General Information of the Molecule (ID: Mol00221)
Name
RAC-alpha serine/threonine-protein kinase (AKT1) ,Homo sapiens
Synonyms
Protein kinase B; PKB; Protein kinase B alpha; PKB alpha; Proto-oncogene c-Akt; RAC-PK-alpha; PKB; RAC
    Click to Show/Hide
Molecule Type
Protein
Gene Name
AKT1
Gene ID
207
Location
chr14:104769349-104795751[-]
Sequence
MSDVAIVKEGWLHKRGEYIKTWRPRYFLLKNDGTFIGYKERPQDVDQREAPLNNFSVAQC
QLMKTERPRPNTFIIRCLQWTTVIERTFHVETPEEREEWTTAIQTVADGLKKQEEEEMDF
RSGSPSDNSGAEEMEVSLAKPKHRVTMNEFEYLKLLGKGTFGKVILVKEKATGRYYAMKI
LKKEVIVAKDEVAHTLTENRVLQNSRHPFLTALKYSFQTHDRLCFVMEYANGGELFFHLS
RERVFSEDRARFYGAEIVSALDYLHSEKNVVYRDLKLENLMLDKDGHIKITDFGLCKEGI
KDGATMKTFCGTPEYLAPEVLEDNDYGRAVDWWGLGVVMYEMMCGRLPFYNQDHEKLFEL
ILMEEIRFPRTLGPEAKSLLSGLLKKDPKQRLGGGSEDAKEIMQHRFFAGIVWQHVYEKK
LSPPFKPQVTSETDTRYFDEEFTAQMITITPPDQDDSMECVDSERRPHFPQFSYSASGTA
    Click to Show/Hide
Function
AKT1 is one of 3 closely related serine/threonine-protein kinases (AKT1, AKT2 and AKT3) called the AKT kinase, and which regulate many processes including metabolism, proliferation, cell survival, growth and angiogenesis. This is mediated through serine and/or threonine phosphorylation of a range of downstream substrates. Over 100 substrate candidates have been reported so far, but for most of them, no isoform specificity has been reported. AKT is responsible of the regulation of glucose uptake by mediating insulin-induced translocation of the SLC2A4/GLUT4 glucose transporter to the cell surface. Phosphorylation of PTPN1 at 'Ser-50' negatively modulates its phosphatase activity preventing dephosphorylation of the insulin receptor and the attenuation of insulin signaling. Phosphorylation of TBC1D4 triggers the binding of this effector to inhibitory 14-3-3 proteins, which is required for insulin-stimulated glucose transport. AKT regulates also the storage of glucose in the form of glycogen by phosphorylating GSK3A at 'Ser-21' and GSK3B at 'Ser-9', resulting in inhibition of its kinase activity. Phosphorylation of GSK3 isoforms by AKT is also thought to be one mechanism by which cell proliferation is driven. AKT regulates also cell survival via the phosphorylation of MAP3K5 (apoptosis signal-related kinase). Phosphorylation of 'Ser-83' decreases MAP3K5 kinase activity stimulated by oxidative stress and thereby prevents apoptosis. AKT mediates insulin-stimulated protein synthesis by phosphorylating TSC2 at 'Ser-939' and 'Thr-1462', thereby activating mTORC1 signaling and leading to both phosphorylation of 4E-BP1 and in activation of RPS6KB1. AKT is involved in the phosphorylation of members of the FOXO factors (Forkhead family of transcription factors), leading to binding of 14-3-3 proteins and cytoplasmic localization. In particular, FOXO1 is phosphorylated at 'Thr-24', 'Ser-256' and 'Ser-319'. FOXO3 and FOXO4 are phosphorylated on equivalent sites. AKT has an important role in the regulation of NF-kappa-B-dependent gene transcription and positively regulates the activity of CREB1 (cyclic AMP (cAMP)-response element binding protein). The phosphorylation of CREB1 induces the binding of accessory proteins that are necessary for the transcription of pro-survival genes such as BCL2 and MCL1. AKT phosphorylates 'Ser-454' on ATP citrate lyase (ACLY), thereby potentially regulating ACLY activity and fatty acid synthesis. Activates the 3B isoform of cyclic nucleotide phosphodiesterase (PDE3B) via phosphorylation of 'Ser-273', resulting in reduced cyclic AMP levels and inhibition of lipolysis. Phosphorylates PIKFYVE on 'Ser-318', which results in increased PI(3)P-5 activity. The Rho GTPase-activating protein DLC1 is another substrate and its phosphorylation is implicated in the regulation cell proliferation and cell growth. AKT