Home>>Signaling Pathways>> Endocrinology and Hormones>> Opioid Receptor>>Cebranopadol ((1α,4α)stereoisomer)

Cebranopadol ((1α,4α)stereoisomer) Sale

(Synonyms: GRT6005 (1α,4α)stereoisomer) 目录号 : GC35638

Cebranopadol (1α,4α)stereoisomer 是 cebranopadol 的立体异构体,活性较低。Cebranopadol 是 ORL-1 的有效激活剂。

Cebranopadol ((1α,4α)stereoisomer) Chemical Structure

Cas No.:863513-93-3

规格 价格 库存 购买数量
2mg
¥1,710.00
现货
5mg
¥3,150.00
现货
10mg 待询 待询
50mg 待询 待询

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:

产品描述

Cebranopadol ((1α,4α)stereoisomer) is a stereoisomer of cebranopadol. Cebranopadol is a potent agonist activity on ORL-1.

[1]. Schunk S, et al. Discovery of a Potent Analgesic NOP and Opioid Receptor Agonist: Cebranopadol. ACS Med Chem Lett. 2014 Jun 24;5(8):857-62.

Chemical Properties

Cas No. 863513-93-3 SDF
别名 GRT6005 (1α,4α)stereoisomer
Canonical SMILES FC1=CC=C2C(C(CCO[C@@]34CC[C@@](N(C)C)(C5=CC=CC=C5)CC4)=C3N2)=C1.[relative stereochemistry]
分子式 C24H27FN2O 分子量 378.48
溶解度 Soluble in DMSO 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 2.6421 mL 13.2107 mL 26.4215 mL
5 mM 0.5284 mL 2.6421 mL 5.2843 mL
10 mM 0.2642 mL 1.3211 mL 2.6421 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

Cebranopadol as a Novel Promising Agent for the Treatment of Pain

Molecules 2022 Jun 21;27(13):3987.PMID:35807228DOI:10.3390/molecules27133987.

Opioids are used to treat pain, but despite their effectiveness, they possess several side effects such as respiratory depression, tolerance and physical dependence. Cebranopadol has been evaluated as a solution to this problem. The compound acts on the mu opioid receptor and the nociceptin/orphanin receptor and these receptors co-activation can reduce opioid side-effects without compromising analgesia. In the present review, we have compiled information on the effects of Cebranopadol, its pharmacokinetics, and clinical trials involving Cebranopadol, to further explore its promise in pain management.

Cebranopadol: novel dual opioid/NOP receptor agonist analgesic

J Clin Pharm Ther 2017 Feb;42(1):8-17.PMID:27778406DOI:10.1111/jcpt.12461.

What is known and objective: Chronic pain presents a difficult clinical challenge because of the limited efficacy, the limiting adverse-effect profile or the abuse potential of current analgesic options. Cebranopadol is a novel new agent in clinical trials that combines dual agonist action at opioid and nociceptin/orphanin FQ peptide (NOP) receptors. It is the first truly unique, centrally acting analgesic in several years. We here review the basic and clinical pharmacology of Cebranopadol. Methods: Published literature and Internet sources were searched to identify information related to the basic science (pharmacology and medicinal chemistry) and development (clinical trial) information on the mechanism of dual opioid and NOP receptor pharmacologic action in general, and for Cebranopadol in particular. The identified sources were reviewed and the information synthesized. Results: The preclinical testing of Cebranopadol has characterized it as a dual opioid and NOP receptor agonist that displays antinociceptive and antihyperalgesic action in a variety of acute and chronic pain models in animals. Unlike most current traditional opioids, it is generally more potent against neuropathic than nociceptive pain. Several phase 2 clinical trials have been completed. What is new and conclusion: Despite the medical need, a truly novel centrally acting analgesic has not been developed in many years. Cebranopadol represents a truly novel mechanistic approach. Its actual place in pain pharmacotherapy awaits the results of phase 3 clinical trials.

Cebranopadol : a first-in-class potent analgesic agent with agonistic activity at nociceptin/orphanin FQ and opioid receptors

Expert Opin Investig Drugs 2015 Jun;24(6):837-44.PMID:25865744DOI:10.1517/13543784.2015.1036985.

Introduction: Pain is a syndrome of various clinical disorders, which arises from various pathological conditions and which presents significant challenges in both its diagnosis and treatment. There is currently a strong medical demand to develop new therapies with a higher efficacy and a better tolerability profile. Areas covered: In this review, the authors report on the available data for the pharmacological properties of Cebranopadol (GRT6005), a first in-class, potent analgesic compound which acts as an agonist of nociceptin/orphanin FQ peptide (NOP) and opioid receptors. They highlight the in vitro receptor binding studies, as well as the in vivo preclinical results on the analgesic efficacy of Cebranopadol obtained in several rodent pain models. The authors also briefly summarize the available data from clinical trials with Cebranopadol. Expert opinion: Cebranopadol displays analgesic, antiallodynic and antihyperalgesic properties in several rat models of acute nociceptive, inflammatory, cancer and neuropathic pain. In contrast to classical opioids, it has a higher analgesic potency in models of neuropathic pain than in acute nociceptive pain. Even at higher analgesic doses, Cebranopadol does not induce motor coordination deficits or respiratory depression in rats. Hence, it seems to possess a broader therapeutic window than classical opioids. While it is particularly interesting as a novel, potent bifunctional agonist of NOP/opioid receptors, the outcome of its ongoing and planned clinical trials will be crucial for its future development and potential application in humans.

