General Information of the Molecule (ID: Mol00122)
Name
Serine/threonine-protein kinase mTOR (mTOR) ,Homo sapiens
Synonyms
FK506-binding protein 12-rapamycin complex-associated protein 1; FKBP12-rapamycin complex-associated protein; Mammalian target of rapamycin; mTOR; Mechanistic target of rapamycin; Rapamycin and FKBP12 target 1; Rapamycin target protein 1; FRAP; FRAP1; FRAP2; RAFT1; RAPT1
    Click to Show/Hide
Molecule Type
Protein
Gene Name
MTOR
Gene ID
2475
Location
chr1:11106535-11262551[-]
Sequence
MLGTGPAAATTAATTSSNVSVLQQFASGLKSRNEETRAKAAKELQHYVTMELREMSQEES
TRFYDQLNHHIFELVSSSDANERKGGILAIASLIGVEGGNATRIGRFANYLRNLLPSNDP
VVMEMASKAIGRLAMAGDTFTAEYVEFEVKRALEWLGADRNEGRRHAAVLVLRELAISVP
TFFFQQVQPFFDNIFVAVWDPKQAIREGAVAALRACLILTTQREPKEMQKPQWYRHTFEE
AEKGFDETLAKEKGMNRDDRIHGALLILNELVRISSMEGERLREEMEEITQQQLVHDKYC
KDLMGFGTKPRHITPFTSFQAVQPQQSNALVGLLGYSSHQGLMGFGTSPSPAKSTLVESR
CCRDLMEEKFDQVCQWVLKCRNSKNSLIQMTILNLLPRLAAFRPSAFTDTQYLQDTMNHV
LSCVKKEKERTAAFQALGLLSVAVRSEFKVYLPRVLDIIRAALPPKDFAHKRQKAMQVDA
TVFTCISMLARAMGPGIQQDIKELLEPMLAVGLSPALTAVLYDLSRQIPQLKKDIQDGLL
KMLSLVLMHKPLRHPGMPKGLAHQLASPGLTTLPEASDVGSITLALRTLGSFEFEGHSLT
QFVRHCADHFLNSEHKEIRMEAARTCSRLLTPSIHLISGHAHVVSQTAVQVVADVLSKLL
VVGITDPDPDIRYCVLASLDERFDAHLAQAENLQALFVALNDQVFEIRELAICTVGRLSS
MNPAFVMPFLRKMLIQILTELEHSGIGRIKEQSARMLGHLVSNAPRLIRPYMEPILKALI
LKLKDPDPDPNPGVINNVLATIGELAQVSGLEMRKWVDELFIIIMDMLQDSSLLAKRQVA
LWTLGQLVASTGYVVEPYRKYPTLLEVLLNFLKTEQNQGTRREAIRVLGLLGALDPYKHK
VNIGMIDQSRDASAVSLSESKSSQDSSDYSTSEMLVNMGNLPLDEFYPAVSMVALMRIFR
DQSLSHHHTMVVQAITFIFKSLGLKCVQFLPQVMPTFLNVIRVCDGAIREFLFQQLGMLV
SFVKSHIRPYMDEIVTLMREFWVMNTSIQSTIILLIEQIVVALGGEFKLYLPQLIPHMLR
VFMHDNSPGRIVSIKLLAAIQLFGANLDDYLHLLLPPIVKLFDAPEAPLPSRKAALETVD
RLTESLDFTDYASRIIHPIVRTLDQSPELRSTAMDTLSSLVFQLGKKYQIFIPMVNKVLV
RHRINHQRYDVLICRIVKGYTLADEEEDPLIYQHRMLRSGQGDALASGPVETGPMKKLHV
STINLQKAWGAARRVSKDDWLEWLRRLSLELLKDSSSPSLRSCWALAQAYNPMARDLFNA
AFVSCWSELNEDQQDELIRSIELALTSQDIAEVTQTLLNLAEFMEHSDKGPLPLRDDNGI
VLLGERAAKCRAYAKALHYKELEFQKGPTPAILESLISINNKLQQPEAAAGVLEYAMKHF
GELEIQATWYEKLHEWEDALVAYDKKMDTNKDDPELMLGRMRCLEALGEWGQLHQQCCEK
WTLVNDETQAKMARMAAAAAWGLGQWDSMEEYTCMIPRDTHDGAFYRAVLALHQDLFSLA
QQCIDKARDLLDAELTAMAGESYSRAYGAMVSCHMLSELEEVIQYKLVPERREIIRQIWW
ERLQGCQRIVEDWQKILMVRSLVVSPHEDMRTWLKYASLCGKSGRLALAHKTLVLLLGVD
PSRQLDHPLPTVHPQVTYAYMKNMWKSARKIDAFQHMQHFVQTMQQQAQHAIATEDQQHK
QELHKLMARCFLKLGEWQLNLQGINESTIPKVLQYYSAATEHDRSWYKAWHAWAVMNFEA
VLHYKHQNQARDEKKKLRHASGANITNATTAATTAATATTTASTEGSNSESEAESTENSP
TPSPLQKKVTEDLSKTLLMYTVPAVQGFFRSISLSRGNNLQDTLRVLTLWFDYGHWPDVN
EALVEGVKAIQIDTWLQVIPQLIARIDTPRPLVGRLIHQLLTDIGRYHPQALIYPLTVAS
KSTTTARHNAANKILKNMCEHSNTLVQQAMMVSEELIRVAILWHEMWHEGLEEASRLYFG
ERNVKGMFEVLEPLHAMMERGPQTLKETSFNQAYGRDLMEAQEWCRKYMKSGNVKDLTQA
WDLYYHVFRRISKQLPQLTSLELQYVSPKLLMCRDLELAVPGTYDPNQPIIRIQSIAPSL
QVITSKQRPRKLTLMGSNGHEFVFLLKGHEDLRQDERVMQLFGLVNTLLANDPTSLRKNL
SIQRYAVIPLSTNSGLIGWVPHCDTLHALIRDYREKKKILLNIEHRIMLRMAPDYDHLTL
MQKVEVFEHAVNNTAGDDLAKLLWLKSPSSEVWFDRRTNYTRSLAVMSMVGYILGLGDRH
PSNLMLDRLSGKILHIDFGDCFEVAMTREKFPEKIPFRLTRMLTNAMEVTGLDGNYRITC
HTVMEVLREHKDSVMAVLEAFVYDPLLNWRLMDTNTKGNKRSRTRTDSYSAGQSVEILDG
VELGEPAHKKTGTTVPESIHSFIGDGLVKPEALNKKAIQIINRVRDKLTGRDFSHDDTLD
VPTQVELLIKQATSHENLCQCYIGWCPFW
    Click to Show/Hide
Function
