General Information of Drug Off-Target (DOT) (ID: OTA8N1QI)

DOT Name Proliferation marker protein Ki-67 (MKI67)
Synonyms Antigen identified by monoclonal antibody Ki-67; Antigen KI-67; Antigen Ki67
Gene Name MKI67
UniProt ID
KI67_HUMAN
PDB ID
1R21; 2AFF; 5J28
Pfam ID
PF00498 ; PF08065 ; PF15276
Sequence
MWPTRRLVTIKRSGVDGPHFPLSLSTCLFGRGIECDIRIQLPVVSKQHCKIEIHEQEAIL
HNFSSTNPTQVNGSVIDEPVRLKHGDVITIIDRSFRYENESLQNGRKSTEFPRKIREQEP
ARRVSRSSFSSDPDEKAQDSKAYSKITEGKVSGNPQVHIKNVKEDSTADDSKDSVAQGTT
NVHSSEHAGRNGRNAADPISGDFKEISSVKLVSRYGELKSVPTTQCLDNSKKNESPFWKL
YESVKKELDVKSQKENVLQYCRKSGLQTDYATEKESADGLQGETQLLVSRKSRPKSGGSG
HAVAEPASPEQELDQNKGKGRDVESVQTPSKAVGASFPLYEPAKMKTPVQYSQQQNSPQK
HKNKDLYTTGRRESVNLGKSEGFKAGDKTLTPRKLSTRNRTPAKVEDAADSATKPENLSS
KTRGSIPTDVEVLPTETEIHNEPFLTLWLTQVERKIQKDSLSKPEKLGTTAGQMCSGLPG
LSSVDINNFGDSINESEGIPLKRRRVSFGGHLRPELFDENLPPNTPLKRGEAPTKRKSLV
MHTPPVLKKIIKEQPQPSGKQESGSEIHVEVKAQSLVISPPAPSPRKTPVASDQRRRSCK
TAPASSSKSQTEVPKRGGRKSGNLPSKRVSISRSQHDILQMICSKRRSGASEANLIVAKS
WADVVKLGAKQTQTKVIKHGPQRSMNKRQRRPATPKKPVGEVHSQFSTGHANSPCTIIIG
KAHTEKVHVPARPYRVLNNFISNQKMDFKEDLSGIAEMFKTPVKEQPQLTSTCHIAISNS
ENLLGKQFQGTDSGEEPLLPTSESFGGNVFFSAQNAAKQPSDKCSASPPLRRQCIRENGN
VAKTPRNTYKMTSLETKTSDTETEPSKTVSTANRSGRSTEFRNIQKLPVESKSEETNTEI
VECILKRGQKATLLQQRREGEMKEIERPFETYKENIELKENDEKMKAMKRSRTWGQKCAP
MSDLTDLKSLPDTELMKDTARGQNLLQTQDHAKAPKSEKGKITKMPCQSLQPEPINTPTH
TKQQLKASLGKVGVKEELLAVGKFTRTSGETTHTHREPAGDGKSIRTFKESPKQILDPAA
RVTGMKKWPRTPKEEAQSLEDLAGFKELFQTPGPSEESMTDEKTTKIACKSPPPESVDTP
TSTKQWPKRSLRKADVEEEFLALRKLTPSAGKAMLTPKPAGGDEKDIKAFMGTPVQKLDL
AGTLPGSKRQLQTPKEKAQALEDLAGFKELFQTPGHTEELVAAGKTTKIPCDSPQSDPVD
TPTSTKQRPKRSIRKADVEGELLACRNLMPSAGKAMHTPKPSVGEEKDIIIFVGTPVQKL
DLTENLTGSKRRPQTPKEEAQALEDLTGFKELFQTPGHTEEAVAAGKTTKMPCESSPPES
ADTPTSTRRQPKTPLEKRDVQKELSALKKLTQTSGETTHTDKVPGGEDKSINAFRETAKQ
KLDPAASVTGSKRHPKTKEKAQPLEDLAGLKELFQTPVCTDKPTTHEKTTKIACRSQPDP
VDTPTSSKPQSKRSLRKVDVEEEFFALRKRTPSAGKAMHTPKPAVSGEKNIYAFMGTPVQ
KLDLTENLTGSKRRLQTPKEKAQALEDLAGFKELFQTRGHTEESMTNDKTAKVACKSSQP
DPDKNPASSKRRLKTSLGKVGVKEELLAVGKLTQTSGETTHTHTEPTGDGKSMKAFMESP
KQILDSAASLTGSKRQLRTPKGKSEVPEDLAGFIELFQTPSHTKESMTNEKTTKVSYRAS
QPDLVDTPTSSKPQPKRSLRKADTEEEFLAFRKQTPSAGKAMHTPKPAVGEEKDINTFLG
TPVQKLDQPGNLPGSNRRLQTRKEKAQALEELTGFRELFQTPCTDNPTTDEKTTKKILCK
SPQSDPADTPTNTKQRPKRSLKKADVEEEFLAFRKLTPSAGKAMHTPKAAVGEEKDINTF
