General Information of Drug Off-Target (DOT) (ID: OT78QCN8)

DOT Name GTPase KRas (KRAS)
Synonyms EC 3.6.5.2; K-Ras 2; Ki-Ras; c-K-ras; c-Ki-ras
Gene Name KRAS
Related Disease
Cardiofaciocutaneous syndrome 1 ( )
Cardiofaciocutaneous syndrome 2 ( )
Noonan syndrome ( )
Noonan syndrome 3 ( )
Cardiofaciocutaneous syndrome ( )
Linear nevus sebaceous syndrome ( )
Costello syndrome ( )
UniProt ID
RASK_HUMAN
3D Structure
Download
2D Sequence (FASTA)
Download
3D Structure (PDB)
Download
PDB ID
1D8D ; 1D8E ; 1KZO ; 1KZP ; 1N4P ; 1N4Q ; 1N4R ; 1N4S ; 3GFT ; 4DSN ; 4DSO ; 4EPR ; 4EPT ; 4EPV ; 4EPW ; 4EPX ; 4EPY ; 4L8G ; 4LDJ ; 4LPK ; 4LRW ; 4LUC ; 4LV6 ; 4LYF ; 4LYH ; 4LYJ ; 4M1O ; 4M1S ; 4M1T ; 4M1W ; 4M1Y ; 4M21 ; 4M22 ; 4NMM ; 4OBE ; 4PZY ; 4PZZ ; 4Q01 ; 4Q02 ; 4Q03 ; 4QL3 ; 4TQ9 ; 4TQA ; 4WA7 ; 5F2E ; 5KYK ; 5MLA ; 5MLB ; 5O2S ; 5O2T ; 5OCG ; 5OCO ; 5OCT ; 5TAR ; 5TB5 ; 5UFE ; 5UFQ ; 5UK9 ; 5UQW ; 5US4 ; 5USJ ; 5V6S ; 5V6V ; 5V71 ; 5V9L ; 5V9O ; 5V9U ; 5VBM ; 5VP7 ; 5VPI ; 5VPY ; 5VPZ ; 5VQ0 ; 5VQ1 ; 5VQ2 ; 5VQ6 ; 5VQ8 ; 5W22 ; 5WHA ; 5WHB ; 5WHD ; 5WHE ; 5WLB ; 5WPM ; 5XCO ; 5YXZ ; 5YY1 ; 6ARK ; 6ASA ; 6ASE ; 6B0V ; 6B0Y ; 6BOF ; 6BP1 ; 6CC9 ; 6CCH ; 6CCX ; 6CU6 ; 6E6F ; 6E6G ; 6EPL ; 6EPM ; 6EPN ; 6EPO ; 6EPP ; 6F76 ; 6FA1 ; 6FA2 ; 6FA3 ; 6FA4 ; 6GJ5 ; 6GJ6 ; 6GJ7 ; 6GJ8 ; 6GOD ; 6GOE ; 6GOF ; 6GOG ; 6GOM ; 6GQT ; 6GQW ; 6GQX ; 6GQY ; 6H46 ; 6H47 ; 6JTN ; 6JTO ; 6JTP ; 6M9W ; 6MBQ ; 6MBT ; 6MBU ; 6MNX ; 6MQG ; 6MQN ; 6MS9 ; 6MTA ; 6N2J ; 6N2K ; 6O36 ; 6O46 ; 6O4Y ; 6O4Z ; 6O51 ; 6O53 ; 6OB2 ; 6OB3 ; 6OIM ; 6P0Z ; 6P8W ; 6P8X ; 6P8Y ; 6P8Z ; 6PGO ; 6PGP ; 6PQ3 ; 6PTS ; 6PTW ; 6QUU ; 6QUV ; 6QUW ; 6QUX ; 6T5B ; 6T5U ; 6T5V ; 6TAM ; 6TAN ; 6USX ; 6USZ ; 6UT0 ; 6V5L ; 6V65 ; 6V6F ; 6VC8 ; 6VJJ ; 6W4E ; 6W4F ; 6WGN ; 6WS2 ; 6WS4 ; 6XGU ; 6XGV ; 6XHA ; 6XHB ; 6YR8 ; 6YXW ; 6ZL5 ; 6ZLI ; 7A1W ; 7A1X ; 7A1Y ; 7A47 ; 7ACA ; 7ACF ; 7ACH ; 7ACQ ; 7C40 ; 7C41 ; 7EW9 ; 7EWA ; 7EWB ; 7EYX ; 7F0W ; 7KFZ ; 7KMR ; 7KYZ ; 7LC1 ; 7LC2 ; 7LGI ; 7LZ5 ; 7MDP ; 7MQU ; 7NY8 ; 7O70 ; 7OK3 ; 7OK4 ; 7OO7 ; 7Q9U ; 7R0M ; 7R0N ; 7R0Q ; 7ROV ; 7RP2 ; 7RP3 ; 7RP4 ; 7RPZ ; 7RSC ; 7RSE ; 7RT1 ; 7RT2 ; 7RT3 ; 7RT4 ; 7RT5 ; 7SCW ; 7SCX ; 7STF ; 7T1F ; 7T47 ; 7TLE ; 7TLG ; 7TLK ; 7U8H ; 7VVB ; 7W5R ; 7YCC ; 7YCE ; 7YUZ ; 7YV1 ; 8AFB ; 8AFC ; 8AFD ; 8AQ5 ; 8AQ7 ; 8AZR ; 8AZV ; 8AZX ; 8AZY ; 8AZZ ; 8B00 ; 8B6I ; 8B78 ; 8CX5 ; 8DNI ; 8DNJ ; 8DNK ; 8DVG ; 8EBZ ; 8ECR ; 8EDY ; 8EER ; 8EIE ; 8EPW ; 8EZG ; 8F0M ; 8FMJ ; 8FMK ; 8G42 ; 8G47 ; 8G4F ; 8G4H ; 8G9P ; 8G9Q ; 8I5E ; 8JJS ; 8ONV ; 8QU8 ; 8QUG ; 8QVU ; 8QW6 ; 8QW7 ; 8STM ; 8STN ; 8TXE ; 8TXG ; 8TXH
EC Number
3.6.5.2
Pfam ID
PF00071
Sequence
MTEYKLVVVGAGGVGKSALTIQLIQNHFVDEYDPTIEDSYRKQVVIDGETCLLDILDTAG
QEEYSAMRDQYMRTGEGFLCVFAINNTKSFEDIHHYREQIKRVKDSEDVPMVLVGNKCDL
PSRTVDTKQAQDLARSYGIPFIETSAKTRQRVEDAFYTLVREIRQYRLKKISKEEKTPGC
VKIKKCIIM
Function
Ras proteins bind GDP/GTP and possess intrinsic GTPase activity. Plays an important role in the regulation of cell proliferation. Plays a role in promoting oncogenic events by inducing transcriptional silencing of tumor suppressor genes (TSGs) in colorectal cancer (CRC) cells in a ZNF304-dependent manner.
