General Information of Drug Combination (ID: DCHOPE8)

Drug Combination Name
Venetoclax Daunorubicin
Indication
Disease Entry Status REF
Acute Myeloid Leukemia Phase 1 [1]
Component Drugs Venetoclax   DM8I94Y Daunorubicin   DMQUSBT
Small molecular drug Small molecular drug
2D MOL 2D MOL
3D MOL 3D MOL

Molecular Interaction Atlas of This Drug Combination

Molecular Interaction Atlas (MIA)
Indication(s) of Venetoclax
Disease Entry ICD 11 Status REF
Chronic lymphocytic leukaemia 2A82.0 Approved [2]
Venetoclax Interacts with 1 DTP Molecule(s)
DTP Name DTP ID UniProt ID Mode of Action REF
P-glycoprotein 1 (ABCB1) DTUGYRD MDR1_HUMAN Substrate [6]
------------------------------------------------------------------------------------
Venetoclax Interacts with 11 DOT Molecule(s)
DOT Name DOT ID UniProt ID Mode of Action REF
Poly polymerase 1 (PARP1) OT310QSG PARP1_HUMAN Increases Cleavage [7]
Apoptosis regulator Bcl-2 (BCL2) OT9DVHC0 BCL2_HUMAN Decreases Activity [8]
Receptor-type tyrosine-protein kinase FLT3 (FLT3) OTMSRYMK FLT3_HUMAN Decreases Expression [9]
Caspase-3 (CASP3) OTIJRBE7 CASP3_HUMAN Increases Cleavage [7]
Apoptosis regulator BAX (BAX) OTAW0V4V BAX_HUMAN Affects Folding [10]
Bcl-2-like protein 1 (BCL2L1) OTRC5K9O B2CL1_HUMAN Decreases Expression [10]
Induced myeloid leukemia cell differentiation protein Mcl-1 (MCL1) OT2YYI1A MCL1_HUMAN Decreases Expression [9]
Phorbol-12-myristate-13-acetate-induced protein 1 (PMAIP1) OTXEE550 APR_HUMAN Increases Expression [10]
Bcl-2 homologous antagonist/killer (BAK1) OTDP6ILW BAK_HUMAN Affects Folding [10]
Broad substrate specificity ATP-binding cassette transporter ABCG2 (ABCG2) OTW8V2V1 ABCG2_HUMAN Affects Binding [11]
Bcl-2-binding component 3, isoforms 3/4 (BBC3) OTUAXDAY BBC3B_HUMAN Affects Response To Substance [12]
------------------------------------------------------------------------------------
⏷ Show the Full List of 11 DOT(s)
Indication(s) of Daunorubicin
Disease Entry ICD 11 Status REF
Acute monocytic leukemia N.A. Approved [3]
Acute myelogenous leukaemia 2A41 Approved [3]
Acute myeloid leukaemia 2A60 Approved [4]
Kaposi sarcoma 2B57 Approved [2]
Leukemia N.A. Approved [3]
Solid tumour/cancer 2A00-2F9Z Phase 2 [5]
Daunorubicin Interacts with 2 DTT Molecule(s)
DTT Name DTT ID UniProt ID Mode of Action REF
DNA replication (DNA repli) TTABD5E NOUNIPROTAC Modulator [19]
Human Deoxyribonucleic acid (hDNA) TTUTN1I NOUNIPROTAC Intercalator [20]
------------------------------------------------------------------------------------
Daunorubicin Interacts with 4 DTP Molecule(s)
DTP Name DTP ID UniProt ID Mode of Action REF
Multidrug resistance-associated protein 1 (ABCC1) DTSYQGK MRP1_HUMAN Substrate [21]
P-glycoprotein 1 (ABCB1) DTUGYRD MDR1_HUMAN Substrate [22]
Breast cancer resistance protein (ABCG2) DTI7UX6 ABCG2_HUMAN Substrate [23]
Multidrug resistance-associated protein 6 (ABCC6) DT582KR MRP6_HUMAN Substrate [24]
------------------------------------------------------------------------------------
Daunorubicin Interacts with 6 DME Molecule(s)
DME Name DME ID UniProt ID Mode of Action REF
Cytochrome P450 3A4 (CYP3A4) DE4LYSA CP3A4_HUMAN Metabolism [25]
Cytochrome P450 1A2 (CYP1A2) DEJGDUW CP1A2_HUMAN Metabolism [25]
Cytochrome P450 1A1 (CYP1A1) DE6OQ3W CP1A1_HUMAN Metabolism [25]
NADPH-dependent carbonyl reductase 1 (CBR1) DE9JFMC CBR1_HUMAN Metabolism [26]
NADPH-dependent carbonyl reductase 3 (CBR3) DEIVKZ8 CBR3_HUMAN Metabolism [27]
NADPH-cytochrome P450 reductase (CPR) DE3N2FM NCPR_HUMAN Metabolism [28]
------------------------------------------------------------------------------------
⏷ Show the Full List of 6 DME(s)
Daunorubicin Interacts with 158 DOT Molecule(s)
