Size | Price | |
---|---|---|
500mg | ||
1g | ||
Other Sizes |
Purity: ≥98%
JTE 013 (JTE-013) is a novel potent and selective S1P2 (sphingosine-1-phosphate 2) antagonist with IC50 of 17.6 nM. It binds to the human and rat receptors with IC50 values of 17 and 22 nM, respectively, and with IC50 values >10 µM for human S1P1 and S1P3. It reverses the inhibitory effects of S1P2 signaling on cell migration of vascular ECs and smooth muscle cells. It also regulates endothelial tight junctions and barrier function in vitro. Blockage of S1P2 signaling by JTE-013 significantly enhances the effects of S1P on the increase of TEER, an in vitro measurement of endothelial integrity, as well as the formation of TJs in senescent ECs.
Targets |
S1P2 (Sphingosine-1-Phosphate 2; EDG-5); human S1P2 (IC50 = 17 nM); rat S1P2 (IC50 = 22 nM)
|
||
---|---|---|---|
ln Vitro |
|
||
ln Vivo |
|
||
Enzyme Assay |
Fluorescent Imaging Plate Reader Assay.[1]
The calcium flux assay on a FLIPRTETRA instrument [fluorescent imaging plate reader (FLIPR) assay] was performed by a CRO company to profile test compounds for dose-dependent agonist and antagonist activities on S1P1–5. Briefly, the agonist assay was conducted on a FLIPRTETRA instrument, in which the test compounds, vehicle controls, and the reference agonist S1P were added to the assay plate after a fluorescence baseline was established. A duration of 180 seconds was used to assess each compound’s ability to activate each S1PR. Upon completion of the agonist assay, the assay plate was removed from the FLIPRTETRA instrument and incubated at 25°C for 7 minutes. After that, the assay plate was placed back in the FLIPRTETRA instrument and the antagonist assay was initiated. Using EC80 potency values determined during the agonist assay, all preincubated sample compound wells were challenged with EC80 concentration of the reference agonist S1P after establishment of a fluorescence baseline. Another duration of 180 seconds was used to assess each compound’s ability to inhibit each S1PR. All assay plate data were subjected to appropriate baseline corrections. After baseline corrections were applied, maximum fluorescence values were exported and data were processed to calculate the percentage of activation (relative to Emax reference agonist S1P and vehicle control values) and the percentage of inhibition (relative to EC80 and vehicle control values). All dose-response curves were generated using GraphPad Prism software |
||
Cell Assay |
Cell Line: SK-N-AS cells
Concentration: 50, 100, 150, 200 μM Incubation Time: 1-3 days Result: Reduced cell viability. Migration Assay. [1] The migration assay was performed in a 96-well chemotaxis microchamber), as described previously (Li et al., 2009b). Briefly, a polycarbonate filter (8-µm pore size) was coated with 50 µg/ml fibronectin. S1P was diluted and added into the lower chamber at 85 µl per well. GB cells were serum starved for 2 hours prior to trypsinization and were pretreated with or without JTE-013 and AB1 for 10 minutes. They were then placed in the upper compartment at 5 × 104 cells per well in 0.39 ml medium and allowed to migrate 5 hours at 37°C. The filter was then fixed overnight at 4°C and the nonmigrated cells were removed with a cotton swab. Attached cells were stained with 0.1% crystal violet and eluted with 10% acetic acid in 96-well plates. The absorbance was measured at 595 nm. Methylthiazolyldiphenyl-Tetrazolium Bromide Assay.[1] The viability of SK-N-AS cells treated with JTE-013 or AB1 was determined by the methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay, as previously described (Li et al., 2013). Briefly, SK-N-AS cells were seeded in 96-well plates and treated with different concentrations of JTE-013 or AB1 for different times, followed by incubation of MTT at 37°C for 2 hours. The insoluble formazan formed in viable cells were dissolved by dimethylsulfoxide and the absorbance was measured at 595 nm by using a Bio-Rad Microplate Reader. Results are presented as the percentage of cell viability relative to the nondrug-treated controls. |
