ref 72 A MDR1 (ABCB1) gene single nucleotide polymorphism predicts outcome of temozolomide treatment in glioblastoma patients. Ann Oncol. 2009 Jan;20(1):175-81. doi: 10.1093/annonc/mdn548. Epub 2008 Aug 7.
Ref 73 Exosomal transfer of long non-coding RNA SBF2-AS1 enhances chemoresistance to temozolomide in glioblastoma. J Exp Clin Cancer Res. 2019 Apr 16;38(1):166. doi: 10.1186/s13046-019-1139-6.
Ref 74 The silencing of LncRNA-H19 decreases chemoresistance of human glioma cells to temozolomide by suppressing epithelial-mesenchymal transition via the Wnt/Beta-Catenin pathway. Onco Targets Ther. 2018 Jan 11;11:313-321. doi: 10.2147/OTT.S154339. eCollection 2018.
Ref 75 Clonal analysis in recurrent astrocytic, oligoastrocytic and oligodendroglial tumors implicates IDH1- mutation as common tumor initiating event. PLoS One. 2012;7(7):e41298. doi: 10.1371/journal.pone.0041298. Epub 2012 Jul 23.
Ref 76 Induction of the unfolded protein response drives enhanced metabolism and chemoresistance in glioma cells. PLoS One. 2013 Aug 15;8(8):e73267. doi: 10.1371/journal.pone.0073267. eCollection 2013.
Ref 77 miR-126-3p sensitizes glioblastoma cells to temozolomide by inactivating Wnt/Beta-catenin signaling via targeting SOX2. Life Sci. 2019 Jun 1;226:98-106. doi: 10.1016/j.lfs.2019.04.023. Epub 2019 Apr 10.
Ref 78 Methylation mediated silencing of miR-23b expression and its role in glioma stem cells. Neurosci Lett. 2012 Oct 24;528(2):185-9. doi: 10.1016/j.neulet.2012.08.055. Epub 2012 Sep 5.
Ref 79 Targeted nanocomplex carrying siRNA against MALAT1 sensitizes glioblastoma to temozolomide. Nucleic Acids Res. 2018 Feb 16;46(3):1424-1440. doi: 10.1093/nar/gkx1221.
Ref 80 miR 146b 5p suppresses glioblastoma cell resistance to temozolomide through targeting TRAF6. Oncol Rep. 2017 Nov;38(5):2941-2950. doi: 10.3892/or.2017.5970. Epub 2017 Sep 19.
Ref 81 MiR-181b modulates chemosensitivity of glioblastoma multiforme cells to temozolomide by targeting the epidermal growth factor receptor. J Neurooncol. 2017 Jul;133(3):477-485. doi: 10.1007/s11060-017-2463-3. Epub 2017 May 13.
Ref 82 MiR-198 enhances temozolomide sensitivity in glioblastoma by targeting MGMT. J Neurooncol. 2017 May;133(1):59-68. doi: 10.1007/s11060-017-2425-9. Epub 2017 Apr 19.
Ref 83 miR-124 suppresses glioblastoma growth and potentiates chemosensitivity by inhibiting AURKA. Biochem Biophys Res Commun. 2017 Apr 22;486(1):43-48. doi: 10.1016/j.bbrc.2017.02.120. Epub 2017 Feb 24.
Ref 84 MiR-433-3p suppresses cell growth and enhances chemosensitivity by targeting CREB in human glioma. Oncotarget. 2017 Jan 17;8(3):5057-5068. doi: 10.18632/oncotarget.13789.
Ref 85 MicroRNA-101 reverses temozolomide resistance by inhibition of GSK3Beta in glioblastoma. Oncotarget. 2016 Nov 29;7(48):79584-79595. doi: 10.18632/oncotarget.12861.
Ref 86 Up-regulation of miR-370-3p restores glioblastoma multiforme sensitivity to temozolomide by influencing MGMT expression. Sci Rep. 2016 Sep 6;6:32972. doi: 10.1038/srep32972.
Ref 87 Downregulation of miR-196b Promotes Glioma Cell Sensitivity to Temozolomide Chemotherapy and Radiotherapy. Ann Clin Lab Sci. 2018 Nov;48(6):719-725.
Ref 88 MiR-7-5p suppresses stemness and enhances temozolomide sensitivity of drug-resistant glioblastoma cells by targeting Yin Yang 1. Exp Cell Res. 2019 Feb 1;375(1):73-81. doi: 10.1016/j.yexcr.2018.12.016. Epub 2018 Dec 23.
Ref 89 MiR-181b-5p modulates chemosensitivity of glioma cells to temozolomide by targeting Bcl-2. Biomed Pharmacother. 2019 Jan;109:2192-2202. doi: 10.1016/j.biopha.2018.11.074. Epub 2018 Nov 27.