DOT Name DOT ID UniProt ID Mode of Action REF
Carbonyl reductase 3 (CBR3) OT9G6SF0 CBR3_HUMAN Affects Response To Substance [27]
Broad substrate specificity ATP-binding cassette transporter ABCG2 (ABCG2) OTW8V2V1 ABCG2_HUMAN Increases Expression [29]
ATP-dependent translocase ABCB1 (ABCB1) OTEJROBO MDR1_HUMAN Decreases Expression [30]
Multidrug resistance-associated protein 1 (ABCC1) OTGUN89S MRP1_HUMAN Decreases Response To Substance [31]
Heterogeneous nuclear ribonucleoprotein D0 (HNRNPD) OT5UO1FA HNRPD_HUMAN Affects Response To Substance [32]
Inhibitor of nuclear factor kappa-B kinase subunit epsilon (IKBKE) OT5VYOSM IKKE_HUMAN Affects Response To Substance [32]
Cytochrome P450 1B1 (CYP1B1) OTYXFLSD CP1B1_HUMAN Affects Response To Substance [32]
SEC14-like protein 3 (SEC14L3) OTGQ8GWM S14L3_HUMAN Increases ADR [32]
Retinoic acid receptor gamma (RARG) OT8LHMDH RARG_HUMAN Increases Response To Substance [33]
NAD(P)H dehydrogenase 1 (NQO1) OTZGGIVK NQO1_HUMAN Decreases Expression [34]
Voltage-dependent T-type calcium channel subunit alpha-1G (CACNA1G) OTGKBRE4 CAC1G_HUMAN Decreases Expression [13]
Tyrosine-protein kinase JAK2 (JAK2) OTBIDOOR JAK2_HUMAN Decreases Expression [13]
Sodium channel subunit beta-2 (SCN2B) OTFAHJ38 SCN2B_HUMAN Decreases Expression [13]
LIM domain-binding protein 3 (LDB3) OTGQL1AM LDB3_HUMAN Decreases Expression [13]
Creatine kinase M-type (CKM) OTME0KO7 KCRM_HUMAN Decreases Expression [13]
Glutathione peroxidase 1 (GPX1) OTE2O72Q GPX1_HUMAN Increases Expression [13]
Glycogen phosphorylase, muscle form (PYGM) OTWLVWTV PYGM_HUMAN Decreases Expression [13]
Dystrophin (DMD) OTD21T5J DMD_HUMAN Decreases Expression [13]
Myosin-7 (MYH7) OT4Z9T8N MYH7_HUMAN Decreases Expression [13]
Myosin-6 (MYH6) OT3YNCH1 MYH6_HUMAN Decreases Expression [13]
Troponin T, slow skeletal muscle (TNNT1) OT8PBOAR TNNT1_HUMAN Decreases Expression [13]
Receptor tyrosine-protein kinase erbB-3 (ERBB3) OTRSST0A ERBB3_HUMAN Decreases Expression [13]
Sarcoplasmic reticulum histidine-rich calcium-binding protein (HRC) OTDLEJ4T SRCH_HUMAN Decreases Expression [13]
Tumor necrosis factor receptor superfamily member 6 (FAS) OTP9XG86 TNR6_HUMAN Increases Expression [13]
Sodium/potassium-transporting ATPase subunit alpha-2 (ATP1A2) OTCF8OWW AT1A2_HUMAN Decreases Expression [13]
Potassium voltage-gated channel subfamily KQT member 1 (KCNQ1) OT8SPJNX KCNQ1_HUMAN Decreases Expression [13]
cAMP-dependent protein kinase catalytic subunit PRKX (PRKX) OTDAC6Z0 PRKX_HUMAN Increases Expression [13]
Myomesin-2 (MYOM2) OTD2UOXW MYOM2_HUMAN Decreases Expression [13]
Inward rectifier potassium channel 2 (KCNJ2) OT2OQEZS KCNJ2_HUMAN Increases Expression [13]
Actin, alpha skeletal muscle (ACTA1) OTOVGLPG ACTS_HUMAN Decreases Expression [13]
Iroquois-class homeodomain protein IRX-4 (IRX4) OT0TV6WK IRX4_HUMAN Decreases Expression [13]
Ankyrin-2 (ANK2) OTWB4R1Y ANK2_HUMAN Decreases Expression [13]
Apoptosis regulator BAX (BAX) OTAW0V4V BAX_HUMAN Increases Expression [13]
Potassium voltage-gated channel subfamily H member 2 (KCNH2) OTZX881H KCNH2_HUMAN Decreases Expression [13]
Dual specificity protein phosphatase 4 (DUSP4) OT6WAO12 DUS4_HUMAN Increases Expression [13]
Sodium channel protein type 5 subunit alpha (SCN5A) OTGYZWR6 SCN5A_HUMAN Decreases Expression [13]
Protein phosphatase 1 regulatory subunit 3A (PPP1R3A) OTJL9VYP PPR3A_HUMAN Decreases Expression [13]
Caveolae-associated protein 4 (CAVIN4) OTB1DM2C CAVN4_HUMAN Decreases Expression [13]
Nebulin-related-anchoring protein (NRAP) OTO6H3YF NRAP_HUMAN Decreases Expression [13]
Ryanodine receptor 2 (RYR2) OT0PF19E RYR2_HUMAN Decreases Expression [13]