KEGG Pathway
EGFR tyrosine ki.se inhibitor resistance (hsa01521 )
Endocrine resistance (hsa01522 )
MAPK sig.ling pathway (hsa04010 )
ErbB sig.ling pathway (hsa04012 )
Ras sig.ling pathway (hsa04014 )
Rap1 sig.ling pathway (hsa04015 )
Chemokine sig.ling pathway (hsa04062 )
FoxO sig.ling pathway (hsa04068 )
Sphingolipid sig.ling pathway (hsa04071 )
Phospholipase D sig.ling pathway (hsa04072 )
Mitophagy - animal (hsa04137 )
Autophagy - animal (hsa04140 )
mTOR sig.ling pathway (hsa04150 )
PI3K-Akt sig.ling pathway (hsa04151 )
Apoptosis (hsa04210 )
Longevity regulating pathway (hsa04211 )
Longevity regulating pathway - multiple species (hsa04213 )
Cellular senescence (hsa04218 )
Axon guidance (hsa04360 )
VEGF sig.ling pathway (hsa04370 )
Apelin sig.ling pathway (hsa04371 )
Gap junction (hsa04540 )
Sig.ling pathways regulating pluripotency of stem cells (hsa04550 )
C-type lectin receptor sig.ling pathway (hsa04625 )
.tural killer cell mediated cytotoxicity (hsa04650 )
T cell receptor sig.ling pathway (hsa04660 )
B cell receptor sig.ling pathway (hsa04662 )
Fc epsilon RI sig.ling pathway (hsa04664 )
Thermogenesis (hsa04714 )
Long-term potentiation (hsa04720 )
Neurotrophin sig.ling pathway (hsa04722 )
Cholinergic sy.pse (hsa04725 )
Serotonergic sy.pse (hsa04726 )
Long-term depression (hsa04730 )
Regulation of actin cytoskeleton (hsa04810 )
Insulin sig.ling pathway (hsa04910 )
GnRH sig.ling pathway (hsa04912 )
Progesterone-mediated oocyte maturation (hsa04914 )
Estrogen sig.ling pathway (hsa04915 )
Melanogenesis (hsa04916 )
Prolactin sig.ling pathway (hsa04917 )
Thyroid hormone sig.ling pathway (hsa04919 )
Oxytocin sig.ling pathway (hsa04921 )
Relaxin sig.ling pathway (hsa04926 )
GnRH secretion (hsa04929 )
AGE-RAGE sig.ling pathway in diabetic complications (hsa04933 )
Growth hormone synthesis, secretion and action (hsa04935 )
Aldosterone-regulated sodium reabsorption (hsa04960 )
Alzheimer disease (hsa05010 )
Pathways of neurodegeneration - multiple diseases (hsa05022 )
Alcoholism (hsa05034 )
Hepatitis C (hsa05160 )
Hepatitis B (hsa05161 )
Human cytomegalovirus infection (hsa05163 )
Human papillomavirus infection (hsa05165 )
Human T-cell leukemia virus 1 infection (hsa05166 )
Kaposi sarcoma-associated herpesvirus infection (hsa05167 )
Human immunodeficiency virus 1 infection (hsa05170 )
Pathways in cancer (hsa05200 )
Viral carcinogenesis (hsa05203 )
Proteoglycans in cancer (hsa05205 )
MicroR.s in cancer (hsa05206 )
Chemical carcinogenesis - receptor activation (hsa05207 )
Chemical carcinogenesis - reactive oxygen species (hsa05208 )
Colorectal cancer (hsa05210 )
Re.l cell carcinoma (hsa05211 )
Pancreatic cancer (hsa05212 )
Endometrial cancer (hsa05213 )
Glioma (hsa05214 )
Prostate cancer (hsa05215 )
Thyroid cancer (hsa05216 )
Melanoma (hsa05218 )
Bladder cancer (hsa05219 )
