Home>>Signaling Pathways>> Apoptosis>> Other Apoptosis>>17-AAG (KOS953)

17-AAG (KOS953) Sale

(Synonyms: 坦螺旋霉素; 17-AAG; NSC 330507; CP 127374) 目录号 : GC11720

An inhibitor of Hsp90

17-AAG (KOS953) Chemical Structure

Cas No.:75747-14-7

规格 价格 库存 购买数量
10mM (in 1mL DMSO)
¥483.00
现货
10mg
¥462.00
现货
50mg
¥1,313.00
现货
100mg
¥1,722.00
现货
200mg
¥2,552.00
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

Cell experiment [1]:

Cell lines

HO-8910 cells

Preparation Method

HO-8910 cells were treated with different concentrations of Hsp90 inhibitor 17-AAG, and cells treated without 17-AAG were regarded as the control group.

Reaction Conditions

17-AAG 0.25、0.5 and 5 μM for 4, 8, 12, and 24 h

Applications

The levels of Rad50 and Ku80 decreased after 8 h in cells treated with relative low concentration of 17-AAG (0.5 μM). In the groups treated with relative high concentration of 17-AAG (5 μM), Rad50 significantly decreased after 4 h, and Ku80 also decreased after 8 h, indicating that DSBs repair proteins were downregulated in a dose and time-depend manner and Hsp90 inhibition could affect DSBs repair.

Animal experiment [2]:

Animal models

Female 4–5weeks old, 18–20 g BALB/c nude mice

Preparation Method

The mice were randomly assigned to three groups (n = 4/group). The luciferase-expressing HO-8910 cells were injected intraperitoneally into groups. After two weeks, mice were intraperitoneally injected with DDP, DDP combined with 17-AAG (50 mg/kg),once a week for up to 6 weeks.

Dosage form

50 mg/kg, once a week for up to 6 weeks

Applications

A combination of 17-AAG and DDP could inhibit tumor growth more efficiently, The survival time of the mice was significantly prolonged, supporting the notion that 17-AAG combined with DDP could prolong the survival time of mice.

References:

[1]. Wang Y, Chen Q, et,al. Lamin-A interacting protein Hsp90 is required for DNA damage repair and chemoresistance of ovarian cancer cells. Cell Death Dis. 2021 Aug 12;12(8):786. doi: 10.1038/s41419-021-04074-z. PMID: 34381017; PMCID: PMC8358027.

产品描述

17-AAG(Geldanamycin), a natural benzoquinone ansamycin antibiotic, is the first established inhibitor of Hsp90. It inhibits Hsp90's ATPase function through binding to its amino-terminal domain[5].

17-AAG as a potent HSP90 inhibitor with an IC50 value of 6 nM in BT474 cells, inhibited the binding of HSP90 to HIF-1α[2]. In a concentration- and time-dependent manner, inhibition of Hsp90 by 17-AAG decreased Akt and eNOS expression. Inhibition of eNOS expression by 17-AAG occurred at the transcriptional level. Furthermore, treatment with 17-AAG decreased basal and vascular endothelial growth factor-stimulated Akt and eNOS phosphorylation. This corresponded with decreased NO production and inhibition of endothelial cell migration and angiogenesis[3].17AAG topically applied to mouse skin, inhibits UVR-induced development of cutaneous squamous cell carcinoma (SCC) [7].

Therapeutic potential of 17-AAG was investigated using nude mice intraperitoneal xenograft. A combination of 17-AAG and DDP could inhibit tumor growth more efficiently, The survival time of the mice was significantly prolonged, supporting the notion that 17-AAG combined with DDP could prolong the survival time of mice[1].17-AAG is able to inhibit the growth of both human glioma cell lines and glioma stem cells in vitro . In addition, 17-AAG can inhibit the growth of intracranial tumors and can synergize with radiation both in tissue culture and in intracranial tumors. This compound was not found to synergize with temozolomide in any of our models of gliomas[6].17-AAG plus trastuzumab is well tolerated and has antitumor activity in patients with HER-2+ breast cancer whose tumors have progressed during treatment with trastuzumab. These data suggest that Hsp90 function can be inhibited in vivo to a degree sufficient to cause inhibition of tumor growth[4].

