规格 | 价格 | 库存 | 数量 |
---|---|---|---|
10 mM * 1 mL in DMSO |
|
||
1mg |
|
||
5mg |
|
||
25mg |
|
||
50mg |
|
||
100mg |
|
||
250mg |
|
||
500mg |
|
||
1g |
|
||
5g |
|
||
Other Sizes |
|
靶点 |
ALK (IC50 = 0.2 nM); InsR (IC50 = 7 nM); IGF-1R (IC50 = 8 nM); STK22D (IC50 = 23 nM); FLT3 (IC50 = 60 nM); FGFR2 (IC50 = 260 nM)
|
---|---|
体外研究 (In Vitro) |
体外活性: Ceritinib(原名 LDK378;商品名:Zykadia)是一种新型、有效、选择性的 ALK(间变性淋巴瘤激酶阳性)抑制剂,在无细胞试验中 IC50 为 0.2 nM,显示其活性是 ALK 的 40 倍和 35 倍。分别针对 IGF-1R 和 InsR 的选择性。色瑞替尼于 2014 年 4 月获得 FDA 批准用于治疗非小细胞肺癌(NSCLC)。在 I 期试验中,Ceritinib 在 78 名间变性 ALK+ 转移性非小细胞肺癌患者中显示出显着的临床反应,这些患者在克唑替尼治疗期间或治疗后病情进展或之前未接受过克唑替尼治疗。 LDK378 在 Ba/F3-NPM-ALK 和 Karpas290 细胞中显示出良好的抗增殖活性,IC50 分别为 26.0 nM 和 22.8 nM,而 Ba/F3-Tel-InsR 和 Ba/F3-WT 中的 IC50 分别为 319.5 nM 和 2477 nM细胞。激酶测定:除大肠杆菌中产生的未标记 ERK2 外,所有激酶均使用杆状病毒表达技术表达为组氨酸或 GST 标记融合蛋白。激酶活性在 LabChip 迁移率变动测定中进行测量。该测定在 30°C 下进行 60 分钟。 LDK378 对酶活性的影响是从 LDK378 不存在和存在下的线性进展曲线获得的,并且通常通过一次读数(终点测量)来确定。细胞测定:将表达荧光素酶的细胞与连续稀释的 LDK378 或 DMSO 一起孵育 2-3 天。荧光素酶表达用作细胞增殖/存活的衡量标准,并使用 Bright-Glo 荧光素酶测定系统进行评估。 IC50 值是使用 XLFit 软件生成的。
|
体内研究 (In Vivo) |
LDK378 旨在降低形成反应性代谢物的可能性,并在肝微粒体中显示出不可检测的谷胱甘肽 (GSH) 加合物水平 (<1%)。 LDK378具有较好的代谢稳定性,具有中等程度的CYP3A4(咪达唑仑底物)抑制作用和hERG抑制作用。与肝血流相比,LDK378 在动物(小鼠、大鼠、狗和猴)中表现出较低的血浆清除率,在小鼠、大鼠、狗和猴中的口服生物利用度高于 55%。 LDK378 在 Karpas299 和 H2228 大鼠异种移植模型中诱导剂量依赖性生长抑制和肿瘤消退,且没有体重减轻。长期剂量高达 100 mg/kg 时,LDK378 对小鼠的胰岛素水平或血浆葡萄糖利用率没有影响。
|
酶活实验 |
所有激酶均使用杆状病毒表达技术表达为 GST 或组氨酸标记的融合蛋白,但未标记的 ERK2 除外,它是在大肠杆菌中产生的。使用 LabChip 迁移率变动测定来测定激酶活性。测定在 30°C 下运行 60 分钟。通常可以通过分析存在和不存在 LDK378 的情况下的线性进展曲线,从单个读数(终点测量)确定 LDK378 对酶活性的影响。
酶激酶谱描述[1] 除了在大肠杆菌中产生的未标记的ERK2外,所有激酶都使用杆状病毒表达技术以组氨酸或gst标记的融合蛋白表达。AURORA-A、JAK2、MK2、SYK、PKA、ERK2购自公司,所有其他激酶均由公司提供。激酶活性通过LabChip移动位移法测定。实验在30°C下进行60分钟。 化合物对酶活性的影响由无化合物和有化合物情况下的线性进展曲线得到,通常由一次读数(终点测量)确定。 GSH-Trapping测定[1] 为了表征代谢激活,将化合物的10 mmol/L DMSO原液与人肝微粒体(50 μL)在37℃下孵育60 min,含1 mg蛋白/mL,磷酸盐缓冲液。在180 μL乙腈/水(比例1:1)混合物中加入0.5 mM的20 μL DMSO原液4微升,加入50 μL 1 mg/mL肝微粒体磷酸盐缓冲液中,37℃预孵育3 min。预孵育后,加入50 μL NADPH (1 mmol/L)、UDPGA (1 mmol/L)、MgCl (2 mmol/L)和还原性乙酯GSH (2 mmol/L)开始反应。60 min后,用200 μL的冰凉乙腈停止反应。反应混合物保存在- 20°C。离心10000g, 5min,取300 μL上清250 μL。取20 μL溶液进行分析。采用Agilent HP1100泵,Phenyl HexylRP柱,150 mm × 2.0 mm,粒径4.6 μm进行液相色谱分离。采用梯度流动相编程,流速为350 μL/min。洗脱液A为含0.1%甲酸的ml - q水。洗脱液B为含0.1%甲酸的乙腈。流动相在6分钟内从5% B到95% B呈线性梯度,在95% B下保持2分钟,总运行时间为10分钟。柱出水直接引入三重四极杆质谱仪或离子阱质谱仪的离子源。所采用的电离技术为正电喷雾(ES)。TS-Quantum被用于产物离子扫描模式,利用Q2(碰撞室)的碰撞诱导解离。碰撞气体是氩气。碰撞能量设定为30 eV。 溶解度测定[1] 在加入500 μL pH 6.8缓冲液之前,在三个小玻璃瓶中各加入100微升1mm DMSO溶液,蒸发至干燥。振荡24小时后,溶液通过0.4 μm改性PCTE膜的MultiScreen可溶性96孔板真空过滤,并将每种滤液的等分液转移到UV板上进行定量,如Uvarova等人所述。 代谢清除测定[1] 代谢清除率测定采用Richmond等人描述的方法进行。 CYP抑制试验[1] 样品在DMSO中配制成10 mM溶液,然后使用Bell等人描述的通用LC-MS /MS方法进行检测和分析。 |
细胞实验 |
LDK378 或 DMSO 连续稀释并与荧光素酶表达细胞一起孵育两到三天。使用 Bright-Glo 荧光素酶测定系统,测量荧光素酶表达作为细胞增殖和存活的指标。 XLFit 程序用于生成 IC50 值。
