Home>>Signaling Pathways>> GPCR/G protein>> CXCR>>AMG 487

AMG 487 Sale

目录号 : GC11939

A CXCR3 antagonist

AMG 487 Chemical Structure

Cas No.:473719-41-4

规格 价格 库存 购买数量
10mM (in 1mL DMSO)
¥1,338.00
现货
2mg
¥770.00
现货
5mg
¥1,008.00
现货
10mg
¥1,428.00
现货
50mg
¥5,376.00
现货
100mg
¥8,232.00
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.

101

客户使用产品发表文献 1

产品文档

Quality Control & SDS

View current batch:

实验参考方法

Cell experiment [1]:

Cell lines

C26 cells and HT29 cells

Preparation Method

Cells treated or not with 1 µM AMG487 or dimethyl sulfoxide (DMSO) vehicle were placed in the upper well and various concentrations of human or mouse rCXCL9, rCXCL10 and rCXCL11 were added to the lower wells. After incubation of the plates for 18 h (C26 cells) or for 40 h (HT29 cells) at 37 C in 5% CO2 atmosphere, non-migrated cells were removed from the upper well and the migrated cells collected on the lower side of the insert were stained using crystal violet dye and enumerated.

Reaction Conditions

1 µM AMG487;18 h (C26 cells) or 40 h (HT29 cells)

Applications

AMG 487, the CXCR3 antagonist, significantly inhibited the migratory responses of the cells to each of the three ligands(rCXCL9, rCXCL10 and rCXCL11).

Animal experiment [2]:

Animal models

SCID and Balb/c mice

Preparation Method

For the induction of pulmonary metastases, HT29 cells or C26 cells were delivered by intravenous tail injection into SCID or Balb/c mice, respectively. For the polymetastatic liver model, HT29 cells or C26 cells were injected intravenously into the portal vein of Nude or Balb/c mice, respectively. The curative treatment of the mice-bearing metastases was achieved by treating the animals twice daily on days +5 to +12 (syngeneic C26 model) or on days +5 to +23 (immunodeficient HT29 model) with subcutaneous injections of 5 mg/kg of AMG 487 or vehicle. The preventive treatment to antagonise CXCR3 was performed by incubating the colon cancer cells with 1 µM of AMG 487 or vehicle for 18 h in vitro before their injection into mice and by treating concomitantly the animals twice daily on days 1 and 0 with subcutaneous injections of 5 mg/kg of AMG487 or vehicle.

Dosage form

SCID :1 µM AMG 487;s.c.7-17day.

Applications

The AMG 487-treated mice exhibited fewer pulmonary nodules than the control mice in both the models: a 62% reduction in the human mode and 42% less nodules in the mouse model. The inhibitory effect of AMG 487 treatment was also detectable in the cumulative tumour volume with a reduction by 58% in the lungs of the HT29-injected immunodeficient mice and by 51% in the lungs of the C26-injected syngeneic mice.

References:

[1]. Cambien B, Karimdjee BF, et,al.Organ-specific inhibition of metastatic colon carcinoma by CXCR3 antagonism. Br J Cancer. 2009 Jun 2;100(11):1755-64. doi: 10.1038/sj.bjc.6605078. Epub 2009 May 12. PMID: 19436305; PMCID: PMC2695685.

产品描述

AMG 487 is an oral, selective chemokine receptor 3 (CXCR3) antagonist that inhibits CXCL10 and CXCL11 binding to CXCR3 with IC50 values of 8.0 and 8.2 nM, respectively[1].

