General Information of Drug Off-Target (DOT) (ID: OTB5DYYZ)

DOT Name Cyclin-dependent kinase 2 (CDK2)
Synonyms EC 2.7.11.22; Cell division protein kinase 2; p33 protein kinase
Gene Name CDK2
UniProt ID
CDK2_HUMAN
3D Structure
Download
2D Sequence (FASTA)
Download
3D Structure (PDB)
Download
PDB ID
1AQ1 ; 1B38 ; 1B39 ; 1BUH ; 1CKP ; 1DI8 ; 1DM2 ; 1E1V ; 1E1X ; 1E9H ; 1F5Q ; 1FIN ; 1FQ1 ; 1FVT ; 1FVV ; 1G5S ; 1GIH ; 1GII ; 1GIJ ; 1GY3 ; 1GZ8 ; 1H00 ; 1H01 ; 1H07 ; 1H08 ; 1H0V ; 1H0W ; 1H1P ; 1H1Q ; 1H1R ; 1H1S ; 1H24 ; 1H25 ; 1H26 ; 1H27 ; 1H28 ; 1HCK ; 1HCL ; 1JST ; 1JSU ; 1JSV ; 1JVP ; 1KE5 ; 1KE6 ; 1KE7 ; 1KE8 ; 1KE9 ; 1OGU ; 1OI9 ; 1OIQ ; 1OIR ; 1OIT ; 1OIU ; 1OIY ; 1OKV ; 1OKW ; 1OL1 ; 1OL2 ; 1P2A ; 1P5E ; 1PF8 ; 1PKD ; 1PW2 ; 1PXI ; 1PXJ ; 1PXK ; 1PXL ; 1PXM ; 1PXN ; 1PXO ; 1PXP ; 1PYE ; 1QMZ ; 1R78 ; 1URC ; 1URW ; 1V1K ; 1VYW ; 1VYZ ; 1W0X ; 1W8C ; 1W98 ; 1WCC ; 1Y8Y ; 1Y91 ; 1YKR ; 2A0C ; 2A4L ; 2B52 ; 2B53 ; 2B54 ; 2B55 ; 2BHE ; 2BHH ; 2BKZ ; 2BPM ; 2BTR ; 2BTS ; 2C4G ; 2C5N ; 2C5O ; 2C5V ; 2C5X ; 2C5Y ; 2C68 ; 2C69 ; 2C6I ; 2C6K ; 2C6L ; 2C6M ; 2C6O ; 2C6T ; 2CCH ; 2CCI ; 2CJM ; 2CLX ; 2DS1 ; 2DUV ; 2EXM ; 2FVD ; 2G9X ; 2I40 ; 2IW6 ; 2IW8 ; 2IW9 ; 2J9M ; 2JGZ ; 2R3F ; 2R3G ; 2R3H ; 2R3I ; 2R3J ; 2R3K ; 2R3L ; 2R3M ; 2R3N ; 2R3O ; 2R3P ; 2R3Q ; 2R3R ; 2R64 ; 2UUE ; 2UZB ; 2UZD ; 2UZE ; 2UZL ; 2UZN ; 2UZO ; 2V0D ; 2V22 ; 2VTA ; 2VTH ; 2VTI ; 2VTJ ; 2VTL ; 2VTM ; 2VTN ; 2VTO ; 2VTP ; 2VTQ ; 2VTR ; 2VTS ; 2VTT ; 2VU3 ; 2VV9 ; 2W05 ; 2W06 ; 2W17 ; 2W1H ; 2WEV ; 2WFY ; 2WHB ; 2WIH ; 2WIP ; 2WMA ; 2WMB ; 2WPA ; 2WXV ; 2X1N ; 2XMY ; 2XNB ; 3BHT ; 3BHU ; 3BHV ; 3DDP ; 3DDQ ; 3DOG ; 3EID ; 3EJ1 ; 3EOC ; 3EZR ; 3EZV ; 3F5X ; 3FZ1 ; 3IG7 ; 3IGG ; 3LE6 ; 3LFN ; 3LFQ ; 3LFS ; 3MY5 ; 3NS9 ; 3PJ8 ; 3PXF ; 3PXQ ; 3PXR ; 3PXY ; 3PXZ ; 3PY0 ; 3PY1 ; 3QHR ; 3QHW ; 3QL8 ; 3QQF ; 3QQG ; 3QQH ; 3QQJ ; 3QQK ; 3QQL ; 3QRT ; 3QRU ; 3QTQ ; 3QTR ; 3QTS ; 3QTU ; 3QTW ; 3QTX ; 3QTZ ; 3QU0 ; 3QWJ ; 3QWK ; 3QX2 ; 3QX4 ; 3QXO ; 3QXP ; 3QZF ; 3QZG ; 3QZH ; 3QZI ; 3R1Q ; 3R1S ; 3R1Y ; 3R28 ; 3R6X ; 3R71 ; 3R73 ; 3R7E ; 3R7I ; 3R7U ; 3R7V ; 3R7Y ; 3R83 ; 3R8L ; 3R8M ; 3R8P ; 3R8U ; 3R8V ; 3R8Z ; 3R9D ; 3R9H ; 3R9N ; 3R9O ; 3RAH ; 3RAI ; 3RAK ; 3RAL ; 3RJC ; 3RK5 ; 3RK7 ; 3RK9 ; 3RKB ; 3RM6 ; 3RM7 ; 3RMF ; 3RNI ; 3ROY ; 3RPO ; 3RPR ; 3RPV ; 3RPY ; 3RZB ; 3S00 ; 3S0O ; 3S1H ; 3S2P ; 3SQQ ; 3SW4 ; 3SW7 ; 3TI1 ; 3TIY ; 3TIZ ; 3TNW ; 3ULI ; 3UNJ ; 3UNK ; 3WBL ; 4ACM ; 4BCK ; 4BCM ; 4BCN ; 4BCO ; 4BCP ; 4BCQ ; 4BGH ; 4BZD ; 4CFM ; 4CFN ; 4CFU ; 4CFV ; 4CFW ; 4CFX ; 4D1X ; 4D1Z ; 4EK3 ; 4EK4 ; 4EK5 ; 4EK6 ; 4EK8 ; 4EOI ; 4EOJ ; 4EOK ; 4EOL ; 4EOM ; 4EON ; 4EOO ; 4EOP ; 4EOQ ; 4EOR ; 4EOS ; 4ERW ; 4EZ3 ; 4EZ7 ; 4FKG ; 4FKI ; 4FKJ ; 4FKL ; 4FKO ; 4FKP ; 4FKQ ; 4FKR ; 4FKS ; 4FKT ; 4FKU ; 4FKV ; 4FKW ; 4FX3 ; 4GCJ ; 4I3Z ; 4II5 ; 4KD1 ; 4LYN ; 4NJ3 ; 4RJ3 ; 5A14 ; 5AND ; 5ANE ; 5ANG ; 5ANI ; 5ANJ ; 5ANK ; 5ANO ; 5CYI ; 5D1J ; 5FP5 ; 5FP6 ; 5IEV ; 5IEX ; 5IEY ; 5IF1 ; 5JQ5 ; 5JQ8 ; 5K4J ; 5L2W ; 5LMK ; 5MHQ ; 5NEV ; 5OO0 ; 5OO1 ; 5OO3 ; 5OSJ ; 5OSM ; 5UQ1 ; 5UQ2 ; 5UQ3 ; 6ATH ; 6GUB ; 6GUC ; 6GUE ; 6GUF ; 6GUH ; 6GUK ; 6GVA ; 6INL ; 6JGM ; 6OQI ; 6P3W ; 6Q3B ; 6Q3C ; 6Q3F ; 6Q48 ; 6Q49 ; 6Q4A ; 6Q4B ; 6Q4C ; 6Q4D ; 6Q4E ; 6Q4F ; 6Q4G ; 6Q4H ; 6Q4I ; 6Q4J ; 6Q4K ; 6RIJ ; 6SG4 ; 6YL1 ; 6YL6 ; 6YLK ; 7ACK ; 7B5L ; 7B5R ; 7B7S ; 7E34 ; 7KJS ; 7M2F ; 7MKX ; 7NVQ ; 7QHL ; 7RA5 ; 7RWE ; 7RWF ; 7RXO ; 7S4T ; 7S7A ; 7S84 ; 7S85 ; 7S9X ; 7SA0 ; 7UG1 ; 7UXI ; 7UXK ; 7VDU ; 7ZPC ; 8B54 ; 8BYA ; 8BZO ; 8CUR ; 8ERD ; 8ERN ; 8FOW ; 8FP0 ; 8FP5 ; 8H6P ; 8H6T ; 8OR0 ; 8OR4 ; 8OY2
