Preferred name
TOPOTECAN
Synonyms
TOPOTECAN HYDROCHLORIDE ()
SKF 104864A ()
NSC 609669 ()
Topotecan HCl ()
NSC609699 ()
Nogitecan HCl ()
SKFS 104864A ()
Hycamtin ()
Topotecan (HCl) ()
Topotecan HCl (Hycamtin) ()
Hycamptin ()
Hycamptamine ()
9-Dimethylaminomethyl-10-hydroxycamptothecin, HCl salt ()
SKF-S 104864-A ()
1H-Pyrano[3',4':6,7]indolizino[1,2-b]quinoline- 3,14(4H,12H)-dione, 10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-, monohydrochloride, (4S)- ()
SKF 104864A (Hydrochloride) ()
NSC 609669 (Hydrochloride) ()
Topetecan ()
Topotecan (Hydrochloride) ()
Topotecan (NSC609699) HCl ()
Nogitecan HCl, SKFS 104864A,Hycamtin, NSC 609699 ()
NSC609699,Nogitecan,SKFS 104864A ()
NSC-609699 ()
Topotecane ()
NSC-641007 ()
SKF-104864 ()
Ff-10850 (liposomal topotecan) ()
Topotecan ()
SK-S-104864-A ()
Evotopin ()
Topotecan teva ()
SK&F S-104864-A ()
SK&F-S-104864A ()
Topotecan hospira ()
Topotecan actavis ()
NSC-759263 ()
Nogitecan hydrochloride ()
Potactasol ()
Topotecan (as hydrochloride) ()
Topotecan-d6 ()
P&D ID
PD003417
CAS
123948-87-8
119413-54-6
1044904-10-0
Tags
available
nuisance
drug
probe
Approved by
FDA
EMA
First approval
1996
Drug indication
Small-cell lung cancer
Ovarian cancer
Solid tumour/cancer
Antineoplastic (DNA topoisomerase I Inhibitor)
Drug Status
withdrawn
approved
investigational
Max Phase
4.0
Probe control not defined
3
0
No structurally similar probes found
SMILES
CC[C@@]1(O)C(=O)OCc2c1cc1n(c2=O)Cc2cc3c(CN(C)C)c(O)ccc3nc2-1
InChI
InChI=1S/C23H23N3O5/c1-4-23(30)16-8-18-20-12(9-26(18)21(28)15(16)11-31-22(23)29)7-13-14(10-25(2)3)19(27)6-5-17(13)24-20/h5-8,27,30H,4,9-11H2,1-3H3/t23-/m0/s1
InChIkey
UCFGDBYHRUNTLO-QHCPKHFHSA-N
MOL
TOPOTECAN
RDKit 2D
31 35 0 0 0 0 0 0 0 0999 V2000
4.0586 0.4512 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
2.6491 0.9642 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
1.5000 0.0000 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
2.6491 -0.9642 0.0000 O 0 0 0 0 0 0 0 0 0 0 0 0
0.7500 -1.2990 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
1.5000 -2.5981 0.0000 O 0 0 0 0 0 0 0 0 0 0 0 0
-0.7500 -1.2990 0.0000 O 0 0 0 0 0 0 0 0 0 0 0 0
-1.5000 0.0000 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
-0.7500 1.2990 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
0.7500 1.2990 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
1.5000 2.5981 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
0.7500 3.8971 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
-0.7500 3.8971 0.0000 N 0 0 0 0 0 0 0 0 0 0 0 0
-1.5000 2.5981 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
-3.0000 2.5981 0.0000 O 0 0 0 0 0 0 0 0 0 0 0 0
-1.2135 5.3237 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
0.0000 6.2054 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
0.1568 7.6972 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
1.5271 8.3073 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
1.6839 9.7990 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
0.4704 10.6807 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
0.6272 12.1725 0.0000 N 0 0 0 0 0 0 0 0 0 0 0 0
-0.5864 13.0542 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
1.9975 12.7826 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
3.0542 10.4092 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
3.2110 11.9009 0.0000 O 0 0 0 0 0 0 0 0 0 0 0 0
4.2677 9.5275 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
4.1110 8.0357 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
2.7406 7.4256 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
2.5838 5.9338 0.0000 N 0 0 0 0 0 0 0 0 0 0 0 0
1.2135 5.3237 0.0000 C 0 0 0 0 0 0 0 0 0 0 0 0
1 2 1 0
2 3 1 0
3 4 1 6
3 5 1 0
5 6 2 0
5 7 1 0
7 8 1 0
8 9 1 0
9 10 2 0
10 11 1 0
11 12 2 0
12 13 1 0
13 14 1 0
14 15 2 0
13 16 1 0
16 17 1 0
17 18 2 0
18 19 1 0
19 20 2 0
20 21 1 0
21 22 1 0
22 23 1 0
22 24 1 0
20 25 1 0
25 26 1 0