Functional Profile of Systemic and Intrathecal Cebranopadol in Nonhuman Primates

Anesthesiology 2021 Sep 1;135(3):482-493.PMID:34237134DOI:10.1097/ALN.0000000000003848.

Background: Cebranopadol, a mixed nociceptin/opioid receptor full agonist, can effectively relieve pain in rodents and humans. However, it is unclear to what degree different opioid receptor subtypes contribute to its antinociception and whether Cebranopadol lacks acute opioid-associated side effects in primates. The authors hypothesized that coactivation of nociceptin receptors and μ receptors produces analgesia with reduced side effects in nonhuman primates. Methods: The antinociceptive, reinforcing, respiratory-depressant, and pruritic effects of Cebranopadol in adult rhesus monkeys (n = 22) were compared with μ receptor agonists fentanyl and morphine using assays, including acute thermal nociception, IV drug self-administration, telemetric measurement of respiratory function, and itch-scratching responses. Results: Subcutaneous Cebranopadol (ED50, 2.9 [95% CI, 1.8 to 4.6] μg/kg) potently produced antinociception compared to fentanyl (15.8 [14.6 to 17.1] μg/kg). Pretreatment with antagonists selective for nociceptin and μ receptors, but not δ and κ receptor antagonists, caused rightward shifts of the antinociceptive dose-response curve of Cebranopadol with dose ratios of 2 and 9, respectively. Cebranopadol produced reinforcing effects comparable to fentanyl, but with decreased reinforcing strength, i.e., Cebranopadol (mean ± SD, 7 ± 3 injections) versus fentanyl (12 ± 3 injections) determined by a progressive-ratio schedule of reinforcement. Unlike fentanyl (8 ± 2 breaths/min), systemic Cebranopadol at higher doses did not decrease the respiratory rate (17 ± 2 breaths/min). Intrathecal Cebranopadol (1 μg) exerted full antinociception with minimal scratching responses (231 ± 137 scratches) in contrast to intrathecal morphine (30 μg; 3,009 ± 1,474 scratches). Conclusions: In nonhuman primates, the μ receptor mainly contributed to cebranopadol-induced antinociception. Similar to nociceptin/μ receptor partial agonists, Cebranopadol displayed reduced side effects, such as a lack of respiratory depression and pruritus. Although Cebranopadol showed reduced reinforcing strength, its detectable reinforcing effects and strength warrant caution, which is critical for the development and clinical use of Cebranopadol.

Cebranopadol reduces cocaine self-administration in male rats: Dose, treatment and safety consideration

Neuropharmacology 2020 Aug 1;172:108128.PMID:32389751DOI:10.1016/j.neuropharm.2020.108128.

As a novel first-in-class potent analgesic acting as an agonist of multiple opioid receptors, Cebranopadol showed high efficacy and good tolerability in a broad range of preclinical models and clinical trials related to pain. In the present study, to evaluate the efficacy and safety of Cebranopadol as a potential treatment of cocaine dependence, we tested the effects of Cebranopadol with single and repeated doses (25, 50, 75, or 100 μg/kg, oral gavage) using rat models of cocaine fixed-ratio (FR) self-administration (SA), cocaine progressive-ratio (PR) SA, and sucrose pellet SA. In single-dosing treatment paradigm, Cebranopadol significantly and dose-dependently reduced cocaine SA under FR and PR schedules and suppressed food intake under FR schedule without causing apparent side effects. In repeated-dosing treatment scheme, i.e. daily administration of 25, 50, 75, or 100 μg/kg Cebranopadol for a week, the similar reduction in cocaine intake was detected, while non-negligible complications/side effects were observed at repeated high doses (75 and 100 μg/kg). The observed side effects were similar to the common toxic signs elicited by heroin at high doses, although Cebranopadol did not fully substitute heroin's discriminative stimulant effects in our drug discriminative tests. These results demonstrated that the most appropriate oral dose of Cebranopadol to balance the efficacy and safety is 50 μg/kg. Collectively, although Cebranopadol may serve as a new treatment for cocaine dependence, more consideration, cautiousness, and a clear optimal dose window to dissociate its therapeutic effects from opioid side effects/complications in male and female subjects will be necessary to increase its practical clinical utility.