GI50决心[3] 为了计算单个化合物[ Ceritinib (LDK-378)]的一半最大生长抑制浓度(GI50),神经母细胞瘤肿瘤细胞以100 μL的总体积接种于96孔板中,并允许贴壁过夜。第二天,将化合物(例如Ceritinib (LDK-378)溶解在DMSO中)以100 μL的浓度梯度分为6个重复添加到孔中,其中包括仅DMSO的对照。将细胞暴露于药物72小时后,用10%三氯乙酸固定细胞,并用1%醋酸磺胺B染色,估计处理孔与对照孔的细胞数量。采用石墨板棱镜,通过非线性回归分析,计算出使细胞生长比对照抑制50%的药物浓度。 蛋白裂解物的制备[3] 细胞系通过刮削收集,在500 g下旋转5分钟,在磷酸盐缓冲盐水中洗涤一次,细胞颗粒在CHAPS裂解缓冲液中重悬[50 mm Tris/HCl pH 8.0, 1 mm EDTA, 150 mm NaCl, 1% CHAPS, 0.2 mm PMSF, 1:50磷酸酶抑制剂鸡尾酒2和3,1:100蛋白酶抑制剂鸡尾酒]。冷冻组织样品在CHAPS裂解缓冲液中均质,作为细胞裂解液。在冰上孵育30分钟后,裂解物在16000 g下旋转15分钟,收集上清。用BCA蛋白法测定蛋白浓度,并与牛丝氨酸白蛋白标准对照。 |
动物实验 |
Studies on in vivo PK are carried out on dogs, rats, mice, and cynomolgus monkeys. Male Balb/c mice are given cetinib (LDK378) (HCl salt) orally via gavage at a dose of 20 mg/kg (n=3) and intravenously via tail vein at a dose of 5 mg/kg (n= 3). Sprague-Dawley rats are dosed with Ceritinib (LDK378) (HCl salt) intravenously via the tail vein at 3 mg/kg (n=3) and orally via gavage at 10 mg/kg (n=3) using the same formulation. Serial blood samples are taken over the course of 24 hours following dosage at prearranged times. Ceritinib (phosphate salt) is given as a single intravenous (n = 2) or oral (n = 3) dose to male beagle dogs. The intravenous solution has a dosage of 5 mg/kg, while the oral suspension has a dosage of 20 mg/kg. A single intravenous (n = 2) or oral (n = 3) dose of Ceritinib (free base) is given to male Cynomologus monkeys. The intravenous solution has a dose of 5 mg/kg, while the oral suspension has a dose of 60 mg/kg. For plasma, blood is drawn at prearranged intervals over a period of 144 hours following dosage.
PK Studies[1] In vivo PK studies were conducted in mice, rats, dogs, and cynomolgus monkeys. 15b (HCl salt) was formulated as a solution in 75% PEG300/25% D5W and administered to male Balb/c mice intravenously via tail vein at 5 mg/kg (n = 3) and orally via gavage at 20 mg/kg (n = 3). By use of the same formulation, 15b (HCl salt) was dosed to Sprague–Dawley rats intravenously via the tail vein at 3 mg/kg (n = 3) and orally via gavage at 10 mg/kg (n = 3). Blood samples were collected serially at scheduled times over 24 h after dosing. Male beagle dogs received a single intravenous (n = 2) or oral (n = 3) dose of 15b (phosphate salt) as an intravenous solution in 30% propylene glycol/5% Solutol buffer at 5 mg/kg and an oral suspension in suspension in 0.5% (w/v) aqueous methylcellulose/0.5% Tween 80 at 20 mg/kg, respectively. Male cynomologus monkeys received single intravenous (n = 2) or oral (n = 3) dose of 15b (free base) as an intravenous solution in 30% propylene glycol/5% solutol at 5 mg/kg and an oral suspension in 0.5% (w/v) methylcellulose at 60 mg/kg, respectively. Blood samples for plasma were collected at prescheduled times over 144 h after dosing. In Vivo Experiments[1] RNU nude rats bearing the Karpas299 tumors were randomized into five groups (n = 6 rats per group) with an average tumor size of 326 ± 128 mm3. 