25 27 2 0
27 28 1 0
28 29 2 0
29 30 1 0
30 31 2 0
10 3 1 0
31 12 1 0
14 9 1 0
31 17 1 0
29 19 1 0
M END
> <ID>
PD003417
> <Name>
TOPOTECAN
(extracted from source data)
MOA
Topotecan has the same mechanism of action as irinotecan and is believed to exert its cytotoxic effects during the S-phase of DNA synthesis. Topoisomerase I relieves torsional strain in DNA by inducing reversible single strand breaks. Topotecan binds to the topoisomerase I-DNA complex and prevents religation of these single strand breaks. This ternary complex interferes with the moving replication fork, which leads to the induction of replication arrest and lethal double-stranded breaks in DNA. As mammalian cells cannot efficiently repair these double strand breaks, the formation of this ternary complex eventually leads to apoptosis (programmed cell death).; ; Topotecan mimics a DNA base pair and binds at the site of DNA cleavage by intercalating between the upstream (−1) and downstream (+1) base pairs. Intercalation displaces the downstream DNA, thus preventing religation of the cleaved strand. By specifically binding to the enzyme–substrate complex, Topotecan acts as an uncompetitive inhibitor.
DESCRIPTION
Topotecan is a semisynthetic analogue of the natural plant alkaloid camptothecin (PubChem CID 24360). Camptothecin was first isolated from the bark and stem of Camptotheca acuminata, the Chinsese happy tree. Topotecan is an anticancer drug which causes DNA strand breaks and leads to cell-cycle arrest and cell death.
Marketed formulations may contain topotecan hydrochloride (PubChem CID 60699). (GtoPdb)
Marketed formulations may contain topotecan hydrochloride (PubChem CID 60699). (GtoPdb)
DESCRIPTION
inhibitor of topoisomerase I
(Informer Set)
DESCRIPTION
Inhibitor of UCHL5 and USP14
(Tocris Bioactive Compound Library)
26
7.15
EPAS1,HIF1A Hypoxia inducible factors; HIF-1-alpha, HIF-2-alpha
0
1
6.6
7.57
HIF1A Hypoxia-inducible factor 1-alpha
CELL-BASED 2.6x
11
3
6.6
LMNA Prelamin-A/C
BIOCHEM 1x
8
1
6.23
TOP2A,TOP2B DNA topoisomerase II
0
1
5.97
5.88
TOP1 DNA topoisomerase 1 inhibitor
1
13
5.89
SLC47A1 Multidrug and toxin extrusion protein 1
1
1
5.82
CYP3A4 Cytochrome P450 3A4
7
1
5.67
ORF 73
0
1
5.55
RMLC dTDP-4-dehydrorhamnose 3,5-epimerase
0
1
5.5
LEF Lethal factor
1
1
5.44
LANA1 Protein LANA1
0
1
5.43
KEAP1,NFE2L2 Keap1/Nrf2
0
1
5.32
RORC Nuclear receptor ROR-gamma
3
1
5.25
CASP1 Caspase-1
10
1
5.25
CASP7 Caspase-7
6
1
5.2
BRCA1 Breast cancer type 1 susceptibility protein
27
1
5.16
GLF UDP-galactopyranose mutase
0
1
5.07
SLC47A2 Multidrug and toxin extrusion protein 2
1
1
4.9
HBB Hemoglobin subunit beta
9
1
4.9
KMT2A,MEN1 Menin/Histone-lysine N-methyltransferase MLL
0
1
4.55
MAPT Microtubule-associated protein tau
3
1
4.55
IMPA1 Inositol monophosphatase 1
1
1
4.35
KMT2A Histone-lysine N-methyltransferase 2A
5
1
4.21
SLC22A2 Solute carrier family 22 member 2
5
1
TOP1MT DNA topoisomerase I, mitochondrial inhibitor
0
0
EPAS1 Endothelial PAS domain-containing protein 1
0
94
Abacavir transmembrane transport
SLC22A2
Activation of AMPK downstream of NMDARs
MAPT
Activation of caspases through apoptosome-mediated cleavage
CASP7
Aflatoxin activation and detoxification
CYP3A4
Apoptotic cleavage of cellular proteins
CASP7
Aspirin ADME
CYP3A4
Atorvastatin ADME
CYP3A4
Biosynthesis of maresin-like SPMs
CYP3A4
Breakdown of the nuclear lamina
LMNA
Caspase-mediated cleavage of cytoskeletal proteins
MAPT
CASP7
Cellular response to hypoxia
HIF1A
Chaperone Mediated Autophagy
HBB
Circadian Clock
HIF1A
Cytoprotection by HMOX1
HBB
Defective DNA double strand break response due to BARD1 loss of function
BRCA1
Defective DNA double strand break response due to BRCA1 loss of function
BRCA1
Defective HDR through Homologous Recombination Repair (HRR) due to PALB2 loss of BRCA1 binding function
BRCA1
Defective HDR through Homologous Recombination Repair (HRR) due to PALB2 loss of BRCA2/RAD51/RAD51C binding function
