Home>>Signaling Pathways>> Chromatin/Epigenetics>> Epigenetic Reader Domain>>OTX-015

OTX-015 Sale

(Synonyms: OTX 015;OTX015) 目录号 : GC17973

A BRD2, BRD3, and BRD4 inhibitor

OTX-015 Chemical Structure

Cas No.:202590-98-5

规格 价格 库存 购买数量
10mM (in 1mL DMSO)
¥643.00
现货
2mg
¥300.00
现货
5mg
¥594.00
现货
10mg
¥900.00
现货
50mg
¥2,460.00
现货
100mg
¥4,200.00
现货
200mg
¥6,900.00
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

CD4+ T cells

Preparation Method

CD4+ lymphocytes isolated from healthy donors were incubated with prostratin, OTX-015 or OTX-015/prostratin for 48 h and immunostained with anti-CD25, anti-CD69, anti-HLA-DR, anti-CD4, anti-CCR5 or anti-CXCR4 antibodies for 20 min at 4 °C.

Reaction Conditions

5 μM for 48 hours

Applications

Primary CD4+ T cells were treated with OTX-015 for 48 h and evaluated for the expression of CD4, CCR5 and CXCR4 using flow cytometry. OTX-015 treatment did not cause an increase in the expression of these HIV receptors/co-receptors, suggesting that OTX-015 may not pose the risk of increasing the susceptibility of CD4+ T cells to HIV-1 infection during the reactivation of HIV-1 latency.

Animal experiment [2]:

Animal models

Female nude Foxn1 mice 6-week-old

Preparation Method

mice were randomized (nine animals/group) to one of the following experimental groups: vehicle (for OTX-015, water, twice daily, oral; for everolimus vehicle, 5% Tween-80/5% polyethylene glycol 400, thrice weekly, intraperitoneal); 50 mg/kg OTX-015, twice daily, oral; 2 mg/kg everolimus, thrice weekly, intraperitoneal; 50 mg/kg OTX-015 + 2 mg/kg everolimus, according to the single agent dosing schedules.

Dosage form

50 mg/kg OTX-015, twice daily, oral

Applications

OTX-015-treated mice showed a substantial reduction in tumor mass with respect to the control group (p < 0.05) from 7 days after treatment start. The best T/C value was 41.3% recorded on day 23.

References:

[1]. Lu P, et al. The BET inhibitor OTX-015 reactivates latent HIV-1 through P-TEFb. Sci Rep. 2016 Apr 12;6:24100

[2]. Vázquez R, et al. The bromodomain inhibitor OTX-015 (MK-8628) exerts anti-tumor activity in triple-negative breast cancer models as single agent and in combination with RAD001. Oncotarget. 2017 Jan 31;8(5):7598-7613.

产品描述

OTX-015 inhibits the binding of BRD2, BRD3, and BRD4 to acetylated histone 4 in a concentration-dependent manner, with IC50 values from 92-112nM [1].

OTX-015 significant growth inhibition was mesured in six of nine AML cell lines and all four ALL cell lines: HEL IC50 value as 248 nM, NB4 IC50 value as 233 nM, NOMO-1 IC50 value as 229 nM, KG1 IC50 value as 198 nM, OCI-AML3 IC50 value as 60 nM, Kasumi IC50 value as 17 nM, JURKAT IC50 value as 249 nM, BV-173 IC50 value as 161 nM, TOM-1 IC50 value as 133 nM, RS4-11 IC50 value as 34 nM[2]. GI50 values obtained with OTX-015 in vitro in glioblastoma cell lines were equivalent to those achieved in plasma from patients treated at nontoxic doses in the ongoing Phase Ib clinical study in patients with hematologic malignancies, as described in a clinical pharmacokinetics evaluation. OTX-015 displayed higher potency (GI50 618.1 nM) in glioblastoma cell line panel. OTX-015 exerted antiproliferative effects and delayed tumor growth in lymphoma and neuroblastoma cells, together with a downregulation of C-MYC, MYCN and genes associated with superenhancers[3].

OTX-015, offering a significant survival advantage in vivo in the orthotopic human glioblastoma model and slowing tumor progression in the heterotopic model with all administration regimens. Furthermore, no toxicity was seen with any of the OTX-015 dosing regimens, in direct contrast to treatment with temozolomide which induced pronounced weight loss. Synergistic and additive activity of OTX-015 in combination with several antitumor agents currently used to manage GBM patients, improving mouse survival with simultaneous combination of OTX-015 and temozolomide in the absence of toxicity, supporting the incorporation of OTX-015 as an epigenetic modulator in combination with temozolomide in glioblastoma [3].

References:
[1]. J. Kay Noel, et al. Abstract C244: Development of the BET bromodomain inhibitor OTX015. Mol Cancer Ther November 2013 12; C244.
[2]. Marie-Magdelaine Coudé, et al. BET inhibitor OTX015 targets BRD2 and BRD4 and decreases c-MYC in acute leukemia cells. Oncotarget. 2015 Jul 10; 6(19): 17698–17712.
[3]. Berenguer-Daize C, et al. OTX015 (MK-8628), a novel BET inhibitor, displays in vitro and in vivo antitumor effects alone and in combination with conventional therapies in glioblastoma models. Int J Cancer. 2016;139(9):2047–2055.

OTX-015 以浓度依赖性方式抑制 BRD2、BRD3 和 BRD4 与乙酰化组蛋白 4 的结合,IC50 值为 92-112nM [1].