15b (phosphate salt) was formulated in 0.5% MC/0.5% Tween 80 and administered by oral gavage at a dosing volume of 10 μL/g of an animal body weight. Animals in each group received vehicle or 6.25, 12.5, 25, 50 mg/kg 15b every day for 14 consecutive days. RNU nude rats bearing the H2228 tumors were randomized into four groups (n = 4 rats per group) with an average tumor volume of 371 ± 139 mm3. 15b (phosphate salt) was formulated in 0.5% MC/0.5% Tween 80 and administered by oral gavage at a dosing volume of 10 μL/g of an animal body weight. Animals in each group received vehicle or 5, 10, 25 mg/kg 15b (phosphate salt) every day for 14 consecutive days. RNU nude rats bearing the Karpas299 tumors were dosed with 15b (phosphate salt) at 50 mg/kg. Tumor and plasma samples were collected 7, 24, 48, and 72 h after dosing. Two tumor pieces were collected from each animal, one piece for protein extraction and the other for PK analysis. Proteins were extracted from tumor samples and then subjected to SDS–PAGE followed by Western blot with phospho-STAT3 antibody (pSTAT3, Tyr705). HOMA-IR[1] Homeostatic model assessment (HOMA) of insulin resistance (IR) is a technical method for assessing IR from basal (fasting) glucose and insulin or C-peptide concentrations. The model is widely used to estimate insulin resistance. Groups of wild-type mice (n = 8 mice per group) were randomized into treatment groups based on their initial body weight. Mice were housed four per cage and dosed with vehicle orally, or ALK inhibitor (15b or Ceritinib (LDK-378)), orally once per day for 7 days. On day 7, compound was administered 180 min prior to a 3 g/kg glucose bolus. OGTT evaluations were performed in conscious mice that were 11 weeks of age. The mice were fasted by removing food at 6 p.m. the day before. A baseline blood sample was taken at t = −180 min, and the mice were then dosed orally with the compounds. A baseline blood sample was taken at t = 0 min, and the animals were then administered an oral glucose bolus (3 g/kg) immediately. Blood was obtained (via tail bleeding) to measure blood glucose (using glucometer). A single drop of blood from the tail was measured for glucose using a glucometer at t = −180, 0, 20, 40, 60, 120 min. Approximately 40 μL samples of blood were collected separately for insulin analysis 3 h prior to dosing (on day 0 and day 7) into chilled collection tubes containing EDTA. Plasma was isolated and stored at −70 °C until further analysis. The homeostatic model assessment-insulin resistance index (HOMA-IR) was used as a measure of insulin resistance and was