Ref 90 Chemo-resistance of A172 glioblastoma cells is controlled by miR-1271-regulated Bcl-2. Biomed Pharmacother. 2018 Dec;108:734-740. doi: 10.1016/j.biopha.2018.08.102. Epub 2018 Sep 21.
Ref 91 Inhibition of microRNA-299-5p sensitizes glioblastoma cells to temozolomide via the MAPK/ERK signaling pathway. Biosci Rep. 2018 Sep 12;38(5):BSR20181051. doi: 10.1042/BSR20181051. Print 2018 Oct 31.
Ref 92 miR-1268a regulates ABCC1 expression to mediate temozolomide resistance in glioblastoma. J Neurooncol. 2018 Jul;138(3):499-508. doi: 10.1007/s11060-018-2835-3. Epub 2018 Jun 6.
Ref 93 miR-29c contribute to glioma cells temozolomide sensitivity by targeting O6-methylguanine-DNA methyltransferases indirectely. Oncotarget. 2016 Aug 2;7(31):50229-50238. doi: 10.18632/oncotarget.10357.
Ref 94 Long noncoding RNA RP11-838N2.4 enhances the cytotoxic effects of temozolomide by inhibiting the functions of miR-10a in glioblastoma cell lines. Oncotarget. 2016 Jul 12;7(28):43835-43851. doi: 10.18632/oncotarget.9699.
Ref 95 MiR-203 sensitizes glioma cells to temozolomide and inhibits glioma cell invasion by targeting E2F3. Mol Med Rep. 2015 Apr;11(4):2838-44. doi: 10.3892/mmr.2014.3101. Epub 2014 Dec 16.
Ref 96 miR-128 and miR-149 enhance the chemosensitivity of temozolomide by Rap1B-mediated cytoskeletal remodeling in glioblastoma. Oncol Rep. 2014 Sep;32(3):957-64. doi: 10.3892/or.2014.3318. Epub 2014 Jul 10.
Ref 97 miR-125b controls apoptosis and temozolomide resistance by targeting TNFAIP3 and NKIRAS2 in glioblastomas. Cell Death Dis. 2014 Jun 5;5(6):e1279. doi: 10.1038/cddis.2014.245.
Ref 98 miR-181 subunits enhance the chemosensitivity of temozolomide by Rap1B-mediated cytoskeleton remodeling in glioblastoma cells. Med Oncol. 2014 Apr;31(4):892. doi: 10.1007/s12032-014-0892-9. Epub 2014 Feb 27.
Ref 99 miR-125b inhibitor may enhance the invasion-prevention activity of temozolomide in glioblastoma stem cells by targeting PIAS3. BioDrugs. 2014 Feb;28(1):41-54. doi: 10.1007/s40259-013-0053-2.
Ref 100 microRNA-17 regulates the expression of ATG7 and modulates the autophagy process, improving the sensitivity to temozolomide and low-dose ionizing radiation treatments in human glioblastoma cells. Cancer Biol Ther. 2013 Jul;14(7):574-86. doi: 10.4161/cbt.24597. Epub 2013 May 10.
Ref 101 miR-181b modulates glioma cell sensitivity to temozolomide by targeting MEK1. Cancer Chemother Pharmacol. 2013 Jul;72(1):147-58. doi: 10.1007/s00280-013-2180-3. Epub 2013 May 5.
Ref 102 MicroRNA-21 inhibition enhances in vitro chemosensitivity of temozolomide-resistant glioblastoma cells. Anticancer Res. 2012 Jul;32(7):2835-41.
Ref 103 MiR-200c-based metabolic modulation in glioblastoma cells as a strategy to overcome tumor chemoresistance. Hum Mol Genet. 2021 Nov 16;30(23):2315-2331. doi: 10.1093/hmg/ddab193.
Ref 104 MiR-144 overexpression as a promising therapeutic strategy to overcome glioblastoma cell invasiveness and resistance to chemotherapy. Hum Mol Genet. 2019 Aug 15;28(16):2738-2751. doi: 10.1093/hmg/ddz099.
Ref 105 MiR-26b reverses temozolomide resistance via targeting Wee1 in glioma cells. Cell Cycle. 2017 Oct 18;16(20):1954-1964. doi: 10.1080/15384101.2017.1367071. Epub 2017 Sep 12.
Ref 106 Exosomal miR-221 targets DNM3 to induce tumor progression and temozolomide resistance in glioma. J Neurooncol. 2017 Jan;131(2):255-265. doi: 10.1007/s11060-016-2308-5. Epub 2016 Nov 11.