Serine/threonine protein kinase which is a central regulator of cellular metabolism, growth and survival in response to hormones, growth factors, nutrients, energy and stress signals. MTOR directly or indirectly regulates the phosphorylation of at least 800 proteins. Functions as part of 2 structurally and functionally distinct signaling complexes mTORC1 and mTORC2 (mTOR complex 1 and 2). Activated mTORC1 up-regulates protein synthesis by phosphorylating key regulators of mRNA translation and ribosome synthesis. This includes phosphorylation of EIF4EBP1 and release of its inhibition toward the elongation initiation factor 4E (eiF4E). Moreover, phosphorylates and activates RPS6KB1 and RPS6KB2 that promote protein synthesis by modulating the activity of their downstream targets including ribosomal protein S6, eukaryotic translation initiation factor EIF4B, and the inhibitor of translation initiation PDCD4. This also includes mTORC1 signaling cascade controlling the MiT/TFE factors TFEB and TFE3: in the presence of nutrients, mediates phosphorylation of TFEB and TFE3, promoting their cytosolic retention and inactivation. Upon starvation or lysosomal stress, inhibition of mTORC1 induces dephosphorylation and nuclear translocation of TFEB and TFE3, promoting their transcription factor activity. Stimulates the pyrimidine biosynthesis pathway, both by acute regulation through RPS6KB1-mediated phosphorylation of the biosynthetic enzyme CAD, and delayed regulation, through transcriptional enhancement of the pentose phosphate pathway which produces 5-phosphoribosyl-1-pyrophosphate (PRPP), an allosteric activator of CAD at a later step in synthesis, this function is dependent on the mTORC1 complex. Regulates ribosome synthesis by activating RNA polymerase III-dependent transcription through phosphorylation and inhibition of MAF1 an RNA polymerase III-repressor. In parallel to protein synthesis, also regulates lipid synthesis through SREBF1/SREBP1 and LPIN1. To maintain energy homeostasis mTORC1 may also regulate mitochondrial biogenesis through regulation of PPARGC1A. mTORC1 also negatively regulates autophagy through phosphorylation of ULK1. Under nutrient sufficiency, phosphorylates ULK1 at 'Ser-758', disrupting the interaction with AMPK and preventing activation of ULK1. Also prevents autophagy through phosphorylation of the autophagy inhibitor DAP. Also prevents autophagy by phosphorylating RUBCNL/Pacer under nutrient-rich conditions. Prevents autophagy by mediating phosphorylation of AMBRA1, thereby inhibiting AMBRA1 ability to mediate ubiquitination of ULK1 and interaction between AMBRA1 and PPP2CA. mTORC1 exerts a feedback control on upstream growth factor signaling that includes phosphorylation and activation of GRB10 a INSR-dependent signaling suppressor. Among other potential targets mTORC1 may phosphorylate CLIP1 and regulate microtubules. As part of the mTORC2 complex MTOR may regulate other cellular processes including survival and organization of the cytoskeleton. Plays a critical role in the phosphorylation at 'Ser-473' of AKT1, a pro-survival effector of phosphoinositide 3-kinase, facilitating its activation by PDK1. mTORC2 may regulate the actin cytoskeleton, through phosphorylation of PRKCA, PXN and activation of the Rho-type guanine nucleotide exchange factors RHOA and RAC1A or RAC1B. mTORC2 also regulates the phosphorylation of SGK1 at 'Ser-422'. Regulates osteoclastogenesis by adjusting the expression of CEBPB isoforms. Plays an important regulatory role in the circadian clock function; regulates period length and rhythm amplitude of the suprachiasmatic nucleus (SCN) and liver clocks. Phosphorylates SQSTM1, promoting interaction between SQSTM1 and KEAP1 and subsequent inactivation of the BCR(KEAP1) complex.