References:
[1]: Wang Y, Chen Q, et,al. Lamin-A interacting protein Hsp90 is required for DNA damage repair and chemoresistance of ovarian cancer cells. Cell Death Dis. 2021 Aug 12;12(8):786. doi: 10.1038/s41419-021-04074-z. PMID: 34381017; PMCID: PMC8358027.
[2]: Kamal A, Thao L, et,al. A high-affinity conformation of Hsp90 confers tumour selectivity on Hsp90 inhibitors. Nature. 2003 Sep 25;425(6956):407-10. doi: 10.1038/nature01913. PMID: 14508491.
[3]: Sun J, Liao JK. Induction of angiogenesis by heat shock protein 90 mediated by protein kinase Akt and endothelial nitric oxide synthase. Arterioscler Thromb Vasc Biol. 2004 Dec;24(12):2238-44. doi: 10.1161/01.ATV.0000147894.22300.4c. Epub 2004 Oct 14. PMID: 15486309; PMCID: PMC2633590.
[4]: Modi S, Stopeck AT, et,al. Combination of trastuzumab and tanespimycin (17-AAG, KOS-953) is safe and active in trastuzumab-refractory HER-2 overexpressing breast cancer: a phase I dose-escalation study. J Clin Oncol. 2007 Dec 1;25(34):5410-7. doi: 10.1200/JCO.2007.11.7960. PMID: 18048823.
[5]: Neckers L, Schulte TW, et,al. Geldanamycin as a potential anti-cancer agent: its molecular target and biochemical activity. Invest New Drugs. 1999;17(4):361-73. doi: 10.1023/a:1006382320697. PMID: 10759403.
[6]: Sauvageot CM, Weatherbee JL, et,al. Efficacy of the HSP90 inhibitor 17-AAG in human glioma cell lines and tumorigenic glioma stem cells. Neuro Oncol. 2009 Apr;11(2):109-21. doi: 10.1215/15228517-2008-060. Epub 2008 Aug 5. PMID: 18682579; PMCID: PMC2718982.
[7]: Singh A, Singh A, et,al. Topically applied Hsp90 inhibitor 17AAG inhibits UVR-induced cutaneous squamous cell carcinomas. J Invest Dermatol. 2015 Apr;135(4):1098-1107. doi: 10.1038/jid.2014.460. Epub 2014 Oct 22. PMID: 25337691; PMCID: PMC4366283.

17-AAG(格尔德霉素)是一种天然的苯醌安沙霉素抗生素,是第一个确定的 Hsp90 抑制剂。它通过与其氨基末端结构域结合来抑制 Hsp90 的 ATPase 功能[5]

17-AAG 作为一种有效的 HSP90 抑制剂,在 BT474 细胞中的 IC50 值为 6 nM,可抑制 HSP90 与 HIF-1α[2] 的结合。以浓度和时间依赖性方式,17-AAG 对 Hsp90 的抑制作用降低了 Akt 和 eNOS 的表达。 17-AAG 对 eNOS 表达的抑制发生在转录水平。此外,用 17-AAG 治疗可降低基础和血管内皮生长因子刺激的 Akt 和 eNOS 磷酸化。这对应于 NO 生成减少以及内皮细胞迁移和血管生成的抑制[3]。17AAG 局部应用于小鼠皮肤,抑制 UVR 诱导的皮肤鳞状细胞癌 (SCC) 的发展[7 ].

使用裸鼠腹膜内异种移植物研究了 17-AAG 的治疗潜力。 17-AAG与DDP联用可更有效地抑制肿瘤生长,小鼠的生存时间明显延长,支持17-AAG与DDP联用可延长小鼠生存时间的观点[1].17-AAG 能够在体外抑制人神经胶质瘤细胞系和神经胶质瘤干细胞的生长。此外,17-AAG 可抑制颅内肿瘤的生长,并可在组织培养和颅内肿瘤中与放疗协同作用。在我们的任何神经胶质瘤模型中均未发现该化合物与替莫唑胺有协同作用[6]。17-AAG 加曲妥珠单抗耐受性良好,在 HER-2 患者中具有抗肿瘤活性+< /sup> 曲妥珠单抗治疗期间肿瘤进展的乳腺癌。这些数据表明,Hsp90 功能可在体内被抑制到足以抑制肿瘤生长的程度[4]

Chemical Properties

Cas No. 75747-14-7 SDF
别名 坦螺旋霉素; 17-AAG; NSC 330507; CP 127374
化学名 [(3R,5S,6R,7S,8E,10S,11S,12Z,14E)-6-hydroxy-5,11-dimethoxy-3,7,9,15-tetramethyl-16,20,22-trioxo-21-(prop-2-enylamino)-17-azabicyclo[16.3.1]docosa-1(21),8,12,14,18-pentaen-10-yl] carbamate
Canonical SMILES CC1CC(C(C(C=C(C(C(C=CC=C(C(=O)NC2=CC(=O)C(=C(C1)C2=O)NCC=C)C)OC)OC(=O)N)C)C)O)OC
分子式 C31H43N3O8 分子量 585.7
溶解度 ≥ 24.95 mg/mL in DMSO, ≥ 9.56 mg/mL in EtOH with ultrasonic 储存条件 4°C, protect from light
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 1.7074 mL 8.5368 mL 17.0736 mL
5 mM 0.3415 mL 1.7074 mL 3.4147 mL
10 mM 0.1707 mL 0.8537 mL 1.7074 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