BRCA1
Defective homologous recombination repair (HRR) due to BRCA1 loss of function
BRCA1
Depolymerization of the Nuclear Lamina
LMNA
Deregulated CDK5 triggers multiple neurodegenerative pathways in Alzheimer's disease models
LMNA
Erythrocytes take up carbon dioxide and release oxygen
HBB
Erythrocytes take up oxygen and release carbon dioxide
HBB
Factors involved in megakaryocyte development and platelet production
HBB
Formation of WDR5-containing histone-modifying complexes
KMT2A
G2/M DNA damage checkpoint
BRCA1
HDR through Homologous Recombination (HRR)
BRCA1
HDR through Single Strand Annealing (SSA)
BRCA1
Heme signaling
HBB
Homologous DNA Pairing and Strand Exchange
BRCA1
Impaired BRCA2 binding to PALB2
BRCA1
Impaired BRCA2 binding to RAD51
BRCA1
Initiation of Nuclear Envelope (NE) Reformation
LMNA
Interleukin-1 processing
CASP1
Interleukin-37 signaling
CASP1
Interleukin-4 and Interleukin-13 signaling
RORC
HIF1A
KEAP1-NFE2L2 pathway
BRCA1
Late endosomal microautophagy
HBB
Meiotic recombination
BRCA1
Meiotic synapsis
BRCA1
LMNA
Metalloprotease DUBs
BRCA1
Na+/Cl- dependent neurotransmitter transporters
SLC22A2
Neddylation
BRCA1
HIF1A
Neurotransmitter clearance
SLC22A2
Neutrophil degranulation
HBB
NOD1/2 Signaling Pathway
CASP1
Nonhomologous End-Joining (NHEJ)
BRCA1
Norepinephrine Neurotransmitter Release Cycle
SLC22A2
NOTCH1 Intracellular Domain Regulates Transcription
HIF1A
Nuclear Envelope Breakdown
LMNA
Nuclear Receptor transcription pathway
RORC
Organic cation transport
SLC22A2
Oxygen-dependent proline hydroxylation of Hypoxia-inducible Factor Alpha
HIF1A
Phase I - Functionalization of compounds
CYP3A4
PKMTs methylate histone lysines
KMT2A
PKR-mediated signaling
MAPT
Prednisone ADME
CYP3A4
Presynaptic phase of homologous DNA pairing and strand exchange
BRCA1
Processing of DNA double-strand break ends
BRCA1
PTK6 Expression
HIF1A
PTK6 promotes HIF1A stabilization
HIF1A
Purinergic signaling in leishmaniasis infection
CASP1
Pyroptosis
CASP1
Recruitment and ATM-mediated phosphorylation of repair and signaling proteins at DNA double strand breaks
BRCA1
Regulation of gene expression by Hypoxia-inducible Factor
HIF1A
Regulation of MITF-M-dependent genes involved in DNA replication, damage repair and senescence
BRCA1
Regulation of TP53 Activity through Phosphorylation
BRCA1
Resolution of D-loop Structures through Holliday Junction Intermediates
BRCA1
Resolution of D-loop Structures through Synthesis-Dependent Strand Annealing (SDSA)
BRCA1
RUNX1 regulates genes involved in megakaryocyte differentiation and platelet function
KMT2A
RUNX1 regulates transcription of genes involved in differentiation of HSCs
KMT2A
RUNX3 Regulates Immune Response and Cell Migration
RORC
SARS-CoV-1 activates/modulates innate immune responses
CASP1
Scavenging of heme from plasma
HBB
Signaling by BRAF and RAF1 fusions
LMNA
SMAC (DIABLO) binds to IAPs
CASP7
SMAC(DIABLO)-mediated dissociation of IAP:caspase complexes
CASP7
SMAC, XIAP-regulated apoptotic response
CASP7
STAT3 nuclear events downstream of ALK signaling
HIF1A
SUMOylation of DNA damage response and repair proteins
BRCA1
SUMOylation of DNA replication proteins
TOP1
Synthesis of IP2, IP, and Ins in the cytosol
IMPA1
The AIM2 inflammasome
CASP1
The IPAF inflammasome
CASP1
The NLRP3 inflammasome
CASP1
TP53 Regulates Transcription of Caspase Activators and Caspases
CASP1
TP53 Regulates Transcription of DNA Repair Genes
BRCA1
Transcriptional Regulation by E2F6
BRCA1
Transcriptional regulation of granulopoiesis
KMT2A
Transport of bile salts and organic acids, metal ions and amine compounds
SLC47A1
SLC47A2
Ub-specific processing proteases
HIF1A
Uptake and function of anthrax toxins
LEF
XBP1(S) activates chaperone genes
LMNA
Xenobiotics
CYP3A4
104
2023
N(14)-substituted evodiamine derivatives as dual topoisomerase 1/tubulin-Inhibiting anti-gastrointestinal tumor agents.