    Click to Show/Hide
Uniprot ID
MTOR_HUMAN
Ensembl ID
ENSG00000198793
HGNC ID
HGNC:3942
        Click to Show/Hide the Complete Species Lineage
Kingdom: Metazoa
Phylum: Chordata
Class: Mammalia
Order: Primates
Family: Hominidae
Genus: Homo
Species: Homo sapiens
Type(s) of Resistant Mechanism of This Molecule
  ADTT: Aberration of the Drug's Therapeutic Target
  RTDM: Regulation by the Disease Microenvironment
  UAPP: Unusual Activation of Pro-survival Pathway
Drug Resistance Data Categorized by Drug
Approved Drug(s)
10 drug(s) in total
Click to Show/Hide the Full List of Drugs
Cisplatin
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Regulation by the Disease Microenvironment (RTDM) Click to Show/Hide
Disease Class: Lung cancer [1]
Resistant Disease Lung cancer [ICD-11: 2C25.5]
Resistant Drug Cisplatin
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis; qRT-PCR; Luciferase reporter assay
Experiment for
Drug Resistance
CCK8 assay; Flow cytometric analysis
Mechanism Description Cisplatin-resistant lung cancer cell-derived exosomes increase cisplatin resistance of recipient cells in exosomal miR100-5p-dependent manner, and mTOR acts as a target gene of miR100-5p.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Colorectal cancer [2]
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Resistant Drug Cisplatin
Molecule Alteration Expression
Up-regulation
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
Experiment for
Molecule Alteration
Western blot analysis; RT-qPCR
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description miR-1271 enhances the sensitivity of colorectal cancer cells to cisplatin via downregulating mTOP.
Disease Class: Epithelial ovarian cancer [3]
Resistant Disease Epithelial ovarian cancer [ICD-11: 2B5D.0]
Resistant Drug Cisplatin
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
SkOV3/DDP cells Ovary Homo sapiens (Human) CVCL_0532
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Crystal violet staining assay
Mechanism Description miR100 resensitizes resistant epithelial ovarian cancer to cisplatin probably by inhibiting cell proliferation, inducing apoptosis and arresting cell cycle and by targeted downregulation of mTOR and PLk1 expression.
Disease Class: Gastric cancer [4]
Resistant Disease Gastric cancer [ICD-11: 2B72.1]
Resistant Drug Cisplatin
Molecule Alteration Expression
Up-regulation
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
mTOR/HIF-1alpha /P-gp/MRP1 signaling pathway Regulation hsa04150
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
BGC823 cells Gastric Homo sapiens (Human) CVCL_3360
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description Overexpression of long non-coding RNA PVT1 in gastric cancer cells promotes the development of multidrug resistance.PVT-1 was highly expressed in gastric cancer tissues of cisplatin-resistant patients and cisplatin-resistant cells. While, PVT1 overexpression exhibit the anti-apoptotic property in BGC823 and SGC7901 cells transfected with LV-PVT1-GFP and treated with cisplatin. Moreover, qRT-PCR and western blotting revealed that PVT1 up-regulation increased the expression of MDR1, MRP, mTOR and HIF-1alpha. Overexpression of LncRNA PVT1 in gastric carcinoma promotes the development of MDR, suggesting an efficacious target for reversing MDR in gastric cancer therapy.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Cholangiocarcinoma [5]
Sensitive Disease Cholangiocarcinoma [ICD-11: 2C12.0]
Sensitive Drug Cisplatin
Molecule Alteration Expression
Down-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation mTOR signaling pathway Inhibition hsa04150
In Vitro Model GBC-SD cells Gallbladder Homo sapiens (Human) CVCL_6903
RBE cells Liver Homo sapiens (Human) CVCL_4896
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometric analysis
Mechanism Description miR199a-3p enhances cisplatin sensitivity of cholangiocarcinoma cells by inhibiting mTOR signaling pathway and expression of MDR1. miR199a-3p could increase the cisplatin sensitivity of cholangiocarcinoma cell lines by regulating mTOR expression.
Disease Class: Oral squamous cell carcinoma [6]
Sensitive Disease Oral squamous cell carcinoma [ICD-11: 2B6E.0]
Sensitive Drug Cisplatin
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell autophagy Inhibition hsa04140
Cell invasion Inhibition hsa05200
Cell migration Inhibition hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model CAL-27 cells Tongue Homo sapiens (Human) CVCL_1107
KB cells Gastric Homo sapiens (Human) CVCL_0372
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description After HOTAIR silence, autophagy was inhibited with the downregulated expression of MAP1LC3B (microtubule-associated protein 1 light chain 3B), beclin1, and autophagy-related gene (ATG) 3 and ATG7. The expressions of mTOR increased, which promoted the sensitivity to cisplatin.
Disease Class: Gastric cancer [7]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Sensitive Drug Cisplatin
Molecule Alteration Expression
Down-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
mTOR signaling pathway Regulation hsa04150
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Expression
Experiment for
Drug Resistance
WST-1 kit assay
Mechanism Description miR-218 increased chemosensitivity of gastric cancer cells to cisplatin via its target mTOR inhibitor.