EC Number
2.7.11.22
Pfam ID
PF00069
Sequence
MENFQKVEKIGEGTYGVVYKARNKLTGEVVALKKIRLDTETEGVPSTAIREISLLKELNH
PNIVKLLDVIHTENKLYLVFEFLHQDLKKFMDASALTGIPLPLIKSYLFQLLQGLAFCHS
HRVLHRDLKPQNLLINTEGAIKLADFGLARAFGVPVRTYTHEVVTLWYRAPEILLGCKYY
STAVDIWSLGCIFAEMVTRRALFPGDSEIDQLFRIFRTLGTPDEVVWPGVTSMPDYKPSF
PKWARQDFSKVVPPLDEDGRSLLSQMLHYDPNKRISAKAALAHPFFQDVTKPVPHLRL
Function
Serine/threonine-protein kinase involved in the control of the cell cycle; essential for meiosis, but dispensable for mitosis. Phosphorylates CTNNB1, USP37, p53/TP53, NPM1, CDK7, RB1, BRCA2, MYC, NPAT, EZH2. Triggers duplication of centrosomes and DNA. Acts at the G1-S transition to promote the E2F transcriptional program and the initiation of DNA synthesis, and modulates G2 progression; controls the timing of entry into mitosis/meiosis by controlling the subsequent activation of cyclin B/CDK1 by phosphorylation, and coordinates the activation of cyclin B/CDK1 at the centrosome and in the nucleus. Crucial role in orchestrating a fine balance between cellular proliferation, cell death, and DNA repair in human embryonic stem cells (hESCs). Activity of CDK2 is maximal during S phase and G2; activated by interaction with cyclin E during the early stages of DNA synthesis to permit G1-S transition, and subsequently activated by cyclin A2 (cyclin A1 in germ cells) during the late stages of DNA replication to drive the transition from S phase to mitosis, the G2 phase. EZH2 phosphorylation promotes H3K27me3 maintenance and epigenetic gene silencing. Phosphorylates CABLES1. Cyclin E/CDK2 prevents oxidative stress-mediated Ras-induced senescence by phosphorylating MYC. Involved in G1-S phase DNA damage checkpoint that prevents cells with damaged DNA from initiating mitosis; regulates homologous recombination-dependent repair by phosphorylating BRCA2, this phosphorylation is low in S phase when recombination is active, but increases as cells progress towards mitosis. In response to DNA damage, double-strand break repair by homologous recombination a reduction of CDK2-mediated BRCA2 phosphorylation. Phosphorylation of RB1 disturbs its interaction with E2F1. NPM1 phosphorylation by cyclin E/CDK2 promotes its dissociates from unduplicated centrosomes, thus initiating centrosome duplication. Cyclin E/CDK2-mediated phosphorylation of NPAT at G1-S transition and until prophase stimulates the NPAT-mediated activation of histone gene transcription during S phase. Required for vitamin D-mediated growth inhibition by being itself inactivated. Involved in the nitric oxide- (NO) mediated signaling in a nitrosylation/activation-dependent manner. USP37 is activated by phosphorylation and thus triggers G1-S transition. CTNNB1 phosphorylation regulates insulin internalization. Phosphorylates FOXP3 and negatively regulates its transcriptional activity and protein stability. Phosphorylates CDK2AP2. Phosphorylates ERCC6 which is essential for its chromatin remodeling activity at DNA double-strand breaks.