plays a role as key modulator of the AKT-mTOR signaling pathway controlling the tempo of the process of newborn neurons integration during adult neurogenesis, including correct neuron positioning, dendritic development and synapse formation. Signals downstream of phosphatidylinositol 3-kinase (PI(3)K) to mediate the effects of various growth factors such as platelet-derived growth factor (PDGF), epidermal growth factor (EGF), insulin and insulin-like growth factor I (IGF-I). AKT mediates the antiapoptotic effects of IGF-I. Essential for the SPATA13-mediated regulation of cell migration and adhesion assembly and disassembly. May be involved in the regulation of the placental development. Phosphorylates STK4/MST1 at 'Thr-120' and 'Thr-387' leading to inhibition of its: kinase activity, nuclear translocation, autophosphorylation and ability to phosphorylate FOXO3. Phosphorylates STK3/MST2 at 'Thr-117' and 'Thr-384' leading to inhibition of its: cleavage, kinase activity, autophosphorylation at Thr-180, binding to RASSF1 and nuclear translocation. Phosphorylates SRPK2 and enhances its kinase activity towards SRSF2 and ACIN1 and promotes its nuclear translocation. Phosphorylates RAF1 at 'Ser-259' and negatively regulates its activity. Phosphorylation of BAD stimulates its pro-apoptotic activity. Phosphorylates KAT6A at 'Thr-369' and this phosphorylation inhibits the interaction of KAT6A with PML and negatively regulates its acetylation activity towards p53/TP53. Phosphorylates palladin (PALLD), modulating cytoskeletal organization and cell motility. Phosphorylates prohibitin (PHB), playing an important role in cell metabolism and proliferation. Phosphorylates CDKN1A, for which phosphorylation at 'Thr-145' induces its release from CDK2 and cytoplasmic relocalization. These recent findings indicate that the AKT1 isoform has a more specific role in cell motility and proliferation. Phosphorylates CLK2 thereby controlling cell survival to ionizing radiation. Phosphorylates PCK1 at 'Ser-90', reducing the binding affinity of PCK1 to oxaloacetate and changing PCK1 into an atypical protein kinase activity using GTP as donor. Also acts as an activator of TMEM175 potassium channel activity in response to growth factors: forms the lysoK(GF) complex together with TMEM175 and acts by promoting TMEM175 channel activation, independently of its protein kinase activity.
    Click to Show/Hide
Uniprot ID
AKT1_HUMAN
Ensembl ID
ENSG00000142208
HGNC ID
HGNC:391
        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
  UAPP: Unusual Activation of Pro-survival Pathway
Drug Resistance Data Categorized by Drug
Approved Drug(s)
3 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
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Cervical cancer [1]
Resistant Disease Cervical cancer [ICD-11: 2C77.0]
Resistant Drug Cisplatin
Molecule Alteration Expression
Up-regulation
Experimental Note Identified from the Human Clinical Data
In Vitro Model Hela cells Cervix uteri Homo sapiens (Human) CVCL_0030
Siha cells Cervix uteri Homo sapiens (Human) CVCL_0032
Caski cells Uterus Homo sapiens (Human) CVCL_1100
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description GAS5 knockdown (SiHa/cDDP-GAS5-siRNA) and SiHa cells with miR21 overexpression (SiHa/cDDP-miR21) had an up-regulated level of pAkt.
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 [2]
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 proliferation Activation 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.
Gefitinib
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: Non-small cell lung cancer [3]
Resistant Disease Non-small cell lung cancer [ICD-11: 2C25.Y]
Resistant Drug Gefitinib
Molecule Alteration Mutation
.