calculated using the formula HOMA-IR = (FPG × FPI)/22.5 where FPG (mM) is the fasting plasma glucose concentration and FPI (μU/mL) is the fasting plasma insulin concentration. Insulin levels were determined using a detection assay kit from Mesoscale Discovery (MSD): catalog no. K112BZC-2. Higher values indicate insulin resistance. Mouse models[1] Th‐ALK F1174L/MYCN tumor‐bearing animals were enrolled into therapeutic trials when their abdominal tumors reached 5 mm in diameter according to palpation. Volumetric MRI was performed as previously described (Jamin et al., 2013), with each animal undergoing imaging on day 0 and day 7. The tumor volume at each time point was then calculated. For in vivo oral dosing on days 1–7, crizotinib was dissolved in sterile water with 10% Tween 20. Ceritinib (LDK-378) was dissolved in 0.5% methylcellulose, 0.5% Tween 80 with sterile water. Two hours following the final dose of either compound, tumor tissue was excised and snap‐frozen prior to analysis. |
药代性质 (ADME/PK) |
Absorption, Distribution and Excretion
After oral administration of ceritinib, peak concentrations were achieved after approximately 4 to 6 hours. Following oral administration of a single 750 mg radiolabeled ceritinib dose, 92.3% of the administered dose was recovered in the feces (with 68% as unchanged parent compound) while 1.3% of the administered dose was recovered in the urine. The apparent volume of distribution (Vd/F) is 4230 L following a single 750 mg dose. The geometric mean apparent clearance (CL/F) of ceritinib was lower at steady-state (33.2 L/h) after 750 mg daily dosing than after a single 750 mg dose (88.5 L/h). Metabolism / Metabolites In vitro studies demonstrated that CYP3A was the major enzyme involved in the metabolic clearance of ceritinib. Following oral administration of a single 750 mg radiolabeled ceritinib dose, ceritinib as the parent compound was the main circulating component (82%) in human plasma. Biological Half-Life The terminal half life is 41 hours. |
毒性/毒理 (Toxicokinetics/TK) |
Hepatotoxicity
Elevations in serum aminotransferase levels are common during ceritinib therapy occurring in 20% to 50% of patients, but rising above 5 times the upper limit of the normal range in only 1% to 2%. Hepatic failure is said to have occurred in 0.2% of patients and to have resulted in several fatalities. Hepatotoxicity appears to be a class effect among ALK inhibitors, although liver injury appears to be more frequent and more severe with crizotinib than ceritinib or alectinib. Specific details of the liver injury associated with ceritinib such as latency, serum enzyme pattern, clinical features and course, have not been published. Other ALK inhibitors typically cause liver injury arising within