KEGG Pathway
FoxO sig.ling pathway (hsa04068 )
Cell cycle (hsa04110 )
Oocyte meiosis (hsa04114 )
p53 sig.ling pathway (hsa04115 )
PI3K-Akt sig.ling pathway (hsa04151 )
Cellular senescence (hsa04218 )
Progesterone-mediated oocyte maturation (hsa04914 )
Cushing syndrome (hsa04934 )
Hepatitis C (hsa05160 )
Hepatitis B (hsa05161 )
Measles (hsa05162 )
Human papillomavirus infection (hsa05165 )
Human T-cell leukemia virus 1 infection (hsa05166 )
Epstein-Barr virus infection (hsa05169 )
Pathways in cancer (hsa05200 )
Viral carcinogenesis (hsa05203 )
Prostate cancer (hsa05215 )
Small cell lung cancer (hsa05222 )
Gastric cancer (hsa05226 )
Reactome Pathway
Telomere Extension By Telomerase (R-HSA-171319 )
Activation of ATR in response to replication stress (R-HSA-176187 )
Regulation of APC/C activators between G1/S and early anaphase (R-HSA-176408 )
SCF(Skp2)-mediated degradation of p27/p21 (R-HSA-187577 )
Senescence-Associated Secretory Phenotype (SASP) (R-HSA-2559582 )
DNA Damage/Telomere Stress Induced Senescence (R-HSA-2559586 )
Processing of DNA double-strand break ends (R-HSA-5693607 )
TP53 Regulates Transcription of Genes Involved in G1 Cell Cycle Arrest (R-HSA-6804116 )
Regulation of TP53 Activity through Phosphorylation (R-HSA-6804756 )
Regulation of TP53 Degradation (R-HSA-6804757 )
G2 Phase (R-HSA-68911 )
Orc1 removal from chromatin (R-HSA-68949 )
Activation of the pre-replicative complex (R-HSA-68962 )
CDK-mediated phosphorylation and removal of Cdc6 (R-HSA-69017 )
Phosphorylation of proteins involved in G1/S transition by active Cyclin E (R-HSA-69200 )
Cyclin E associated events during G1/S transition (R-HSA-69202 )
Cyclin D associated events in G1 (R-HSA-69231 )
Cyclin A/B1/B2 associated events during G2/M transition (R-HSA-69273 )
p53-Dependent G1 DNA Damage Response (R-HSA-69563 )
Cyclin A (R-HSA-69656 )
PTK6 Regulates Cell Cycle (R-HSA-8849470 )
Meiotic recombination (R-HSA-912446 )
Transcriptional regulation of granulopoiesis (R-HSA-9616222 )
Defective binding of RB1 mutants to E2F1,(E2F2, E2F3) (R-HSA-9661069 )
Factors involved in megakaryocyte development and platelet production (R-HSA-983231 )
G0 and Early G1 (R-HSA-1538133 )

Molecular Interaction Atlas (MIA) of This DOT

Molecular Interaction Atlas (MIA) Jump to Detail Molecular Interaction Atlas of This DOT
This DOT Affected the Drug Response of 3 Drug(s)
Drug Name Drug ID Highest Status Interaction REF
Doxorubicin DMVP5YE Approved Cyclin-dependent kinase 2 (CDK2) increases the response to substance of Doxorubicin. [90]
Mitomycin DMH0ZJE Approved Cyclin-dependent kinase 2 (CDK2) affects the response to substance of Mitomycin. [91]
Vinblastine DM5TVS3 Approved Cyclin-dependent kinase 2 (CDK2) affects the response to substance of Vinblastine. [91]
------------------------------------------------------------------------------------
94 Drug(s) Affected the Gene/Protein Processing of This DOT
Drug Name Drug ID Highest Status Interaction REF
Valproate DMCFE9I Approved Valproate decreases the expression of Cyclin-dependent kinase 2 (CDK2). [1]
Ciclosporin DMAZJFX Approved Ciclosporin decreases the expression of Cyclin-dependent kinase 2 (CDK2). [2]
Cisplatin DMRHGI9 Approved Cisplatin increases the expression of Cyclin-dependent kinase 2 (CDK2). [4]
Estradiol DMUNTE3 Approved Estradiol decreases the expression of Cyclin-dependent kinase 2 (CDK2). [5]
Ivermectin DMDBX5F Approved Ivermectin decreases the expression of Cyclin-dependent kinase 2 (CDK2). [6]
Quercetin DM3NC4M Approved Quercetin decreases the expression of Cyclin-dependent kinase 2 (CDK2). [8]
Arsenic trioxide DM61TA4 Approved Arsenic trioxide decreases the expression of Cyclin-dependent kinase 2 (CDK2). [9]
Hydrogen peroxide DM1NG5W Approved Hydrogen peroxide decreases the expression of Cyclin-dependent kinase 2 (CDK2). [10]
Calcitriol DM8ZVJ7 Approved Calcitriol decreases the expression of Cyclin-dependent kinase 2 (CDK2). [11]
Testosterone DM7HUNW Approved Testosterone decreases the expression of Cyclin-dependent kinase 2 (CDK2). [11]
Carbamazepine DMZOLBI Approved Carbamazepine affects the expression of Cyclin-dependent kinase 2 (CDK2). [12]
Methotrexate DM2TEOL Approved Methotrexate increases the expression of Cyclin-dependent kinase 2 (CDK2). [13]
Selenium DM25CGV Approved Selenium decreases the expression of Cyclin-dependent kinase 2 (CDK2). [14]
Progesterone DMUY35B Approved Progesterone decreases the activity of Cyclin-dependent kinase 2 (CDK2). [15]
Fluorouracil DMUM7HZ Approved Fluorouracil increases the expression of Cyclin-dependent kinase 2 (CDK2). [16]
Fulvestrant DM0YZC6 Approved Fulvestrant increases the expression of Cyclin-dependent kinase 2 (CDK2). [17]
Dexamethasone DMMWZET Approved Dexamethasone decreases the expression of Cyclin-dependent kinase 2 (CDK2). [18]
Folic acid DMEMBJC Approved Folic acid decreases the expression of Cyclin-dependent kinase 2 (CDK2). [19]
Demecolcine DMCZQGK Approved Demecolcine decreases the expression of Cyclin-dependent kinase 2 (CDK2). [20]
Niclosamide DMJAGXQ Approved Niclosamide decreases the expression of Cyclin-dependent kinase 2 (CDK2). [21]
Cannabidiol DM0659E Approved Cannabidiol decreases the expression of Cyclin-dependent kinase 2 (CDK2). [22]
Bortezomib DMNO38U Approved Bortezomib decreases the activity of Cyclin-dependent kinase 2 (CDK2). [23]
Troglitazone DM3VFPD Approved Troglitazone decreases the expression of Cyclin-dependent kinase 2 (CDK2). [24]