Chronic myeloid leukemia (hsa05220 )
Acute myeloid leukemia (hsa05221 )
Non-small cell lung cancer (hsa05223 )
Breast cancer (hsa05224 )
Hepatocellular carcinoma (hsa05225 )
Gastric cancer (hsa05226 )
Central carbon metabolism in cancer (hsa05230 )
Choline metabolism in cancer (hsa05231 )
PD-L1 expression and PD-1 checkpoint pathway in cancer (hsa05235 )
Lipid and atherosclerosis (hsa05417 )
Reactome Pathway
Activation of RAS in B cells (R-HSA-1169092 )
Constitutive Signaling by Ligand-Responsive EGFR Cancer Variants (R-HSA-1236382 )
SHC1 events in ERBB2 signaling (R-HSA-1250196 )
SHC1 events in ERBB4 signaling (R-HSA-1250347 )
Signaling by SCF-KIT (R-HSA-1433557 )
Signalling to RAS (R-HSA-167044 )
p38MAPK events (R-HSA-171007 )
GRB2 events in EGFR signaling (R-HSA-179812 )
SHC1 events in EGFR signaling (R-HSA-180336 )
Downstream signal transduction (R-HSA-186763 )
GRB2 events in ERBB2 signaling (R-HSA-1963640 )
Tie2 Signaling (R-HSA-210993 )
EGFR Transactivation by Gastrin (R-HSA-2179392 )
DAP12 signaling (R-HSA-2424491 )
SHC-related events triggered by IGF1R (R-HSA-2428933 )
FCERI mediated MAPK activation (R-HSA-2871796 )
NCAM signaling for neurite out-growth (R-HSA-375165 )
Ras activation upon Ca2+ influx through NMDA receptor (R-HSA-442982 )
VEGFR2 mediated cell proliferation (R-HSA-5218921 )
CD209 (DC-SIGN) signaling (R-HSA-5621575 )
Constitutive Signaling by EGFRvIII (R-HSA-5637810 )
SHC-mediated cascade (R-HSA-5654688 )
FRS-mediated FGFR1 signaling (R-HSA-5654693 )
SHC-mediated cascade (R-HSA-5654699 )
FRS-mediated FGFR2 signaling (R-HSA-5654700 )
SHC-mediated cascade (R-HSA-5654704 )
FRS-mediated FGFR3 signaling (R-HSA-5654706 )
FRS-mediated FGFR4 signaling (R-HSA-5654712 )
SHC-mediated cascade (R-HSA-5654719 )
Signaling by FGFR2 in disease (R-HSA-5655253 )
Signaling by FGFR4 in disease (R-HSA-5655291 )
Signaling by FGFR1 in disease (R-HSA-5655302 )
Signaling by FGFR3 in disease (R-HSA-5655332 )
Regulation of RAS by GAPs (R-HSA-5658442 )
RAF activation (R-HSA-5673000 )
RAF/MAP kinase cascade (R-HSA-5673001 )
MAP2K and MAPK activation (R-HSA-5674135 )
Negative regulation of MAPK pathway (R-HSA-5675221 )
Signaling by moderate kinase activity BRAF mutants (R-HSA-6802946 )
Signaling by high-kinase activity BRAF mutants (R-HSA-6802948 )
Signaling by BRAF and RAF1 fusions (R-HSA-6802952 )
RAS signaling downstream of NF1 loss-of-function variants (R-HSA-6802953 )
Paradoxical activation of RAF signaling by kinase inactive BRAF (R-HSA-6802955 )
Insulin receptor signalling cascade (R-HSA-74751 )
PTK6 Regulates RHO GTPases, RAS GTPase and MAP kinases (R-HSA-8849471 )
MET activates RAS signaling (R-HSA-8851805 )
RUNX3 regulates p14-ARF (R-HSA-8951936 )
Activated NTRK2 signals through RAS (R-HSA-9026519 )
Erythropoietin activates RAS (R-HSA-9027284 )
Activated NTRK2 signals through FRS2 and FRS3 (R-HSA-9028731 )
Activated NTRK3 signals through RAS (R-HSA-9034864 )
FLT3 Signaling (R-HSA-9607240 )