VGTPVEKLDLLGNLPGSKRRPQTPKEKAKALEDLAGFKELFQTPGHTEESMTDDKITEVS
CKSPQPDPVKTPTSSKQRLKISLGKVGVKEEVLPVGKLTQTSGKTTQTHRETAGDGKSIK
AFKESAKQMLDPANYGTGMERWPRTPKEEAQSLEDLAGFKELFQTPDHTEESTTDDKTTK
IACKSPPPESMDTPTSTRRRPKTPLGKRDIVEELSALKQLTQTTHTDKVPGDEDKGINVF
RETAKQKLDPAASVTGSKRQPRTPKGKAQPLEDLAGLKELFQTPICTDKPTTHEKTTKIA
CRSPQPDPVGTPTIFKPQSKRSLRKADVEEESLALRKRTPSVGKAMDTPKPAGGDEKDMK
AFMGTPVQKLDLPGNLPGSKRWPQTPKEKAQALEDLAGFKELFQTPGTDKPTTDEKTTKI
ACKSPQPDPVDTPASTKQRPKRNLRKADVEEEFLALRKRTPSAGKAMDTPKPAVSDEKNI
NTFVETPVQKLDLLGNLPGSKRQPQTPKEKAEALEDLVGFKELFQTPGHTEESMTDDKIT
EVSCKSPQPESFKTSRSSKQRLKIPLVKVDMKEEPLAVSKLTRTSGETTQTHTEPTGDSK
SIKAFKESPKQILDPAASVTGSRRQLRTRKEKARALEDLVDFKELFSAPGHTEESMTIDK
NTKIPCKSPPPELTDTATSTKRCPKTRPRKEVKEELSAVERLTQTSGQSTHTHKEPASGD
EGIKVLKQRAKKKPNPVEEEPSRRRPRAPKEKAQPLEDLAGFTELSETSGHTQESLTAGK
ATKIPCESPPLEVVDTTASTKRHLRTRVQKVQVKEEPSAVKFTQTSGETTDADKEPAGED
KGIKALKESAKQTPAPAASVTGSRRRPRAPRESAQAIEDLAGFKDPAAGHTEESMTDDKT
TKIPCKSSPELEDTATSSKRRPRTRAQKVEVKEELLAVGKLTQTSGETTHTDKEPVGEGK
GTKAFKQPAKRKLDAEDVIGSRRQPRAPKEKAQPLEDLASFQELSQTPGHTEELANGAAD
SFTSAPKQTPDSGKPLKISRRVLRAPKVEPVGDVVSTRDPVKSQSKSNTSLPPLPFKRGG
GKDGSVTGTKRLRCMPAPEEIVEELPASKKQRVAPRARGKSSEPVVIMKRSLRTSAKRIE
PAEELNSNDMKTNKEEHKLQDSVPENKGISLRSRRQNKTEAEQQITEVFVLAERIEINRN
EKKPMKTSPEMDIQNPDDGARKPIPRDKVTENKRCLRSARQNESSQPKVAEESGGQKSAK
VLMQNQKGKGEAGNSDSMCLRSRKTKSQPAASTLESKSVQRVTRSVKRCAENPKKAEDNV
CVKKIRTRSHRDSEDI
Function
Required to maintain individual mitotic chromosomes dispersed in the cytoplasm following nuclear envelope disassembly. Associates with the surface of the mitotic chromosome, the perichromosomal layer, and covers a substantial fraction of the chromosome surface. Prevents chromosomes from collapsing into a single chromatin mass by forming a steric and electrostatic charge barrier: the protein has a high net electrical charge and acts as a surfactant, dispersing chromosomes and enabling independent chromosome motility. Binds DNA, with a preference for supercoiled DNA and AT-rich DNA. Does not contribute to the internal structure of mitotic chromosomes. May play a role in chromatin organization. It is however unclear whether it plays a direct role in chromatin organization or whether it is an indirect consequence of its function in maintaining mitotic chromosomes dispersed (Probable).