Rosiglitazone DMILWZR Approved Rosiglitazone decreases the expression of Cyclin-dependent kinase 2 (CDK2). [25]
Aspirin DM672AH Approved Aspirin decreases the expression of Cyclin-dependent kinase 2 (CDK2). [26]
Etoposide DMNH3PG Approved Etoposide increases the expression of Cyclin-dependent kinase 2 (CDK2). [27]
Paclitaxel DMLB81S Approved Paclitaxel increases the activity of Cyclin-dependent kinase 2 (CDK2). [28]
Diclofenac DMPIHLS Approved Diclofenac affects the expression of Cyclin-dependent kinase 2 (CDK2). [12]
Piroxicam DMTK234 Approved Piroxicam decreases the expression of Cyclin-dependent kinase 2 (CDK2). [29]
Dasatinib DMJV2EK Approved Dasatinib decreases the expression of Cyclin-dependent kinase 2 (CDK2). [30]
Malathion DMXZ84M Approved Malathion increases the expression of Cyclin-dependent kinase 2 (CDK2). [31]
Indomethacin DMSC4A7 Approved Indomethacin decreases the activity of Cyclin-dependent kinase 2 (CDK2). [32]
Azacitidine DMTA5OE Approved Azacitidine decreases the expression of Cyclin-dependent kinase 2 (CDK2). [33]
Simvastatin DM30SGU Approved Simvastatin decreases the expression of Cyclin-dependent kinase 2 (CDK2). [34]
Sulindac DM2QHZU Approved Sulindac decreases the activity of Cyclin-dependent kinase 2 (CDK2). [32]
Capsaicin DMGMF6V Approved Capsaicin decreases the expression of Cyclin-dependent kinase 2 (CDK2). [35]
Palbociclib DMD7L94 Approved Palbociclib decreases the activity of Cyclin-dependent kinase 2 (CDK2). [36]
Pioglitazone DMKJ485 Approved Pioglitazone decreases the expression of Cyclin-dependent kinase 2 (CDK2). [25]
Acocantherin DM7JT24 Approved Acocantherin decreases the expression of Cyclin-dependent kinase 2 (CDK2). [37]
Acetic Acid, Glacial DM4SJ5Y Approved Acetic Acid, Glacial increases the expression of Cyclin-dependent kinase 2 (CDK2). [38]
Motexafin gadolinium DMEJKRF Approved Motexafin gadolinium increases the expression of Cyclin-dependent kinase 2 (CDK2). [38]
Sorafenib DMS8IFC Approved Sorafenib decreases the expression of Cyclin-dependent kinase 2 (CDK2). [39]
Imatinib DM7RJXL Approved Imatinib decreases the expression of Cyclin-dependent kinase 2 (CDK2). [40]
Ritonavir DMU764S Approved Ritonavir decreases the expression of Cyclin-dependent kinase 2 (CDK2). [41]
Docetaxel DMDI269 Approved Docetaxel increases the activity of Cyclin-dependent kinase 2 (CDK2). [23]
Cholecalciferol DMGU74E Approved Cholecalciferol decreases the activity of Cyclin-dependent kinase 2 (CDK2). [42]
Lovastatin DM9OZWQ Approved Lovastatin decreases the activity of Cyclin-dependent kinase 2 (CDK2). [43]
Orlistat DMRJSP8 Approved Orlistat decreases the expression of Cyclin-dependent kinase 2 (CDK2). [44]
Hesperetin DMKER83 Approved Hesperetin decreases the expression of Cyclin-dependent kinase 2 (CDK2). [46]
Nelfinavir mesylate DMFX6G8 Approved Nelfinavir mesylate decreases the activity of Cyclin-dependent kinase 2 (CDK2). [47]
Omacetaxine mepesuccinate DMPU2WX Approved Omacetaxine mepesuccinate increases the expression of Cyclin-dependent kinase 2 (CDK2). [48]
Salbutamol DMN9CWF Approved Salbutamol increases the expression of Cyclin-dependent kinase 2 (CDK2). [49]
Mechlorethamine DM0CVXA Approved Mechlorethamine increases the activity of Cyclin-dependent kinase 2 (CDK2). [50]
Dronedarone DMA8FS5 Approved Dronedarone decreases the expression of Cyclin-dependent kinase 2 (CDK2). [51]
Amlodipine DMBDAZV Approved Amlodipine decreases the activity of Cyclin-dependent kinase 2 (CDK2). [52]
Deferasirox DM6ETS0 Approved Deferasirox decreases the activity of Cyclin-dependent kinase 2 (CDK2). [54]
Dihydrotestosterone DM3S8XC Phase 4 Dihydrotestosterone increases the expression of Cyclin-dependent kinase 2 (CDK2). [55]
Berberine DMC5Q8X Phase 4 Berberine decreases the expression of Cyclin-dependent kinase 2 (CDK2). [56]
Silymarin DMXBYQR Phase 4 Silymarin decreases the expression of Cyclin-dependent kinase 2 (CDK2). [57]
Resveratrol DM3RWXL Phase 3 Resveratrol decreases the expression of Cyclin-dependent kinase 2 (CDK2). [58]
Curcumin DMQPH29 Phase 3 Curcumin decreases the expression of Cyclin-dependent kinase 2 (CDK2). [39]
Seocalcitol DMKL9QO Phase 3 Seocalcitol decreases the activity of Cyclin-dependent kinase 2 (CDK2). [42]
HMPL-004 DM29XGY Phase 3 HMPL-004 decreases the expression of Cyclin-dependent kinase 2 (CDK2). [59]
I3C DMIGFOR Phase 3 I3C decreases the activity of Cyclin-dependent kinase 2 (CDK2). [60]
Triptolide DMCMDVR Phase 3 Triptolide decreases the expression of Cyclin-dependent kinase 2 (CDK2). [61]
5-methoxypsoralen DME2A8X Phase 3 5-methoxypsoralen decreases the expression of Cyclin-dependent kinase 2 (CDK2). [62]
Genistein DM0JETC Phase 2/3 Genistein increases the expression of Cyclin-dependent kinase 2 (CDK2). [63]
Thymoquinone DMVDTR2 Phase 2/3 Thymoquinone decreases the expression of Cyclin-dependent kinase 2 (CDK2). [64]
GSK2110183 DMZHB37 Phase 2 GSK2110183 decreases the expression of Cyclin-dependent kinase 2 (CDK2). [65]
PEITC DMOMN31 Phase 2 PEITC decreases the expression of Cyclin-dependent kinase 2 (CDK2). [67]
Flavopiridol DMKSUOI Phase 2 Flavopiridol decreases the activity of Cyclin-dependent kinase 2 (CDK2). [68]
Plevitrexed DM7Y60I Phase 2 Plevitrexed increases the expression of Cyclin-dependent kinase 2 (CDK2). [69]
2-hydroxyoleic acid DMCLKA4 Phase 1/2 2-hydroxyoleic acid decreases the expression of Cyclin-dependent kinase 2 (CDK2). [70]
Benzo(a)pyrene DMN7J43 Phase 1 Benzo(a)pyrene decreases the expression of Cyclin-dependent kinase 2 (CDK2). [71]