Growth/differentiation factor 15 (GDF15) OTWQN50N GDF15_HUMAN Increases Expression [13]
Histone-lysine N-methyltransferase PRDM16 (PRDM16) OT0BGA27 PRD16_HUMAN Decreases Expression [13]
Voltage-dependent calcium channel subunit alpha-2/delta-2 (CACNA2D2) OTFJXVQQ CA2D2_HUMAN Decreases Expression [13]
Atrial natriuretic peptide-converting enzyme (CORIN) OT4SK7DK CORIN_HUMAN Decreases Expression [13]
DNA fragmentation factor subunit alpha (DFFA) OTFPFMT8 DFFA_HUMAN Increases Degradation [35]
Agrin (AGRN) OTWJENAZ AGRIN_HUMAN Increases Expression [36]
Etoposide-induced protein 2.4 homolog (EI24) OTD4NOYS EI24_HUMAN Increases Expression [36]
Tax1-binding protein 3 (TAX1BP3) OTQ6IB4T TX1B3_HUMAN Increases Expression [36]
Aurora kinase A (AURKA) OTMX0HYT AURKA_HUMAN Decreases Expression [37]
Protein Mdm4 (MDM4) OTU10BSF MDM4_HUMAN Decreases Expression [38]
Baculoviral IAP repeat-containing protein 5 (BIRC5) OTILXZYL BIRC5_HUMAN Increases Expression [39]
Cytoplasmic protein NCK2 (NCK2) OTUYPF55 NCK2_HUMAN Decreases Expression [36]
Protein regulator of cytokinesis 1 (PRC1) OTHD0XS0 PRC1_HUMAN Decreases Expression [36]
GTPase NRas (NRAS) OTVQ1DG3 RASN_HUMAN Increases Expression [38]
Estrogen receptor (ESR1) OTKLU61J ESR1_HUMAN Decreases Activity [14]
Glucocorticoid receptor (NR3C1) OTCI2YDI GCR_HUMAN Decreases Activity [40]
Cellular tumor antigen p53 (TP53) OTIE1VH3 P53_HUMAN Increases Expression [38]
Plasminogen activator inhibitor 1 (SERPINE1) OTT0MPQ3 PAI1_HUMAN Decreases Expression [41]
Endothelin-1 (EDN1) OTZCACEG EDN1_HUMAN Decreases Expression [42]
Retinoblastoma-associated protein (RB1) OTQJUJMZ RB_HUMAN Increases Expression [38]
Annexin A2 (ANXA2) OTFNS0CC ANXA2_HUMAN Decreases Expression [43]
Transcription factor Sp1 (SP1) OTISPT4X SP1_HUMAN Decreases Expression [44]
Interleukin-6 receptor subunit alpha (IL6R) OTCQL07Z IL6RA_HUMAN Decreases Expression [15]
Annexin A4 (ANXA4) OTUCRYXL ANXA4_HUMAN Increases Expression [36]
Poly polymerase 1 (PARP1) OT310QSG PARP1_HUMAN Increases Cleavage [45]
Interleukin-8 (CXCL8) OTS7T5VH IL8_HUMAN Decreases Expression [15]
Androgen receptor (AR) OTUBKAZZ ANDR_HUMAN Decreases Activity [14]
Apoptosis regulator Bcl-2 (BCL2) OT9DVHC0 BCL2_HUMAN Decreases Expression [46]
Microtubule-associated protein tau (MAPT) OTMTP2Z7 TAU_HUMAN Decreases Expression [47]
C-C motif chemokine 4 (CCL4) OT6B8P25 CCL4_HUMAN Decreases Expression [15]
C-C motif chemokine 2 (CCL2) OTAD2HEL CCL2_HUMAN Decreases Expression [15]
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) OTPWI88U CEAM1_HUMAN Increases Expression [36]
Tissue factor (F3) OT3MSU3B TF_HUMAN Increases Expression [48]
Histone H2AX (H2AX) OT18UX57 H2AX_HUMAN Increases Expression [44]
Cyclic AMP-responsive element-binding protein 1 (CREB1) OT1MDLA1 CREB1_HUMAN Decreases Phosphorylation [44]
Natriuretic peptides B (NPPB) OTSN2IPY ANFB_HUMAN Increases Expression [49]
Sphingomyelin phosphodiesterase (SMPD1) OTZSMA54 ASM_HUMAN Increases Expression [50]
Syndecan-1 (SDC1) OTWKRVZC SDC1_HUMAN Increases Expression [36]
NADPH:adrenodoxin oxidoreductase, mitochondrial (FDXR) OTPDCK1F ADRO_HUMAN Increases Expression [36]
Ribosomal protein S6 kinase beta-1 (RPS6KB1) OTAELNGX KS6B1_HUMAN Increases Phosphorylation [51]
G1/S-specific cyclin-E1 (CCNE1) OTLD7UID CCNE1_HUMAN Affects Expression [38]
Mitogen-activated protein kinase 3 (MAPK3) OTCYKGKO MK03_HUMAN Decreases Phosphorylation [44]
Mitogen-activated protein kinase 1 (MAPK1) OTH85PI5 MK01_HUMAN Decreases Phosphorylation [44]
G1/S-specific cyclin-D3 (CCND3) OTNKPQ22 CCND3_HUMAN Increases Expression [38]
M-phase inducer phosphatase 1 (CDC25A) OTSLKKCO MPIP1_HUMAN Increases Expression [38]