Deng J, Long L et al. Eur J Med Chem
2022
Evodiamine-Inspired Topoisomerase-Histone Deacetylase Dual Inhibitors: Novel Orally Active Antitumor Agents for Leukemia Therapy.
Wu S, Huang Y et al. J Med Chem
2022
Design, synthesis, and biological evaluation of novel 7-substituted 10,11-methylenedioxy-camptothecin derivatives against drug-resistant small-cell lung cancer in vitro and in vivo.
Zhang G, Yin R et al. Eur J Med Chem
2022
Design, synthesis and bioactivity evaluation of novel evodiamine derivatives with excellent potency against gastric cancer.
Liang Z, Lei F et al. Eur J Med Chem
2022
Design, synthesis and bioactivity evaluation of favorable evodiamine derivative scaffold for developing cancer therapy.
Liang Z, Wang Y et al. Eur J Med Chem
2021
Synthesis of Methoxy-, Methylenedioxy-, Hydroxy-, and Halo-Substituted Benzophenanthridinone Derivatives as DNA Topoisomerase IB (TOP1) and Tyrosyl-DNA Phosphodiesterase 1 (TDP1) Inhibitors and Their Biological Activity for Drug-Resistant Cancer.
Hu DX, Tang WL et al. J Med Chem
2021
Design, synthesis and bioactivity evaluation of novel N-phenyl-substituted evodiamine derivatives as potent anti-tumor agents.
Hao X, Deng J et al. Bioorg Med Chem
2021
N-2-(phenylamino) benzamide derivatives as novel anti-glioblastoma agents: Synthesis and biological evaluation.
Li J, Hu X et al. Eur J Med Chem
2021
Quinoline anticancer agents active on DNA and DNA-interacting proteins: From classical to emerging therapeutic targets.
Lauria A, La Monica G et al. Eur J Med Chem
2020
Design, synthesis and antineoplastic activity of novel 20(S)-acylthiourea derivatives of camptothecin.
Yang,CJ.; Li,B. et al. Eur J Med Chem
2020
Development of a metabolically stable topoisomerase I poison as anticancer agent.
Kundu B,Sarkar D et al. Eur J Med Chem
2019
Discovery and Mechanistic Study of Tailor-Made Quinoline Derivatives as Topoisomerase 1 Poison with Potent Anticancer Activity.
Kundu B, Das SK et al. J Med Chem
2019
Design, Synthesis and Anticancer Evaluation of New Substituted Thiophene-Quinoline Derivatives.
Othman DIA, Selim KB et al. Bioorg Med Chem
2018
Structure-Based Drug Design and Identification of H2O-Soluble and Low Toxic Hexacyclic Camptothecin Derivatives with Improved Efficacy in Cancer and Lethal Inflammation Models in Vivo.
Pan P, Chen J et al. J Med Chem
2018
Preparation, characterisation and biological evaluation of new N-phenyl amidobenzenesulfonates and N-phenyl ureidobenzenesulfonates inducing DNA double-strand breaks. Part 3. Modulation of ring A.
Gagné-Boulet M, Bouzriba C et al. Eur J Med Chem
2018
Quest for Efficacious Next-Generation Taxoid Anticancer Agents and Their Tumor-Targeted Delivery.
Ojima I, Wang X et al. J Nat Prod
2018
Small Molecule Inhibition of MicroRNA miR-21 Rescues Chemosensitivity of Renal-Cell Carcinoma to Topotecan.
Naro Y, Ankenbruck N et al. J Med Chem
2017
Design, synthesis and potent cytotoxic activity of novel 7-(N-[(substituted-sulfonyl)piperazinyl]-methyl)-camptothecin derivatives.
Zhu GX, Cheng PL et al. Bioorg Med Chem Lett
2017
Design, synthesis, and cytotoxic activity of novel 7-substituted camptothecin derivatives incorporating piperazinyl-sulfonylamidine moieties.