Molecular Interaction Atlas (MIA) of This DOT

Molecular Interaction Atlas (MIA) Jump to Detail Molecular Interaction Atlas of This DOT
87 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 Proliferation marker protein Ki-67 (MKI67). [1]
Ciclosporin DMAZJFX Approved Ciclosporin decreases the expression of Proliferation marker protein Ki-67 (MKI67). [2]
Tretinoin DM49DUI Approved Tretinoin decreases the expression of Proliferation marker protein Ki-67 (MKI67). [3]
Acetaminophen DMUIE76 Approved Acetaminophen decreases the expression of Proliferation marker protein Ki-67 (MKI67). [4]
Doxorubicin DMVP5YE Approved Doxorubicin decreases the expression of Proliferation marker protein Ki-67 (MKI67). [5]
Cupric Sulfate DMP0NFQ Approved Cupric Sulfate affects the expression of Proliferation marker protein Ki-67 (MKI67). [6]
Cisplatin DMRHGI9 Approved Cisplatin decreases the expression of Proliferation marker protein Ki-67 (MKI67). [7]
Estradiol DMUNTE3 Approved Estradiol increases the expression of Proliferation marker protein Ki-67 (MKI67). [8]
Ivermectin DMDBX5F Approved Ivermectin decreases the expression of Proliferation marker protein Ki-67 (MKI67). [9]
Arsenic trioxide DM61TA4 Approved Arsenic trioxide decreases the expression of Proliferation marker protein Ki-67 (MKI67). [11]
Calcitriol DM8ZVJ7 Approved Calcitriol decreases the expression of Proliferation marker protein Ki-67 (MKI67). [12]
Testosterone DM7HUNW Approved Testosterone decreases the expression of Proliferation marker protein Ki-67 (MKI67). [12]
Triclosan DMZUR4N Approved Triclosan decreases the expression of Proliferation marker protein Ki-67 (MKI67). [13]
Carbamazepine DMZOLBI Approved Carbamazepine affects the expression of Proliferation marker protein Ki-67 (MKI67). [14]
Methotrexate DM2TEOL Approved Methotrexate decreases the expression of Proliferation marker protein Ki-67 (MKI67). [15]
Marinol DM70IK5 Approved Marinol increases the expression of Proliferation marker protein Ki-67 (MKI67). [16]
Progesterone DMUY35B Approved Progesterone increases the expression of Proliferation marker protein Ki-67 (MKI67). [17]
Fluorouracil DMUM7HZ Approved Fluorouracil decreases the expression of Proliferation marker protein Ki-67 (MKI67). [5]
Dexamethasone DMMWZET Approved Dexamethasone decreases the expression of Proliferation marker protein Ki-67 (MKI67). [18]
Folic acid DMEMBJC Approved Folic acid affects the expression of Proliferation marker protein Ki-67 (MKI67). [19]
Niclosamide DMJAGXQ Approved Niclosamide decreases the expression of Proliferation marker protein Ki-67 (MKI67). [20]
Bortezomib DMNO38U Approved Bortezomib decreases the expression of Proliferation marker protein Ki-67 (MKI67). [21]
Troglitazone DM3VFPD Approved Troglitazone decreases the expression of Proliferation marker protein Ki-67 (MKI67). [22]
Rosiglitazone DMILWZR Approved Rosiglitazone decreases the expression of Proliferation marker protein Ki-67 (MKI67). [23]
Azathioprine DMMZSXQ Approved Azathioprine decreases the expression of Proliferation marker protein Ki-67 (MKI67). [24]
Ethanol DMDRQZU Approved Ethanol decreases the expression of Proliferation marker protein Ki-67 (MKI67). [25]
Piroxicam DMTK234 Approved Piroxicam decreases the expression of Proliferation marker protein Ki-67 (MKI67). [26]
Dasatinib DMJV2EK Approved Dasatinib decreases the expression of Proliferation marker protein Ki-67 (MKI67). [27]
Gemcitabine DMSE3I7 Approved Gemcitabine decreases the expression of Proliferation marker protein Ki-67 (MKI67). [28]
Mifepristone DMGZQEF Approved Mifepristone decreases the expression of Proliferation marker protein Ki-67 (MKI67). [29]
Lucanthone DMZLBUO Approved Lucanthone decreases the expression of Proliferation marker protein Ki-67 (MKI67). [30]
Methamphetamine DMPM4SK Approved Methamphetamine decreases the expression of Proliferation marker protein Ki-67 (MKI67). [31]
Palbociclib DMD7L94 Approved Palbociclib decreases the expression of Proliferation marker protein Ki-67 (MKI67). [32]
Thalidomide DM70BU5 Approved Thalidomide decreases the expression of Proliferation marker protein Ki-67 (MKI67). [33]
Bicalutamide DMZMSPF Approved Bicalutamide decreases the expression of Proliferation marker protein Ki-67 (MKI67). [34]
Docetaxel DMDI269 Approved Docetaxel decreases the expression of Proliferation marker protein Ki-67 (MKI67). [35]
Propofol DMB4OLE Approved Propofol decreases the expression of Proliferation marker protein Ki-67 (MKI67). [36]
Melatonin DMKWFBT Approved Melatonin decreases the expression of Proliferation marker protein Ki-67 (MKI67). [37]
Sevoflurane DMC9O43 Approved Sevoflurane increases the expression of Proliferation marker protein Ki-67 (MKI67). [36]
Adenosine DMM2NSK Approved Adenosine affects the expression of Proliferation marker protein Ki-67 (MKI67). [38]