days or weeks of starting therapy, and presenting abruptly with hepatocellular enzyme elevations and a moderate-to-severe course. Immunoallergic and autoimmune features are not common. The rate of clinically significant liver injury and hepatic failure is increased in patients with preexisting cirrhosis or hepatic impairment due to liver tumor burden. Recurrence upon reexposure has been reported. Likelihood score: D (possible cause of clinically apparent liver injury). Effects During Pregnancy and Lactation ◉ Summary of Use during Lactation No information is available on the clinical use of ceritinib during breastfeeding. Because ceritinib is 97% bound to plasma proteins, the amount in milk is likely to be low. The manufacturer recommends that breastfeeding be discontinued during ceritinib therapy and for 2 weeks after the final dose. ◉ Effects in Breastfed Infants Relevant published information was not found as of the revision date. ◉ Effects on Lactation and Breastmilk Relevant published information was not found as of the revision date. Protein Binding Ceritinib is 97% bound to human plasma proteins, independent of drug concentration. |
参考文献 |
|
其他信息 |
Ceritinib is a member of the class of aminopyrimidines that is 2,6-diamino-5-chloropyrimidine in which the amino groups at positions 2 and 6 are respectively carrying 2-methoxy-4-(piperidin-4-yl)-5-methylphenyl and 2-(isopropylsulfonyl)phenyl substituents. Used for the treatment of ALK-positive metastatic non-small cell lung cancer. It has a role as an antineoplastic agent and an EC 2.7.10.1 (receptor protein-tyrosine kinase) inhibitor. It is an aminopyrimidine, an aromatic ether, an organochlorine compound, a secondary amino compound, a member of piperidines and a sulfone.
Ceritinib is used for the treatment of adults with anaplastic lymphoma kinase (ALK)-positive metastatic non-small cell lung cancer (NSCLC) following failure (secondary to resistance or intolerance) of prior crizotinib therapy. About 4% of patients with NSCLC have a chromosomal rearrangement that generates a fusion gene between EML4 (echinoderm microtubule-associated protein-like 4) and ALK (anaplastic lymphoma kinase), which results in constitutive kinase activity that contributes to carcinogenesis and seems to drive the malignant phenotype. Ceritinib exerts its therapeutic effect by inhibiting autophosphorylation of ALK, ALK-mediated phosphorylation of the downstream signaling protein STAT3, and proliferation of ALK-dependent cancer cells. Following treatment with crizotinib (a first-generation ALK inhibitor), most tumours develop drug resistance due to mutations in key "gatekeeper" residues of the enzyme. This occurrence led to development of novel second-generation ALK inhibitors such