Yang CJ, Song ZL et al. Bioorg Med Chem Lett
2017
Design, semisynthesis and potent cytotoxic activity of novel 10-fluorocamptothecin derivatives.
Yang CJ, Song ZL et al. Bioorg Med Chem Lett
2017
Synthesis and antitumor activity of novel substituted uracil-1'(N)-acetic acid ester derivatives of 20(S)-camptothecins.
Li DZ, Zhang QZ et al. Eur J Med Chem
2016
Design, synthesis and biological evaluation of 3-substituted indenoisoquinoline derivatives as topoisomerase I inhibitors.
Zhao Q, Xu X et al. Bioorg. Med. Chem. Lett.
2016
Discovery of indeno[1,2-b]quinoxaline derivatives as potential anticancer agents.
Tseng CH, Chen YR et al. Eur. J. Med. Chem.
2015
Discovery of potent indenoisoquinoline topoisomerase I poisons lacking the 3-nitro toxicophore.
Beck DE, Abdelmalak M et al. J. Med. Chem.
2015
Synthesis and biological evaluation of novel N-phenyl ureidobenzenesulfonate derivatives as potential anticancer agents. Part 2. Modulation of the ring B.
Gagne-Boulet M, Moussa H et al. Eur. J. Med. Chem.
2015
Scaffold Diversity Inspired by the Natural Product Evodiamine: Discovery of Highly Potent and Multitargeting Antitumor Agents.
Wang S, Fang K et al. J. Med. Chem.
2015
Synthesis of novel 10-hydroxycamptothecin derivatives utilizing topotecan hydrochloride as ortho-quinonemethide precursor.
Tan H, Wang G et al. Bioorg. Med. Chem.
2014
Synthesis, antiproliferative and anti-dengue virus evaluations of 2-aroyl-3-arylquinoline derivatives.
Tseng CH, Lin CK et al. Eur. J. Med. Chem.
2014
Novel N-substituted sophoridinol derivatives as anticancer agents.
Bi CW, Zhang CX et al. Eur. J. Med. Chem.
2014
Synthesis and biological evaluation of hydroxycinnamic acid hydrazide derivatives as inducer of caspase-3.
Wu ZR, Liu J et al. Eur. J. Med. Chem.
2014
Design and synthesis of new 7-(N-substituted-methyl)-camptothecin derivatives as potent cytotoxic agents.
Zhao XB, Goto M et al. Bioorg. Med. Chem. Lett.
2014
Design and synthesis of novel spin-labeled camptothecin derivatives as potent cytotoxic agents.
Zhao XB, Wu D et al. Bioorg. Med. Chem.
2014
Tetracycline analogues with a selective inhibitory effect on HIF-1
Bendiabdellah Y, Rahman KM et al. MedChemComm
2014
A promising camptothecin derivative: Semisynthesis, antitumor activity and intestinal permeability.
Rodriguez-Berna G, Mangas-Sanjuan V et al. Eur. J. Med. Chem.
2013
Semisynthesis, cytotoxic activity, and oral availability of new lipophilic 9-substituted camptothecin derivatives.
Rodriguez-Berna G, Cabanas MJ et al. ACS Med. Chem. Lett.
2013
Synthesis and antiproliferative evaluation of 3-phenylquinolinylchalcone derivatives against non-small cell lung cancers and breast cancers.
Tseng CH, Chen YL et al. Eur. J. Med. Chem.
2013
Evolution in medicinal chemistry of E-ring-modified Camptothecin analogs as anticancer agents.
Huang Q, Wang L et al. Eur. J. Med. Chem.
2013
Discovery of potent, selective multidrug and toxin extrusion transporter 1 (MATE1, SLC47A1) inhibitors through prescription drug profiling and computational modeling.
Wittwer MB, Zur AA et al. J. Med. Chem.
2012
Camptothecins in tumor homing via an RGD sequence mimetic.
Alloatti D, Giannini G et al. Bioorg. Med. Chem. Lett.
2012
Synthesis and preliminary bioevaluation of novel E-ring modified acetal analog of camptothecin as cytotoxic agents.
Zhu L, Zhuang C et al. Eur. J. Med. Chem.
2012
New tricks for an old natural product: discovery of highly potent evodiamine derivatives as novel antitumor agents by systemic structure-activity relationship analysis and biological evaluations.
Dong G, Wang S et al. J. Med. Chem.
2011
14-Aminocamptothecins: their synthesis, preclinical activity, and potential use for cancer treatment.
Duan JX, Cai X et al. J. Med. Chem.
2011
Synthesis and antiproliferative evaluation of 6-aryl-11-iminoindeno[1,2-c]quinoline derivatives.