Ref 107 Tumour exosomes from cells harbouring PTPRZ1-MET fusion contribute to a malignant phenotype and temozolomide chemoresistance in glioblastoma .Oncogene. 2017 Sep 21;36(38):5369-5381. doi: 10.1038/onc.2017.134. Epub 2017 May 15. 10.1038/onc.2017.134
Ref 108 MicroRNA-21 targets LRRFIP1 and contributes to VM-26 resistance in glioblastoma multiforme. Brain Res. 2009 Aug 25;1286:13-8. doi: 10.1016/j.brainres.2009.06.053. Epub 2009 Jun 24.
Ref 109 MiR-181b sensitizes glioma cells to teniposide by targeting MDM2. BMC Cancer. 2014 Aug 25;14:611. doi: 10.1186/1471-2407-14-611.
Ref 110 Neocortical interneurons: from diversity, strength. Cell. 2010 Jul 23;142(2):189-93. doi: 10.1016/j.cell.2010.07.005.
Ref 111 Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations. Nat Genet. 2015 Aug;47(8):864-71. doi: 10.1038/ng.3333. Epub 2015 Jun 29.
Ref 112 MicroRNA-326 sensitizes human glioblastoma cells to curcumin via the SHH/GLI1 signaling pathway. Cancer Biol Ther. 2018 Apr 3;19(4):260-270. doi: 10.1080/15384047.2016.1250981. Epub 2018 Feb 22.
Ref 113 Durable Control of Metastatic AKT1-Mutant WHO Grade 1 Meningothelial Meningioma by the AKT Inhibitor, AZD5363J Natl Cancer Inst. 2017 Mar 1;109(3):1-4. doi: 10.1093/jnci/djw320.
Ref 114 Longitudinal analysis of treatment-induced genomic alterations in gliomas. Genome Med. 2017 Feb 2;9(1):12. doi: 10.1186/s13073-017-0401-9.
Ref 115 AG-221, a First-in-Class Therapy Targeting Acute Myeloid Leukemia Harboring Oncogenic IDH2 MutationsCancer Discov. 2017 May;7(5):478-493. doi: 10.1158/2159-8290.CD-16-1034. Epub 2017 Feb 13.
Ref 116 Selumetinib in paediatric patients with BRAF-aberrant or neurofibromatosis type 1-associated recurrent, refractory, or progressive low-grade glioma: a multicentre, phase 2 trialLancet Oncol. 2019 Jul;20(7):1011-1022. doi: 10.1016/S1470-2045(19)30277-3. Epub 2019 May 28.
Ref 117 mir-218-2 promotes glioblastomas growth, invasion and drug resistance by targeting CDC27. Oncotarget. 2017 Jan 24;8(4):6304-6318. doi: 10.18632/oncotarget.13850.
Ref 118 Hedgehog pathway inhibitor saridegib (IPI-926) increases lifespan in a mouse medulloblastoma modelProc Natl Acad Sci U S A. 2012 May 15;109(20):7859-64. doi: 10.1073/pnas.1114718109. Epub 2012 May 1.
Ref 119 First-in-Class ERK1/2 Inhibitor Ulixertinib (BVD-523) in Patients with MAPK Mutant Advanced Solid Tumors: Results of a Phase I Dose-Escalation and Expansion StudyCancer Discov. 2018 Feb;8(2):184-195. doi: 10.1158/2159-8290.CD-17-1119. Epub 2017 Dec 15.
Ref 120 Targeting a Plk1-Controlled Polarity Checkpoint in Therapy-Resistant Glioblastoma-Propagating CellsCancer Res. 2015 Dec 15;75(24):5355-66. doi: 10.1158/0008-5472.CAN-14-3689. Epub 2015 Nov 16.
Ref 121 Targeted therapy for BRAFV600E malignant astrocytomaClin Cancer Res. 2011 Dec 15;17(24):7595-604. doi: 10.1158/1078-0432.CCR-11-1456. Epub 2011 Oct 28.
Ref 122 An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cellsScience. 2013 May 3;340(6132):626-30. doi: 10.1126/science.1236062. Epub 2013 Apr 4.
Ref 123 The second-generation ALK inhibitor alectinib effectively induces apoptosis in human neuroblastoma cells and inhibits tumor growth in a TH-MYCN transgenic neuroblastoma mouse modelCancer Lett. 2017 Aug 1;400:61-68. doi: 10.1016/j.canlet.2017.04.022. Epub 2017 Apr 26.
Ref 124 Inhibition of glutaminase preferentially slows growth of glioma cells with mutant IDH1Cancer Res. 2010 Nov 15;70(22):8981-7. doi: 10.1158/0008-5472.CAN-10-1666. Epub 2010 Nov 2.
Ref 125 Targeting the mTOR Complex by Everolimus in NRAS Mutant NeuroblastomaPLoS One. 2016 Jan 28;11(1):e0147682. doi: 10.1371/journal.pone.0147682. eCollection 2016.