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation ERK/MAPKsignaling pathway Activation hsa04210
In Vitro Model NSCLC cells Lung Homo sapiens (Human) N.A.
In Vivo Model A retrospective survey in conducting clinical studies Homo sapiens
Experiment for
Molecule Alteration
Next-generation sequencing assay
Experiment for
Drug Resistance
Liquid biopsy; ATP-binding pocket affinity comparison assay
Mechanism Description Known mechanisms are secondary resistance mutations occurring in the ATP-binding domain (such as T790M and C797S), mutation or amplification of bypass signallings (such as AXL, Hh, ERBb2, CRIPTO, etc), activating mutations in the downstream pathways (PI3k, AkT, MEk, RAF), low levels of mRNA or polymorphisms of the pro-apoptotic protein BIM, induction of a transcription programme for EMT and phenotypical changes, or induction of elevated tumour PD-L1 levels.
Clinical Trial Drug(s)
5 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
Disease Class: Breast adenocarcinoma [4]
Sensitive Disease Breast adenocarcinoma [ICD-11: 2C60.1]
Sensitive Drug Capivasertib
Molecule Alteration Duplication
p.P68_C77 (c.202_231)
Experimental Note Identified from the Human Clinical Data
Mechanism Description The duplication p.P68_C77 (c.202_231) in gene AKT1 cause the sensitivity of Capivasertib by aberration of the drug's therapeutic target.
Disease Class: Brain glioma [5]
Sensitive Disease Brain glioma [ICD-11: 2A00.0]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
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
Disease Class: Solid tumour/cancer [6]
Sensitive Disease Solid tumour/cancer [ICD-11: 2A00-2F9Z]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Capivasertib by aberration of the drug's therapeutic target
Disease Class: Breast adenocarcinoma [7]
Sensitive Disease Breast adenocarcinoma [ICD-11: 2C60.1]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Capivasertib by aberration of the drug's therapeutic target
Disease Class: Endometrial adenocarcinoma [7]
Sensitive Disease Endometrial adenocarcinoma [ICD-11: 2C76.0]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Capivasertib by aberration of the drug's therapeutic target
Disease Class: Endometrial adenocarcinoma [8]
Sensitive Disease Endometrial adenocarcinoma [ICD-11: 2C76.0]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Breast .
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Capivasertib by aberration of the drug's therapeutic target
Disease Class: HER2 negative breast cancer [8]
Sensitive Disease HER2 negative breast cancer [ICD-11: 2C60.11]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Breast .
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Capivasertib by aberration of the drug's therapeutic target
Disease Class: Parotid gland cancer [8]
Sensitive Disease Parotid gland cancer [ICD-11: 2B67.0]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Breast .
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Capivasertib by aberration of the drug's therapeutic target
Disease Class: Leiomyosarcoma [8]
Sensitive Disease Leiomyosarcoma [ICD-11: 2B58.0]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Breast .
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Capivasertib by aberration of the drug's therapeutic target
Disease Class: HER2 negative breast cancer [8]
Sensitive Disease HER2 negative breast cancer [ICD-11: 2C60.11]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Breast .
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Capivasertib by aberration of the drug's therapeutic target
Disease Class: Cervical cancer [9]
Sensitive Disease Cervical cancer [ICD-11: 2C77.0]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Breast .
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Capivasertib by aberration of the drug's therapeutic target
Disease Class: ER positive breast cancer [9]
Sensitive Disease ER positive breast cancer [ICD-11: 2C60.6]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Breast .
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Capivasertib by aberration of the drug's therapeutic target
Disease Class: Granulosa cell tumor [9]
Sensitive Disease Granulosa cell tumor [ICD-11: 2F76.0]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Breast .
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Capivasertib by aberration of the drug's therapeutic target
Disease Class: Lung adenocarcinoma [9]
Sensitive Disease Lung adenocarcinoma [ICD-11: 2C25.0]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Breast .
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Capivasertib by aberration of the drug's therapeutic target
Disease Class: Ovarian cancer [9]
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.Q79K (c.235C>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Breast .