Tseng CH, Tzeng CC et al. Bioorg. Med. Chem.
2011
Synthesis and antitumor activity of 10-arylcamptothecin derivatives.
Jiao Y, Liu H et al. Bioorg. Med. Chem. Lett.
2011
Synthesis and biological evaluation of novel 7-acyl homocamptothecins as Topoisomerase I inhibitors.
Liu W, Zhu L et al. Eur. J. Med. Chem.
2011
Synthesis and topoisomerase I inhibitory activity of a novel diazaindeno[2,1-b]phenanthrene analogue of Lamellarin D.
Cananzi S, Merlini L et al. Bioorg. Med. Chem.
2010
Cytotoxicity and topo I targeting activity of substituted 10--nitrogenous heterocyclic aromatic group derivatives of SN-38.
Li QY, Deng XQ et al. Eur. J. Med. Chem.
2010
Molecular docking study on anticancer activity of plant-derived natural products
Phosrithong N, Ungwitayatorn J Med Chem Res
2010
Phosphate ester derivatives of homocamptothecin: synthesis, solution stabilities and antitumor activities.
Miao Z, Zhang J et al. Bioorg. Med. Chem.
2010
Synthesis and evaluation of 9-benzylideneamino derivatives of homocamptothecin as potent inhibitors of DNA topoisomerase I.
Guo W, Miao Z et al. Eur. J. Med. Chem.
2009
Synthesis and antitumor activity of novel 20s-camptothecin analogues.
Li Q, Lv H et al. Bioorg. Med. Chem. Lett.
2009
Discovery of a novel series of quinolone and naphthyridine derivatives as potential topoisomerase I inhibitors by scaffold modification.
You QD, Li ZY et al. J. Med. Chem.
2009
7-Cycloalkylcamptothecin derivatives: Preparation and biological evaluation.
Li M, Jin W et al. Bioorg. Med. Chem. Lett.
2009
Synthesis and antiproliferative evaluation of 6-arylindeno[1,2-c]quinoline derivatives.
Tseng CH, Chen YL et al. Bioorg. Med. Chem.
2009
Cytotoxicity and TOP1-targeting activity of 8- and 9-amino derivatives of 5-butyl- and 5-(2-N,N-dimethylamino)ethyl-5H-dibenzo[c,h][1,6]naphthyridin-6-ones.
Sharma L, Tsai YC et al. Eur. J. Med. Chem.
2009
Semisynthesis, biological activity, and molecular modeling studies of C-ring-modified camptothecins.
Samori C, Guerrini A et al. J. Med. Chem.
2008
New homocamptothecins: synthesis, antitumor activity, and molecular modeling.
Miao Z, Sheng C et al. Bioorg. Med. Chem.
2008
Novel hexacyclic camptothecin derivatives. Part 1: synthesis and cytotoxicity of camptothecins with an A-ring fused 1,3-oxazine ring.
Wang S, Li Y et al. Bioorg. Med. Chem. Lett.
2008
E-ring-modified 7-oxyiminomethyl camptothecins: Synthesis and preliminary in vitro and in vivo biological evaluation.
Giannini G, Marzi M et al. Bioorg. Med. Chem. Lett.
2008
Semi-synthesis and biological activity of gamma-lactones analogs of camptothecin.
Li M, Tang W et al. Bioorg. Med. Chem. Lett.
2008
Synthesis and cytotoxic activity of a new series of topoisomerase I inhibitors.
Dallavalle S, Gattinoni S et al. Bioorg. Med. Chem. Lett.
2008
Synthesis and cytotoxic activity of new 9-substituted camptothecins.
Dallavalle S, Rocchetta DG et al. Bioorg. Med. Chem. Lett.
2008
Synthesis of N-substituted 5-[2-(N-alkylamino)ethyl]dibenzo[c,h][1,6]naphthyridines as novel topoisomerase I-targeting antitumor agents.
Feng W, Satyanarayana M et al. Bioorg. Med. Chem.
2008
11-Substituted 2,3-dimethoxy-8,9-methylenedioxybenzo[i]phenanthridine derivatives as novel topoisomerase I-targeting agents.
Feng W, Satyanarayana M et al. Bioorg. Med. Chem.
2007
Synthesis and topoisomerase poisoning activity of A-ring and E-ring substituted luotonin A derivatives.
Nacro K, Zha CC et al. Bioorg. Med. Chem.
2007
Structures, biogenesis, and biological activities of pyrano[4,3-c]isochromen-4-one derivatives from the Fungus Phellinus igniarius.
Wang Y, Shang XY et al. J. Nat. Prod.
2006
Mono-, Bi-, and triphenanthrenes from the tubers of Cremastra appendiculata.