(+)-JQ1 DM1CZSJ Phase 1 (+)-JQ1 decreases the expression of Cyclin-dependent kinase 2 (CDK2). [72]
TAK-114 DMTXE19 Phase 1 TAK-114 decreases the expression of Cyclin-dependent kinase 2 (CDK2). [74]
AMG 900 DMASGXJ Phase 1 AMG 900 decreases the expression of Cyclin-dependent kinase 2 (CDK2). [75]
PMID28460551-Compound-2 DM4DOUB Patented PMID28460551-Compound-2 decreases the expression of Cyclin-dependent kinase 2 (CDK2). [76]
PMID28870136-Compound-52 DMFDERP Patented PMID28870136-Compound-52 decreases the activity of Cyclin-dependent kinase 2 (CDK2). [77]
PMID26394986-Compound-22 DM43Z1G Patented PMID26394986-Compound-22 decreases the expression of Cyclin-dependent kinase 2 (CDK2). [78]
Flavonoid derivative 1 DMCQP0B Patented Flavonoid derivative 1 decreases the activity of Cyclin-dependent kinase 2 (CDK2). [79]
Steroid derivative 1 DMB0NVQ Patented Steroid derivative 1 decreases the expression of Cyclin-dependent kinase 2 (CDK2). [80]
KENPAULLONE DMAGVXW Patented KENPAULLONE decreases the activity of Cyclin-dependent kinase 2 (CDK2). [81]
Perillyl alcohol DMFWC3O Discontinued in Phase 2 Perillyl alcohol decreases the expression of Cyclin-dependent kinase 2 (CDK2). [82]
Lexacalcitol DMJ3ZT8 Discontinued in Phase 2 Lexacalcitol decreases the activity of Cyclin-dependent kinase 2 (CDK2). [42]
LG100268 DM41RK2 Discontinued in Phase 1 LG100268 decreases the expression of Cyclin-dependent kinase 2 (CDK2). [25]
THAPSIGARGIN DMDMQIE Preclinical THAPSIGARGIN decreases the expression of Cyclin-dependent kinase 2 (CDK2). [83]
Celastrol DMWQIJX Preclinical Celastrol decreases the expression of Cyclin-dependent kinase 2 (CDK2). [84]
Dioscin DM5H2W9 Preclinical Dioscin decreases the expression of Cyclin-dependent kinase 2 (CDK2). [85]
BMS-3870032 DMCWIBU Preclinical BMS-3870032 decreases the activity of Cyclin-dependent kinase 2 (CDK2). [86]
NS398 DMINUWH Terminated NS398 decreases the activity of Cyclin-dependent kinase 2 (CDK2). [32]
EMBELIN DMFZO4Y Terminated EMBELIN decreases the expression of Cyclin-dependent kinase 2 (CDK2). [87]
ABT-100 DMPFLS5 Terminated ABT-100 decreases the activity of Cyclin-dependent kinase 2 (CDK2). [88]
Bisphenol A DM2ZLD7 Investigative Bisphenol A decreases the expression of Cyclin-dependent kinase 2 (CDK2). [89]
------------------------------------------------------------------------------------
⏷ Show the Full List of 94 Drug(s)
1 Drug(s) Affected the Protein Interaction/Cellular Processes of This DOT
Drug Name Drug ID Highest Status Interaction REF
Tretinoin DM49DUI Approved Tretinoin decreases the degradation of Cyclin-dependent kinase 2 (CDK2). [3]
------------------------------------------------------------------------------------
5 Drug(s) Affected the Post-Translational Modifications of This DOT
Drug Name Drug ID Highest Status Interaction REF
Arsenic DMTL2Y1 Approved Arsenic affects the methylation of Cyclin-dependent kinase 2 (CDK2). [7]
Ciprofloxacin XR DM2NLS9 Approved Ciprofloxacin XR decreases the phosphorylation of Cyclin-dependent kinase 2 (CDK2). [45]
Tetracaine DM9J6C2 Approved Tetracaine decreases the phosphorylation of Cyclin-dependent kinase 2 (CDK2). [53]
phorbol 12-myristate 13-acetate DMJWD62 Phase 2 phorbol 12-myristate 13-acetate decreases the phosphorylation of Cyclin-dependent kinase 2 (CDK2). [66]
TAK-243 DM4GKV2 Phase 1 TAK-243 increases the sumoylation of Cyclin-dependent kinase 2 (CDK2). [73]
------------------------------------------------------------------------------------

References

1 Effects of valproic acid and levetiracetam on viability and cell cycle regulatory genes expression in the OVCAR-3 cell line. Pharmacol Rep. 2012;64(1):157-65. doi: 10.1016/s1734-1140(12)70742-9.
2 Comparison of HepG2 and HepaRG by whole-genome gene expression analysis for the purpose of chemical hazard identification. Toxicol Sci. 2010 May;115(1):66-79.
3 The ubiquitin-proteasome pathway plays essential roles in ATRA-induced leukemia cells G0/G1 phase arrest and transition into granulocytic differentiation. Cancer Biol Ther. 2010 Dec 1;10(11):1157-67. doi: 10.4161/cbt.10.11.13556. Epub 2010 Dec 1.
4 Low doses of cisplatin induce gene alterations, cell cycle arrest, and apoptosis in human promyelocytic leukemia cells. Biomark Insights. 2016 Aug 24;11:113-21.
5 Estrogen Regulates MAPK-Related Genes through Genomic and Nongenomic Interactions between IGF-I Receptor Tyrosine Kinase and Estrogen Receptor-Alpha Signaling Pathways in Human Uterine Leiomyoma Cells. J Signal Transduct. 2012;2012:204236. doi: 10.1155/2012/204236. Epub 2012 Oct 9.
6 Quantitative proteomics reveals a broad-spectrum antiviral property of ivermectin, benefiting for COVID-19 treatment. J Cell Physiol. 2021 Apr;236(4):2959-2975. doi: 10.1002/jcp.30055. Epub 2020 Sep 22.
7 Prenatal arsenic exposure and the epigenome: identifying sites of 5-methylcytosine alterations that predict functional changes in gene expression in newborn cord blood and subsequent birth outcomes. Toxicol Sci. 2015 Jan;143(1):97-106. doi: 10.1093/toxsci/kfu210. Epub 2014 Oct 10.
8 Inhibition of prostate cancer cell colony formation by the flavonoid quercetin correlates with modulation of specific regulatory genes. Clin Diagn Lab Immunol. 2004 Jan;11(1):63-9. doi: 10.1128/cdli.11.1.63-69.2004.
9 Essential role of cell cycle regulatory genes p21 and p27 expression in inhibition of breast cancer cells by arsenic trioxide. Med Oncol. 2011 Dec;28(4):1225-54.