Peroxisome proliferator-activated receptor gamma (PPARG) OTHMARHO PPARG_HUMAN Decreases Activity [14]
Cyclin-dependent kinase inhibitor 1 (CDKN1A) OTQWHCZE CDN1A_HUMAN Increases Expression [38]
Tumor necrosis factor ligand superfamily member 9 (TNFSF9) OTV9L89D TNFL9_HUMAN Increases Expression [36]
Serine/threonine-protein kinase mTOR (MTOR) OTHH8KU7 MTOR_HUMAN Increases Phosphorylation [51]
Caspase-3 (CASP3) OTIJRBE7 CASP3_HUMAN Increases Activity [45]
Cyclin-dependent kinase inhibitor 2A (CDKN2A) OTN0ZWAE CDN2A_HUMAN Affects Expression [38]
Cyclin-dependent kinase 4 inhibitor B (CDKN2B) OTAG24N1 CDN2B_HUMAN Increases Expression [38]
Dual specificity mitogen-activated protein kinase kinase 3 (MAP2K3) OTI2OREX MP2K3_HUMAN Increases Expression [36]
Dual specificity protein kinase CLK2 (CLK2) OT1CRSY2 CLK2_HUMAN Increases Expression [38]
Caspase-9 (CASP9) OTD4RFFG CASP9_HUMAN Increases Cleavage [44]
Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform (PPP2CA) OT83PT85 PP2AA_HUMAN Increases Expression [44]
Cytochrome c (CYCS) OTBFALJD CYC_HUMAN Affects Expression [39]
Transcription factor Sp3 (SP3) OTYDQZ1T SP3_HUMAN Increases Expression [50]
Peroxisome proliferator-activated receptor delta (PPARD) OTI4WTOP PPARD_HUMAN Decreases Activity [14]
Antigen peptide transporter 1 (TAP1) OTJL27PW TAP1_HUMAN Increases Expression [36]
CCAAT/enhancer-binding protein zeta (CEBPZ) OT11BATG CEBPZ_HUMAN Decreases Expression [36]
Protein kinase C delta type (PRKCD) OTSEH90E KPCD_HUMAN Increases Cleavage [52]
Induced myeloid leukemia cell differentiation protein Mcl-1 (MCL1) OT2YYI1A MCL1_HUMAN Decreases Expression [44]
Serine-protein kinase ATM (ATM) OTQVOHLT ATM_HUMAN Increases Expression [38]
Eukaryotic translation initiation factor 4E-binding protein 1 (EIF4EBP1) OTHBQVD5 4EBP1_HUMAN Increases Phosphorylation [51]
Retinoblastoma-binding protein 5 (RBBP5) OT12W2MK RBBP5_HUMAN Affects Expression [38]
Transcription factor E2F5 (E2F5) OT1XWING E2F5_HUMAN Increases Expression [38]
Cyclin-G2 (CCNG2) OTII38K2 CCNG2_HUMAN Increases Expression [53]
Quinone oxidoreductase PIG3 (TP53I3) OTSCM68G QORX_HUMAN Increases Expression [36]
EPM2A-interacting protein 1 (EPM2AIP1) OT21ALNF EPMIP_HUMAN Increases Expression [36]
E3 ubiquitin-protein ligase RBBP6 (RBBP6) OTTVG4HU RBBP6_HUMAN Increases Expression [38]
Bile acid receptor (NR1H4) OTWZLPTB NR1H4_HUMAN Decreases Activity [14]
Mitogen-activated protein kinase kinase kinase 5 (MAP3K5) OTQR6ENW M3K5_HUMAN Decreases Expression [36]
BRCA1-associated RING domain protein 1 (BARD1) OTTC0Z9Y BARD1_HUMAN Affects Expression [38]
Serine/threonine-protein kinase PLK3 (PLK3) OT19CT2Z PLK3_HUMAN Increases Expression [36]
NADPH oxidase 4 (NOX4) OTTYQ097 NOX4_HUMAN Increases Expression [44]
Sphingomyelin phosphodiesterase 3 (SMPD3) OTHQBETH NSMA2_HUMAN Increases Expression [50]
F-box/WD repeat-containing protein 1A (BTRC) OT2EZDGR FBW1A_HUMAN Increases Expression [44]
Calcium-binding protein 39 (CAB39) OT2CL9ST CAB39_HUMAN Decreases Expression [36]
Tyrosine-protein kinase ABL1 (ABL1) OT09YVXH ABL1_HUMAN Decreases Response To Substance [54]
Integrin-linked kinase-associated serine/threonine phosphatase 2C (ILKAP) OT2A6FSA ILKAP_HUMAN Affects Response To Substance [17]
Kinesin-like protein KIF1A (KIF1A) OT3JVEGV KIF1A_HUMAN Affects Response To Substance [17]
Phosphatidylinositol 3-kinase regulatory subunit alpha (PIK3R1) OT5BZ1J9 P85A_HUMAN Affects Response To Substance [17]
Solute carrier family 28 member 3 (SLC28A3) OT5TLTYU S28A3_HUMAN Increases ADR [16]
Protein GAPT (GAPT) OT6C2WXX GAPT_HUMAN Affects Response To Substance [17]
RAC-alpha serine/threonine-protein kinase (AKT1) OT8H2YY7 AKT1_HUMAN Decreases Response To Substance [55]