Berberine regulates the Notch1/PTEN/PI3K/AKT/mTOR pathway and acts synergistically with 17-AAG and SAHA in SW480 colon cancer cells

Pharm Biol2021 Dec;59(1):21-30.PMID: 33417512DOI: 10.1080/13880209.2020.1865407

Context: Berberine (BBR) is used to treat diarrhoea and gastroenteritis in the clinic. It was found to have anticolon cancer effects.
Objective: To study the anticolon cancer mechanism of BBR by connectivity map (CMAP) analysis.
Materials and methods: CMAP based mechanistic prediction was conducted by comparing gene expression profiles of 10 μM BBR treated MCF-7 cells with that of clinical drugs such as helveticoside, ianatoside C, pyrvinium, gossypol and trifluoperazine. The treatment time was 12 h and two biological replications were performed. The DMSO-treated cells were selected as a control. The interaction between 100 μM BBR and target protein was measured by cellular thermal shift assay. The protein expression of 1-9 μM BBR treated SW480 cells were measured by WB assay. Apoptosis, cell cycle arrest, mitochondrial membrane potential (MMP) of 1-9 μM BBR treated SW480 cells were measured by flow cytometry and Hoechst 33342 staining methods.
Results: CMAP analysis found 14 Hsp90, HDAC, PI3K or mTOR protein inhibitors have similar functions with BBR. The experiments showed that BBR inhibited SW480 cells proliferation with IC50 of 3.436 μM, induced apoptosis, autophage, MMP depolarization and arrested G1 phase of cell cycle at 1.0 μM. BBR dose-dependently up-regulated PTEN, while inhibited Notch1, PI3K, Akt and mTOR proteins at 1.0-9.0 μM (p < 0.05). BBR also acted synergistically with Hsp90 and HDAC inhibitor (0.01 μM) in SW480 cells at 0.5 and 1.0 μM.
Discussion and conclusions: The integrative gene expression-based chemical genomic method using CMAP analysis may be applicable for mechanistic studies of other multi-targets drugs.

Spotlight on 17-AAG as an Hsp90 inhibitor for molecular targeted cancer treatment

Chem Biol Drug Des2019 May;93(5):760-786.PMID: 30697932DOI: 10.1111/cbdd.13486

Hsp90 is a ubiquitous chaperone with important roles in the organization and maturation of client proteins that are involved in the progression and survival of cancer cells. Multiple oncogenic pathways can be affected by inhibition of Hsp90 function through degradation of its client proteins. That makes Hsp90 a therapeutic target for cancer treatment. 17-allylamino-17-demethoxy-geldanamycin (17-AAG) is a potent Hsp90 inhibitor that binds to Hsp90 and inhibits its chaperoning function, which results in the degradation of Hsp90's client proteins. There have been several preclinical studies of 17-AAG as a single agent or in combination with other anticancer agents for a wide range of human cancers. Data from various phases of clinical trials show that 17-AAG can be given safely at biologically active dosages with mild toxicity. Even though 17-AAG has suitable pharmacological potency, its low water solubility and high hepatotoxicity could significantly restrict its clinical use. Nanomaterials-based drug delivery carriers may overcome these drawbacks. In this paper, we review preclinical and clinical research on 17-AAG as a single agent and in combination with other anticancer agents. In addition, we highlight the potential of using nanocarriers and nanocombination therapy to improve therapeutic effects of 17-AAG.

Effects of 17-AAG on the RIP1/RIP3/MLKL pathway during the development of heart failure following myocardial infarction in rats

J Pharmacol Sci2021 Oct;147(2):192-199.PMID: 34384567DOI: 10.1016/j.jphs.2021.06.009