10 Down-regulation and decreased activity of cyclin-dependent kinase 2 in H2O2-induced premature senescence. Int J Biochem Cell Biol. 2003 Feb;35(2):246-54. doi: 10.1016/s1357-2725(02)00129-2.
11 Effects of 1alpha,25 dihydroxyvitamin D3 and testosterone on miRNA and mRNA expression in LNCaP cells. Mol Cancer. 2011 May 18;10:58.
12 Drug-induced endoplasmic reticulum and oxidative stress responses independently sensitize toward TNF-mediated hepatotoxicity. Toxicol Sci. 2014 Jul;140(1):144-59. doi: 10.1093/toxsci/kfu072. Epub 2014 Apr 20.
13 Peripheral blood expression of nuclear factor-kappab-regulated genes is associated with rheumatoid arthritis disease activity and responds differentially to anti-tumor necrosis factor-alpha versus methotrexate. J Rheumatol. 2007 Sep;34(9):1817-22. Epub 2007 Aug 1.
14 Selenite and selenomethionine promote HL-60 cell cycle progression. J Nutr. 2002 Apr;132(4):674-9. doi: 10.1093/jn/132.4.674.
15 Progesterone inhibits human endothelial cell proliferation through a p53-dependent pathway. Cell Mol Life Sci. 2008 Nov;65(23):3839-50. doi: 10.1007/s00018-008-8441-3.
16 Transcriptional profiling of MCF7 breast cancer cells in response to 5-Fluorouracil: relationship with cell cycle changes and apoptosis, and identification of novel targets of p53. Int J Cancer. 2006 Sep 1;119(5):1164-75.
17 Estrogen receptor regulates E2F1 expression to mediate tamoxifen resistance. Mol Cancer Res. 2010 Mar;8(3):343-52. doi: 10.1158/1541-7786.MCR-09-0395. Epub 2010 Mar 9.
18 CDK2 is involved in the S-phase lengthening induced by glucocorticoids in normal human lymphocytes. Eur J Cell Biol. 2003 May;82(5):253-61. doi: 10.1078/0171-9335-00298.
19 Folic acid inhibits COLO-205 colon cancer cell proliferation through activating the FR/c-SRC/ERK1/2/NFB/TP53 pathway: in vitro and in vivo studies. Sci Rep. 2015 Jun 9;5:11187. doi: 10.1038/srep11187.
20 Characterization of formaldehyde's genotoxic mode of action by gene expression analysis in TK6 cells. Arch Toxicol. 2013 Nov;87(11):1999-2012.
21 Targeting of cell cycle and let-7a/STAT3 pathway by niclosamide inhibits proliferation, migration and invasion in oral squamous cell carcinoma cells. Biomed Pharmacother. 2017 Dec;96:434-442. doi: 10.1016/j.biopha.2017.09.149. Epub 2017 Oct 12.
22 Cannabidiol-induced transcriptomic changes and cellular senescence in human Sertoli cells. Toxicol Sci. 2023 Feb 17;191(2):227-238. doi: 10.1093/toxsci/kfac131.
23 The proteasome inhibitor bortezomib enhances the activity of docetaxel in orthotopic human pancreatic tumor xenografts. Mol Cancer Ther. 2004 Jan;3(1):59-70.
24 Effects of ciglitazone and troglitazone on the proliferation of human stomach cancer cells. World J Gastroenterol. 2009 Jan 21;15(3):310-20.
25 Thiazolidinediones inhibit proliferation of microvascular and macrovascular cells by a PPARgamma-independent mechanism. Diabetologia. 2005 Mar;48(3):586-94. doi: 10.1007/s00125-005-1672-z. Epub 2005 Feb 24.
26 Lunasin, a novel seed peptide, sensitizes human breast cancer MDA-MB-231 cells to aspirin-arrested cell cycle and induced apoptosis. Chem Biol Interact. 2010 Jul 30;186(2):127-34. doi: 10.1016/j.cbi.2010.04.027. Epub 2010 May 21.
27 New role for nuclear hormone receptors and coactivators in regulation of BRCA1-mediated DNA repair in breast cancer cell lines. Breast Cancer Res. 2006;8(1):R1. doi: 10.1186/bcr1362. Epub 2005 Dec 9.
28 Sequential dependent enhancement of caspase activation and apoptosis by flavopiridol on paclitaxel-treated human gastric and breast cancer cells. Clin Cancer Res. 1999 Jul;5(7):1876-83.
29 Apoptosis induced by piroxicam plus cisplatin combined treatment is triggered by p21 in mesothelioma. PLoS One. 2011;6(8):e23569.
30 Dasatinib reverses cancer-associated fibroblasts (CAFs) from primary lung carcinomas to a phenotype comparable to that of normal fibroblasts. Mol Cancer. 2010 Jun 27;9:168.
31 Malathion induced cancer-linked gene expression in human lymphocytes. Environ Res. 2020 Mar;182:109131. doi: 10.1016/j.envres.2020.109131. Epub 2020 Jan 10.
32 Nonsteroidal anti-inflammatory drugs inhibit growth of human neuroendocrine tumor cells via G1 cell-cycle arrest. Int J Cancer. 2003 Dec 10;107(5):844-53. doi: 10.1002/ijc.11446.
33 Chronic azacitidine treatment results in differentiating effects, sensitizes against bicalutamide in androgen-independent prostate cancer cells. Prostate. 2008 May 15;68(7):793-801. doi: 10.1002/pros.20748.
34 Simvastatin inhibits cell growth and induces apoptosis and G0/G1 cell cycle arrest in hepatic cancer cells. Int J Mol Med. 2010 Nov;26(5):735-41. doi: 10.3892/ijmm_00000520.
35 [Induction of cell cycle arrest in bladder cancer RT4 cells by capsaicin]. Zhonghua Yi Xue Za Zhi. 2010 May 11;90(18):1230-3.
36 Pharmacologic inhibition of CDK4/6: mechanistic evidence for selective activity or acquired resistance in acute myeloid leukemia. Blood. 2007 Sep 15;110(6):2075-83. doi: 10.1182/blood-2007-02-071266. Epub 2007 May 30.
37 Targeting the Na(+)/K(+)-ATPase alpha1 subunit of hepatoma HepG2 cell line to induce apoptosis and cell cycle arresting. Biol Pharm Bull. 2010;33(5):743-51. doi: 10.1248/bpb.33.743.