||
Animal Protocol |
|
||
References | |||
Additional Infomation |
The bioactive lipid sphingosine-1-phosphate (S1P) and its receptors (S1P1-5) play critical roles in many pathologic processes, including cancer. The S1P axis has become a bona fide therapeutic target in cancer. JTE-013 [N-(2,6-dichloro-4-pyridinyl)-2-[1,3-dimethyl-4-(1-methylethyl)-1H-pyrazolo[3,4-b]pyridin-6-yl]-hydrazinecarboxamide], a known S1P2 antagonist, suffers from instability in vivo. Structurally modified, more potent, and stable S1P2 inhibitors would be desirable pharmacological tools. One of the JTE-013 derivatives, AB1 [N-(1H-4-isopropyl-1-allyl-3-methylpyrazolo[3,4-b]pyridine-6-yl)-amino-N'-(2,6-dichloropyridine-4-yl) urea], exhibited improved S1P2 antagonism compared with JTE-013. Intravenous pharmacokinetics indicated enhanced stability or slower clearance of AB1 in vivo. Migration assays in glioblastoma showed that AB1 was slightly more effective than JTE-013 in blocking S1P2-mediated inhibition of cell migration. Functional studies in the neuroblastoma (NB) cell line SK-N-AS showed that AB1 displayed potency at least equivalent to JTE-013 in affecting signaling molecules downstream of S1P2. Similarly, AB1 inhibition of the growth of SK-N-AS tumor xenografts was improved compared with JTE-013. Cell viability assays excluded that this enhanced AB1 effect is caused by inhibition of cancer cell survival. Both JTE-013 and AB1 trended to inhibit (C-C motif) ligand 2 expression and were able to significantly inhibit subsequent tumor-associated macrophage infiltration in NB xenografts. Interestingly, AB1 was more effective than JTE-013 in inhibiting the expression of the profibrotic mediator connective tissue growth factor. The terminal deoxynucleotidyl transferase-mediated digoxigenin-deoxyuridine nick-end labeling assay and cleaved caspase-3 detection further demonstrated that apoptosis was increased in AB1-treated NB xenografts compared with JTE-013. Overall, the modification of JTE-013 to produce the AB1 compound improved potency, intravenous pharmacokinetics, cellular activity, and antitumor activity in NB and may have enhanced clinical and experimental applicability. [1]
In summary, here we report the novel modification of the S1P2 antagonist JTE-013 to produce AB1. AB1 has moderately improved potency and intravenous pharmacokinetics that demonstrate better stability. In the context of NB, it also appears to have better cellular activity and antitumor activity. On the basis of these findings, we conclude that AB1 may have enhanced clinical and experimental applicability, overcoming some of the shortcomings of JTE-013. [1] Sphingosine-1-phosphate (S1P) regulates various molecular and cellular events in cultured endothelial cells, such as cytoskeletal restructuring, cell-extracellular matrix interactions, and intercellular junction interactions. We utilized the venular leakage model of the cremaster muscle vascular bed in Sprague-Dawley rats to investigate the role of S1P signaling in regulation of microvascular permeability. S1P signaling is mediated by the S1P family of G protein-coupled receptors (S1P(1-5) receptors). S1P(1) and S1P(2) receptors, which transduce stimulatory and inhibitory signaling, respectively, are expressed in the endothelium of the cremaster muscle vasculature. S1P