Constitutive Signaling by Overexpressed ERBB2 (R-HSA-9634285 )
Estrogen-stimulated signaling through PRKCZ (R-HSA-9634635 )
RAS processing (R-HSA-9648002 )
Signaling downstream of RAS mutants (R-HSA-9649948 )
Signaling by RAF1 mutants (R-HSA-9656223 )
Signaling by ERBB2 KD Mutants (R-HSA-9664565 )
Signaling by ERBB2 ECD mutants (R-HSA-9665348 )
Signaling by ERBB2 TMD/JMD mutants (R-HSA-9665686 )
Signaling by phosphorylated juxtamembrane, extracellular and kinase domain KIT mutants (R-HSA-9670439 )
Signaling by PDGFRA transmembrane, juxtamembrane and kinase domain mutants (R-HSA-9673767 )
Signaling by PDGFRA extracellular domain mutants (R-HSA-9673770 )
Signaling by CSF3 (G-CSF) (R-HSA-9674555 )
Signaling by CSF1 (M-CSF) in myeloid cells (R-HSA-9680350 )
Signaling by FLT3 fusion proteins (R-HSA-9703465 )
Signaling by FLT3 ITD and TKD mutants (R-HSA-9703648 )
Signaling by RAS GAP mutants (R-HSA-9753510 )
Signaling by RAS GTPase mutants (R-HSA-9753512 )
SOS-mediated signalling (R-HSA-112412 )

Molecular Interaction Atlas (MIA) of This DOT

7 Disease(s) Related to This DOT
Disease Name Disease ID Evidence Level Mode of Inheritance REF
Cardiofaciocutaneous syndrome 1 DISV4HQA Definitive Autosomal dominant [1]
Cardiofaciocutaneous syndrome 2 DISJMMVB Definitive Autosomal dominant [2]
Noonan syndrome DIS7Q7DN Definitive Autosomal dominant [3]
Noonan syndrome 3 DIS4MLJP Definitive Autosomal dominant [4]
Cardiofaciocutaneous syndrome DISZJKSC Strong Autosomal dominant [3]
Linear nevus sebaceous syndrome DIS6VQ2J Strong Autosomal dominant [5]
Costello syndrome DISXVJH3 Disputed Autosomal dominant [3]
------------------------------------------------------------------------------------
⏷ Show the Full List of 7 Disease(s)
Molecular Interaction Atlas (MIA) Jump to Detail Molecular Interaction Atlas of This DOT
This DOT Affected the Drug Response of 15 Drug(s)
Drug Name Drug ID Highest Status Interaction REF
Methotrexate DM2TEOL Approved GTPase KRas (KRAS) increases the response to substance of Methotrexate. [37]
Sulindac DM2QHZU Approved GTPase KRas (KRAS) affects the response to substance of Sulindac. [38]
Gefitinib DM15F0X Approved GTPase KRas (KRAS) decreases the response to substance of Gefitinib. [39]
Crizotinib DM4F29C Approved GTPase KRas (KRAS) decreases the response to substance of Crizotinib. [40]
Lapatinib DM3BH1Y Approved GTPase KRas (KRAS) decreases the response to substance of Lapatinib. [39]
Vemurafenib DM62UG5 Approved GTPase KRas (KRAS) affects the response to substance of Vemurafenib. [41]
Pemetrexed DMMX2E6 Approved GTPase KRas (KRAS) increases the response to substance of Pemetrexed. [37]
Dabrafenib DMX6OE3 Approved GTPase KRas (KRAS) affects the response to substance of Dabrafenib. [41]
Sotorasib DMLSV74 Approved GTPase KRas (KRAS) increases the response to substance of Sotorasib. [42]
Trimetrexate DMDEA85 Approved GTPase KRas (KRAS) increases the response to substance of Trimetrexate. [37]