Ximelegatran DMU8ANS Approved Ximelegatran decreases the expression of Proliferation marker protein Ki-67 (MKI67). [39]
Propranolol DM79NTF Approved Propranolol decreases the expression of Proliferation marker protein Ki-67 (MKI67). [40]
LY2835219 DM93VBZ Approved LY2835219 decreases the expression of Proliferation marker protein Ki-67 (MKI67). [41]
Exemestane DM9HPW3 Approved Exemestane decreases the expression of Proliferation marker protein Ki-67 (MKI67). [42]
Omacetaxine mepesuccinate DMPU2WX Approved Omacetaxine mepesuccinate decreases the expression of Proliferation marker protein Ki-67 (MKI67). [43]
Ketamine DMT5HA4 Approved Ketamine increases the expression of Proliferation marker protein Ki-67 (MKI67). [44]
Ropivacaine DMSPJG2 Approved Ropivacaine decreases the expression of Proliferation marker protein Ki-67 (MKI67). [45]
Olaparib DM8QB1D Approved Olaparib decreases the expression of Proliferation marker protein Ki-67 (MKI67). [46]
Letrozole DMH07Y3 Approved Letrozole decreases the expression of Proliferation marker protein Ki-67 (MKI67). [42]
Levonorgestrel DM1DP7T Approved Levonorgestrel decreases the expression of Proliferation marker protein Ki-67 (MKI67). [47]
OPC-34712 DMHG57U Approved OPC-34712 decreases the expression of Proliferation marker protein Ki-67 (MKI67). [48]
Anastrozole DMNP60F Approved Anastrozole decreases the expression of Proliferation marker protein Ki-67 (MKI67). [49]
Resveratrol DM3RWXL Phase 3 Resveratrol decreases the expression of Proliferation marker protein Ki-67 (MKI67). [50]
Curcumin DMQPH29 Phase 3 Curcumin decreases the expression of Proliferation marker protein Ki-67 (MKI67). [51]
Chlorpromazine DMBGZI3 Phase 3 Trial Chlorpromazine decreases the expression of Proliferation marker protein Ki-67 (MKI67). [52]
Triptolide DMCMDVR Phase 3 Triptolide decreases the expression of Proliferation marker protein Ki-67 (MKI67). [53]
Remdesivir DMBFZ6L Phase 3 Trial Remdesivir decreases the expression of Proliferation marker protein Ki-67 (MKI67). [54]
OSI-906 DMHKZLF Phase 3 OSI-906 decreases the expression of Proliferation marker protein Ki-67 (MKI67). [55]
Thymoquinone DMVDTR2 Phase 2/3 Thymoquinone decreases the expression of Proliferation marker protein Ki-67 (MKI67). [56]
INK128 DMGO7QT Phase 2 INK128 decreases the expression of Proliferation marker protein Ki-67 (MKI67). [58]
STX-140 DMJK5CT Phase 2 STX-140 decreases the expression of Proliferation marker protein Ki-67 (MKI67). [59]
ME-344 DM6JN19 Phase 1/2 ME-344 decreases the expression of Proliferation marker protein Ki-67 (MKI67). [60]
INCB054329 DM7TZL0 Phase 1/2 INCB054329 decreases the expression of Proliferation marker protein Ki-67 (MKI67). [46]
(+)-JQ1 DM1CZSJ Phase 1 (+)-JQ1 decreases the expression of Proliferation marker protein Ki-67 (MKI67). [61]
Leflunomide DMR8ONJ Phase 1 Trial Leflunomide decreases the expression of Proliferation marker protein Ki-67 (MKI67). [62]
Aminoguanidine DMJQDUC Phase 1 Aminoguanidine decreases the expression of Proliferation marker protein Ki-67 (MKI67). [63]
PMID28460551-Compound-2 DM4DOUB Patented PMID28460551-Compound-2 decreases the expression of Proliferation marker protein Ki-67 (MKI67). [64]
PMID26394986-Compound-22 DM43Z1G Patented PMID26394986-Compound-22 decreases the expression of Proliferation marker protein Ki-67 (MKI67). [65]
Piperazinyl methyl quinazolinone derivative 2 DM913KS Patented Piperazinyl methyl quinazolinone derivative 2 decreases the expression of Proliferation marker protein Ki-67 (MKI67). [66]
THAPSIGARGIN DMDMQIE Preclinical THAPSIGARGIN decreases the expression of Proliferation marker protein Ki-67 (MKI67). [67]
NS398 DMINUWH Terminated NS398 decreases the expression of Proliferation marker protein Ki-67 (MKI67). [11]
EMBELIN DMFZO4Y Terminated EMBELIN decreases the expression of Proliferation marker protein Ki-67 (MKI67). [68]
ABT-737 DML0DBV Terminated ABT-737 decreases the expression of Proliferation marker protein Ki-67 (MKI67). [69]
Bisphenol A DM2ZLD7 Investigative Bisphenol A decreases the expression of Proliferation marker protein Ki-67 (MKI67). [70]
Trichostatin A DM9C8NX Investigative Trichostatin A decreases the expression of Proliferation marker protein Ki-67 (MKI67). [71]
Coumestrol DM40TBU Investigative Coumestrol increases the expression of Proliferation marker protein Ki-67 (MKI67). [72]
Deguelin DMXT7WG Investigative Deguelin decreases the expression of Proliferation marker protein Ki-67 (MKI67). [73]
methyl p-hydroxybenzoate DMO58UW Investigative methyl p-hydroxybenzoate increases the expression of Proliferation marker protein Ki-67 (MKI67). [74]
geraniol DMS3CBD Investigative geraniol decreases the expression of Proliferation marker protein Ki-67 (MKI67). [76]
