Home>>Signaling Pathways>> Cell Cycle/Checkpoint>> Kinesin>>PF-2771

PF-2771 Sale

目录号 : GC34297

PF-2771是一种选择性的着丝粒蛋白E(CENP-E)抑制剂,IC50值为16.1nM,具有抗肿瘤活性。

PF-2771 Chemical Structure

Cas No.:2070009-55-9

规格 价格 库存 购买数量
10mM (in 1mL DMSO)
¥2,745.00
现货
5mg
¥2,250.00
现货
10mg
¥3,600.00
现货
50mg
¥10,800.00
现货
100mg
¥13,514.00
现货

电话:400-920-5774 Email: sales@glpbio.cn

Customer Reviews

Based on customer reviews.

Sample solution is provided at 25 µL, 10mM.

产品文档

Quality Control & SDS

View current batch:

实验参考方法

Kinase experiment:

Microtubule-activated CENP-E kinesin ATPase activity is measured spectrophotometrically by coupling the hydrolysis of ATP to ADP to NADH oxidation (decrease in 340 nm absorbance) through the activities of pyruvate kinase (PK) and lactate dehydrogenase (LDH). Reactions contained 2 mM phosphoenolpyruvate, 0.28 mM NADH, 5 mM MgCl2, 1 mM DTT, 15 μM taxol, 0.7 μM MT (preformed porcine microtubules), 50 μM ATP, 10 U/mL PK, and 10 U/mL LDH in 15 mM PIPES buffer (pH 7.0). Reactions are initiated with a 20 nM CENP-E addition (Human: 1-342; WT) at 30°C. The IC50 values are determined by a nonlinear, least squares fit of the data to the four-parameter dose-response curve equation. PF-2771 kinesin biochemical selectivity toward other kinesins is tested in triplicate at both 1 and 10 μM PF-2771 in a variant of the CENP-E enzymatic assays that have the following enzyme concentrations: 200 nM chromokinesin motor domain, 200 nM Eg5 motor domain, 226 nM MCAK motor domain. PF-2771 biochemical mechanism is determined by measuring CENP-E enzymatic activity as a function of ATP and PF-2771 concentrations (0, 2.5, 5, 10, 20, 30, 45 nM PF-2771; 1,000, 500, 250, 125, 62.5, 31.2, 15.6, 7.81, 3.90, 1.95, 0.977 μM ATP)[1].

Cell experiment:

PF-2771 is added to cells seeded in 96-well plates. The number of cells seeded (1,000-3,000) depended on growth characteristics of each cell type and normalized proliferation rates. Ten different concentrations of compound (PF-2771) used are based on a half-log increment between 1 nM and either 1 or 25 μM. Cells are incubated at 37°C for 7 days before assessing viability with the CellTiter-Glo reagent. Untreated control cells are 80% to 90% confluent after 7 days of culture. Data are fitted into a sigmoidal curve-fitting program to calculate IC50 values[1].

Animal experiment:

HCC1806 tumor cells (3 × 106) are implanted in the mammary fat pad of CB17/lcr.Cg-PrkdcscidLystbg female mice. PF-2771 is administered intraperitoneally (i.p.) to groups of 12 mice at 3, 10, and 30 mg/kg every day for 14 days and at 100 mg/kg every day for 4 days followed by 3 days off and then another 4-day cycle. Tumor volumes are recorded twice weekly by calipers with the final measurement taken 3 days after the last dose. Tumor growth inhibition (TGI) is calculated using the formula 100 × (1 − ΔT/ΔC), where ΔT (treated) and ΔC (control) are the mean tumor volume changes between 1 day after the last dose and the first-day treatment. Time-to-progression endpoint and associated tumor growth delay determinations are calculated using median days to reach to two doublings of initial tumor size. Statistical comparisons are made using one-way ANOVA with Dunnett posttests. Other tumor models had similar methodology except where noted. PDX-AA1077 is a patient-derived xenograft model developed from tumor tissue from a triple-negative patient engrafted to the mammary glands of NSG female mice to create passage 1 tumor-bearing mice. Surgically resected tumor tissue is cut into 2- to 4-mm3 fragments and subcutaneously implanted into the flank of SCID-bg mice. Mice with palpable tumors are randomized before dosing. An HCC1599 tumor cell line xenograft model is established from tumor fragments of established tumors and reimplanted into flank of female SCID mice. When average tumor size reached 250 mm3, animals are randomized into five groups of 10 mice. Animals are treated with vehicle (daily dosing), 10 mg/kg docetaxel (once per week dosing), 25 mg/kg docetaxel (once per week dosing), 20 mg/kg paclitaxel (twice per week dosing), or 100 mg/kg of PF-2771 (every day, i.p.)[1].