Disease Class: Gastric cancer [8]
Sensitive Disease Gastric cancer [ICD-11: 2B72.1]
Sensitive Drug Cisplatin
Molecule Alteration Expression
Down-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
IGF1R/IRS1 signaling pathway Regulation hsa04212
In Vitro Model SGC7901 cells Gastric Homo sapiens (Human) CVCL_0520
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Transwell assay
Mechanism Description Enforced miR-1271 expression repressed the protein levels of its targets, inhibited proliferation of SGC7901/DDP cells, and sensitized SGC7901/DDP cells to DDP-induced apoptosis. Overall, on the basis of the results of our study, we proposed that miR-1271 could regulate cisplatin resistance in human gastric cancer cells, at least partially, via targeting the IGF1R/IRS1 pathway.
Disease Class: Chondrosarcoma [9]
Sensitive Disease Chondrosarcoma [ICD-11: 2B50.0]
Sensitive Drug Cisplatin
Molecule Alteration Expression
Down-regulation
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation mTOR signaling pathway Inhibition hsa04150
In Vitro Model C-28/l2 cells Cartilage Homo sapiens (Human) CVCL_0187
CHON-001 cells Cartilage Homo sapiens (Human) CVCL_C462
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description mTOR is frequently activated in multiple carcinoma. The overexpression of miR-100 significantly down-regulated mTOR proteins and inhibition of miR-100 restored the expression of mTOR in CH-2879 cells, the present studies highlight miR-100 as a tumor suppressor in chondrosarcoma contributing to anti-chemoresistance.
Doxorubicin
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Hepatocellular cancer [10]
Sensitive Disease Hepatocellular cancer [ICD-11: 2C12.4]
Sensitive Drug Doxorubicin
Molecule Alteration Expression
Down-regulation
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
mTOR signaling pathway Inhibition hsa04150
In Vitro Model Huh-7 cells Liver Homo sapiens (Human) CVCL_0336
HepG2 cells Liver Homo sapiens (Human) CVCL_0027
SNU475 cells Liver Homo sapiens (Human) CVCL_0497
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
Cell invasion assay
Mechanism Description The mTOR pathway is activated by multiple extracellular signals, such as growth factors, nutrients, amino acids, hormones, and mitogens leading to the phosphorylation of the translational regulator, phospho-p70S6 kinase, which, in turn, regulates cell proliferation, regulates protein synthesis, and allows progression from the G1 to the S phase of the cell cycle. There is an inverse correlation linking miR-199a-3p and mTOR. miR-199a-3p restoration blocks the G1-S transition of the cell cycle, impairs invasion capability, and sensitizes HCC cells to doxorubicin challenge.
Everolimus
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Disease Class: Thyroid carcinoma [11]
Resistant Disease Thyroid carcinoma [ICD-11: 2D10.4]
Resistant Drug Everolimus
Molecule Alteration Missense mutation
p.F2108L
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation mTOR signaling pathway Activation hsa04150
Experiment for
Molecule Alteration
Whole-exome sequencing assay; Whole-genome sequencing assay
Experiment for
Drug Resistance
Computerized tomography assay
Mechanism Description On the basis of these findings, we hypothesized that mTORF2108L causes resistance to allosteric mTOR inhibition by preventing the binding of the drug to the protein.
Fluorouracil
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Colorectal cancer [12]
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Resistant Drug Fluorouracil
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation AKT/mTOR signaling pathway Activation hsa04150
Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
In Vitro Model SW480 cells Colon Homo sapiens (Human) CVCL_0546
DLD1 cells Colon Homo sapiens (Human) CVCL_0248
SW620 cells Colon Homo sapiens (Human) CVCL_0547
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
LOVO cells Colon Homo sapiens (Human) CVCL_0399
HT-29 cells Colon Homo sapiens (Human) CVCL_0320
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 Overexpressed RP11-708H21.4 suppresses CRC cell proliferation through inducing G1 arrest. Moreover, up-regulation of RP11-708H21.4 inhibits cell migration and invasion, causes cell apoptosis, and enhances 5-FU sensitivity of CRC cells.
Disease Class: Colorectal cancer [13]
Resistant Disease Colorectal cancer [ICD-11: 2B91.1]
Resistant Drug Fluorouracil
Molecule Alteration Expression
Up-regulation
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell viability Activation hsa05200
In Vitro Model HCT116 cells Colon Homo sapiens (Human) CVCL_0291
HCT-8 cells Colon Homo sapiens (Human) CVCL_2478
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; Flow cytometry assay
Mechanism Description The overexpression of PVT1 increased the mRNA and protein expression levels of multidrug resistance associated protein 1, P glycoprotein, serine/threonine protein kinase mTOR and apoptosis regulator Bcl2.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Colon cancer [14]
Sensitive Disease Colon cancer [ICD-11: 2B90.1]
Sensitive Drug Fluorouracil
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation miR338-3p/mTOR signaling pathway Activation hsa05206
In Vitro Model HT29 Cells Colon Homo sapiens (Human) CVCL_A8EZ
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
Experiment for
Molecule Alteration
Western blot analysis; Dual luciferase reporter assay
Experiment for
Drug Resistance
MTT assay; Flow cytometric analysis
Mechanism Description miR338-3p increased 5-FU resistance by reducing the expression of its target gene, mTOR; and miR338-3p inhibitor sensitized HT29 (mutant p53) and HCT116 p53-/- (deficient p53) cells by activating mTOR; and miR338-3p-mTOR-autophagy was in the competition with 5-FU-induced apoptosis and contributed to the subsequent 5-FU resistance. (Inhibition of mTOR induces autophagy and depresses apoptosis to confer resistance to 5-FU).