as ceritinib to overcome crizotinib resistance. The FDA approved ceritinib in April 2014 due to a surprisingly high response rate (56%) towards crizotinib-resistant tumours and has designated it with orphan drug status. Ceritinib is a Kinase Inhibitor. The mechanism of action of ceritinib is as a Tyrosine Kinase Inhibitor, and Cytochrome P450 3A Inhibitor, and Cytochrome P450 2C9 Inhibitor. Ceritinib is a small molecule tyrosine kinase receptor inhibitor and antineoplastic agent that is used in the therapy of selected forms of advanced non-small cell lung cancer (NSCLC). Ceritinib is associated with a moderate rate of serum aminotransferase elevations during therapy and rare instances of clinically apparent acute liver injury. Ceritinib is an orally available inhibitor of the receptor tyrosine kinase activity of anaplastic lymphoma kinase (ALK) with antineoplastic activity. Upon administration, ceritinib binds to and inhibits wild-type ALK kinase, ALK fusion proteins and ALK point mutation variants. Inhibition of ALK leads to both the disruption of ALK-mediated signaling and the inhibition of cell growth in ALK-overexpressing tumor cells. ALK belongs to the insulin receptor superfamily and plays an important role in nervous system development. ALK dysregulation and gene rearrangements are associated with a variety of tumor cell types. Drug Indication Ceritinib is a kinase inhibitor indicated for the treatment of patients with anaplastic lymphoma kinase (ALK)-positive metastatic non-small cell lung cancer (NSCLC) who have progressed on or are intolerant to crizotinib. This indication is approved under accelerated approval based on tumor response rate and duration of response. An improvement in survival or disease-related symptoms has not been established. Continued approval for this indication may be contingent upon verification and description of clinical benefit in confirmatory trials. FDA Label Zykadia is indicated for the treatment of adult patients with anaplastic lymphoma kinase (ALK) positive advanced non small cell lung cancer (NSCLC) previously treated with crizotinib. Mechanism of Action Ceritinib inhibits Anaplastic lymphoma kinase (ALK) also known as ALK tyrosine kinase receptor or CD246 (cluster of differentiation 246), which is an enzyme that in humans is encoded by the ALK gene. About 4-5% of NSCLCs have a chromosomal rearrangement that generates a fusion gene between EML4 (echinoderm microtubule-associated protein-like 4) and ALK (anaplastic lymphoma kinase), which results in constitutive kinase activity that contributes to carcinogenesis and seems to drive the malignant phenotype. Ceritinib exerts its therapeutic effect by inhibiting autophosphorylation of ALK, ALK-mediated phosphorylation of the downstream signaling protein STAT3, and proliferation of ALK-dependent cancer cells. Ceritinib has been shown to inhibit in vitro proliferation of cell lines expressing EML4-ALK and NPM-ALK fusion proteins and demonstrated dose-dependent inhibition of EML4-ALK-positive NSCLC xenograft growth in mice and rats. |