Mechanism Description The missense mutation p.Q79K (c.235C>A) in gene AKT1 cause the sensitivity of Capivasertib by aberration of the drug's therapeutic target
Disease Class: ER negative breast cancer [9]
Sensitive Disease ER negative breast cancer [ICD-11: 2C60.7]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model Breast .
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Capivasertib by aberration of the drug's therapeutic target
Disease Class: Breast adenocarcinoma [10]
Sensitive Disease Breast adenocarcinoma [ICD-11: 2C60.1]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
In Vitro Model MCF10A cells Breast Homo sapiens (Human) CVCL_0598
In Vivo Model Male nude mouse (nu/nu:Alpk) xenograft model Mus musculus
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
Disease Class: Ovarian cancer [7]
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Sensitive Drug Capivasertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
Experiment for
Molecule Alteration
Whole-exome sequencing
Ipatasertib
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: HER2 negative breast cancer [11]
Sensitive Disease HER2 negative breast cancer [ICD-11: 2C60.11]
Sensitive Drug Ipatasertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
Cell Pathway Regulation AKT signaling pathway Inhibition hsa04151
In Vivo Model SCID beige mouse PDX model Mus musculus
Experiment for
Drug Resistance
CellTitre-Glo assay; Wallac multilabel reader assay
Mechanism Description Ipatasertib (GDC-0068) is a novel selective ATP-competitive small-molecule inhibitor of AKT that preferentially targets active phosphorylated AKT (pAKT) and is potent in cell lines with evidence of AKT activation.
Miransertib
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: Ovarian cancer [12]
Sensitive Disease Ovarian cancer [ICD-11: 2C73.0]
Sensitive Drug Miransertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Miransertib by aberration of the drug's therapeutic target
Disease Class: Endometrial adenocarcinoma [13]
Sensitive Disease Endometrial adenocarcinoma [ICD-11: 2C76.0]
Sensitive Drug Miransertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model DU-145 cells Prostate Homo sapiens (Human) CVCL_0105
LNCaP cells Prostate Homo sapiens (Human) CVCL_0395
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
Hela cells Cervix uteri Homo sapiens (Human) CVCL_0030
T47D cells Breast Homo sapiens (Human) CVCL_0553
NCI-H460 cells Lung Homo sapiens (Human) CVCL_0459
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
HCC70 cells Breast Homo sapiens (Human) CVCL_1270
A2058 cells Skin Homo sapiens (Human) CVCL_1059
Caco-2 cells Colon Homo sapiens (Human) CVCL_0025
MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
NCI-60 cells N.A. Homo sapiens (Human) N.A.
KU-19 cells Blood Bos taurus (Bovine) CVCL_VN09
EVSA-T cells Ascites Homo sapiens (Human) CVCL_1207
CAL-120 cells Pleural effusion Homo sapiens (Human) CVCL_1104
BT-549 cells Breast Homo sapiens (Human) CVCL_1092
BT-474 cells Breast Homo sapiens (Human) CVCL_0179
BT-20 cells Mammary gland Homo sapiens (Human) CVCL_0178
B16F10 cells Skin Mus musculus (Mouse) CVCL_0159
In Vivo Model Female NMRI (nu/nu) mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Miransertib by aberration of the drug's therapeutic target
Uprosertib
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: Solid tumour/cancer [14]
Sensitive Disease Solid tumour/cancer [ICD-11: 2A00-2F9Z]
Sensitive Drug Uprosertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SkOV3 cells Ovary Homo sapiens (Human) CVCL_0532
BT474 cells Breast Homo sapiens (Human) CVCL_0179
In Vivo Model Female nu/nu CD-1 mouse xenograft model Mus musculus
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of Uprosertib by aberration of the drug's therapeutic target
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Melanoma [15]
Sensitive Disease Melanoma [ICD-11: 2C30.0]
Sensitive Drug Uprosertib
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model 19 drug na?ve cell lines and four sub-lines N.A. . N.A.