38 Motexafin gadolinium and zinc induce oxidative stress responses and apoptosis in B-cell lymphoma lines. Cancer Res. 2005 Dec 15;65(24):11676-88.
39 Novel carbocyclic curcumin analog CUR3d modulates genes involved in multiple apoptosis pathways in human hepatocellular carcinoma cells. Chem Biol Interact. 2015 Dec 5;242:107-22.
40 Imatinib mesylate, a new kid on the block for the treatment of anti-neutrophil cytoplasmic autoantibodies-associated vasculitis?. Clin Exp Immunol. 2008 Mar;151(3):391-8. doi: 10.1111/j.1365-2249.2007.03572.x. Epub 2008 Jan 10.
41 Ritonavir blocks AKT signaling, activates apoptosis and inhibits migration and invasion in ovarian cancer cells. Mol Cancer. 2009 Apr 22;8:26. doi: 10.1186/1476-4598-8-26.
42 Inhibition of MG-63 cell cycle progression by synthetic vitamin D3 analogs mediated by p27, Cdk2, cyclin E, and the retinoblastoma protein. Biochem Pharmacol. 2003 Aug 1;66(3):495-504. doi: 10.1016/s0006-2952(03)00283-1.
43 Lovastatin inhibits G1/S transition of normal human B-lymphocytes independent of apoptosis. Exp Cell Res. 1999 Oct 10;252(1):144-53. doi: 10.1006/excr.1999.4608.
44 Orlistat Displays Antitumor Activity and Enhances the Efficacy of Paclitaxel in Human Hepatoma Hep3B Cells. Chem Res Toxicol. 2019 Feb 18;32(2):255-264. doi: 10.1021/acs.chemrestox.8b00269. Epub 2019 Jan 22.
45 Ciprofloxacin mediated cell growth inhibition, S/G2-M cell cycle arrest, and apoptosis in a human transitional cell carcinoma of the bladder cell line. Clin Cancer Res. 2000 Mar;6(3):891-900.
46 Hesperetin induced G1-phase cell cycle arrest in human breast cancer MCF-7 cells: involvement of CDK4 and p21. Nutr Cancer. 2007;59(1):115-9. doi: 10.1080/01635580701419030.
47 HIV protease inhibitor nelfinavir inhibits growth of human melanoma cells by induction of cell cycle arrest. Cancer Res. 2007 Feb 1;67(3):1221-7. doi: 10.1158/0008-5472.CAN-06-3377.
48 Homoharringtonine suppresses LoVo cell growth by inhibiting EphB4 and the PI3K/AKT and MAPK/EKR1/2 signaling pathways. Food Chem Toxicol. 2020 Feb;136:110960. doi: 10.1016/j.fct.2019.110960. Epub 2019 Nov 11.
49 Mechanisms by which S-albuterol induces human bronchial smooth muscle cell proliferation. Int Arch Allergy Immunol. 2008;146(4):321-33. doi: 10.1159/000121466. Epub 2008 Mar 21.
50 G2 delay induced by nitrogen mustard in human cells affects cyclin A/cdk2 and cyclin B1/cdc2-kinase complexes differently. J Biol Chem. 1993 Apr 15;268(11):8298-308.
51 The role of hepatic cytochrome P450s in the cytotoxicity of dronedarone. Arch Toxicol. 2018 Jun;92(6):1969-1981. doi: 10.1007/s00204-018-2196-x. Epub 2018 Apr 3.
52 G1 cell cycle arrest by amlodipine, a dihydropyridine Ca2+ channel blocker, in human epidermoid carcinoma A431 cells. Biochem Pharmacol. 2007 Apr 1;73(7):943-53. doi: 10.1016/j.bcp.2006.12.011. Epub 2006 Dec 14.
53 Tetracaine hydrochloride induces cell cycle arrest in melanoma by downregulating hnRNPA1. Toxicol Appl Pharmacol. 2022 Jan 1;434:115810. doi: 10.1016/j.taap.2021.115810. Epub 2021 Nov 23.
54 Iron chelators ICL670 and 311 inhibit HIV-1 transcription. Virology. 2007 Oct 25;367(2):324-33. doi: 10.1016/j.virol.2007.06.011. Epub 2007 Jul 13.
55 Bone morphogenetic protein-2 induces hypophosphorylation of Rb protein and repression of E2F in androgen-treated LNCaP human prostate cancer cells. Int J Mol Med. 2005 Feb;15(2):253-8.
56 Berberine, a natural product, induces G1-phase cell cycle arrest and caspase-3-dependent apoptosis in human prostate carcinoma cells. Mol Cancer Ther. 2006 Feb;5(2):296-308. doi: 10.1158/1535-7163.MCT-05-0448.
57 Identifying the differential effects of silymarin constituents on cell growth and cell cycle regulatory molecules in human prostate cancer cells. Int J Cancer. 2008 Jul 1;123(1):41-50. doi: 10.1002/ijc.23485.
58 Influence of resveratrol on rheumatoid fibroblast-like synoviocytes analysed with gene chip transcription. Phytomedicine. 2013 Feb 15;20(3-4):310-8. doi: 10.1016/j.phymed.2012.09.020. Epub 2012 Nov 6.
59 Inhibition of cell-cycle progression in human colorectal carcinoma Lovo cells by andrographolide. Chem Biol Interact. 2008 Aug 11;174(3):201-10. doi: 10.1016/j.cbi.2008.06.006. Epub 2008 Jun 20.
60 N-Alkoxy derivatization of indole-3-carbinol increases the efficacy of the G1 cell cycle arrest and of I3C-specific regulation of cell cycle gene transcription and activity in human breast cancer cells. Biochem Pharmacol. 2008 Feb 1;75(3):713-24. doi: 10.1016/j.bcp.2007.09.024. Epub 2007 Oct 2.
61 Triptolide inhibits the growth and metastasis of solid tumors. Mol Cancer Ther. 2003 Jan;2(1):65-72.
62 Bergapten induces G1 arrest and pro-apoptotic cascade in colorectal cancer cells associating with p53/p21/PTEN axis. Environ Toxicol. 2019 Mar;34(3):303-311. doi: 10.1002/tox.22685. Epub 2018 Dec 21.
63 Convergent transcriptional profiles induced by endogenous estrogen and distinct xenoestrogens in breast cancer cells. Carcinogenesis. 2006 Aug;27(8):1567-78.
64 Thymoquinone suppresses invasion and metastasis in bladder cancer cells by reversing EMT through the Wnt/-catenin signaling pathway. Chem Biol Interact. 2020 Apr 1;320:109022. doi: 10.1016/j.cbi.2020.109022. Epub 2020 Feb 27.