AMG 487(1 µM AMG 487;18 h (C26 cells) or 40 h (HT29 cells)), the CXCR3 antagonist, significantly inhibited the migratory responses of the cells to each of the three ligands(rCXCL9, rCXCL10 and rCXCL11)[2].

the AMG 487-treated mice(SCID mice:1 µM AMG 487;s.c.7-17day. Balb/c mice:5 mg/kg: s.c.˙twice daily.) exhibited fewer pulmonary nodules than the control mice in both the models: a 62% reduction in the human mode and 42% less nodules in the mouse model. The inhibitory effect of AMG 487 treatment was also detectable in the cumulative tumour volume with a reduction by 58% in the lungs of the HT29-injected immunodeficient mice and by 51% in the lungs of the C26-injected syngeneic mice[2]. During in vitro differentiation, the addition of AMG 487(3- 60µM) decreased the expression of DC co-stimulatory markers, indicating that DC was in a semi-mature state[4]. 66.1 tumor cells were pretreated with AMG 487 prior to i.v. injection into immune-competent female mice. Antagonism of CXCR3 on 66.1 tumor cells inhibited experimental lung metastasis, and this antimetastatic activity was compromised in mice depleted of natural killer cells. Systemic administration of AMG 487(5 mg/kg;s.c.;twice daily) also inhibited experimental lung metastasis[3]. In mice, injection of AMG-487 (i.c.v.)significantly attenuated the accumulation of c-fos in neurons induced by polyinosinic-polycytidylic acid (PIC)[5]. WT mice treated with AMG-487 (5 µg/g twice a day) showed a 45% reduction in bone loss and decreased osteoclasts after LPS injections[6]. Meanwhile, AMG 487 treatment significantly reduced bone loss in mice[7].

References:
[1]. Johnson M, Li AR,et,al. Discovery and optimization of a series of quinazolinone-derived antagonists of CXCR3. Bioorg Med Chem Lett. 2007 Jun 15;17(12):3339-43. doi: 10.1016/j.bmcl.2007.03.106. Epub 2007 Apr 6. PMID: 17448658.
[2]. Cambien B, Karimdjee BF, et,al. Organ-specific inhibition of metastatic colon carcinoma by CXCR3 antagonism. Br J Cancer. 2009 Jun 2;100(11):1755-64. doi: 10.1038/sj.bjc.6605078. Epub 2009 May 12. PMID: 19436305; PMCID: PMC2695685.
[3]. Walser TC, Rifat S, et,al. Antagonism of CXCR3 inhibits lung metastasis in a murine model of metastatic breast cancer. Cancer Res. 2006 Aug 1;66(15):7701-7. doi: 10.1158/0008-5472.CAN-06-0709. PMID: 16885372.
[4]. Qin C, Liu H, et,al.In Vitro Immunological Effects of CXCR3 Inhibitor AMG487 on Dendritic Cells. Arch Immunol Ther Exp (Warsz). 2020 Apr 1;68(2):11. doi: 10.1007/s00005-020-00577-3. PMID: 32239302.
[5]. Petrisko TJ, Konat GW. Peripheral viral challenge increases c-fos level in cerebral neurons. Metab Brain Dis. 2021 Oct;36(7):1995-2002. doi: 10.1007/s11011-021-00819-z. Epub 2021 Aug 18. PMID: 34406561.
[6]. Hiyari S, Green E, et,al. Genomewide Association Study Identifies Cxcl Family Members as Partial Mediators of LPS-Induced Periodontitis. J Bone Miner Res. 2018 Aug;33(8):1450-1463. doi: 10.1002/jbmr.3440. Epub 2018 May 22. PMID: 29637625; PMCID: PMC8434897.
[7]. Lari S, Hiyari S, et,al. Local delivery of a CXCR3 antagonist decreases the progression of bone resorption induced by LPS injection in a murine model. Clin Oral Investig. 2022 Aug;26(8):5163-5169. doi: 10.1007/s00784-022-04484-z. Epub 2022 Apr 25. PMID: 35462591; PMCID: PMC9710470.