Afimoxifene DMFORDT Phase 2 GTPase KRas (KRAS) affects the response to substance of Afimoxifene. [43]
GDC0941 DM1YAK6 Phase 2 GTPase KRas (KRAS) increases the response to substance of GDC0941. [44]
PF-04691502 DMS610L Phase 2 GTPase KRas (KRAS) affects the response to substance of PF-04691502. [45]
(+)-JQ1 DM1CZSJ Phase 1 GTPase KRas (KRAS) increases the response to substance of (+)-JQ1. [46]
AZ-628 DMEWQHP Investigative GTPase KRas (KRAS) increases the response to substance of AZ-628. [47]
------------------------------------------------------------------------------------
⏷ Show the Full List of 15 Drug(s)
This DOT Affected the Biotransformations of 1 Drug(s)
Drug Name Drug ID Highest Status Interaction REF
Guanosine-5'-Triphosphate DMV2OJX Investigative GTPase KRas (KRAS) increases the hydrolysis of Guanosine-5'-Triphosphate. [36]
------------------------------------------------------------------------------------
28 Drug(s) Affected the Gene/Protein Processing of This DOT
Drug Name Drug ID Highest Status Interaction REF
Valproate DMCFE9I Approved Valproate increases the expression of GTPase KRas (KRAS). [6]
Doxorubicin DMVP5YE Approved Doxorubicin decreases the expression of GTPase KRas (KRAS). [7]
Cisplatin DMRHGI9 Approved Cisplatin decreases the expression of GTPase KRas (KRAS). [8]
Estradiol DMUNTE3 Approved Estradiol decreases the expression of GTPase KRas (KRAS). [9]
Ivermectin DMDBX5F Approved Ivermectin decreases the expression of GTPase KRas (KRAS). [10]
Arsenic DMTL2Y1 Approved Arsenic increases the mutagenesis of GTPase KRas (KRAS). [11]
Quercetin DM3NC4M Approved Quercetin increases the expression of GTPase KRas (KRAS). [12]
Carbamazepine DMZOLBI Approved Carbamazepine affects the expression of GTPase KRas (KRAS). [13]
Marinol DM70IK5 Approved Marinol increases the expression of GTPase KRas (KRAS). [14]
Zoledronate DMIXC7G Approved Zoledronate increases the expression of GTPase KRas (KRAS). [15]
Fluorouracil DMUM7HZ Approved Fluorouracil decreases the expression of GTPase KRas (KRAS). [16]
Isotretinoin DM4QTBN Approved Isotretinoin decreases the expression of GTPase KRas (KRAS). [17]
Hydroquinone DM6AVR4 Approved Hydroquinone increases the expression of GTPase KRas (KRAS). [18]
Testosterone enanthate DMB6871 Approved Testosterone enanthate affects the expression of GTPase KRas (KRAS). [19]
Diclofenac DMPIHLS Approved Diclofenac affects the expression of GTPase KRas (KRAS). [13]
Sevoflurane DMC9O43 Approved Sevoflurane decreases the expression of GTPase KRas (KRAS). [22]
Tiazofurin DM5JWV9 Approved Tiazofurin decreases the expression of GTPase KRas (KRAS). [23]
Urethane DM7NSI0 Phase 4 Urethane decreases the expression of GTPase KRas (KRAS). [24]
Resveratrol DM3RWXL Phase 3 Resveratrol decreases the expression of GTPase KRas (KRAS). [25]
Curcumin DMQPH29 Phase 3 Curcumin decreases the expression of GTPase KRas (KRAS). [26]
Genistein DM0JETC Phase 2/3 Genistein decreases the expression of GTPase KRas (KRAS). [27]