References:

[1]. Kung PP, et al. Chemogenetic evaluation of the mitotic kinesin CENP-E reveals a critical role in triple-negative breast cancer. Mol Cancer Ther. 2014 Aug;13(8):2104-15.

产品描述

PF-2771 is a potent and selective centromere protein E (CENP-E) inhibitor, inhibiting CENP-E motor activity with an IC50 of 16.1 nM; PF-2771 is used as an anticancer agent.

PF-2771 is a potent and selective CENP-E inhibitor, inhibiting CENP-E motor activity with an IC50 of 16.1 nM. PF-2771 shows no inhibitory effect on the ATPase activities of highly related kinesins (0% inhibition of Eg5/KSP, chromokinesin, and MCAK at both 1 or 10 μM PF-2771). PF-2771 exhibits inactive activity against 74 protein kinases (all 5 μM). PF-2771 (100 nM) reusults in a chromosomal congression defect in MDA-MB-468 cells[1].

PF-2771 (100 mg/kg, every day i.p.) potently inhibits CENP-E motor function, and causes tumor regression in SCID mice bearing AA1077 mammary tumors[1].

[1]. Kung PP, et al. Chemogenetic evaluation of the mitotic kinesin CENP-E reveals a critical role in triple-negative breast cancer. Mol Cancer Ther. 2014 Aug;13(8):2104-15.

Chemical Properties

Cas No. 2070009-55-9 SDF
Canonical SMILES ClC1=CC(C(N[C@@H](CC2=CC=C(C3=CN(C)C(C(C)=O)=N3)C=C2)CNC(CN(C)C)=O)=O)=CC=C1OC(C)C
分子式 C29H36ClN5O4 分子量 554.08
溶解度 DMSO : ≥ 32 mg/mL (57.75 mM) 储存条件 Store at -20°C
General tips 请根据产品在不同溶剂中的溶解度选择合适的溶剂配制储备液;一旦配成溶液,请分装保存,避免反复冻融造成的产品失效。
储备液的保存方式和期限:-80°C 储存时,请在 6 个月内使用,-20°C 储存时,请在 1 个月内使用。
为了提高溶解度,请将管子加热至37℃,然后在超声波浴中震荡一段时间。
Shipping Condition 评估样品解决方案:配备蓝冰进行发货。所有其他可用尺寸:配备RT,或根据请求配备蓝冰。

溶解性数据

制备储备液
1 mg 5 mg 10 mg
1 mM 1.8048 mL 9.024 mL 18.0479 mL
5 mM 0.361 mL 1.8048 mL 3.6096 mL
10 mM 0.1805 mL 0.9024 mL 1.8048 mL
  • 摩尔浓度计算器

  • 稀释计算器

  • 分子量计算器

质量
=
浓度
x
体积
x
分子量
 
 
 
*在配置溶液时,请务必参考产品标签上、MSDS / COA(可在Glpbio的产品页面获得)批次特异的分子量使用本工具。

计算

动物体内配方计算器 (澄清溶液)