AMG 487 是一种口服的选择性趋化因子受体 3 (CXCR3) 拮抗剂,可抑制 CXCL10 和 CXCL11 与 CXCR3 的结合,IC50 值分别为 8.0 和 8.2 nM[1]

AMG 487(1 77777#181;M AMG 487;18 小时(C26 细胞)或 40 小时(HT29 细胞)),CXCR3 拮抗剂,显着抑制细胞对三种配体(rCXCL9、rCXCL10)中每一种的迁移反应和 rCXCL11)[2]

AMG 487 处理的小鼠(SCID 小鼠:1 µM AMG 487;s.c.7-17day。Balb/c 小鼠:5 mg /kg:每天两次。)在两种模型中都比对照小鼠表现出更少的肺部结节:人类模式减少了 62%,小鼠模型中的结节减少了 42%。 AMG 487 治疗的抑制作用也可在累积肿瘤体积中检测到,注射 HT29 的免疫缺陷小鼠的肺部减少了 58%,注射了 C26 的同基因小鼠的肺部减少了 51%[2 ]。在体外分化过程中,AMG 487(3-60µM)的加入降低了DC共刺激标志物的表达,表明DC处于半成熟状态[4]。 66.1 个肿瘤细胞在静脉注射前用 AMG 487 预处理。注射到具有免疫能力的雌性小鼠体内。 CXCR3 对 66.1 肿瘤细胞的拮抗作用抑制了实验性肺转移,并且这种抗转移活性在自然杀伤细胞耗尽的小鼠中受到损害。 AMG 487(5 mg/kg;s.c.;每日两次)的全身给药也抑制了实验性肺转移[3]。在小鼠中,注射 AMG-487 (i.c.v.) 可显着减弱聚肌胞苷酸 (PIC) 诱导的神经元中 c-fos 的积累[5]。用 AMG-487(5 77777#181;g/g,每天两次)处理的 WT 小鼠在注射 LPS 后骨质流失减少了 45%,破骨细胞也减少了[6]。同时,AMG 487 治疗显着减少了小鼠的骨质流失[7]

Chemical Properties

Cas No. 473719-41-4 SDF
化学名 N-[(1R)-1-[3-(4-ethoxyphenyl)-4-oxopyrido[2,3-d]pyrimidin-2-yl]ethyl]-N-(pyridin-3-ylmethyl)-2-[4-(trifluoromethoxy)phenyl]acetamide
Canonical SMILES CCOC1=CC=C(C=C1)N2C(=O)C3=C(N=CC=C3)N=C2C(C)N(CC4=CN=CC=C4)C(=O)CC5=CC=C(C=C5)OC(F)(F)F
分子式 C32H28F3N5O4 分子量 603.59
溶解度 ≥ 122.2 mg/mL in DMSO, ≥ 122 mg/mL in EtOH 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 1.6568 mL 8.2838 mL 16.5675 mL
5 mM 0.3314 mL 1.6568 mL 3.3135 mL
10 mM 0.1657 mL 0.8284 mL 1.6568 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

An inhibitory metabolite leads to dose- and time-dependent pharmacokinetics of (R)-N-{1-[3-(4-ethoxy-phenyl)-4-oxo-3,4-dihydro-pyrido[2,3-d]pyrimidin-2-yl]-ethyl}-N-pyridin-3-yl-methyl-2-(4-trifluoromethoxy-phenyl)-acetamide (AMG 487) in human subjects after multiple dosing