分子式 |
C28H36CLN5O3S
|
---|---|
分子量 |
558.14
|
精确质量 |
557.222
|
元素分析 |
C, 60.25; H, 6.50; Cl, 6.35; N, 12.55; O, 8.60; S, 5.75
|
CAS号 |
1032900-25-6
|
相关CAS号 |
Ceritinib dihydrochloride;1380575-43-8;Ceritinib-d7;1632484-77-5; 032900-25-6; 2055376-76-4; 1380575-43-8 (2HCl); 2055376-74-2 (mesylate); 1190399-48-4 (xHCl)
|
PubChem CID |
57379345
|
外观&性状 |
white solid powder
|
密度 |
1.3±0.1 g/cm3
|
沸点 |
720.7±70.0 °C at 760 mmHg
|
闪点 |
389.6±35.7 °C
|
蒸汽压 |
0.0±2.3 mmHg at 25°C
|
折射率 |
1.595
|
LogP |
5.03
|
tPSA |
116.85
|
氢键供体(HBD)数目 |
3
|
氢键受体(HBA)数目 |
8
|
可旋转键数目(RBC) |
9
|
重原子数目 |
38
|
分子复杂度/Complexity |
835
|
定义原子立体中心数目 |
0
|
SMILES |
CC(C)OC1=CC(C2CCNCC2)=C(C)C=C1NC3=NC=C(Cl)C(NC4=CC=CC=C4S(=O)(C(C)C)=O)=N3
|
InChi Key |
VERWOWGGCGHDQE-UHFFFAOYSA-N
|
InChi Code |
InChI=1S/C28H36ClN5O3S/c1-17(2)37-25-15-21(20-10-12-30-13-11-20)19(5)14-24(25)33-28-31-16-22(29)27(34-28)32-23-8-6-7-9-26(23)38(35,36)18(3)4/h6-9,14-18,20,30H,10-13H2,1-5H3,(H2,31,32,33,34)
|
化学名 |
5-chloro-2-N-(5-methyl-4-piperidin-4-yl-2-propan-2-yloxyphenyl)-4-N-(2-propan-2-ylsulfonylphenyl)pyrimidine-2,4-diamine
|
别名 |
LDK 378; Ceritinib; LDK378; LDK-378; ZYKADIA; LDK-378; NVP-LDK378-NX; 5-chloro-N2-(2-isopropoxy-5-methyl-4-(piperidin-4-yl)phenyl)-N4-(2-(isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine; trade name: Zykadia
|
HS Tariff Code |
2934.99.9001
|
存储方式 |
Powder -20°C 3 years 4°C 2 years In solvent -80°C 6 months -20°C 1 month |
运输条件 |
Room temperature (This product is stable at ambient temperature for a few days during ordinary shipping and time spent in Customs)
|
溶解度 (体外实验) |
|
|||
---|---|---|---|---|
溶解度 (体内实验) |
配方 1 中的溶解度: ≥ 0.5 mg/mL (0.90 mM) (饱和度未知) in 10% DMSO + 40% PEG300 + 5% Tween80 + 45% Saline (这些助溶剂从左到右依次添加,逐一添加), 澄清溶液。
例如,若需制备1 mL的工作液,可将100 μL 5.0 mg/mL澄清DMSO储备液加入400 μL PEG300中,混匀;然后向上述溶液中加入50 μL Tween-80,混匀;加入450 μL生理盐水定容至1 mL。 *生理盐水的制备:将 0.9 g 氯化钠溶解在 100 mL ddH₂O中,得到澄清溶液。 配方 2 中的溶解度: ≥ 0.5 mg/mL (0.90 mM) (饱和度未知) in 10% DMSO + 90% (20% SBE-β-CD in Saline) (这些助溶剂从左到右依次添加,逐一添加), 澄清溶液。 例如,若需制备1 mL的工作液,可将 100 μL 5.0 mg/mL 澄清 DMSO 储备液加入 900 μL 20% SBE-β-CD 生理盐水溶液中,混匀。 *20% SBE-β-CD 生理盐水溶液的制备(4°C,1 周):将 2 g SBE-β-CD 溶解于 10 mL 生理盐水中,得到澄清溶液。 View More