Rapamycin Immunosuppressant Drug DM678IB Investigative Rapamycin Immunosuppressant Drug decreases the expression of Proliferation marker protein Ki-67 (MKI67). [69]
Cordycepin DM72Y01 Investigative Cordycepin increases the expression of Proliferation marker protein Ki-67 (MKI67). [38]
BRN-3548355 DM4KXT0 Investigative BRN-3548355 increases the expression of Proliferation marker protein Ki-67 (MKI67). [77]
Wogonin DMGCF51 Investigative Wogonin decreases the expression of Proliferation marker protein Ki-67 (MKI67). [78]
Icariside II DM3DB8X Investigative Icariside II decreases the expression of Proliferation marker protein Ki-67 (MKI67). [79]
ABBV-744 DMTEA9C Investigative ABBV-744 decreases the expression of Proliferation marker protein Ki-67 (MKI67). [80]
Silmitasertib DMQIBZD Investigative Silmitasertib increases the expression of Proliferation marker protein Ki-67 (MKI67). [81]
2-chloro-5-nitro-N-phenylbenzamide DMUGQIV Investigative 2-chloro-5-nitro-N-phenylbenzamide increases the expression of Proliferation marker protein Ki-67 (MKI67). [82]
------------------------------------------------------------------------------------
⏷ Show the Full List of 87 Drug(s)
4 Drug(s) Affected the Post-Translational Modifications of This DOT
Drug Name Drug ID Highest Status Interaction REF
Quercetin DM3NC4M Approved Quercetin affects the phosphorylation of Proliferation marker protein Ki-67 (MKI67). [10]
PMID28870136-Compound-52 DMFDERP Patented PMID28870136-Compound-52 affects the phosphorylation of Proliferation marker protein Ki-67 (MKI67). [10]
Coumarin DM0N8ZM Investigative Coumarin affects the phosphorylation of Proliferation marker protein Ki-67 (MKI67). [10]
Hexadecanoic acid DMWUXDZ Investigative Hexadecanoic acid increases the phosphorylation of Proliferation marker protein Ki-67 (MKI67). [75]
------------------------------------------------------------------------------------
1 Drug(s) Affected the Protein Interaction/Cellular Processes of This DOT
Drug Name Drug ID Highest Status Interaction REF
DNCB DMDTVYC Phase 2 DNCB affects the binding of Proliferation marker protein Ki-67 (MKI67). [57]
------------------------------------------------------------------------------------

References

1 Stem cell transcriptome responses and corresponding biomarkers that indicate the transition from adaptive responses to cytotoxicity. Chem Res Toxicol. 2017 Apr 17;30(4):905-922.
2 Inter-laboratory comparison of human renal proximal tubule (HK-2) transcriptome alterations due to Cyclosporine A exposure and medium exhaustion. Toxicol In Vitro. 2009 Apr;23(3):486-99.
3 Effect of retinoic acid on gene expression in human conjunctival epithelium: secretory phospholipase A2 mediates retinoic acid induction of MUC16. Invest Ophthalmol Vis Sci. 2005 Nov;46(11):4050-61.
4 Multiple microRNAs function as self-protective modules in acetaminophen-induced hepatotoxicity in humans. Arch Toxicol. 2018 Feb;92(2):845-858.
5 Cell-type-specific responses to chemotherapeutics in breast cancer. Cancer Res. 2004 Jun 15;64(12):4218-26.
6 Physiological and toxicological transcriptome changes in HepG2 cells exposed to copper. Physiol Genomics. 2009 Aug 7;38(3):386-401.
7 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.
8 Global gene expression profiles induced by phytoestrogens in human breast cancer cells. Endocr Relat Cancer. 2008 Mar;15(1):161-73.
9 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.
10 Quantitative phosphoproteomics reveal cellular responses from caffeine, coumarin and quercetin in treated HepG2 cells. Toxicol Appl Pharmacol. 2022 Aug 15;449:116110. doi: 10.1016/j.taap.2022.116110. Epub 2022 Jun 7.
11 Mitophagy inhibits proliferation by decreasing cyclooxygenase-2 (COX-2) in arsenic trioxide-treated HepG2 cells. Environ Toxicol Pharmacol. 2016 Jul;45:212-21.
12 Effects of 1alpha,25 dihydroxyvitamin D3 and testosterone on miRNA and mRNA expression in LNCaP cells. Mol Cancer. 2011 May 18;10:58.
13 Transcriptome and DNA methylome dynamics during triclosan-induced cardiomyocyte differentiation toxicity. Stem Cells Int. 2018 Oct 29;2018:8608327.
14 Gene Expression Regulation and Pathway Analysis After Valproic Acid and Carbamazepine Exposure in a Human Embryonic Stem Cell-Based Neurodevelopmental Toxicity Assay. Toxicol Sci. 2015 Aug;146(2):311-20. doi: 10.1093/toxsci/kfv094. Epub 2015 May 15.
15 Methotrexate modulates folate phenotype and inflammatory profile in EA.hy 926 cells. Eur J Pharmacol. 2014 Jun 5;732:60-7.
16 THC exposure of human iPSC neurons impacts genes associated with neuropsychiatric disorders. Transl Psychiatry. 2018 Apr 25;8(1):89. doi: 10.1038/s41398-018-0137-3.
17 Elucidating progesterone effects in breast cancer: cross talk with PDGF signaling pathway in smooth muscle cell. J Cell Biochem. 2007 Jan 1;100(1):174-83. doi: 10.1002/jcb.21045.