Experiment for
Molecule Alteration
Western blotting analysis
Experiment for
Drug Resistance
ATP-based luminescent assay; Flow cytometry assay
BAY1125976
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: Anal canal cancer [16]
Sensitive Disease Anal canal cancer [ICD-11: 2C00.0]
Sensitive Drug BAY1125976
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model LNCaP cells Prostate Homo sapiens (Human) CVCL_0395
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
Hela cells Cervix uteri Homo sapiens (Human) CVCL_0030
NCI-H460 cells Lung Homo sapiens (Human) CVCL_0459
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
HCC70 cells Breast Homo sapiens (Human) CVCL_1270
A2058 cells Skin Homo sapiens (Human) CVCL_1059
Caco-2 cells Colon Homo sapiens (Human) CVCL_0025
MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
DU-145 cells Prostate Homo sapiens (Human) CVCL_0105
NCI-60 cells N.A. Homo sapiens (Human) N.A.
EVSA-T cells Ascites Homo sapiens (Human) CVCL_1207
KU-19 cells Blood Bos taurus (Bovine) CVCL_VN09
T47D cells Pleural effusion Homo sapiens (Human) CVCL_0553
CAL-120 cells Pleural effusion Homo sapiens (Human) CVCL_1104
BT-549 cells Breast Homo sapiens (Human) CVCL_1092
BT-474 cells Breast Homo sapiens (Human) CVCL_0179
BT-20 cells Mammary gland Homo sapiens (Human) CVCL_0178
B16F10 cells Skin Mus musculus (Mouse) CVCL_0159
In Vivo Model Female NMRI (nu/nu) mouse xenograft model Mus musculus
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of BAY1125976 by aberration of the drug's therapeutic target
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Disease Class: Breast adenocarcinoma [16]
Sensitive Disease Breast adenocarcinoma [ICD-11: 2C60.1]
Sensitive Drug BAY1125976
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model LNCaP cells Prostate Homo sapiens (Human) CVCL_0395
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
Hela cells Cervix uteri Homo sapiens (Human) CVCL_0030
NCI-H460 cells Lung Homo sapiens (Human) CVCL_0459
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
HCC70 cells Breast Homo sapiens (Human) CVCL_1270
A2058 cells Skin Homo sapiens (Human) CVCL_1059
Caco-2 cells Colon Homo sapiens (Human) CVCL_0025
MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
DU-145 cells Prostate Homo sapiens (Human) CVCL_0105
NCI-60 cells N.A. Homo sapiens (Human) N.A.
EVSA-T cells Ascites Homo sapiens (Human) CVCL_1207
KU-19 cells Blood Bos taurus (Bovine) CVCL_VN09
T47D cells Pleural effusion Homo sapiens (Human) CVCL_0553
CAL-120 cells Pleural effusion Homo sapiens (Human) CVCL_1104
BT-549 cells Breast Homo sapiens (Human) CVCL_1092
BT-474 cells Breast Homo sapiens (Human) CVCL_0179
BT-20 cells Mammary gland Homo sapiens (Human) CVCL_0178
B16F10 cells Skin Mus musculus (Mouse) CVCL_0159
In Vivo Model Female NMRI (nu/nu) mouse xenograft model Mus musculus
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of BAY1125976 by unusual activation of pro-survival pathway
Preclinical Drug(s)
3 drug(s) in total
Click to Show/Hide the Full List of Drugs
Allosteric AKT 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
Disease Class: Breast adenocarcinoma [17]
Sensitive Disease Breast adenocarcinoma [ICD-11: 2C60.1]
Sensitive Drug Allosteric AKT inhibitors
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model Hela cells Cervix uteri Homo sapiens (Human) CVCL_0030
HEK293T cells Kidney Homo sapiens (Human) CVCL_0063
In Vivo Model Female NOD/SCID mouse xenograft model Mus musculus
Experiment for
Drug Resistance
MTT assay
Mechanism Description Deactivation of Akt by a small molecule inhibitor ,SC66, targeting pleckstrin homology domain and facilitating Akt ubiquitination. A cancer-relevant Akt1 (e17k) mutant is unstable, making it intrinsically sensitive to functional inhibition by SC66 in cellular contexts in which the PI3K inhibition has little inhibitory effect.