配方 3 中的溶解度: ≥ 0.5 mg/mL (0.90 mM) (饱和度未知) in 10% DMSO + 90% Corn Oil (这些助溶剂从左到右依次添加,逐一添加), 澄清溶液。 配方 4 中的溶解度: 1% DMSO+30% polyethylene glycol+1% Tween 80: 30 mg/mL 1、请先配制澄清的储备液(如:用DMSO配置50 或 100 mg/mL母液(储备液)); 2、取适量母液,按从左到右的顺序依次添加助溶剂,澄清后再加入下一助溶剂。以 下列配方为例说明 (注意此配方只用于说明,并不一定代表此产品 的实际溶解配方): 10% DMSO → 40% PEG300 → 5% Tween-80 → 45% ddH2O (或 saline); 假设最终工作液的体积为 1 mL, 浓度为5 mg/mL: 取 100 μL 50 mg/mL 的澄清 DMSO 储备液加到 400 μL PEG300 中,混合均匀/澄清;向上述体系中加入50 μL Tween-80,混合均匀/澄清;然后继续加入450 μL ddH2O (或 saline)定容至 1 mL; 3、溶剂前显示的百分比是指该溶剂在最终溶液/工作液中的体积所占比例; 4、 如产品在配制过程中出现沉淀/析出,可通过加热(≤50℃)或超声的方式助溶; 5、为保证最佳实验结果,工作液请现配现用! 6、如不确定怎么将母液配置成体内动物实验的工作液,请查看说明书或联系我们; 7、 以上所有助溶剂都可在 Invivochem.cn网站购买。 |
制备储备液 | 1 mg | 5 mg | 10 mg | |
1 mM | 1.7917 mL | 8.9583 mL | 17.9167 mL | |
5 mM | 0.3583 mL | 1.7917 mL | 3.5833 mL | |
10 mM | 0.1792 mL | 0.8958 mL | 1.7917 mL |
1、根据实验需要选择合适的溶剂配制储备液 (母液):对于大多数产品,InvivoChem推荐用DMSO配置母液 (比如:5、10、20mM或者10、20、50 mg/mL浓度),个别水溶性高的产品可直接溶于水。产品在DMSO 、水或其他溶剂中的具体溶解度详见上”溶解度 (体外)”部分;
2、如果您找不到您想要的溶解度信息,或者很难将产品溶解在溶液中,请联系我们;
3、建议使用下列计算器进行相关计算(摩尔浓度计算器、稀释计算器、分子量计算器、重组计算器等);
4、母液配好之后,将其分装到常规用量,并储存在-20°C或-80°C,尽量减少反复冻融循环。
计算结果:
工作液浓度: mg/mL;
DMSO母液配制方法: mg 药物溶于 μL DMSO溶液(母液浓度 mg/mL)。如该浓度超过该批次药物DMSO溶解度,请首先与我们联系。
体内配方配制方法:取 μL DMSO母液,加入 μL PEG300,混匀澄清后加入μL Tween 80,混匀澄清后加入 μL ddH2O,混匀澄清。
(1) 请确保溶液澄清之后,再加入下一种溶剂 (助溶剂) 。可利用涡旋、超声或水浴加热等方法助溶;
(2) 一定要按顺序加入溶剂 (助溶剂) 。
NCT Number | Recruitment | interventions | Conditions | Sponsor/Collaborators | Start Date | Phases |
NCT03501368 | Active Recruiting |
Drug: Ceritinib Drug: Trametinib |
Melanoma Advanced Melanoma |
H. Lee Moffitt Cancer Center and Research Institute |
June 27, 2018 | Phase 1 |
NCT02321501 | Active Recruiting |
Drug: Ceritinib Drug: Everolimus |
ALK Positive ROS1 Gene Rearrangement |
M.D. Anderson Cancer Center | June 22, 2016 | Phase 1 |
NCT03611738 | Active Recruiting |
Drug: Ceritinib Drug: Docetaxel |
Lung Cancer | H. Lee Moffitt Cancer Center and Research Institute |
February 1, 2019 | Phase 1 |
NCT01828099 | Active Recruiting |
Drug: Ceritinib Drug: Pemetrexed |
Non-Small Cell Lung Cancer/td> | Novartis Pharmaceuticals | July 9, 2013 | Phase 3 |
NCT02584933 | Recruiting | Drug: ceritinib | ALK Positive Malignancies | Novartis Pharmaceuticals | December 11, 2015 | Phase 4 |
Dose–response and time course comparison of ALK inhibition by crizotinib or ceritinib.Mol Oncol.2017 Aug;11(8):996-1006. td> |
Tumor volume measurement and MSD®immunoassay quantitation of Th‐ALKF1174L/MYCNtumors following the treatment with crizotinib or ceritinib. Tumor‐bearing animals were treated with either crizotinib (100mg·kg−1per day, orally) or ceritinib (100mg·kg−1per day, orally) for 7days before animal sacrifice and harvesting of tumor tissue samples. td> |
Basal ALK activity in neuroblastoma cell lines. (A) Immunoblots of lysates from neuroblastoma cell line panel, including lysate from Ba/F3ALK F1174Las a positive control for ALK expression and lysate from Hela cells as negative control for ALK expression.Mol Oncol.2017 Aug;11(8):996-1006. td> |