Glypican-5 (GPC5) OT8NR7GC GPC5_HUMAN Affects Response To Substance [17]
Voltage-dependent L-type calcium channel subunit alpha-1S (CACNA1S) OT96MCM2 CAC1S_HUMAN Affects Response To Substance [17]
Ephexin-1 (NGEF) OT9QUXJ3 NGEF_HUMAN Affects Response To Substance [17]
Spliceosome-associated protein CWC27 homolog (CWC27) OTB0HBP1 CWC27_HUMAN Affects Response To Substance [17]
Cadherin-13 (CDH13) OTD2CYM5 CAD13_HUMAN Affects Response To Substance [17]
Sp110 nuclear body protein (SP110) OTFW6WH7 SP110_HUMAN Affects Response To Substance [17]
Leucine zipper protein 2 (LUZP2) OTGGOSPW LUZP2_HUMAN Affects Response To Substance [17]
Catalase (CAT) OTHEBX9R CATA_HUMAN Affects Response To Substance [56]
Transgelin-3 (TAGLN3) OTJBP3VJ TAGL3_HUMAN Affects Response To Substance [17]
ATP-binding cassette sub-family C member 2 (ABCC2) OTJSIGV5 MRP2_HUMAN Affects Response To Substance [32]
Inositol polyphosphate 4-phosphatase type II (INPP4B) OTLROA7G INP4B_HUMAN Affects Response To Substance [17]
Anoctamin-3 (ANO3) OTM34U6Q ANO3_HUMAN Affects Response To Substance [17]
Protein kinase C zeta type (PRKCZ) OTN2FE42 KPCZ_HUMAN Decreases Response To Substance [57]
RelA-associated inhibitor (PPP1R13L) OTNCPLWE IASPP_HUMAN Decreases Response To Substance [58]
Transmembrane 4 L6 family member 20 (TM4SF20) OTNXU9QW T4S20_HUMAN Affects Response To Substance [17]
Aldo-keto reductase family 1 member B10 (AKR1B10) OTOA4HTH AK1BA_HUMAN Decreases Response To Substance [59]
Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN (PTEN) OTOWDUNT PTEN_HUMAN Increases Response To Substance [60]
Ran-binding protein 17 (RANBP17) OTQEVTYH RBP17_HUMAN Affects Response To Substance [17]
NPC intracellular cholesterol transporter 1 (NPC1) OTRIPICX NPC1_HUMAN Increases Secretion [61]
Aldo-keto reductase family 1 member B1 (AKR1B1) OTRX72TH ALDR_HUMAN Increases Reduction [62]
COX assembly mitochondrial protein 2 homolog (CMC2) OTRYIO1J COXM2_HUMAN Affects Response To Substance [17]
Glutathione S-transferase Mu 1 (GSTM1) OTSBF2MO GSTM1_HUMAN Affects Response To Substance [18]
Protein SPMIP2 (SPMIP2) OTSNYR9R SMIP2_HUMAN Affects Response To Substance [17]
Aldo-keto reductase family 1 member C3 (AKR1C3) OTU2SXBA AK1C3_HUMAN Decreases Response To Substance [63]
Slit homolog 3 protein (SLIT3) OTU8MKEU SLIT3_HUMAN Affects Response To Substance [17]
Microtubule-associated protein 1B (MAP1B) OTVXW089 MAP1B_HUMAN Affects Response To Substance [17]
Antigen peptide transporter 2 (TAP2) OTWSYFI7 TAP2_HUMAN Affects Response To Substance [32]
Citramalyl-CoA lyase, mitochondrial (CLYBL) OTXZLTST CLYBL_HUMAN Affects Response To Substance [17]
Limbic system-associated membrane protein (LSAMP) OTYXVQX2 LSAMP_HUMAN Affects Response To Substance [17]
C-Maf-inducing protein (CMIP) OTZN8Z4A CMIP_HUMAN Affects Response To Substance [17]
Aldo-keto reductase family 1 member A1 (AKR1A1) OTZT9NOW AK1A1_HUMAN Increases Reduction [64]
EGF-containing fibulin-like extracellular matrix protein 1 (EFEMP1) OTZVUOOB FBLN3_HUMAN Affects Response To Substance [65]
------------------------------------------------------------------------------------
⏷ Show the Full List of 158 DOT(s)

References

1 ClinicalTrials.gov (NCT05342584) Venetoclax Plus Intensive Chemotherapy in AML and Advanced MDS
2 Drugs@FDA. U.S. Food and Drug Administration. U.S. Department of Health & Human Services. 2015
3 Daunorubicin FDA Label
4 URL: http://www.guidetopharmacology.org Nucleic Acids Res. 2015 Oct 12. pii: gkv1037. The IUPHAR/BPS Guide to PHARMACOLOGY in 2016: towards curated quantitative interactions between 1300 protein targets and 6000 ligands. (Ligand id: 7063).