Disease Class: Breast adenocarcinoma [17]
Sensitive Disease Breast adenocarcinoma [ICD-11: 2C60.1]
Sensitive Drug Allosteric AKT inhibitors
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model Hela cells Cervix uteri Homo sapiens (Human) CVCL_0030
HEK293 cells Kidney Homo sapiens (Human) CVCL_0045
In Vivo Model Female NOD/SCID mouse xenograft model Mus musculus
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of allosteric AKT inhibitors by aberration of the drug's therapeutic target
ARQ 751
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: Endometrial adenocarcinoma [13]
Sensitive Disease Endometrial adenocarcinoma [ICD-11: 2C76.0]
Sensitive Drug ARQ 751
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model DU-145 cells Prostate Homo sapiens (Human) CVCL_0105
LNCaP cells Prostate Homo sapiens (Human) CVCL_0395
MDA-MB-231 cells Breast Homo sapiens (Human) CVCL_0062
Hela cells Cervix uteri Homo sapiens (Human) CVCL_0030
T47D cells Breast Homo sapiens (Human) CVCL_0553
NCI-H460 cells Lung Homo sapiens (Human) CVCL_0459
MDA-MB-453 cells Breast Homo sapiens (Human) CVCL_0418
MDA-MB-468 cells Breast Homo sapiens (Human) CVCL_0419
HCC70 cells Breast Homo sapiens (Human) CVCL_1270
A2058 cells Skin Homo sapiens (Human) CVCL_1059
Caco-2 cells Colon Homo sapiens (Human) CVCL_0025
MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
ZR75-1 cells Breast Homo sapiens (Human) CVCL_0588
NCI-60 cells N.A. Homo sapiens (Human) N.A.
KU-19 cells Blood Bos taurus (Bovine) CVCL_VN09
EVSA-T cells Ascites Homo sapiens (Human) CVCL_1207
CAL-120 cells Pleural effusion Homo sapiens (Human) CVCL_1104
BT-549 cells Breast Homo sapiens (Human) CVCL_1092
BT-474 cells Breast Homo sapiens (Human) CVCL_0179
BT-20 cells Mammary gland Homo sapiens (Human) CVCL_0178
B16F10 cells Skin Mus musculus (Mouse) CVCL_0159
In Vivo Model Female NMRI (nu/nu) mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
Western blotting analysis
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of ARQ 751 by aberration of the drug's therapeutic target
MK2206
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: Solid tumour/cancer [18]
Resistant Disease Solid tumour/cancer [ICD-11: 2A00-2F9Z]
Resistant Drug MK2206
Molecule Alteration Missense mutation
p.W80A (c.238_239delTGinsGC)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model 3T3-L1 cells Nasopharynx Mus musculus (Mouse) CVCL_0123
Mechanism Description The missense mutation p.W80A (c.238_239delTGinsGC) in gene AKT1 cause the resistance of MK2206 by aberration of the drug's therapeutic target
Disease Class: Breast adenocarcinoma [19]
Resistant Disease Breast adenocarcinoma [ICD-11: 2C60.1]
Resistant Drug MK2206
Molecule Alteration Missense mutation
p.W80A (c.238_239delTGinsGC)
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model H1975 cells Lung Homo sapiens (Human) CVCL_1511
BT474 cells Breast Homo sapiens (Human) CVCL_0179
HCC827 cells Lung Homo sapiens (Human) CVCL_2063
MCF10A cells Breast Homo sapiens (Human) CVCL_0598
HCC4006 cells Lung Homo sapiens (Human) CVCL_1269
MDA-MB-361 cells Breast Homo sapiens (Human) CVCL_0620
GTL-16 cells Gastric Homo sapiens (Human) CVCL_7668
MCF-7 cells Breast Homo sapiens (Human) CVCL_0031
HCT116 cells Colon Homo sapiens (Human) CVCL_0291
MDA-MB-361 cells Breast Homo sapiens (Human) CVCL_0620
H1648 cells Lymph node Homo sapiens (Human) CVCL_1482
EBC1 cells Skin Homo sapiens (Human) CVCL_2891
Mechanism Description The missense mutation p.W80A (c.238_239delTGinsGC) in gene AKT1 cause the resistance of MK2206 by aberration of the drug's therapeutic target
Investigative Drug(s)
2 drug(s) in total
Click to Show/Hide the Full List of Drugs
Non-allosteric AKT 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
Disease Class: Solid tumour/cancer [20]