Ref 126 BRAF Status in Personalizing Treatment Approaches for Pediatric GliomasClin Cancer Res. 2016 Nov 1;22(21):5312-5321. doi: 10.1158/1078-0432.CCR-15-1101. Epub 2016 May 23.
Ref 127 A Rationally Designed Fully Human EGFRvIII:CD3-Targeted Bispecific Antibody Redirects Human T Cells to Treat Patient-derived Intracerebral Malignant GliomaClin Cancer Res. 2018 Aug 1;24(15):3611-3631. doi: 10.1158/1078-0432.CCR-17-0126. Epub 2018 Apr 27.
Ref 128 Anti-tumor effect of a novel PI3-kinase inhibitor, SF1126, in (12) V-Ha-Ras transgenic mouse glioma modelCancer Cell Int. 2014 Nov 12;14(1):105. doi: 10.1186/s12935-014-0105-9. eCollection 2014.
Ref 129 Targeting class IA PI3K isoforms selectively impairs cell growth, survival, and migration in glioblastomaPLoS One. 2014 Apr 9;9(4):e94132. doi: 10.1371/journal.pone.0094132. eCollection 2014.
Ref 130 miR-423-5p knockdown enhances the sensitivity of glioma stem cells to apigenin through the mitochondrial pathway. Tumour Biol. 2017 Apr;39(4):1010428317695526. doi: 10.1177/1010428317695526.
Ref 131 Novel ALK inhibitor AZD3463 inhibits neuroblastoma growth by overcoming crizotinib resistance and inducing apoptosisSci Rep. 2016 Jan 20;6:19423. doi: 10.1038/srep19423.
Ref 132 CEP-28122, a highly potent and selective orally active inhibitor of anaplastic lymphoma kinase with antitumor activity in experimental models of human cancersMol Cancer Ther. 2012 Mar;11(3):670-9. doi: 10.1158/1535-7163.MCT-11-0776. Epub 2011 Dec 27.
Ref 133 Regression of BRAF (V600E) mutant adult glioblastoma after primary combined BRAF-MEK inhibitor targeted therapy: a report of two casesOncotarget. 2019 Jun 4;10(38):3818-3826. eCollection 2019 Jun 4.
Ref 134 Randomized phase II trial of erlotinib versus temozolomide or carmustine in recurrent glioblastoma: EORTC brain tumor group study 26034J Clin Oncol. 2009 Mar 10;27(8):1268-74. doi: 10.1200/JCO.2008.17.5984. Epub 2009 Feb 9.
Ref 135 Molecular study of malignant gliomas treated with epidermal growth factor receptor inhibitors: tissue analysis from North American Brain Tumor Consortium Trials 01-03 and 00-01Clin Cancer Res. 2005 Nov 1;11(21):7841-50. doi: 10.1158/1078-0432.CCR-05-0421.
Ref 136 FGFR-TACC gene fusions in human glioma. Neuro Oncol. 2017 Apr 1;19(4):475-483. doi: 10.1093/neuonc/now240.
Ref 137 Smoothened mutation confers resistance to a Hedgehog pathway inhibitor in medulloblastoma. Science. 2009 Oct 23;326(5952):572-4. doi: 10.1126/science.1179386. Epub 2009 Sep 2.
Ref 138 The Bromodomain protein BRD4 controls HOTAIR, a long noncoding RNA essential for glioblastoma proliferation. Proc Natl Acad Sci U S A. 2015 Jul 7;112(27):8326-31. doi: 10.1073/pnas.1424220112. Epub 2015 Jun 25.
Ref 139 MiR-143 enhances the antitumor activity of shikonin by targeting BAG3 expression in human glioblastoma stem cells. Biochem Biophys Res Commun. 2015 Dec 4-11;468(1-2):105-12. doi: 10.1016/j.bbrc.2015.10.153. Epub 2015 Nov 2.
Ref 140 Inhibition of COX-2, mPGES-1 and CYP4A by isoliquiritigenin blocks the angiogenic Akt signaling in glioma through ceRNA effect of miR-194-5p and lncRNA NEAT1J Exp Clin Cancer Res. 2019 Aug 22;38(1):371. doi: 10.1186/s13046-019-1361-2.
Ref 141 Blocking MIR155HG/miR-155 axis inhibits mesenchymal transition in glioma. Neuro Oncol. 2017 Sep 1;19(9):1195-1205. doi: 10.1093/neuonc/nox017.
Ref 142 A Multicenter, Phase II, Randomized, Noncomparative Clinical Trial of Radiation and Temozolomide with or without Vandetanib in Newly Diagnosed Glioblastoma PatientsClin Cancer Res. 2015 Aug 15;21(16):3610-8. doi: 10.1158/1078-0432.CCR-14-3220. Epub 2015 Apr 24.

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