65 Novel ATP-competitive Akt inhibitor afuresertib suppresses the proliferation of malignant pleural mesothelioma cells. Cancer Med. 2017 Nov;6(11):2646-2659. doi: 10.1002/cam4.1179. Epub 2017 Sep 27.
66 Signal transduction of phorbol 12-myristate 13-acetate (PMA)-induced growth inhibition of human monocytic leukemia THP-1 cells is reactive oxygen dependent. Leuk Res. 2005 Aug;29(8):863-79. doi: 10.1016/j.leukres.2004.12.011. Epub 2005 Feb 24.
67 Phenethyl isothiocyanate alters the gene expression and the levels of protein associated with cell cycle regulation in human glioblastoma GBM 8401 cells. Environ Toxicol. 2017 Jan;32(1):176-187.
68 Flavopiridol down-regulates antiapoptotic proteins and sensitizes human breast cancer cells to epothilone B-induced apoptosis. Cancer Res. 2003 Jan 1;63(1):93-9.
69 P21Cip1 is a critical mediator of the cytotoxic action of thymidylate synthase inhibitors in colorectal carcinoma cells. Cancer Res. 2004 Sep 1;64(17):6296-303. doi: 10.1158/0008-5472.CAN-04-0863.
70 The repression of E2F-1 is critical for the activity of Minerval against cancer. J Pharmacol Exp Ther. 2005 Oct;315(1):466-74. doi: 10.1124/jpet.105.088716. Epub 2005 Jul 18.
71 Comparison of phenotypic and transcriptomic effects of false-positive genotoxins, true genotoxins and non-genotoxins using HepG2 cells. Mutagenesis. 2011 Sep;26(5):593-604.
72 Inhibition of BRD4 attenuates tumor cell self-renewal and suppresses stem cell signaling in MYC driven medulloblastoma. Oncotarget. 2014 May 15;5(9):2355-71.
73 Inhibiting ubiquitination causes an accumulation of SUMOylated newly synthesized nuclear proteins at PML bodies. J Biol Chem. 2019 Oct 18;294(42):15218-15234. doi: 10.1074/jbc.RA119.009147. Epub 2019 Jul 8.
74 Natura-alpha targets forkhead box m1 and inhibits androgen-dependent and -independent prostate cancer growth and invasion. Clin Cancer Res. 2011 Jul 1;17(13):4414-24. doi: 10.1158/1078-0432.CCR-11-0431. Epub 2011 May 23.
75 AMG900 as novel inhibitor of the translationally controlled tumor protein. Chem Biol Interact. 2021 Jan 25;334:109349. doi: 10.1016/j.cbi.2020.109349. Epub 2020 Nov 28.
76 Cell-based two-dimensional morphological assessment system to predict cancer drug-induced cardiotoxicity using human induced pluripotent stem cell-derived cardiomyocytes. Toxicol Appl Pharmacol. 2019 Nov 15;383:114761. doi: 10.1016/j.taap.2019.114761. Epub 2019 Sep 15.
77 Caffeine induces TP53-independent G(1)-phase arrest and apoptosis in human lung tumor cells in a dose-dependent manner. Radiat Res. 2002 Feb;157(2):166-74. doi: 10.1667/0033-7587(2002)157[0166:citigp]2.0.co;2.
78 [Effects of celecoxib on the proliferation and apoptosis of human colorectal cancer cell line HT-29]. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2004 Jun;29(3):261-5.
79 Effects of structurally related flavonoids on cell cycle progression of human melanoma cells: regulation of cyclin-dependent kinases CDK2 and CDK1. Biochem Pharmacol. 2001 May 15;61(10):1205-15. doi: 10.1016/s0006-2952(01)00583-4.
80 2-methoxyestradiol induces mitotic arrest, apoptosis, and synergistic cytotoxicity with arsenic trioxide in human urothelial carcinoma cells. PLoS One. 2013 Aug 13;8(8):e68703. doi: 10.1371/journal.pone.0068703. eCollection 2013.
81 The specificities of protein kinase inhibitors: an update. Biochem J. 2003 Apr 1;371(Pt 1):199-204. doi: 10.1042/BJ20021535.
82 Cell cycle arrest by the isoprenoids perillyl alcohol, geraniol, and farnesol is mediated by p21(Cip1) and p27(Kip1) in human pancreatic adenocarcinoma cells. J Pharmacol Exp Ther. 2007 Mar;320(3):1163-70. doi: 10.1124/jpet.106.111666. Epub 2006 Nov 30.
83 Endoplasmic reticulum stress impairs insulin signaling through mitochondrial damage in SH-SY5Y cells. Neurosignals. 2012;20(4):265-80.
84 Gene expression signature-based chemical genomic prediction identifies a novel class of HSP90 pathway modulators. Cancer Cell. 2006 Oct;10(4):321-30.
85 Dioscin inhibits human endometrial carcinoma proliferation via G0/G1 cell cycle arrest and mitochondrial-dependent signaling pathway. Food Chem Toxicol. 2021 Feb;148:111941. doi: 10.1016/j.fct.2020.111941. Epub 2020 Dec 24.
86 Characterization of molecular and cellular functions of the cyclin-dependent kinase CDK9 using a novel specific inhibitor. Br J Pharmacol. 2014 Jan;171(1):55-68. doi: 10.1111/bph.12408.
87 Embelin suppresses growth of human pancreatic cancer xenografts, and pancreatic cancer cells isolated from KrasG12D mice by inhibiting Akt and Sonic hedgehog pathways. PLoS One. 2014 Apr 2;9(4):e92161.
88 Farnesyltransferase inhibitor, ABT-100, is a potent liver cancer chemopreventive agent. Clin Cancer Res. 2005 Jun 1;11(11):4266-74. doi: 10.1158/1078-0432.CCR-04-2386.
89 Bisphenol A induces DSB-ATM-p53 signaling leading to cell cycle arrest, senescence, autophagy, stress response, and estrogen release in human fetal lung fibroblasts. Arch Toxicol. 2018 Apr;92(4):1453-1469.
90 TNF-alpha promotes Doxorubicin-induced cell apoptosis and anti-cancer effect through downregulation of p21 in p53-deficient tumor cells. Biochem Biophys Res Commun. 2005 May 20;330(4):1034-40. doi: 10.1016/j.bbrc.2005.02.188.
91 Gene expression profiling of 30 cancer cell lines predicts resistance towards 11 anticancer drugs at clinically achieved concentrations. Int J Cancer. 2006 Apr 1;118(7):1699-712. doi: 10.1002/ijc.21570.