18 Dickkopf 1 mediates glucocorticoid-induced changes in human neural progenitor cell proliferation and differentiation. Toxicol Sci. 2012 Feb;125(2):488-95. doi: 10.1093/toxsci/kfr304. Epub 2011 Nov 1.
19 Possible roles for folic acid in the regulation of trophoblast invasion and placental development in normal early human pregnancy. Biol Reprod. 2011 Jun;84(6):1148-53. doi: 10.1095/biolreprod.110.088351. Epub 2011 Feb 23.
20 Computational discovery of niclosamide ethanolamine, a repurposed drug candidate that reduces growth of hepatocellular carcinoma cells initro and in mice by inhibiting cell division cycle 37 signaling. Gastroenterology. 2017 Jun;152(8):2022-2036.
21 Curcumin circumvents chemoresistance in vitro and potentiates the effect of thalidomide and bortezomib against human multiple myeloma in nude mice model. Mol Cancer Ther. 2009 Apr;8(4):959-70. doi: 10.1158/1535-7163.MCT-08-0905.
22 Effects of ciglitazone and troglitazone on the proliferation of human stomach cancer cells. World J Gastroenterol. 2009 Jan 21;15(3):310-20.
23 Rosiglitazone impairs proliferation of human adrenocortical cancer: preclinical study in a xenograft mouse model. Endocr Relat Cancer. 2010 Feb 18;17(1):169-77. doi: 10.1677/ERC-09-0170. Print 2010 Mar.
24 A transcriptomics-based in vitro assay for predicting chemical genotoxicity in vivo. Carcinogenesis. 2012 Jul;33(7):1421-9.
25 Prenatal ethanol exposure-induced a low level of foetal blood cholesterol and its mechanism of IGF1-related placental cholesterol transport dysfunction. Toxicology. 2019 Aug 1;424:152237. doi: 10.1016/j.tox.2019.152237. Epub 2019 Jun 18.
26 Apoptosis induced by piroxicam plus cisplatin combined treatment is triggered by p21 in mesothelioma. PLoS One. 2011;6(8):e23569.
27 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.
28 Targeting PKM2 improves the gemcitabine sensitivity of intrahepatic cholangiocarcinoma cells via inhibiting -catenin signaling pathway. Chem Biol Interact. 2024 Jan 5;387:110816. doi: 10.1016/j.cbi.2023.110816. Epub 2023 Nov 22.
29 The effect of mifepristone on breast cell proliferation in premenopausal women evaluated through fine needle aspiration cytology. Hum Reprod. 2008 Sep;23(9):2072-9. doi: 10.1093/humrep/den228. Epub 2008 Jun 24.
30 Lucanthone is a novel inhibitor of autophagy that induces cathepsin D-mediated apoptosis. J Biol Chem. 2011 Feb 25;286(8):6602-13.
31 Methamphetamine alters the normal progression by inducing cell cycle arrest in astrocytes. PLoS One. 2014 Oct 7;9(10):e109603.
32 Specific inhibition of cyclin-dependent kinase 4/6 by PD 0332991 and associated antitumor activity in human tumor xenografts. Mol Cancer Ther. 2004 Nov;3(11):1427-38.
33 Thalidomide suppresses angiogenesis and immune evasion via lncRNA FGD5-AS1/miR-454-3p/ZEB1 axis-mediated VEGFA expression and PD-1/PD-L1 checkpoint in NSCLC. Chem Biol Interact. 2021 Nov 1;349:109652. doi: 10.1016/j.cbi.2021.109652. Epub 2021 Sep 11.
34 Microarray analysis of bicalutamide action on telomerase activity, p53 pathway and viability of prostate carcinoma cell lines. J Pharm Pharmacol. 2005 Jan;57(1):83-92.
35 Enhanced chemotherapeutic efficacy of docetaxel in human lung cancer cell line via GLUT1 inhibitor. J Biochem Mol Toxicol. 2023 Jun;37(6):e23348. doi: 10.1002/jbt.23348. Epub 2023 Mar 31.
36 Sevoflurane but not propofol enhances ovarian cancer cell biology through regulating cellular metabolic and signaling mechanisms. Cell Biol Toxicol. 2023 Aug;39(4):1395-1411. doi: 10.1007/s10565-022-09766-6. Epub 2022 Oct 8.
37 Melatonin, a novel Sirt1 inhibitor, imparts antiproliferative effects against prostate cancer in vitro in culture and in vivo in TRAMP model. J Pineal Res. 2011 Mar;50(2):140-9. doi: 10.1111/j.1600-079X.2010.00823.x. Epub 2010 Nov 9.
38 Adenosine and Cordycepin Accelerate Tissue Remodeling Process through Adenosine Receptor Mediated Wnt/-Catenin Pathway Stimulation by Regulating GSK3b Activity. Int J Mol Sci. 2021 May 25;22(11):5571. doi: 10.3390/ijms22115571.
39 ATPR induces acute promyelocytic leukemia cells differentiation and growth arrest by blockade of SHP2/Rho/ROCK1 pathway. Toxicol Appl Pharmacol. 2020 Jul 15;399:115053. doi: 10.1016/j.taap.2020.115053. Epub 2020 May 15.