5 Multicentre, phase II study evaluating capecitabine monotherapy in patients with anthracycline- and taxane-pretreated metastatic breast cancer. Eur J Cancer. 2004 Mar;40(4):536-42.
6 Venclexta FDA label
7 ZCL-082, a boron-containing compound, induces apoptosis of non-Hodgkin's lymphoma via targeting p90 ribosomal S6 kinase 1/NF-B signaling pathway. Chem Biol Interact. 2022 Jan 5;351:109770. doi: 10.1016/j.cbi.2021.109770. Epub 2021 Nov 30.
8 Genomics and drug profiling of fatal TCF3-HLF-positive acute lymphoblastic leukemia identifies recurrent mutation patterns and therapeutic options. Nat Genet. 2015 Sep;47(9):1020-1029. doi: 10.1038/ng.3362. Epub 2015 Jul 27.
9 HSP90 Inhibitor PU-H71 in Combination with BH3-Mimetics in the Treatment of Acute Myeloid Leukemia. Curr Issues Mol Biol. 2023 Aug 23;45(9):7011-7026. doi: 10.3390/cimb45090443.
10 Superior efficacy of cotreatment with BET protein inhibitor and BCL2 or MCL1 inhibitor against AML blast progenitor cells. Blood Cancer J. 2019 Jan 15;9(2):4. doi: 10.1038/s41408-018-0165-5.
11 Prospective Drug Candidates as Human Multidrug Transporter ABCG2 Inhibitors: an In Silico Drug Discovery Study. Cell Biochem Biophys. 2021 Jun;79(2):189-200. doi: 10.1007/s12013-021-00985-y. Epub 2021 May 5.
12 Statin-induced Mitochondrial Priming Sensitizes Multiple Myeloma Cells to BCL2 and MCL-1 Inhibitors. Cancer Res Commun. 2023 Dec 8;3(12):2497-2509. doi: 10.1158/2767-9764.CRC-23-0350.
13 Identification of genomic biomarkers for anthracycline-induced cardiotoxicity in human iPSC-derived cardiomyocytes: an in vitro repeated exposure toxicity approach for safety assessment. Arch Toxicol. 2016 Nov;90(11):2763-2777.
14 Quantitative high-throughput profiling of environmental chemicals and drugs that modulate farnesoid X receptor. Sci Rep. 2014 Sep 26;4:6437. doi: 10.1038/srep06437.
15 Cell-based and cytokine-directed chemical screen to identify potential anti-multiple myeloma agents. Leuk Res. 2010 Jul;34(7):917-24. doi: 10.1016/j.leukres.2009.12.002. Epub 2010 Feb 8.
16 Recommendations for genetic testing to reduce the incidence of anthracycline-induced cardiotoxicity. Br J Clin Pharmacol. 2016 Sep;82(3):683-95. doi: 10.1111/bcp.13008. Epub 2016 Jun 30.
17 Mapping genes that contribute to daunorubicin-induced cytotoxicity. Cancer Res. 2007 Jun 1;67(11):5425-33. doi: 10.1158/0008-5472.CAN-06-4431.
18 Pharmacogenetics of outcome in children with acute lymphoblastic leukemia. Blood. 2005 Jun 15;105(12):4752-8. doi: 10.1182/blood-2004-11-4544. Epub 2005 Feb 15.
19 Drugs@FDA. U.S. Food and Drug Administration. U.S. Department of Health & Human Services.
20 31P NMR spectra of ethidium, quinacrine, and daunomycin complexes with poly(adenylic acid).poly(uridylic acid) RNA duplex and calf thymus DNA. Biochemistry. 1989 Apr 4;28(7):2804-12.
21 Vinblastine and sulfinpyrazone export by the multidrug resistance protein MRP2 is associated with glutathione export. Br J Cancer. 2000 Aug;83(3):375-83.
22 Mammalian drug efflux transporters of the ATP binding cassette (ABC) family in multidrug resistance: A review of the past decade. Cancer Lett. 2016 Jan 1;370(1):153-64.
23 Functional characterization of the human multidrug transporter, ABCG2, expressed in insect cells. Biochem Biophys Res Commun. 2001 Jul 6;285(1):111-7.
