(-)-S-Trityl-D-cysteine
Need Assistance?
  • US & Canada:
    +
  • UK: +

(-)-S-Trityl-D-cysteine

* Please kindly note that our products are not to be used for therapeutic purposes and cannot be sold to patients.

(-)-S-Trityl-D-cysteine is an influential anti-angiogenesis agent, garnering significant attention in the research of cancer therapy. Its mechanism primarily focuses on the suppression of tumor growth by effectively targeting the VEGF receptor.

Category
L-Amino Acids
Catalog number
BAT-004202
CAS number
25840-82-8
Molecular Formula
C22H21NO2S
Molecular Weight
363.47
(-)-S-Trityl-D-cysteine
IUPAC Name
(2S)-2-amino-3-tritylsulfanylpropanoic acid
Synonyms
D-Cys(Trt)-OH; (S)-2-Amino-3-(Tritylthio)Propanoic Acid
Appearance
White Powder
Purity
98%
Density
1.232±0.06 g/cm3 (Predicted)
Melting Point
190°C
Boiling Point
524.7±50.0°C (Predicted)
Storage
Store at 2-8°C under inert atmosphere
Solubility
Soluble in Water (Sparingly)
InChI
InChI=1S/C22H21NO2S/c23-20(21(24)25)16-26-22(17-10-4-1-5-11-17,18-12-6-2-7-13-18)19-14-8-3-9-15-19/h1-15,20H,16,23H2,(H,24,25)/t20-/m1/s1
InChI Key
DLMYFMLKORXJPO-HXUWFJFHSA-N
Canonical SMILES
C1=CC=C(C=C1)C(C2=CC=CC=C2)(C3=CC=CC=C3)SCC(C(=O)O)N
1. S-trityl-L-cysteine is a reversible, tight binding inhibitor of the human kinesin Eg5 that specifically blocks mitotic progression
Dimitrios A Skoufias, Salvatore DeBonis, Yasmina Saoudi, Luc Lebeau, Isabelle Crevel, Robert Cross, Richard H Wade, David Hackney, Frank Kozielski J Biol Chem. 2006 Jun 30;281(26):17559-69. doi: 10.1074/jbc.M511735200. Epub 2006 Feb 28.
Human Eg5, responsible for the formation of the bipolar mitotic spindle, has been identified recently as one of the targets of S-trityl-L-cysteine, a potent tumor growth inhibitor in the NCI 60 tumor cell line screen. Here we show that in cell-based assays S-trityl-L-cysteine does not prevent cell cycle progression at the S or G(2) phases but inhibits both separation of the duplicated centrosomes and bipolar spindle formation, thereby blocking cells specifically in the M phase of the cell cycle with monoastral spindles. Following removal of S-trityl-L-cysteine, mitotically arrested cells exit mitosis normally. In vitro, S-trityl-L-cysteine targets the catalytic domain of Eg5 and inhibits Eg5 basal and microtubule-activated ATPase activity as well as mant-ADP release. S-trityl-L-cysteine is a tight binding inhibitor (estimation of K(i,app) <150 nm at 300 mm NaCl and 600 nm at 25 mm KCl). S-trityl-L-cysteine binds more tightly than monastrol because it has both an approximately 8-fold faster association rate and approximately 4-fold slower release rate (6.1 microM(-1) s(-1) and 3.6 s(-1) for S-trityl-L-cysteine versus 0.78 microM(-1) s(-1) and 15 s(-1) for monastrol). S-trityl-L-cysteine inhibits Eg5-driven microtubule sliding velocity in a reversible fashion with an IC(50) of 500 nm. The S and D-enantiomers of S-tritylcysteine are nearly equally potent, indicating that there is no significant stereospecificity. Among nine different human kinesins tested, S-trityl-L-cysteine is specific for Eg5. The results presented here together with the proven effect on human tumor cell line growth make S-trityl-L-cysteine a very attractive starting point for the development of more potent mitotic inhibitors.
2. S-trityl-L-cysteine, a novel Eg5 inhibitor, is a potent chemotherapeutic strategy in neuroblastoma
Wei Wu, Shao Jingbo, Weijue Xu, Jiangbin Liu, Yiming Huang, Qingfeng Sheng, Zhibao Lv Oncol Lett. 2018 Jul;16(1):1023-1030. doi: 10.3892/ol.2018.8755. Epub 2018 May 21.
Eg5 is a member of the kinesin-5 family. It is involved in the formation of the bipolar spindle and serves a crucial role in mitosis; meaning that mitotic activation may serve as a chemotherapeutic strategy. However, the anticancer activity of Eg5 inhibitors in neuroblastoma remains uncharacterized. In the present study, the expression of Eg5 was examined in clinical tissue samples and neuroblastoma cell lines, SK-N-SH, SH-SY5Y and SK-N-BE2. Additionally, the antitumor activity of the Eg5 inhibitor, S-trityl-L-cysteine (STLC), was confirmed in vitro. STLC could mediate cell apoptosis, as well as cell cycle arrest, in a dose-dependent manner, which may contribute toward its antitumor activity. STLC-mediated apoptosis and cell cycle arrest were triggered by activation of the mitogen-activated protein kinase and nuclear factor kB signaling pathways. These results suggested that STLC may have potential in the in vivo treatment of neuroblastoma.
3. Antiproliferative S-Trityl-l-Cysteine -Derived Compounds as SIRT2 Inhibitors: Repurposing and Solubility Enhancement
Mohamed O Radwan, et al. Molecules. 2019 Sep 10;24(18):3295. doi: 10.3390/molecules24183295.
S-trityl-l-cysteine (STLC) is a well-recognized lead compound known for its anticancer activity owing to its potent inhibitory effect on human mitotic kinesin Eg5. STLC contains two free terminal amino and carboxyl groups that play pivotal roles in binding to the Eg5 pocket. On the other hand, such a zwitterion structure complicates the clinical development of STLC because of the solubility issues. Masking either of these radicals reduces or abolishes STLC activity against Eg5. We recently identified and characterized a new class of nicotinamide adenine dinucleotide-dependent deacetylase isoform 2 of sirtuin protein (SIRT2) inhibitors that can be utilized as cytotoxic agents based on an S-trityl-l-histidine scaffold. Herein, we propose new STLC-derived compounds that possess pronounced SIRT2 inhibition effects. These derivatives contain modified amino and carboxyl groups, which conferred STLC with SIRT2 bioactivity, representing an explicit repurposing approach. Compounds STC4 and STC11 exhibited half maximal inhibitory concentration values of 10.8 ± 1.9 and 9.5 ± 1.2 μM, respectively, against SIRT2. Additionally, introduction of the derivatizations in this study addressed the solubility limitations of free STLC, presumably due to interruption of the zwitterion structure. Therefore, we could obtain drug-like STLC derivatives that work by a new mechanism of action. The new derivatives were designed, synthesized, and their structure was confirmed using different spectroscopic approaches. In vitro and cellular bioassays with various cancer cell lines and in silico molecular docking and solubility calculations of the synthesized compounds demonstrated that they warrant attention for further refinement of their bioactivity.
Online Inquiry
Verification code
Inquiry Basket