Sensitive Disease Solid tumour/cancer [ICD-11: 2A00-2F9Z]
Sensitive Drug Non-allosteric AKT inhibitors
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of non-allosteric AKT inhibitors by aberration of the drug's therapeutic target
Disease Class: Solid tumour/cancer [20]
Sensitive Disease Solid tumour/cancer [ICD-11: 2A00-2F9Z]
Sensitive Drug Non-allosteric AKT inhibitors
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
Disease Class: Solid tumour/cancer [20]
Sensitive Disease Solid tumour/cancer [ICD-11: 2A00-2F9Z]
Sensitive Drug Non-allosteric AKT inhibitors
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
PI3K pathway inhibitors
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: Head and neck cancer [21]
Sensitive Disease Head and neck cancer [ICD-11: 2D42.0]
Sensitive Drug PI3K pathway inhibitors
Molecule Alteration Missense mutation
p.E17K (c.49G>A)
Experimental Note Identified from the Human Clinical Data
Mechanism Description The missense mutation p.E17K (c.49G>A) in gene AKT1 cause the sensitivity of PI3K pathway inhibitors by aberration of the drug's therapeutic target
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: 5.17E-11; Fold-change: 1.86E-01; Z-score: 4.96E-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: 7.08E-01; Fold-change: -1.96E-01; Z-score: -4.40E-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: 5.78E-02; Fold-change: 3.86E-01; Z-score: 7.39E-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: 1.80E-01; Fold-change: -2.05E-01; Z-score: -7.62E-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
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: 2.63E-16; Fold-change: -3.05E-01; Z-score: -7.97E-01
The Expression Level of Disease Section Compare with the Adjacent Tissue p-value: 1.55E-17; Fold-change: -6.10E-01; Z-score: -1.18E+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
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: 9.20E-04; Fold-change: 4.25E-01; Z-score: 6.80E-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: 4.76E-37; Fold-change: 5.98E-01; Z-score: 9.55E-01
The Expression Level of Disease Section Compare with the Adjacent Tissue p-value: 1.00E-07; Fold-change: 4.07E-01; Z-score: 8.71E-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
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: 4.44E-02; Fold-change: 4.74E-01; Z-score: 8.15E-01
The Expression Level of Disease Section Compare with the Adjacent Tissue p-value: 8.39E-03; Fold-change: 1.02E+00; Z-score: 1.22E+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: 6.30E-03; Fold-change: 2.34E-01; Z-score: 7.02E-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
Head and neck cancer [ICD-11: 2D42]
Click to Show/Hide
Differential expression of molecule in resistant diseases
The Studied Tissue Head and neck tissue
The Specified Disease Head and neck cancer
The Expression Level of Disease Section Compare with the Healthy Individual Tissue p-value: 3.59E-01; Fold-change: 4.76E-02; Z-score: 1.16E-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
Tissue-specific Molecule Abundances in Healthy Individuals
Click to Show/Hide the Molecule Abundances
References
Ref 1 Long Noncoding RNA GAS5, Which Acts as a Tumor Suppressor via microRNA 21, Regulates Cisplatin Resistance Expression in Cervical Cancer. Int J Gynecol Cancer. 2017 Jul;27(6):1096-1108. doi: 10.1097/IGC.0000000000001028.
Ref 2 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 3 Mechanisms of resistance to EGFR-targeted drugs: lung cancer. ESMO Open. 2016 May 11;1(3):e000060. doi: 10.1136/esmoopen-2016-000060. eCollection 2016.
Ref 4 Accelerating Discovery of Functional Mutant Alleles in CancerCancer Discov. 2018 Feb;8(2):174-183. doi: 10.1158/2159-8290.CD-17-0321. Epub 2017 Dec 15.