24 Characterization of the drug resistance and transport properties of multidrug resistance protein 6 (MRP6, ABCC6). Cancer Res. 2002 Nov 1;62(21):6172-7.
25 The effect of new lipophilic chelators on the activities of cytosolic reductases and P450 cytochromes involved in the metabolism of anthracycline antibiotics: studies in vitro. Physiol Res. 2004;53(6):683-91.
26 Flavonoids as inhibitors of human carbonyl reductase 1. Chem Biol Interact. 2008 Jul 30;174(2):98-108.
27 Naturally occurring variants of human CBR3 alter anthracycline in vitro metabolism. J Pharmacol Exp Ther. 2010 Mar;332(3):755-63.
28 Lack of mechanism-based inactivation of rat hepatic microsomal cytochromes P450 by doxorubicin. Can J Physiol Pharmacol. 1999 Aug;77(8):589-97.
29 Chemotherapeutic drug-induced ABCG2 promoter demethylation as a novel mechanism of acquired multidrug resistance. Neoplasia. 2009 Dec;11(12):1359-70. doi: 10.1593/neo.91314.
30 Reversal of multidrug resistance by magnetic Fe3O4 nanoparticle copolymerizating daunorubicin and MDR1 shRNA expression vector in leukemia cells. Int J Nanomedicine. 2010 Aug 9;5:437-44. doi: 10.2147/ijn.s10083.
31 Cloning and functional characterization of the multidrug resistance-associated protein (MRP1/ABCC1) from the cynomolgus monkey. Mol Cancer Ther. 2003 Mar;2(3):307-16.
32 Genetic variants contributing to daunorubicin-induced cytotoxicity. Cancer Res. 2008 May 1;68(9):3161-8. doi: 10.1158/0008-5472.CAN-07-6381.
33 A coding variant in RARG confers susceptibility to anthracycline-induced cardiotoxicity in childhood cancer. Nat Genet. 2015 Sep;47(9):1079-84. doi: 10.1038/ng.3374. Epub 2015 Aug 3.
34 High-throughput library screening identifies two novel NQO1 inducers in human lung cells. Am J Respir Cell Mol Biol. 2012 Mar;46(3):365-71.
35 Comparison of anthracycline-induced death of human leukemia cells: programmed cell death versus necrosis. Apoptosis. 2002 Dec;7(6):537-48. doi: 10.1023/a:1020647211557.
36 Characterization of DNA reactive and non-DNA reactive anticancer drugs by gene expression profiling. Mutat Res. 2007 Jun 1;619(1-2):16-29. doi: 10.1016/j.mrfmmm.2006.12.007. Epub 2007 Feb 8.
37 BET inhibition as a single or combined therapeutic approach in primary paediatric B-precursor acute lymphoblastic leukaemia. Blood Cancer J. 2013 Jul 19;3(7):e126. doi: 10.1038/bcj.2013.24.
38 Daunorubicin-induced variations in gene transcription: commitment to proliferation arrest, senescence and apoptosis. Biochem J. 2003 Jun 15;372(Pt 3):703-11. doi: 10.1042/BJ20021950.
39 Identification of the key pathway of oxazolinoanthracyclines mechanism of action in cells derived from human solid tumors. Toxicol Appl Pharmacol. 2016 Dec 15;313:159-169. doi: 10.1016/j.taap.2016.10.018. Epub 2016 Oct 22.
40 Discovery of selective glucocorticoid receptor modulators by multiplexed reporter screening. Proc Natl Acad Sci U S A. 2009 Mar 24;106(12):4929-34. doi: 10.1073/pnas.0812308106. Epub 2009 Mar 2.
41 Daunorubicin attenuates tumor necrosis factor-alpha-induced biosynthesis of plasminogen activator inhibitor-1 in human umbilical vein endothelial cells. Biochim Biophys Acta. 2001 Apr 23;1538(2-3):234-41. doi: 10.1016/s0167-4889(01)00073-8.
42 Role of endothelin-1 in the development of a special type of cardiomyopathy. Clin Sci (Lond). 2002 Aug;103 Suppl 48:272S-275S. doi: 10.1042/CS103S272S.
43 [Influence of arsenic trioxide and daunorubicin on the expression of annexin II and fibrinolytic activity in NB4 cells]. Zhonghua Xue Ye Xue Za Zhi. 2010 Dec;31(12):813-6.
44 CREB/Sp1-mediated MCL1 expression and NFB-mediated ABCB1 expression modulate the cytotoxicity of daunorubicin in chronic myeloid leukemia cells. Toxicol Appl Pharmacol. 2022 Jan 15;435:115847. doi: 10.1016/j.taap.2021.115847. Epub 2021 Dec 25.
45 Anthracyclines trigger apoptosis of both G0-G1 and cycling peripheral blood lymphocytes and induce massive deletion of mature T and B cells. Cancer Res. 2000 Apr 1;60(7):1901-7.