MG-132 DMKA2YS Preclinical MG-132 increases the expression of GTPase KRas (KRAS). [29]
MANUMYCIN A DM1SWOY Terminated MANUMYCIN A decreases the activity of GTPase KRas (KRAS). [30]
Bisphenol A DM2ZLD7 Investigative Bisphenol A increases the mutagenesis of GTPase KRas (KRAS). [31]
Coumestrol DM40TBU Investigative Coumestrol decreases the expression of GTPase KRas (KRAS). [32]
3R14S-OCHRATOXIN A DM2KEW6 Investigative 3R14S-OCHRATOXIN A decreases the expression of GTPase KRas (KRAS). [33]
acrolein DMAMCSR Investigative acrolein decreases the expression of GTPase KRas (KRAS). [34]
OXYBENZONE DMMZYX6 Investigative OXYBENZONE increases the expression of GTPase KRas (KRAS). [35]
------------------------------------------------------------------------------------
⏷ Show the Full List of 28 Drug(s)
3 Drug(s) Affected the Post-Translational Modifications of This DOT
Drug Name Drug ID Highest Status Interaction REF
Simvastatin DM30SGU Approved Simvastatin decreases the prenylation of GTPase KRas (KRAS). [20]
Lovastatin DM9OZWQ Approved Lovastatin decreases the prenylation of GTPase KRas (KRAS). [21]
Benzo(a)pyrene DMN7J43 Phase 1 Benzo(a)pyrene affects the methylation of GTPase KRas (KRAS). [28]
------------------------------------------------------------------------------------
1 Drug(s) Affected the Protein Interaction/Cellular Processes of This DOT
Drug Name Drug ID Highest Status Interaction REF
Guanosine-5'-Diphosphate DM0MUKQ Investigative Guanosine-5'-Diphosphate affects the binding of GTPase KRas (KRAS). [36]
------------------------------------------------------------------------------------

References

1 Flexible and scalable diagnostic filtering of genomic variants using G2P with Ensembl VEP. Nat Commun. 2019 May 30;10(1):2373. doi: 10.1038/s41467-019-10016-3.
2 PanelApp crowdsources expert knowledge to establish consensus diagnostic gene panels. Nat Genet. 2019 Nov;51(11):1560-1565. doi: 10.1038/s41588-019-0528-2.
3 Technical standards for the interpretation and reporting of constitutional copy-number variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics (ACMG) and the Clinical Genome Resource (ClinGen). Genet Med. 2020 Feb;22(2):245-257. doi: 10.1038/s41436-019-0686-8. Epub 2019 Nov 6.
4 Germline KRAS and BRAF mutations in cardio-facio-cutaneous syndrome. Nat Genet. 2006 Mar;38(3):294-6. doi: 10.1038/ng1749. Epub 2006 Feb 12.
5 Oculoectodermal syndrome is a mosaic RASopathy associated with KRAS alterations. Am J Med Genet A. 2015 Jul;167(7):1429-35. doi: 10.1002/ajmg.a.37048. Epub 2015 Mar 21.
6 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.
7 Bringing in vitro analysis closer to in vivo: studying doxorubicin toxicity and associated mechanisms in 3D human microtissues with PBPK-based dose modelling. Toxicol Lett. 2018 Sep 15;294:184-192.
8 Activation of AIFM2 enhances apoptosis of human lung cancer cells undergoing toxicological stress. Toxicol Lett. 2016 Sep 6;258:227-236.