In a previous study, we suggested that the Hsp90 inhibitor 17-AAG prevents cardiac dysfunction in the failing heart following myocardial infarction in rats. Although it is assumed that the RIP1/RIP3/MLKL necroptotic pathway, which comprises client proteins for Hsp90, is involved; however, the relationship between the cardioprotective effects of 17-AAG and the activity of the cardiac RIP1/RIP3/MLKL necrosome-associated proteins in the failing heart following myocardial infarction remained unclear. Therefore, the levels of phosphorylated MLKL after myocardial infarction with or without Hsp90 inhibitor treatment were measured. Myocardial infarction was induced by ligation of the coronary artery (CAL) in Wistar rats. 17-AAG was injected from the 2nd to the 8th week after myocardial infarction. The administration of 17-AAG attenuated the cardiac dysfunction, hypertrophy, and fibrosis at the 8th week after CAL, simultaneously lessening the increases in the expression and phosphorylation levels of RIP1, RIP3, and MLKL in the area of the left ventricular muscle without infarct. These results indicate that the activation of the RIP1/RIP3/MLKL pathway is a common event in the development of chronic heart failure. Furthermore, our findings suggest that the effects of 17-AAG treatment on the improvement of cardiac function in rats after myocardial infarction is related to the attenuation of this RIP1/RIP3/MLKL pathway.

17 AAG for HSP90 inhibition in cancer--from bench to bedside

Curr Mol Med2009 Jun;9(5):654-64.PMID: 19601813DOI: 10.2174/156652409788488757

Heat shock protein 90 (HSP90) family of proteins are ubiquitous molecular chaperones that are involved in folding, activation, maturation and assembly of many proteins that include essential mediators of signal transduction and cell cycle progression. They are abundant in eukaryotic cells and localized to the cytoplasm, mitochondria as well as the endoplasmic reticulum under normal conditions, making up 1-2% of all cellular proteins. HSP90 proteins have increased expression in a number of malignancies. A large number of HSP90 client proteins have been shown to be necessary for the development, proliferation and survival of specific types of cancers. HSP90 inhibition can affect multiple oncogenic pathways and involved proteins, therefore make it an attractive target for drug development. This article serves as an overview of the pre-clinical data and clinical trial data on HSP90 inhibitor 17-AAG in different malignancies. 17-AAG has shown significant anti-tumor activity against a spectrum of cancers in the pre-clinical studies and information from various phases of clinical trials is growing. The potential indication of 17-AGG for the treatment of refractory multiple myeloma now awaits for the results of two phase III studies. More work needs to be done before the broader oncological use of HSP90 inhibitors in the area of defining HSP90 client proteins, understanding the mechanism of HSP90 actions, identifying reliable surrogate markers for HSP90 inhibition in vivo and optimizing drug delivery and efficacy.

17‑AAG synergizes with Belinostat to exhibit a negative effect on the proliferation and invasion of MDA‑MB‑231 breast cancer cells

Oncol Rep2020 Jun;43(6):1928-1944.PMID: 32236631DOI: 10.3892/or.2020.7563

Breast cancer is one of the most common malignancies that threaten the health of women. Although there are a few chemotherapies for the clinical treatment of breast cancer, these therapies are faced with the problems of drug‑resistance and metastasis. Drug combination can help to reduce the adverse side effects of chemotherapies using single drugs, and also help to overcome common drug‑resistance during clinical treatment of breast cancer. The present study reported the synergistic effect of the heat shock protein 90 inhibitor 17‑AAG and the histone deacetylase 6 inhibitor Belinostat in triple‑negative breast cancer (TNBC) MDA‑MB‑231 cells, by detection of proliferation, apoptosis and cell cycle arrest following treatment with this combination. Subsequently, RNA sequencing (RNA‑seq) data was collected and analyzed to investigate the synergistic mechanism of this combination. Based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathways revealed by RNA‑seq data analysis, a wound‑healing assay was used to investigate the effect of this combination on the migration of MDA‑MB‑231 cells. Compared with treatment with 17‑AAG or Belinostat alone, both the viability inhibition and apoptosis rate of MDA‑MB‑231 cells were significantly enhanced in the combination group. The combination index values were <1 in three concentration groups. Revealed by the RNA‑seq data analysis, the most significantly enriched KEGG pathways in the combination group were closely associated with cell migration. Based on these findings, the anti‑migration effect of this combination was investigated. It was revealed that the migration of MDA‑MB‑231 cells was significantly suppressed in the combination group compared with in the groups treated with 17‑AAG or Belinostat alone. In terms of specific genes, the mRNA expression levels of TEA domain family proteins were significantly decreased in the combination group, whereas the phosphorylation of YY1 associated protein 1 and modulator of VRAC current 1 was significantly enhanced in the combination group. These alterations may help to explain the anti‑migration effect of this combination. Belinostat has already been approved as a treatment for T‑cell lymphoma and 17‑AAG is undergoing clinical trials. These findings could provide a beneficial reference for the clinical treatment of patients with TNBC.