46 Reversal of multidrug resistance by magnetic Fe3O4 nanoparticle copolymerizating daunorubicin and 5-bromotetrandrine in xenograft nude-mice. Int J Nanomedicine. 2009;4:73-8. doi: 10.2147/ijn.s5093. Epub 2009 Apr 1.
47 Pharmacologic reductions of total tau levels; implications for the role of microtubule dynamics in regulating tau expression. Mol Neurodegener. 2006 Jul 26;1:6. doi: 10.1186/1750-1326-1-6.
48 [Effects of all-trans retinoic acid, arsenic trioxide and daunorubicin on tissue factor expression in NB4 cells]. Zhonghua Xue Ye Xue Za Zhi. 1999 Sep;20(9):453-5.
49 Brain natriuretic peptide is a predictor of anthracycline-induced cardiotoxicity. Acta Haematol. 2000;104(4):158-63. doi: 10.1159/000046508.
50 Transcriptional regulation of neutral sphingomyelinase 2 gene expression of a human breast cancer cell line, MCF-7, induced by the anti-cancer drug, daunorubicin. Biochim Biophys Acta. 2009 Nov-Dec;1789(11-12):681-90. doi: 10.1016/j.bbagrm.2009.08.006. Epub 2009 Aug 19.
51 Antileukaemia effect of rapamycin alone or in combination with daunorubicin on Ph+ acute lymphoblastic leukaemia cell line. Hematol Oncol. 2012 Sep;30(3):123-30. doi: 10.1002/hon.1013. Epub 2011 Sep 4.
52 Regulation of phospholipase D activity and ceramide production in daunorubicin-induced apoptosis in A-431 cells. Biochim Biophys Acta. 2000 Nov 15;1488(3):219-32. doi: 10.1016/s1388-1981(00)00125-6.
53 Cotylenin A, a new differentiation inducer, and rapamycin cooperatively inhibit growth of cancer cells through induction of cyclin G2. Cancer Sci. 2008 Aug;99(8):1693-8. doi: 10.1111/j.1349-7006.2008.00867.x.
54 Resistance of bcr-abl-positive acute lymphoblastic leukemia to daunorubicin is not mediated by mdr1 gene expression. Am J Hematol. 2002 Nov;71(3):172-6. doi: 10.1002/ajh.10212.
55 Daunorubicin induces cell death via activation of apoptotic signalling pathway and inactivation of survival pathway in muscle-derived stem cells. Cell Biol Toxicol. 2012 Apr;28(2):103-14. doi: 10.1007/s10565-011-9210-x. Epub 2012 Jan 18.
56 Down-regulation of catalase gene expression in the doxorubicin-resistant AML subline AML-2/DX100. Biochem Biophys Res Commun. 2001 Feb 16;281(1):109-14. doi: 10.1006/bbrc.2001.4324.
57 4-benzimidazolyl-3-phenylbutanoic acids as novel PIF-pocket-targeting allosteric inhibitors of protein kinase PKC. J Med Chem. 2011 Oct 13;54(19):6714-23. doi: 10.1021/jm2005892. Epub 2011 Sep 14.
58 siRNA-mediated down-regulation of iASPP promotes apoptosis induced by etoposide and daunorubicin in leukemia cells expressing wild-type p53. Leuk Res. 2009 Sep;33(9):1243-8. doi: 10.1016/j.leukres.2009.02.016. Epub 2009 Mar 18.
59 Cancer biomarker AKR1B10 and carbonyl metabolism. Chem Biol Interact. 2009 Mar 16;178(1-3):134-7. doi: 10.1016/j.cbi.2008.10.044. Epub 2008 Nov 5.
60 Involvement of miR-21 in resistance to daunorubicin by regulating PTEN expression in the leukaemia K562 cell line. FEBS Lett. 2011 Jan 21;585(2):402-8. doi: 10.1016/j.febslet.2010.12.027. Epub 2010 Dec 25.
61 Niemann-Pick C1 protein facilitates the efflux of the anticancer drug daunorubicin from cells according to a novel vesicle-mediated pathway. J Pharmacol Exp Ther. 2006 Jan;316(1):242-7. doi: 10.1124/jpet.105.089482. Epub 2005 Sep 20.
62 Major differences exist in the function and tissue-specific expression of human aflatoxin B1 aldehyde reductase and the principal human aldo-keto reductase AKR1 family members. Biochem J. 1999 Oct 15;343 Pt 2(Pt 2):487-504.
63 Aldo-keto reductase 1C3 (AKR1C3): a missing piece of the puzzle in the dinaciclib interaction profile. Arch Toxicol. 2018 Sep;92(9):2845-2857.
64 Aldo-keto reductase 1C2 fails to metabolize doxorubicin and daunorubicin in vitro. Drug Metab Dispos. 2008 Jun;36(6):991-4. doi: 10.1124/dmd.108.020388. Epub 2008 Mar 5.
65 Decreased expression of angiogenesis antagonist EFEMP1 in sporadic breast cancer is caused by aberrant promoter methylation and points to an impact of EFEMP1 as molecular biomarker. Int J Cancer. 2009 Apr 1;124(7):1727-35. doi: 10.1002/ijc.24108.