40 Propranolol inhibits proliferation and induces apoptosis of hemangioma-derived endothelial cells via Akt pathway by down-regulating Ang-2 expression. Chem Biol Interact. 2020 Jan 25;316:108925. doi: 10.1016/j.cbi.2019.108925. Epub 2019 Dec 12.
41 Biological specificity of CDK4/6 inhibitors: dose response relationship, in vivo signaling, and composite response signature. Oncotarget. 2017 Jul 4;8(27):43678-43691. doi: 10.18632/oncotarget.18435.
42 Aromatase inhibitors: cellular and molecular effects. J Steroid Biochem Mol Biol. 2005 May;95(1-5):83-9. doi: 10.1016/j.jsbmb.2005.04.010.
43 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.
44 Ketamine cystitis as a mimic of carcinoma in situ. Histopathology. 2009 Dec;55(6):705-8. doi: 10.1111/j.1365-2559.2009.03437.x.
45 Ropivacaine inhibits proliferation?and invasion?and promotes apoptosis and autophagy in bladder cancer cells via inhibiting PI3K/AKT pathway. J Biochem Mol Toxicol. 2023 Jan;37(1):e23233. doi: 10.1002/jbt.23233. Epub 2022 Oct 3.
46 The BET inhibitor INCB054329 reduces homologous recombination efficiency and augments PARP inhibitor activity in ovarian cancer. Gynecol Oncol. 2018 Jun;149(3):575-584. doi: 10.1016/j.ygyno.2018.03.049. Epub 2018 Mar 20.
47 Effect of menstrual cycle and hormonal treatment on ki-67 and bcl-2 expression and adenomyosis. Gynecol Endocrinol. 2005 Mar;20(3):127-31. doi: 10.1080/09513590400021086.
48 Brexpiprazole suppresses cell proliferation and de novo lipogenesis through AMPK/SREBP1 pathway in colorectal cancer. Environ Toxicol. 2023 Oct;38(10):2352-2360. doi: 10.1002/tox.23871. Epub 2023 Jun 22.
49 Biomarker changes during neoadjuvant anastrozole, tamoxifen, or the combination: influence of hormonal status and HER-2 in breast cancer--a study from the IMPACT trialists. J Clin Oncol. 2005 Apr 10;23(11):2477-92. doi: 10.1200/JCO.2005.07.559. Epub 2005 Mar 14.
50 Resveratrol-induced gene expression profiles in human prostate cancer cells. Cancer Epidemiol Biomarkers Prev. 2005 Mar;14(3):596-604. doi: 10.1158/1055-9965.EPI-04-0398.
51 Curcumin sensitizes TRAIL-resistant xenografts: molecular mechanisms of apoptosis, metastasis and angiogenesis. Mol Cancer. 2008 Jan 29;7:16.
52 Effects of chlorpromazine with and without UV irradiation on gene expression of HepG2 cells. Mutat Res. 2005 Aug 4;575(1-2):47-60. doi: 10.1016/j.mrfmmm.2005.03.002. Epub 2005 Apr 26.
53 Establishment of hypoxia induction in an in vivo animal replacement model for experimental evaluation of pancreatic cancer. Oncol Rep. 2014 Jul;32(1):153-8. doi: 10.3892/or.2014.3196. Epub 2014 May 16.
54 An in vitro study on anti-carcinogenic effect of remdesivir in human ovarian cancer cells via generation of reactive oxygen species. Hum Exp Toxicol. 2022 Jan-Dec;41:9603271221089257. doi: 10.1177/09603271221089257.
55 Screening of Organophosphate Flame Retardants with Placentation-Disrupting Effects in Human Trophoblast Organoid Model and Characterization of Adverse Pregnancy Outcomes in Mice. Environ Health Perspect. 2022 May;130(5):57002. doi: 10.1289/EHP10273. Epub 2022 May 3.
56 Thymoquinone suppresses the proliferation of renal cell carcinoma cells via reactive oxygen species-induced apoptosis and reduces cell stemness. Environ Toxicol. 2019 Nov;34(11):1208-1220. doi: 10.1002/tox.22822. Epub 2019 Jul 12.
57 Proteomic analysis of the cellular response to a potent sensitiser unveils the dynamics of haptenation in living cells. Toxicology. 2020 Dec 1;445:152603. doi: 10.1016/j.tox.2020.152603. Epub 2020 Sep 28.
58 Dual mTOR inhibitor MLN0128 suppresses Merkel cell carcinoma (MCC) xenograft tumor growth. Oncotarget. 2016 Feb 9;7(6):6576-92. doi: 10.18632/oncotarget.5878.
59 In vivo inhibition of angiogenesis by sulphamoylated derivatives of 2-methoxyoestradiol. Br J Cancer. 2007 May 7;96(9):1368-76. doi: 10.1038/sj.bjc.6603727. Epub 2007 Apr 10.
60 Randomized Phase 0/I Trial of the Mitochondrial Inhibitor ME-344 or Placebo Added to Bevacizumab in Early HER2-Negative Breast Cancer. Clin Cancer Res. 2020 Jan 1;26(1):35-45. doi: 10.1158/1078-0432.CCR-19-2023. Epub 2019 Oct 9.