(R)-N-{1-[3-(4-Ethoxy-phenyl)-4-oxo-3,4-dihydro-pyrido[2,3-d]-pyrimidin-2-yl]-ethyl}-N-pyridin-3-yl-methyl-2-(4-trifluoromethoxyphenyl)-acetamide (AMG 487) is a potent and selective orally bioavailable chemokine (C-X-C motif) receptor 3 (CXCR3) antagonist that displays dose- and time-dependent pharmacokinetics in human subjects after multiple oral dosing. Although AMG 487 exhibited linear pharmacokinetics on both days 1 and 7 at the 25-mg dose, dose- and time-dependent kinetics were evident at the two higher doses. Nonlinear kinetics were more pronounced after multiple dosing. Area under the plasma concentration-time curve from 0 to 24 h [AUC((0-24 h))] increased 96-fold with a 10-fold increase in dose on day 7 compared with a 28-fold increase in AUC((0-24 h)) on day 1. These changes were correlated with time- and dose-dependent decreases in the metabolite to parent plasma concentrations, suggesting that these changes result from a decrease in the oral clearance (CL) of AMG 487 (e.g., intestinal/hepatic first-pass metabolism and systemic CL). The biotransformation of AMG 487 is dependent on CYP3A and results in the formation of two primary metabolites, a pyridyl N-oxide AMG 487 (M1) and an O-deethylated AMG 487 (M2). One of these metabolites, M2, undergoes further metabolism by CYP3A. M2 has also been demonstrated to inhibit CYP3A in a competitive (K(i)=0.75 microM) manner as well as via mechanism-based inhibition (unbound K(I)=1.4 microM, k(inact)=0.041 min(-1)). Data from this study implicate M2-mediated CYP3A mechanism-based inhibition as the proximal cause for the time-dependent pharmacokinetics of AMG 487. However, the sequential metabolism of M2, nonlinear AMG 487 pharmacokinetics, and the inability to accurately determine the role of intestinal AMG 487 metabolism complicates the correlation between M2 plasma concentrations and the time-dependent AMG 487 pharmacokinetic changes.

Sequential metabolism of AMG 487, a novel CXCR3 antagonist, results in formation of quinone reactive metabolites that covalently modify CYP3A4 Cys239 and cause time-dependent inhibition of the enzyme

CYP3A4-mediated biotransformation of (R)-N-(1-(3-(4-ethoxyphenyl)-4-oxo-3,4-dihydropyrido[2,3-d]pyrimidin-2-yl)ethyl)-N-(pyridin-3-ylmethyl)-2-(4-(trifluoromethoxy)phenyl)acetamide (AMG 487) was previously shown to generate an inhibitory metabolite linked to dose- and time-dependent pharmacokinetics in humans. Although in vitro activity loss assays failed to demonstrate CYP3A4 time-dependent inhibition (TDI) with AMG 487, its M2 phenol metabolite readily produced TDI when remaining activity was assessed using either midazolam or testosterone (K(I) = 0.73-0.74 μM, k(inact) = 0.088-0.099 min(-1)). TDI investigations using an IC(50) shift method successfully produced inhibition attributable to AMG 487, but only when preincubations were extended from 30 to 90 min. The shift magnitude was ?3× for midazolam activity, but no shift was observed for testosterone activity. Subsequent partition ratio determinations conducted for M2 using recombinant CYP3A4 showed that inactivation was a relatively inefficient process (r = 36). CYP3A4-mediated biotransformation of [(3)H]M2 in the presence of GSH led to identification of two new metabolites, M4 and M5, which shifted focus away from M2 being directly responsible for TDI. M4 (hydroxylated M2) was further metabolized to form reactive intermediates that, upon reaction with GSH, produced isomeric adducts, collectively designated M5. Incubations conducted in the presence of [(18)O]H(2)O confirmed incorporation of oxygen from O(2) for the majority of M4 and M5 formed (>75%). Further evidence of a primary role for M4 in CYP3A4 TDI was generated by protein labeling and proteolysis experiments, in which M4 was found to be covalently bound to Cys239 of CYP3A4. These investigations confirmed a primarily role for M4 in CYP3A4 inactivation, suggesting that a more complex metabolic pathway was responsible for generation of inhibitory metabolites affecting AMG 487 human pharmacokinetics.