Xue Z, Li S et al. J. Nat. Prod.
2006
A nitrogen-containing 3-alkyl-1,4-benzoquinone and a gomphilactone derivative from Embelia ribes.
Lin P, Li S et al. J. Nat. Prod.
2006
Synthesis and cytotoxic activity of polyamine analogues of camptothecin.
Dallavalle S, Giannini G et al. J. Med. Chem.
2005
Synthesis and antitumor activity of 7-ethyl-9-alkyl derivatives of camptothecin.
Gao H, Zhang X et al. Bioorg. Med. Chem. Lett.
2005
5-(2-aminoethyl)dibenzo[c,h][1,6]naphthyridin-6-ones: variation of n-alkyl substituents modulates sensitivity to efflux transporters associated with multidrug resistance.
Ruchelman AL, Houghton PJ et al. J. Med. Chem.
2004
Phelligridins C-F: cytotoxic pyrano[4,3-c][2]benzopyran-1,6-dione and furo[3,2-c]pyran-4-one derivatives from the fungus Phellinus igniarius.
Mo S, Wang S et al. J. Nat. Prod.
2004
Dimethoxybenzo[i]phenanthridine-12-carboxylic acid derivatives and 6H-dibenzo[c,h][2,6]naphthyridin-5-ones with potent topoisomerase I-targeting activity and cytotoxicity.
Ruchelman AL, Zhu S et al. Bioorg. Med. Chem. Lett.
2004
Design, synthesis, and evaluation of 3,4-dihydro-2H-[1,4]diazepino[6,7,1-hi]indol-1-ones as inhibitors of poly(ADP-ribose) polymerase.
Tikhe JG, Webber SE et al. J. Med. Chem.
2004
Laurenditerpenol, a new diterpene from the tropical marine alga Laurenciaintricata that potently inhibits HIF-1 mediated hypoxic signaling in breast tumor cells.
Mohammed KA, Hossain CF et al. J. Nat. Prod.
2004
Imatinib mesylate is a potent inhibitor of the ABCG2 (BCRP) transporter and reverses resistance to topotecan and SN-38 in vitro.
Houghton PJ, Germain GS et al. Cancer Res.
2003
Synthesis and pharmacological evaluation of novel non-lactone analogues of camptothecin.
Hautefaye P, Cimetiere B et al. Bioorg. Med. Chem. Lett.
2003
Regioselective synthesis and cytotoxicities of camptothecin derivatives modified at the 7-, 10- and 20-positions.
Pan XD, Han R et al. Bioorg. Med. Chem. Lett.
2003
Nitro and amino substitution in the D-ring of 5-(2-dimethylaminoethyl)- 2,3-methylenedioxy-5H-dibenzo[c,h][1,6]naphthyridin-6-ones: effect on topoisomerase-I targeting activity and cytotoxicity.
Singh SK, Ruchelman AL et al. J. Med. Chem.
2002
Novel angular benzophenazines: dual topoisomerase I and topoisomerase II inhibitors as potential anticancer agents.
Vicker N, Burgess L et al. J. Med. Chem.
2002
Novel tricyclic poly(ADP-ribose) polymerase-1 inhibitors with potent anticancer chemopotentiating activity: design, synthesis, and X-ray cocrystal structure.
Canan Koch SS, Thoresen LH et al. J. Med. Chem.
2002
Diaza- and triazachrysenes: potent topoisomerase-targeting agents with exceptional antitumor activity against the human tumor xenograft, MDA-MB-435.
Ruchelman AL, Singh SK et al. Bioorg. Med. Chem. Lett.
2001
Novel cytotoxic 7-iminomethyl and 7-aminomethyl derivatives of camptothecin.
Dallavalle S, Ferrari A et al. Bioorg. Med. Chem. Lett.
2000
Novel C-ring analogues of 20(S)-camptothecin. Part 3: synthesis and their in vitro cytotoxicity of A-, B- and C-ring analogues.
Subrahmanyam D, Sarma VM et al. Bioorg. Med. Chem. Lett.
2000
Resistance-modifying agents. 9. Synthesis and biological properties of benzimidazole inhibitors of the DNA repair enzyme poly(ADP-ribose) polymerase.
White AW, Almassy R et al. J. Med. Chem.
2000
Novel 7-substituted camptothecins with potent antitumor activity.
Dallavalle S, Delsoldato T et al. J. Med. Chem.
1999
Novel C-ring analogues of 20(S)-camptothecin-part-2: synthesis and in vitro cytotoxicity of 5-C-substituted 20(S)-camptothecin analogues.
Subrahmanyam D, Venkateswarlu A et al. Bioorg. Med. Chem. Lett.