Disease Class: Nasopharyngeal carcinoma [15]
Sensitive Disease Nasopharyngeal carcinoma [ICD-11: 2B6B.0]
Sensitive Drug Fluorouracil
Molecule Alteration Expression
Down-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell proliferation Inhibition hsa05200
PI3K/AKT/mTOR signaling pathway Inhibition hsa04151
In Vitro Model HNE1 cells Nasopharynx Homo sapiens (Human) CVCL_0308
5-8F cells Nasopharynx Homo sapiens (Human) CVCL_C528
CNE2 cells Nasopharynx Homo sapiens (Human) CVCL_6889
C666-1 cells Throat Homo sapiens (Human) CVCL_7949
CNE1 cells Throat Homo sapiens (Human) CVCL_6888
HONE1 cells Throat Homo sapiens (Human) CVCL_8706
6-10B cells Nasopharynx Homo sapiens (Human) CVCL_C529
SUNE-1 cells Nasopharynx Homo sapiens (Human) CVCL_6946
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay
Mechanism Description miR-3188 regulates nasopharyngeal carcinoma proliferation and chemosensitivity through a FOXO1-modulated positive feedback loop with mTOR-p-PI3k/AkT-c-JUN.
Meclizine
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Glioblastoma multiforme [16]
Sensitive Disease Glioblastoma multiforme [ICD-11: 2A00.03]
Sensitive Drug Meclizine
Molecule Alteration Expression
Down-regulation
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.
Paclitaxel
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Ovarian cancer [17]
Resistant Disease Ovarian cancer [ICD-11: 2C73.0]
Resistant Drug Paclitaxel
Molecule Alteration Missense mutation
p.K1655N
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AXLK signaling pathway Activation hsa01521
In Vitro Model Plasma Blood Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Circulating-free DNA assay; Whole exome sequencing assay
Mechanism Description Quantification of allele fractions in plasma identified increased representation of mutant alleles in association with emergence of therapy resistance.
Disease Class: Ovarian serous carcinoma [17]
Resistant Disease Ovarian serous carcinoma [ICD-11: 2C73.2]
Resistant Drug Paclitaxel
Molecule Alteration Missense mutation
p.K1655N
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
Circulating-free DNA assay; Whole exome sequencing assay
Mechanism Description Quantification of allele fractions in plasma identified increased representation of mutant alleles in association with emergence of therapy resistance.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Breast cancer [18]
Sensitive Disease Breast cancer [ICD-11: 2C60.3]
Sensitive Drug Paclitaxel
Molecule Alteration Expression
Up-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Activation hsa05200
Cell migration Activation hsa04670
Cell proliferation Inhibition hsa05200
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
T47D cells Breast Homo sapiens (Human) CVCL_0553
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
BT549 cells Breast Homo sapiens (Human) CVCL_1092
Hs-578T cells Breast Homo sapiens (Human) CVCL_0332
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-100 expression was significantly downregulated in breast cancer, and the downregulation was more extensive in luminal A breast cancers and was associated with worse patient survival. Ectopic expression of miR-100 sensitized, while inhibition of miR-100 expression desensitized, breast cancer cells to the effect of paclitaxel on cell cycle arrest, multinucleation, apoptosis and tumorigenesis. Expression of genes that are part of a known signature of paclitaxel sensitivity in breast cancer significantly correlated with miR-100 expression. Mechanistically, targeting mTOR appeared to mediate miR-100's function in sensitizing breast cancer cells to paclitaxel, but other mechanisms also seem to be involved, including targeting other molecules such as PLk1.
Propofol
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Cervical cancer [19]
Sensitive Disease Cervical cancer [ICD-11: 2C77.0]
Sensitive Drug Propofol
Molecule Alteration Expression
Down-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell invasion Inhibition hsa05200
Cell proliferation Inhibition hsa05200
mTOR/p70S6k signaling pathway Inhibition hsa04150
In Vitro Model Hela cells Cervix uteri Homo sapiens (Human) CVCL_0030
Caski cells Uterus Homo sapiens (Human) CVCL_1100
C33A cells Uterus Homo sapiens (Human) CVCL_1094
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
MTT assay; Flow cytometry assay
Mechanism Description Propofol promotes cell apoptosis via inhibiting HOTAIR mediated mTOR pathway in cervical cancer.
Sirolimus
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Disease Class: Breast cancer [20]
Resistant Disease Breast cancer [ICD-11: 2C60.3]
Resistant Drug Sirolimus
Molecule Alteration Missense mutation
p.F2108L
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation PIk3CA/AKT/mTOR signaling pathway Activation hsa04211
In Vitro Model MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
Experiment for
Molecule Alteration
Integrated Mutation Profiling of Actionable Cancer Targets assay; Sanger sequencing assay
Experiment for
Drug Resistance
CellTiter-Glo luminescent cell viability assay
Mechanism Description The clinical relevance of these mutations is supported by a case report of a patient who acquired the identical F2108L mTOR mutation after relapse under everolimus treatment.
Temozolomide
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Malignant glioma [21]
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Resistant Drug Temozolomide
Molecule Alteration Expression
Down-regulation
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.
Disease Class: Glioma [22]
Resistant Disease Glioma [ICD-11: 2A00.1]
Resistant Drug Temozolomide
Molecule Alteration Expression
Down-regulation
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.