In Vitro Immunological Effects of CXCR3 Inhibitor AMG487 on Dendritic Cells

AMG 487 is the targeted blocker of chemokine receptor CXCR3 and improves inflammatory symptoms by blocking the inflammatory cycle. Here we investigated whether AMG 487 affects dendritic cell (DC) biology and function. The expression of co-stimulatory markers on DCs was reduced, indicating the semi-mature state of DC when AMG 487 was added throughout the in vitro differentiation period. Additionally, when added solely during the final lipopolysaccharide-induced activation step, AMG 487 inhibited DC activation, as demonstrated by a decreased expression of activation markers. AMG487 also promoted the expression of PD-L2 and impaired the ability to induce antigen-specific T cell responses. Our results demonstrated that AMG 487 significantly affects DC maturity in vitro and function leading to impaired T cell activation, inducing DCs to have characteristics similar to tolerogenic DCs. AMG 487 may directly play an immunomodulatory role during DC development and functional shaping.

Peripheral viral challenge increases c-fos level in cerebral neurons

Peripheral viral infection can substantially alter brain function. We have previously shown that intraperitoneal (i.p.) injection of a viral mimetic, polyinosinic-polycytidylic acid (PIC), engenders hyperexcitability of cerebral neurons. Because neuronal activity is invariably associated with their expression of the Cfos gene, the present study was undertaken to determine whether PIC challenge also increases neuronal c-fos protein level. Female C57BL/6 mice were i.p. injected with PIC, and neuronal c-fos was analyzed in the motor cortex by immunohistochemistry. PIC challenge instigated a robust increase in the number of c-fos-positive neurons. This increase reached approximately tenfold over control at 24 h. Also, the c-fos staining intensity of individual neurons increased. AMG-487, a specific inhibitor of the chemokine receptor CXCR3, profoundly attenuated the accumulation of neuronal c-fos, indicating the activation of CXCL10/CXCR3 axis as the trigger of the process. Together, these results show that the accumulation of c-fos is a viable readout to assess the response of cerebral neurons to peripheral PIC challenge, and to elucidate the underlying molecular mechanisms.

Local delivery of a CXCR3 antagonist decreases the progression of bone resorption induced by LPS injection in a murine model

Objectives: This experimental study was carried out to investigate the effects of locally delivered nanoparticles (AMG-487 NP) containing a CXCR3 antagonist in inhibiting the progression of LPS-induced inflammation, osteoclastic activity, and bone resorption on a murine model.
Materials and methods: Thirty, 7-week-old C57BL/6 J male mice were used. Inflammatory bone loss was induced by Porphyromonas gingivalis-lipopolysaccharide (P.g.-LPS) injections between the first and second maxillary molars, bilaterally, twice a week for 6 weeks (n = 20). AMG-487 NP were incorporated into a liposome carrier and locally delivered on sites where P.g.-LPS was injected. Control mice (n = 10) were injected with vehicle only. Experimental groups included (1) control, (2) LPS, and (3) LPS + NP. At the end of 1 and 6 weeks, mice were euthanized, maxillae harvested, fixed, and stored for further analysis.
Results: Volumetric bone loss analysis revealed, at 1 week, an increase in bone loss in the LPS group (47.9%) compared to control (27.4%) and LPS + NP (27.8%) groups. H&E staining demonstrated reduced inflammatory infiltrate in the LPS + NP group compared to LPS group. At 6 weeks, volumetric bone loss increased in all groups; however, treatment with the CXCR3 antagonist (LPS + NP) significantly reduced bone loss compared to the LPS group. CXCR3 antagonist treatment significantly reduced osteoclast numbers when compared to LPS group at 1 and 6 weeks.
Conclusions: This study showed that local delivery of a CXCR antagonist, via nanoparticles, in a bone resorption model, induced by LPS injection, was effective in reducing inflammation, osteoclast numbers, and bone loss.
Clinical relevance: CXCR3 blockade can be regarded as a novel target for therapeutic intervention of bone loss. It can be a safe and convenient method for periodontitis treatment or prevention applicable in clinical practice.