administration alone via the carotid artery was unable to protect against histamine-induced venular leakage of the cremaster muscle vascular bed in Sprague-Dawley rats. However, activation of S1P(1)-mediated signaling by SEW2871 and FTY720, two agonists of S1P(1), significantly inhibited histamine-induced microvascular leakage. Treatment with VPC 23019 to antagonize S1P(1)-regulated signaling greatly potentiated histamine-induced venular leakage. After inhibition of S1P(2) signaling by JTE-013, a specific antagonist of S1P(2), S1P was able to protect microvascular permeability in vivo. Moreover, endothelial tight junctions and barrier function were regulated by S1P(1)- and S1P(2)-mediated signaling in a concerted manner in cultured endothelial cells. These data suggest that the balance between S1P(1) and S1P(2) signaling regulates the homeostasis of microvascular permeability in the peripheral circulation and, thus, may affect total peripheral vascular resistance.[2] |
Molecular Formula |
C17H19CL2N7O
|
|
---|---|---|
Molecular Weight |
408.29
|
|
Exact Mass |
407.102
|
|
Elemental Analysis |
C, 50.01; H, 4.69; Cl, 17.37; N, 24.01; O, 3.92
|
|
CAS # |
547756-93-4
|
|
Related CAS # |
547756-93-4
|
|
PubChem CID |
25168534
|
|
Appearance |
Typically exists as
White to off-white solid at room temperature
|
|
Density |
1.5±0.1 g/cm3
|
|
Index of Refraction |
1.697
|
|
LogP |
4.42
|
|
Hydrogen Bond Donor Count |
3
|
|
Hydrogen Bond Acceptor Count |
5
|
|
Rotatable Bond Count |
5
|
|
Heavy Atom Count |
27
|
|
Complexity |
504
|
|
Defined Atom Stereocenter Count |
0
|
|
SMILES |
CN1C2=NC(NNC(NC3C(Cl)=CN=CC=3Cl)=O)=CC(CCC)=C2C(C)=N1
|
|
InChi Key |
GDFXUTXWCNQTEF-UHFFFAOYSA-N
|
|
InChi Code |
InChI=1S/C17H19Cl2N7O/c1-4-5-10-6-13(21-16-14(10)9(2)25-26(16)3)23-24-17(27)22-15-11(18)7-20-8-12(15)19/h6-8H,4-5H2,1-3H3,(H,21,23)(H2,20,22,24,27)
|
|
Chemical Name |
1-(3,5-dichloropyridin-4-yl)-3-[(1,3-dimethyl-4-propylpyrazolo[3,4-b]pyridin-6-yl)amino]urea
|
|
Synonyms |
|
|
HS Tariff Code |
2934.99.9001
|
|
Storage |
Powder -20°C 3 years 4°C 2 years In solvent -80°C 6 months -20°C 1 month |
|
Shipping Condition |
Room temperature (This product is stable at ambient temperature for a few days during ordinary shipping and time spent in Customs)
|
Solubility (In Vitro) |
|
|||
---|---|---|---|---|
Solubility (In Vivo) |
|
Preparing Stock Solutions | 1 mg | 5 mg | 10 mg | |
1 mM | 2.4492 mL | 12.2462 mL | 24.4924 mL | |
5 mM | 0.4898 mL | 2.4492 mL | 4.8985 mL | |
10 mM | 0.2449 mL | 1.2246 mL | 2.4492 mL |
*Note: Please select an appropriate solvent for the preparation of stock solution based on your experiment needs. For most products, DMSO can be used for preparing stock solutions (e.g. 5 mM, 10 mM, or 20 mM concentration); some products with high aqueous solubility may be dissolved in water directly. Solubility information is available at the above Solubility Data section. Once the stock solution is prepared, aliquot it to routine usage volumes and store at -20°C or -80°C. Avoid repeated freeze and thaw cycles.
Calculation results
Working concentration: mg/mL;
Method for preparing DMSO stock solution: mg drug pre-dissolved in μL DMSO (stock solution concentration mg/mL). Please contact us first if the concentration exceeds the DMSO solubility of the batch of drug.
Method for preparing in vivo formulation::Take μL DMSO stock solution, next add μL PEG300, mix and clarify, next addμL Tween 80, mix and clarify, next add μL ddH2O,mix and clarify.
(1) Please be sure that the solution is clear before the addition of next solvent. Dissolution methods like vortex, ultrasound or warming and heat may be used to aid dissolving.
(2) Be sure to add the solvent(s) in order.
![]() |
![]() |