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Glioma [21]
Sensitive Disease Glioma [ICD-11: 2A00.1]
Sensitive Drug Temozolomide
Molecule Alteration Expression
Up-regulation
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.
Curcumin
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Lung cancer [23]
Sensitive Disease Lung cancer [ICD-11: 2C25.5]
Sensitive Drug Curcumin
Molecule Alteration Phosphorylation
Down-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell proliferation Inhibition hsa05200
Wnt signaling pathway Inhibition hsa04310
mTOR signaling pathway Inhibition hsa04150
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay; BrdU assay; Flow cytometry assay
Mechanism Description Curcumin inhibited Wnt and mTOR pathways through down-regulation of UCA1.
Clinical Trial Drug(s)
2 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
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Melanoma [24]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.H1968Y (c.5902C>T)
Experimental Note Identified from the Human Clinical Data
In Vitro Model HEK 292T cells Kidney Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
LY-294002
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Aberration of the Drug's Therapeutic Target (ADTT) Click to Show/Hide
Disease Class: Melanoma [24]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug LY-294002
Molecule Alteration Missense mutation
p.H1968Y (c.5902C>T)
Experimental Note Identified from the Human Clinical Data
In Vitro Model HEK 292T cells Kidney Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Preclinical Drug(s)
1 drug(s) in total
Click to Show/Hide the Full List of Drugs
LY-294002/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
Disease Class: Melanoma [24]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug LY-294002/Capivasertib
Molecule Alteration Missense mutation
p.P2213S (c.6637C>T)
Experimental Note Identified from the Human Clinical Data
In Vitro Model HEK 292T cells Kidney Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Disease Class: Melanoma [24]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug LY-294002/Capivasertib
Molecule Alteration Missense mutation
p.P2213S (c.6637C>T)
Experimental Note Identified from the Human Clinical Data
In Vitro Model HEK 292T cells Kidney Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
CCK-8 assay
Investigative Drug(s)
1 drug(s) in total
Click to Show/Hide the Full List of Drugs
Celastrol
Click to Show/Hide
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Lung adenocarcinoma [25]
Sensitive Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Sensitive Drug Celastrol
Molecule Alteration Expression
Down-regulation
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Activation hsa04210
Cell viability Inhibition hsa05200
mTOR signaling pathway Inhibition hsa04150
In Vitro Model A549 cells Lung Homo sapiens (Human) CVCL_0023
LTEP-a-2 cells Lung Homo sapiens (Human) CVCL_6929
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 Combination of celastrol and miR-33a-5p increases the expression of miR-33a-5p to inhibit the mTOR signaling pathway.
Disease- and Tissue-specific Abundances of This Molecule
ICD Disease Classification 02
Click to Show/Hide the Resistance Disease of This Class
Brain cancer [ICD-11: 2A00]
Click to Show/Hide
Differential expression of molecule in resistant diseases
The Studied Tissue Nervous tissue
The Specified Disease Brain cancer
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 3.79E-15; Fold-change: -8.92E-02; Z-score: -5.01E-01
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
Disease-specific Molecule Abundances Click to View the Clearer Original Diagram
The Studied Tissue Brainstem tissue
The Specified Disease Glioma
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 6.62E-01; Fold-change: -3.46E-02; Z-score: -2.66E-01
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
Disease-specific Molecule Abundances Click to View the Clearer Original Diagram
The Studied Tissue White matter
The Specified Disease Glioma
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 4.64E-02; Fold-change: -2.79E-01; Z-score: -9.19E-01
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
Disease-specific Molecule Abundances Click to View the Clearer Original Diagram
The Studied Tissue Brainstem tissue
The Specified Disease Neuroectodermal tumor
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 8.18E-01; Fold-change: 1.05E-02; Z-score: 9.24E-02
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
Disease-specific Molecule Abundances Click to View the Clearer Original Diagram
Oral squamous cell carcinoma [ICD-11: 2B6E]
Click to Show/Hide
Differential expression of molecule in resistant diseases
The Studied Tissue Oral tissue
The Specified Disease Oral squamous cell carcinoma
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 1.20E-01; Fold-change: -1.14E-01; Z-score: -4.34E-01
The Expression Level of Disease Section Compare with the Adjacent Tissue p-value: 9.87E-01; Fold-change: -7.21E-03; Z-score: -3.00E-02
Molecule expression in the normal tissue adjacent to the diseased tissue of patients
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
Disease-specific Molecule Abundances Click to View the Clearer Original Diagram
Gastric cancer [ICD-11: 2B72]
Click to Show/Hide
Differential expression of molecule in resistant diseases
The Studied Tissue Gastric tissue
The Specified Disease Gastric cancer
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 3.80E-01; Fold-change: 1.17E-01; Z-score: 8.60E-01
The Expression Level of Disease Section Compare with the Adjacent Tissue p-value: 7.96E-01; Fold-change: 4.28E-04; Z-score: 2.40E-03