9 Convergent transcriptional profiles induced by endogenous estrogen and distinct xenoestrogens in breast cancer cells. Carcinogenesis. 2006 Aug;27(8):1567-78.
10 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.
11 KRAS-retroviral fusion transcripts and gene amplification in arsenic-transformed, human prostate CAsE-PE cancer cells. Toxicol Appl Pharmacol. 2020 Apr 25;397:115017. doi: 10.1016/j.taap.2020.115017. Online ahead of print.
12 Quercetin and Its Fermented Extract as a Potential Inhibitor of Bisphenol A-Exposed HT-29 Colon Cancer Cells' Viability. Int J Mol Sci. 2023 Mar 15;24(6):5604. doi: 10.3390/ijms24065604.
13 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.
14 Delta9-tetrahydrocannabinol inhibits cytotrophoblast cell proliferation and modulates gene transcription. Mol Hum Reprod. 2006 May;12(5):321-33. doi: 10.1093/molehr/gal036. Epub 2006 Apr 5.
15 Zoledronate dysregulates fatty acid metabolism in renal tubular epithelial cells to induce nephrotoxicity. Arch Toxicol. 2018 Jan;92(1):469-485.
16 Proteomic analysis of antiproliferative effects by treatment of 5-fluorouracil in cervical cancer cells. DNA Cell Biol. 2004 Nov;23(11):769-76.
17 Temporal changes in gene expression in the skin of patients treated with isotretinoin provide insight into its mechanism of action. Dermatoendocrinol. 2009 May;1(3):177-87.
18 Hydroquinone-induced malignant transformation of TK6 cells by facilitating SIRT1-mediated p53 degradation and up-regulating KRAS. Toxicol Lett. 2016 Sep 30;259:133-142. doi: 10.1016/j.toxlet.2016.08.006. Epub 2016 Aug 8.
19 Transcriptional profiling of testosterone-regulated genes in the skeletal muscle of human immunodeficiency virus-infected men experiencing weight loss. J Clin Endocrinol Metab. 2007 Jul;92(7):2793-802. doi: 10.1210/jc.2006-2722. Epub 2007 Apr 17.
20 Cytotoxicity of simvastatin to pancreatic adenocarcinoma cells containing mutant ras gene. Jpn J Cancer Res. 1994 Jun;85(6):633-8. doi: 10.1111/j.1349-7006.1994.tb02406.x.
21 Combining prenylation inhibitors causes synergistic cytotoxicity, apoptosis and disruption of RAS-to-MAP kinase signalling in multiple myeloma cells. Br J Haematol. 2005 Sep;130(6):912-25. doi: 10.1111/j.1365-2141.2005.05696.x.
22 The differential cancer growth associated with anaesthetics in a cancer xenograft model of mice: mechanisms and implications of postoperative cancer recurrence. Cell Biol Toxicol. 2023 Aug;39(4):1561-1575. doi: 10.1007/s10565-022-09747-9. Epub 2022 Aug 12.
23 Regulation of the signal transduction program by drugs. Adv Enzyme Regul. 1997;37:35-55. doi: 10.1016/s0065-2571(96)00025-8.
24 Ethyl carbamate induces cell death through its effects on multiple metabolic pathways. Chem Biol Interact. 2017 Nov 1;277:21-32.
25 Interactive gene expression pattern in prostate cancer cells exposed to phenolic antioxidants. Life Sci. 2002 Mar 1;70(15):1821-39.
26 Synthesis and biological analysis of new curcumin analogues bearing an enhanced potential for the medicinal treatment of cancer. Mol Cancer Ther. 2006 Oct;5(10):2563-71. doi: 10.1158/1535-7163.MCT-06-0174.
27 Regulation of gene expression and inhibition of experimental prostate cancer bone metastasis by dietary genistein. Neoplasia. 2004 Jul-Aug;6(4):354-63. doi: 10.1593/neo.03478.
28 Air pollution and DNA methylation alterations in lung cancer: A systematic and comparative study. Oncotarget. 2017 Jan 3;8(1):1369-1391. doi: 10.18632/oncotarget.13622.
29 Proteasome inhibition creates a chromatin landscape favorable to RNA Pol II processivity. J Biol Chem. 2020 Jan 31;295(5):1271-1287. doi: 10.1074/jbc.RA119.011174. Epub 2019 Dec 5.