61 The BET bromodomain inhibitor JQ1 suppresses growth of pancreatic ductal adenocarcinoma in patient-derived xenograft models. Oncogene. 2016 Feb 18;35(7):833-45.
62 Endoplasmic reticulum stress and MAPK signaling pathway activation underlie leflunomide-induced toxicity in HepG2 Cells. Toxicology. 2017 Dec 1;392:11-21.
63 Aminoguanidine impedes human pancreatic tumor growth and metastasis development in nude mice. World J Gastroenterol. 2009 Mar 7;15(9):1065-71.
64 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.
65 Pre-surgical study of the biological effects of the selective cyclo-oxygenase-2 inhibitor celecoxib in patients with primary breast cancer. Breast Cancer Res Treat. 2010 Oct;123(3):829-36. doi: 10.1007/s10549-010-1100-z. Epub 2010 Aug 10.
66 A novel circular RNA confers trastuzumab resistance in human epidermal growth factor receptor 2-positive breast cancer through regulating ferroptosis. Environ Toxicol. 2022 Jul;37(7):1597-1607. doi: 10.1002/tox.23509. Epub 2022 Mar 2.
67 Endoplasmic reticulum stress impairs insulin signaling through mitochondrial damage in SH-SY5Y cells. Neurosignals. 2012;20(4):265-80.
68 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.
69 Combined Bcl-2/mammalian target of rapamycin inhibition leads to enhanced radiosensitization via induction of apoptosis and autophagy in non-small cell lung tumor xenograft model. Clin Cancer Res. 2009 Oct 1;15(19):6096-105. doi: 10.1158/1078-0432.CCR-09-0589. Epub 2009 Sep 22.
70 Effects of low-dose Bisphenol A on calcium ion influx and on genes of proliferation and differentiation in immortalized human gingival cells in vitro: The role of estrogen receptor beta. Dent Mater. 2017 Sep;33(9):1021-1032. doi: 10.1016/j.dental.2017.06.011. Epub 2017 Jul 9.
71 MCM-2 is a therapeutic target of Trichostatin A in colon cancer cells. Toxicol Lett. 2013 Jul 31;221(1):23-30. doi: 10.1016/j.toxlet.2013.05.643. Epub 2013 Jun 13.
72 Pleiotropic combinatorial transcriptomes of human breast cancer cells exposed to mixtures of dietary phytoestrogens. Food Chem Toxicol. 2009 Apr;47(4):787-95.
73 Deguelin, an Akt inhibitor, down-regulates NF-B signaling and induces apoptosis in colon cancer cells and inhibits tumor growth in mice. Dig Dis Sci. 2012 Nov;57(11):2873-82. doi: 10.1007/s10620-012-2237-x. Epub 2012 May 24.
74 The potential implications of estrogenic and antioxidant-dependent activities of high doses of methyl paraben on MCF7 breast cancer cells. J Biochem Mol Toxicol. 2022 May;36(5):e23012. doi: 10.1002/jbt.23012. Epub 2022 Feb 17.
75 Functional lipidomics: Palmitic acid impairs hepatocellular carcinoma development by modulating membrane fluidity and glucose metabolism. Hepatology. 2017 Aug;66(2):432-448. doi: 10.1002/hep.29033. Epub 2017 Jun 16.
76 Geraniol suppresses prostate cancer growth through down-regulation of E2F8. Cancer Med. 2016 Oct;5(10):2899-2908.
77 Short-term exposure to tobacco toxins alters expression of multiple proliferation gene markers in primary human bronchial epithelial cell cultures. J Oncol. 2011;2011:208563. doi: 10.1155/2011/208563. Epub 2011 Apr 5.
78 Wogonin induces cellular senescence in breast cancer via suppressing TXNRD2 expression. Arch Toxicol. 2020 Oct;94(10):3433-3447. doi: 10.1007/s00204-020-02842-y. Epub 2020 Jul 15.
79 Blockade of epidermal growth factor receptor/mammalian target of rapamycin pathway by Icariside II results in reduced cell proliferation of osteosarcoma cells. Food Chem Toxicol. 2014 Nov;73:7-16. doi: 10.1016/j.fct.2014.08.002. Epub 2014 Aug 10.
80 ABBV-744 induces autophagy in gastric cancer cells by regulating PI3K/AKT/mTOR/p70S6k and MAPK signaling pathways. Neoplasia. 2023 Nov;45:100936. doi: 10.1016/j.neo.2023.100936. Epub 2023 Sep 26.
81 MEK inhibitor PD-0325901 overcomes resistance to CK2 inhibitor CX-4945 and exhibits anti-tumor activity in head and neck cancer. Int J Biol Sci. 2015 Feb 23;11(4):411-22. doi: 10.7150/ijbs.10745. eCollection 2015.
82 Involvement of peroxisome proliferator-activated receptor gamma (PPAR) and autophagic pathways in the mechanism of action of the tris(2,3-dibromopropyl) isocyanurate (TDBP-TAZTO or TBC) flame retardant in the lung adenocarcinoma (A549) cells in vitro. J Appl Toxicol. 2023 Sep;43(9):1358-1367. doi: 10.1002/jat.4470. Epub 2023 Mar 31.