Molecule expression in the normal tissue adjacent to the diseased tissue of patients
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
Disease-specific Molecule Abundances Click to View the Clearer Original Diagram
Colon cancer [ICD-11: 2B90]
Click to Show/Hide
Differential expression of molecule in resistant diseases
The Studied Tissue Colon
The Specified Disease Colon cancer
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 5.38E-04; Fold-change: -6.19E-02; Z-score: -3.14E-01
The Expression Level of Disease Section Compare with the Adjacent Tissue p-value: 2.90E-10; Fold-change: -1.34E-01; Z-score: -6.50E-01
Molecule expression in the normal tissue adjacent to the diseased tissue of patients
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
Disease-specific Molecule Abundances Click to View the Clearer Original Diagram
Liver cancer [ICD-11: 2C12]
Click to Show/Hide
Differential expression of molecule in resistant diseases
The Studied Tissue Liver
The Specified Disease Liver cancer
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 4.03E-01; Fold-change: 6.65E-04; Z-score: 2.97E-03
The Expression Level of Disease Section Compare with the Adjacent Tissue p-value: 4.61E-02; Fold-change: 2.81E-06; Z-score: 1.35E-05
The Expression Level of Disease Section Compare with the Other Disease Section p-value: 1.05E-01; Fold-change: 1.50E-01; Z-score: 9.66E-01
Molecule expression in the normal tissue adjacent to the diseased tissue of patients
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
Molecule expression in tissue other than the diseased tissue of patients
Disease-specific Molecule Abundances Click to View the Clearer Original Diagram
Lung cancer [ICD-11: 2C25]
Click to Show/Hide
Differential expression of molecule in resistant diseases
The Studied Tissue Lung
The Specified Disease Lung cancer
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 3.07E-13; Fold-change: 1.68E-01; Z-score: 7.07E-01
The Expression Level of Disease Section Compare with the Adjacent Tissue p-value: 8.18E-15; Fold-change: 1.59E-01; Z-score: 8.75E-01
Molecule expression in the normal tissue adjacent to the diseased tissue of patients
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
Disease-specific Molecule Abundances Click to View the Clearer Original Diagram
Melanoma [ICD-11: 2C30]
Click to Show/Hide
Differential expression of molecule in resistant diseases
The Studied Tissue Skin
The Specified Disease Melanoma
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 4.51E-02; Fold-change: -9.55E-02; Z-score: -4.47E-01
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
Disease-specific Molecule Abundances Click to View the Clearer Original Diagram
Breast cancer [ICD-11: 2C60]
Click to Show/Hide
Differential expression of molecule in resistant diseases
The Studied Tissue Breast tissue
The Specified Disease Breast cancer
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 1.52E-01; Fold-change: 2.53E-02; Z-score: 1.26E-01
The Expression Level of Disease Section Compare with the Adjacent Tissue p-value: 6.20E-01; Fold-change: 1.71E-02; Z-score: 5.63E-02
Molecule expression in the normal tissue adjacent to the diseased tissue of patients
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
Disease-specific Molecule Abundances Click to View the Clearer Original Diagram
Ovarian cancer [ICD-11: 2C73]
Click to Show/Hide
Differential expression of molecule in resistant diseases
The Studied Tissue Ovary
The Specified Disease Ovarian cancer
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 4.89E-03; Fold-change: 3.07E-01; Z-score: 1.24E+00
The Expression Level of Disease Section Compare with the Adjacent Tissue p-value: 6.56E-04; Fold-change: 1.96E-01; Z-score: 1.46E+00
Molecule expression in the normal tissue adjacent to the diseased tissue of patients
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
Disease-specific Molecule Abundances Click to View the Clearer Original Diagram
Cervical cancer [ICD-11: 2C77]
Click to Show/Hide
Differential expression of molecule in resistant diseases
The Studied Tissue Cervix uteri
The Specified Disease Cervical cancer
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 4.94E-01; Fold-change: 6.08E-04; Z-score: 4.03E-03
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
Disease-specific Molecule Abundances Click to View the Clearer Original Diagram
Thyroid cancer [ICD-11: 2D10]
Click to Show/Hide
Differential expression of molecule in resistant diseases
The Studied Tissue Thyroid
The Specified Disease Thyroid cancer
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 2.00E-07; Fold-change: 9.74E-02; Z-score: 5.71E-01
The Expression Level of Disease Section Compare with the Adjacent Tissue p-value: 2.18E-06; Fold-change: 1.26E-01; Z-score: 7.46E-01
Molecule expression in the normal tissue adjacent to the diseased tissue of patients
Molecule expression in the diseased tissue of patients
Molecule expression in the normal tissue of healthy individuals
Disease-specific Molecule Abundances Click to View the Clearer Original Diagram
Tissue-specific Molecule Abundances in Healthy Individuals
Click to Show/Hide the Molecule Abundances
References
Ref 1 Cisplatin-resistant lung cancer cell-derived exosomes increase cisplatin resistance of recipient cells in exosomal miR-100-5p-dependent manner. Int J Nanomedicine. 2017 May 15;12:3721-3733. doi: 10.2147/IJN.S131516. eCollection 2017.
Ref 2 miR-1271 enhances the sensitivity of colorectal cancer cells to cisplatin. Exp Ther Med. 2019 Jun;17(6):4363-4370. doi: 10.3892/etm.2019.7501. Epub 2019 Apr 18.
Ref 3 miR-100 resensitizes resistant epithelial ovarian cancer to cisplatin. Oncol Rep. 2016 Dec;36(6):3552-3558. doi: 10.3892/or.2016.5140. Epub 2016 Oct 3.