Ref 5 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 6 Safety and tolerability of AZD5363 in Japanese patients with advanced solid tumorsCancer Chemother Pharmacol. 2016 Apr;77(4):787-95. doi: 10.1007/s00280-016-2987-9. Epub 2016 Mar 1.
Ref 7 Preclinical pharmacology of AZD5363, an inhibitor of AKT: pharmacodynamics, antitumor activity, and correlation of monotherapy activity with genetic backgroundMol Cancer Ther. 2012 Apr;11(4):873-87. doi: 10.1158/1535-7163.MCT-11-0824-T. Epub 2012 Jan 31.
Ref 8 Capivasertib Active against AKT1-Mutated CancersCancer Discov. 2019 Jan;9(1):OF7. doi: 10.1158/2159-8290.CD-NB2018-153. Epub 2018 Nov 14.
Ref 9 AKT Inhibition in Solid Tumors With AKT1 MutationsJ Clin Oncol. 2017 Jul 10;35(20):2251-2259. doi: 10.1200/JCO.2017.73.0143. Epub 2017 May 10.
Ref 10 Tumors with AKT1E17K Mutations Are Rational Targets for Single Agent or Combination Therapy with AKT InhibitorsMol Cancer Ther. 2015 Nov;14(11):2441-51. doi: 10.1158/1535-7163.MCT-15-0230. Epub 2015 Sep 8.
Ref 11 A First-in-Human Phase I Study of the ATP-Competitive AKT Inhibitor Ipatasertib Demonstrates Robust and Safe Targeting of AKT in Patients with Solid TumorsCancer Discov. 2017 Jan;7(1):102-113. doi: 10.1158/2159-8290.CD-16-0512. Epub 2016 Nov 21.
Ref 12 First evidence of a therapeutic effect of miransertib in a teenager with Proteus syndrome and ovarian carcinomaAm J Med Genet A. 2019 Jul;179(7):1319-1324. doi: 10.1002/ajmg.a.61160. Epub 2019 May 6.
Ref 13 Targeting AKT1-E17K and the PI3K/AKT Pathway with an Allosteric AKT Inhibitor, ARQ 092PLoS One. 2015 Oct 15;10(10):e0140479. doi: 10.1371/journal.pone.0140479. eCollection 2015.
Ref 14 Discovery of novel AKT inhibitors with enhanced anti-tumor effects in combination with the MEK inhibitorPLoS One. 2014 Jun 30;9(6):e100880. doi: 10.1371/journal.pone.0100880. eCollection 2014.
Ref 15 Effects of AKT inhibitor therapy in response and resistance to BRAF inhibition in melanomaMol Cancer. 2014 Apr 16;13:83. doi: 10.1186/1476-4598-13-83.
Ref 16 BAY 1125976, a selective allosteric AKT1/2 inhibitor, exhibits high efficacy on AKT signaling-dependent tumor growth in mouse modelsInt J Cancer. 2017 Jan 15;140(2):449-459. doi: 10.1002/ijc.30457. Epub 2016 Oct 20.
Ref 17 Deactivation of Akt by a small molecule inhibitor targeting pleckstrin homology domain and facilitating Akt ubiquitinationProc Natl Acad Sci U S A. 2011 Apr 19;108(16):6486-91. doi: 10.1073/pnas.1019062108. Epub 2011 Apr 4.
Ref 18 Development of a new model system to dissect isoform specific Akt signalling in adipocytesBiochem J. 2015 Jun 15;468(3):425-34. doi: 10.1042/BJ20150191. Epub 2015 Apr 9.
Ref 19 A kinase-independent function of AKT promotes cancer cell survivalElife. 2014 Dec 31;3:e03751. doi: 10.7554/eLife.03751.
Ref 20 STATISTICS/DATA TYPE - iGMDR.
Ref 21 Incorporation of Next-Generation Sequencing into Routine Clinical Care to Direct Treatment of Head and Neck Squamous Cell CarcinomaClin Cancer Res. 2016 Jun 15;22(12):2939-49. doi: 10.1158/1078-0432.CCR-15-2314. Epub 2016 Jan 13.

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