30 Manumycin inhibits STAT3, telomerase activity, and growth of glioma cells by elevating intracellular reactive oxygen species generation. Free Radic Biol Med. 2009 Aug 15;47(4):364-74.
31 Mutagenicity of bisphenol A and its suppression by interferon-alpha in human RSa cells. Mutat Res. 2001 Feb 20;490(2):199-207. doi: 10.1016/s1383-5718(00)00161-3.
32 Pleiotropic combinatorial transcriptomes of human breast cancer cells exposed to mixtures of dietary phytoestrogens. Food Chem Toxicol. 2009 Apr;47(4):787-95.
33 Persistence of epigenomic effects after recovery from repeated treatment with two nephrocarcinogens. Front Genet. 2018 Dec 3;9:558.
34 Acrolein decreases endothelial cell migration and insulin sensitivity through induction of let-7a. Toxicol Sci. 2014 Aug 1;140(2):271-82. doi: 10.1093/toxsci/kfu087. Epub 2014 May 8.
35 Chromatin modifiers: A new class of pollutants with potential epigenetic effects revealed by in vitro assays and transcriptomic analyses. Toxicology. 2023 Jan 15;484:153413. doi: 10.1016/j.tox.2022.153413. Epub 2022 Dec 26.
36 A KRAS GTPase K104Q Mutant Retains Downstream Signaling by Offsetting Defects in Regulation. J Biol Chem. 2017 Mar 17;292(11):4446-4456. doi: 10.1074/jbc.M116.762435. Epub 2017 Jan 30.
37 KRAS mutation status is associated with enhanced dependency on folate metabolism pathways in non-small cell lung cancer cells. Mol Cancer Ther. 2014 Jun;13(6):1611-24. doi: 10.1158/1535-7163.MCT-13-0649. Epub 2014 Mar 31.
38 Molecular analysis of sulindac-resistant adenomas in familial adenomatous polyposis. Clin Cancer Res. 2001 Dec;7(12):4000-7.
39 The K-Ras effector p38 MAPK confers intrinsic resistance to tyrosine kinase inhibitors by stimulating EGFR transcription and EGFR dephosphorylation. J Biol Chem. 2017 Sep 8;292(36):15070-15079. doi: 10.1074/jbc.M117.779488. Epub 2017 Jul 24.
40 Mechanisms of resistance to crizotinib in patients with ALK gene rearranged non-small cell lung cancer. Clin Cancer Res. 2012 Mar 1;18(5):1472-82. doi: 10.1158/1078-0432.CCR-11-2906. Epub 2012 Jan 10.
41 Paradoxical activation of MEK/ERK signaling induced by B-Raf inhibition enhances DR5 expression and DR5 activation-induced apoptosis in Ras-mutant cancer cells. Sci Rep. 2016 May 25;6:26803. doi: 10.1038/srep26803.
42 The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature. 2019 Nov;575(7781):217-223. doi: 10.1038/s41586-019-1694-1. Epub 2019 Oct 30.
43 Genome-wide functional screen identifies a compendium of genes affecting sensitivity to tamoxifen. Proc Natl Acad Sci U S A. 2012 Feb 21;109(8):2730-5. doi: 10.1073/pnas.1018872108. Epub 2011 Apr 11.
44 Phosphoinositide 3-kinase (PI3K) pathway alterations are associated with histologic subtypes and are predictive of sensitivity to PI3K inhibitors in lung cancer preclinical models. Clin Cancer Res. 2012 Dec 15;18(24):6771-83. doi: 10.1158/1078-0432.CCR-12-2347. Epub 2012 Nov 7.
45 Synergistic inhibition of ovarian cancer cell growth by combining selective PI3K/mTOR and RAS/ERK pathway inhibitors. Eur J Cancer. 2013 Dec;49(18):3936-44. doi: 10.1016/j.ejca.2013.08.007. Epub 2013 Sep 3.
46 Efficacy of BET bromodomain inhibition in Kras-mutant non-small cell lung cancer. Clin Cancer Res. 2013 Nov 15;19(22):6183-92. doi: 10.1158/1078-0432.CCR-12-3904. Epub 2013 Sep 17.
47 BAY61-3606 affects the viability of colon cancer cells in a genotype-directed manner. PLoS One. 2012;7(7):e41343. doi: 10.1371/journal.pone.0041343. Epub 2012 Jul 18.