第一步:请输入基本实验信息(考虑到实验过程中的损耗,建议多配一只动物的药量)
给药剂量 mg/kg 动物平均体重 g 每只动物给药体积 ul 动物数量
第二步:请输入动物体内配方组成(配方适用于不溶于水的药物;不同批次药物配方比例不同,请联系GLPBIO为您提供正确的澄清溶液配方)
% DMSO % % Tween 80 % saline
计算重置

Research Update

Chemogenetic evaluation of the mitotic kinesin CENP-E reveals a critical role in triple-negative breast cancer

Breast cancer patients with tumors lacking the three diagnostic markers (ER, PR, and HER2) are classified as triple-negative (primarily basal-like) and have poor prognosis because there is no disease-specific therapy available. To address this unmet medical need, gene expression analyses using more than a thousand breast cancer samples were conducted, which identified elevated centromere protein E (CENP-E) expression in the basal-a molecular subtype relative to other subtypes. CENP-E, a mitotic kinesin component of the spindle assembly checkpoint, is shown to be induced in basal-a tumor cell lines by the mitotic spindle inhibitor drug docetaxel. CENP-E knockdown by inducible shRNA reduces basal-a breast cancer cell viability. A potent, selective CENP-E inhibitor (PF-2771) was used to define the contribution of CENP-E motor function to basal-like breast cancer. Mechanistic evaluation of PF-2771 in basal-a tumor cells links CENP-E-dependent molecular events (e.g., phosphorylation of histone H3 Ser-10; phospho-HH3-Ser10) to functional outcomes (e.g., chromosomal congression defects). Across a diverse panel of breast cell lines, CENP-E inhibition by PF-2771 selectively inhibits proliferation of basal breast cancer cell lines relative to premalignant ones and its response correlates with the degree of chromosomal instability. Pharmacokinetic-pharmacodynamic efficacy analysis in a basal-a xenograft tumor model shows that PF-2771 exposure is well correlated with increased phospho-HH3-Ser10 levels and tumor growth regression. Complete tumor regression is observed in a patient-derived, basal-a breast cancer xenograft tumor model treated with PF-2771. Tumor regression is also observed with PF-2771 in a taxane-resistant basal-a model. Taken together, CENP-E may be an effective therapeutic target for patients with triple-negative/basal-a breast cancer.

Development of a novel HAC-based "gain of signal" quantitative assay for measuring chromosome instability (CIN) in cancer cells

Accumulating data indicates that chromosome instability (CIN) common to cancer cells can be used as a target for cancer therapy. At present the rate of chromosome mis-segregation is quantified by laborious techniques such as coupling clonal cell analysis with karyotyping or fluorescence in situ hybridization (FISH). Recently, a novel assay was developed based on the loss of a non-essential human artificial chromosome (HAC) carrying a constitutively expressed EGFP transgene ("loss of signal" assay). Using this system, anticancer drugs can be easily ranked on by their effect on HAC loss. However, it is problematic to covert this "loss of signal" assay into a high-throughput screen to identify drugs and mutations that increase CIN levels. To address this point, we re-designed the HAC-based assay. In this new system, the HAC carries a constitutively expressed shRNA against the EGFP transgene integrated into human genome. Thus, cells that inherit the HAC display no green fluorescence, while cells lacking the HAC do. We verified the accuracy of this "gain of signal" assay by measuring the level of CIN induced by known antimitotic drugs and added to the list of previously ranked CIN inducing compounds, two newly characterized inhibitors of the centromere-associated protein CENP-E, PF-2771 and GSK923295 that exhibit the highest effect on chromosome instability measured to date. The "gain of signal" assay was also sensitive enough to detect increase of CIN after siRNA depletion of known genes controlling mitotic progression through distinct mechanisms. Hence this assay can be utilized in future experiments to uncover novel human CIN genes, which will provide novel insight into the pathogenesis of cancer. Also described is the possible conversion of this new assay into a high-throughput screen using a fluorescence microplate reader to characterize chemical libraries and identify new conditions that modulate CIN level.