1998
Homocamptothecins: synthesis and antitumor activity of novel E-ring-modified camptothecin analogues.
Lavergne O, Lesueur-Ginot L et al. J. Med. Chem.
1997
BN 80245: An E-ring modified camptothecin with potent antiproliferative and topoisomerase I inhibitory activities
Lavergne O, Lesueur-Ginot L et al. Bioorg. Med. Chem. Lett.
1991
Synthesis of water-soluble (aminoalkyl)camptothecin analogues: inhibition of topoisomerase I and antitumor activity.
Kingsbury WD, Boehm JC et al. J. Med. Chem.
DrugMatrix in vitro pharmacology data (dataset)
PUBCHEM_BIOASSAY: qHTS Assay for Inhibitors and Activators of N370S glucocerebrosidase as a Potential Chaperone Treatment of Gaucher Disease: Purified N370S Glucocerebrosidase. (Class of assay: confirmatory) [Related pubchem assays: 2101 ]
AID 2597
80
37
A Collection of Useful Nuisance Compounds (CONS) for Interrogation of Bioassay Integrity
AdooQ Bioactive Compound Library
ChEMBL Approved Drugs
ChEMBL Drugs
CZ-OPENSCREEN Bioactive Library
Drug Repurposing Hub
DrugBank
DrugBank Approved Drugs
DrugCentral
DrugCentral Approved Drugs
Enamine BioReference Compounds
Guide to Pharmacology
Informer Set
LINCS compound set
LSP-MoA library (Laboratory of Systems Pharmacology)
Mcule NIBR MoA Box Subset
MedChem Express Bioactive Compound Library
Natural product-based probes and drugs
NCATS Inxight Approved Drugs
NIH Approved Oncology Drugs
NIH Clinical Collections (NCC)
Novartis Chemogenetic Library (NIBR MoA Box)
NPC Screening Collection
Nuisance compounds in cellular assays
Pandemic Response Box
Prestwick Chemical Library
Probe Miner (suitable probes)
ReFrame library
Selleckchem Bioactive Compound Library
TargetMol Bioactive Compound Library
The Spectrum Collection
Tocris Bioactive Compound Library
Withdrawn 2.0
ZINC Tool Compounds
12
AdooQ Bioactive Compound Library
Enamine BioReference Compounds
Mcule NIBR MoA Box Subset
MedChem Express Bioactive Compound Library
MolPort
Prestwick Chemical Library
Selleckchem Bioactive Compound Library
TargetMol Bioactive Compound Library
The Spectrum Collection
Tocris Bioactive Compound Library
87
GtoPdb 7101
ZINC ZINC000001611274
ChEBI 63632
ClinicalTrials.gov TOPOTECAN HYDRO... HYCAMPTIN SKF-104864 HYCAMTIN NOGITECAN HYDRO... NSC-609699 TOPOTECAN TOPOTECAN HOSPIRA POTACTASOL
COCONUT CNP0074922.1
DrugBank DB01030
DrugCentral 2707
IBM NIH C491BF277F0255B...
KEGG C11158
LINCS LSM-5662
MolPort MolPort-003-850-580 Molport-003-850-580 MolPort-003-666-754 Molport-003-666-754 MolPort-003-850-581 Molport-003-850-581
NIH NCC SAM001246651
PDB TTC
PharmGKB PA451729
PubChem DOTF 12014351
Selleck Topotecan-Hydro...
(calculated by RDKit )
Molecular Weight
421.16
Hydrogen Bond Acceptors
8
Hydrogen Bond Donors
2
Rotatable Bonds
3
Ring Count
5
Aromatic Ring Count
3
cLogP
1.85
TPSA
104.89
Fraction CSP3
0.35
Chiral centers
1.0
Largest ring
6.0
QED
0.49
1
Intercalation
Nuisance compounds
(extracted from source data)
Target
TOP1
Autophagy
Topo I (DU-145 Luc cells)
Topo I (MCF-7 Luc cells)
TOP1, TOP1MT
Topoisomerase
Apoptosis related,Autophagy,Topoisomerase
Compound status
FDA
Pathway
Cell Cycle/DNA Damage
DNA Damage/DNA Repair
Apoptosis
Primary Target
DNA Topoisomerases
MOA
Inhibitor
DNA Topoisomerase I Inhibitors
Topoisomerase inhibitor
Member status
member
Indication
small cell lung cancer, cervical cancer
ATC
L01XX17
Biosynthetic Origin
Alkaloid
Therapeutic Indication
Anticancer
Therapeutic Class
Anticancer Agents
Nuisance MOA
Intercalation
Targets
Hypoxia-inducible factor 1 alpha
DNA topoisomerase I