Ref 4 Overexpression of long non-coding RNA PVT1 in gastric cancer cells promotes the development of multidrug resistance. Biochem Biophys Res Commun. 2015 Jul 3;462(3):227-32. doi: 10.1016/j.bbrc.2015.04.121. Epub 2015 May 5.
Ref 5 MiR-199a-3p enhances cisplatin sensitivity of cholangiocarcinoma cells by inhibiting mTOR signaling pathway and expression of MDR1. Oncotarget. 2017 May 16;8(20):33621-33630. doi: 10.18632/oncotarget.16834.
Ref 6 RNA interference of long noncoding RNA HOTAIR suppresses autophagy and promotes apoptosis and sensitivity to cisplatin in oral squamous cell carcinoma. J Oral Pathol Med. 2018 Nov;47(10):930-937. doi: 10.1111/jop.12769. Epub 2018 Aug 27.
Ref 7 MicroRNA-218 is upregulated in gastric cancer after cytoreductive surgery and hyperthermic intraperitoneal chemotherapy and increases chemosensitivity to cisplatin. World J Gastroenterol. 2014 Aug 28;20(32):11347-55. doi: 10.3748/wjg.v20.i32.11347.
Ref 8 miR-1271 regulates cisplatin resistance of human gastric cancer cell lines by targeting IGF1R, IRS1, mTOR, and BCL2. Anticancer Agents Med Chem. 2014;14(6):884-91. doi: 10.2174/1871520614666140528161318.
Ref 9 MicroRNA-100 resensitizes resistant chondrosarcoma cells to cisplatin through direct targeting of mTOR. Asian Pac J Cancer Prev. 2014;15(2):917-23. doi: 10.7314/apjcp.2014.15.2.917.
Ref 10 MiR-199a-3p regulates mTOR and c-Met to influence the doxorubicin sensitivity of human hepatocarcinoma cells. Cancer Res. 2010 Jun 15;70(12):5184-93. doi: 10.1158/0008-5472.CAN-10-0145. Epub 2010 May 25.
Ref 11 Response and acquired resistance to everolimus in anaplastic thyroid cancer. N Engl J Med. 2014 Oct 9;371(15):1426-33. doi: 10.1056/NEJMoa1403352.
Ref 12 Down-regulation of long non-coding RNA RP11-708H21.4 is associated with poor prognosis for colorectal cancer and promotes tumorigenesis through regulating AKT/mTOR pathway. Oncotarget. 2017 Apr 25;8(17):27929-27942. doi: 10.18632/oncotarget.15846.
Ref 13 Knockdown of long non coding RNA PVT1 reverses multidrug resistance in colorectal cancer cells. Mol Med Rep. 2018 Jun;17(6):8309-8315. doi: 10.3892/mmr.2018.8907. Epub 2018 Apr 20.
Ref 14 miR-338-3p confers 5-fluorouracil resistance in p53 mutant colon cancer cells by targeting the mammalian target of rapamycin. Exp Cell Res. 2017 Nov 15;360(2):328-336. doi: 10.1016/j.yexcr.2017.09.023. Epub 2017 Sep 18.
Ref 15 miR-3188 regulates nasopharyngeal carcinoma proliferation and chemosensitivity through a FOXO1-modulated positive feedback loop with mTOR-p-PI3K/AKT-c-JUN. Nat Commun. 2016 Apr 20;7:11309. doi: 10.1038/ncomms11309.
Ref 16 Novel mTORC1 Inhibitors Kill Glioblastoma Stem Cells .Pharmaceuticals (Basel). 2020 Nov 24;13(12):419. doi: 10.3390/ph13120419. 10.3390/ph13120419
Ref 17 Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013 May 2;497(7447):108-12. doi: 10.1038/nature12065. Epub 2013 Apr 7.
Ref 18 MicroRNA 100 sensitizes luminal A breast cancer cells to paclitaxel treatment in part by targeting mTOR. Oncotarget. 2016 Feb 2;7(5):5702-14. doi: 10.18632/oncotarget.6790.
Ref 19 Propofol promotes cell apoptosis via inhibiting HOTAIR mediated mTOR pathway in cervical cancer. Biochem Biophys Res Commun. 2015 Dec 25;468(4):561-7. doi: 10.1016/j.bbrc.2015.10.129. Epub 2015 Oct 31.
Ref 20 Overcoming mTOR resistance mutations with a new-generation mTOR inhibitor. Nature. 2016 Jun 9;534(7606):272-6. doi: 10.1038/nature17963. Epub 2016 May 18.
Ref 21 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 22 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 23 Curcumin inhibits proliferation and enhances apoptosis in A549 cells by downregulating lncRNA UCA1. Pharmazie. 2018 Jul 1;73(7):402-407. doi: 10.1691/ph.2018.8402.
Ref 24 Analysis of mTOR Gene Aberrations in Melanoma Patients and Evaluation of Their Sensitivity to PI3K-AKT-mTOR Pathway InhibitorsClin Cancer Res. 2016 Feb 15;22(4):1018-27. doi: 10.1158/1078-0432.CCR-15-1110. Epub 2015 Oct 21.
Ref 25 miR-33a-5p enhances the sensitivity of lung adenocarcinoma cells to celastrol by regulating mTOR signaling. Int J Oncol. 2018 Apr;52(4):1328-1338. doi: 10.3892/ijo.2018.4276. Epub 2018 Feb 14.

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