OTX-015 在九个 AML 细胞系中的六个和所有四个 ALL 细胞系中测量到显着的生长抑制:HEL IC50 值为 248 nM,NB4 IC50值为 233 nM,NOMO-1 IC50 值为 229 nM,KG1 IC50 值为 198 nM,OCI-AML3 IC50 值为Kasumi IC50 值为 60 nM,Kasumi IC50 值为 17 nM,JURKAT IC50 值为 249 nM,BV-173 IC50 值为 161 nM , TOM-1 IC50 值为 133 nM, RS4-11 IC50 值为 34 nM[2]。如临床药代动力学评估所述,在胶质母细胞瘤细胞系中体外使用 OTX-015 获得的 GI50 值与在血液系统恶性肿瘤患者中正在进行的 Ib 期临床研究中以无毒剂量治疗的患者血浆中获得的值相同。 OTX-015 在胶质母细胞瘤细胞系组中显示出更高的效力 (GI50 618.1 nM)。 OTX-015 在淋巴瘤和神经母细胞瘤细胞中发挥抗增殖作用并延缓肿瘤生长,同时下调 C-MYC、MYCN 和与超增强子相关的基因[3]

OTX-015,在原位人胶质母细胞瘤模型中提供显着的体内生存优势,并在所有给药方案的异位模型中减缓肿瘤进展。此外,在任何 OTX-015 给药方案中都没有发现毒性,这与使用替莫唑胺治疗导致明显的体重减轻形成鲜明对比。 OTX-015 与目前用于治疗 GBM 患者的几种抗肿瘤药物的协同和相加活性,在没有毒性的情况下同时结合 OTX-015 和替莫唑胺提高小鼠存活率,支持将 OTX-015 作为表观遗传调节剂纳入与替莫唑胺联合治疗胶质母细胞瘤[3]

Chemical Properties

Cas No. 202590-98-5 SDF
别名 OTX 015;OTX015
Canonical SMILES CC1=C(SC2=C1C(=NC(C3=NN=C(N32)C)CC(=O)NC4=CC=C(C=C4)O)C5=CC=C(C=C5)Cl)C
分子式 C25H22ClN5O2S 分子量 491.99
溶解度 ≥ 24.6 mg/mL in DMSO, ≥ 106 mg/mL in EtOH with gentle warming 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 2.0326 mL 10.1628 mL 20.3256 mL
5 mM 0.4065 mL 2.0326 mL 4.0651 mL
10 mM 0.2033 mL 1.0163 mL 2.0326 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

Potent BRD4 inhibitor suppresses cancer cell-macrophage interaction

Nat Commun2020 Apr 14;11(1):1833.PMID: 32286255DOI: 10.1038/s41467-020-15290-0

Small molecule inhibitor of the bromodomain and extraterminal domain (BET) family proteins is a promising option for cancer treatment. However, current BET inhibitors are limited by their potency or oral bioavailability. Here we report the discovery and characterization of NHWD-870, a BET inhibitor that is more potent than three major clinical stage BET inhibitors BMS-986158, OTX-015, and GSK-525762. NHWD-870 causes tumor shrinkage or significantly suppresses tumor growth in nine xenograft or syngeneic models. In addition to its ability to downregulate c-MYC and directly inhibit tumor cell proliferation, NHWD-870 blocks the proliferation of tumor associated macrophages (TAMs) through multiple mechanisms, partly by reducing the expression and secretion of macrophage colony-stimulating factor CSF1 by tumor cells. NHWD-870 inhibits CSF1 expression through suppressing BRD4 and its target HIF1α. Taken together, these results reveal a mechanism by which BRD4 inhibition suppresses tumor growth, and support further development of NHWD-870 to treat solid tumors.

Synergistic efficacy of inhibiting MYCN and mTOR signaling against neuroblastoma

BMC Cancer2021 Sep 26;21(1):1061.PMID: 34565342DOI: 10.1186/s12885-021-08782-9

Background: Neuroblastoma (NB) patients with MYCN amplification or overexpression respond poorly to current therapies and exhibit extremely poor clinical outcomes. PI3K-mTOR signaling-driven deregulation of protein synthesis is very common in NB and various other cancers that promote MYCN stabilization. In addition, both the MYCN and mTOR signaling axes can directly regulate a common translation pathway that leads to increased protein synthesis and cell proliferation. However, a strategy of concurrently targeting MYCN and mTOR signaling in NB remains unexplored. This study aimed to investigate the therapeutic potential of targeting dysregulated protein synthesis pathways by inhibiting the MYCN and mTOR pathways together in NB.
Methods: Using small molecule/pharmacologic approaches, we evaluated the effects of combined inhibition of MYCN transcription and mTOR signaling on NB cell growth/survival and associated molecular mechanism(s) in NB cell lines. We used two well-established BET (bromodomain extra-terminal) protein inhibitors (JQ1, OTX-015), and a clinically relevant mTOR inhibitor, temsirolimus, to target MYCN transcription and mTOR signaling, respectively. The single agent and combined efficacies of these inhibitors on NB cell growth, apoptosis, cell cycle and neurospheres were assessed using MTT, Annexin-V, propidium-iodide staining and sphere assays, respectively. Effects of inhibitors on global protein synthesis were quantified using a fluorescence-based (FamAzide)-based protein synthesis assay. Further, we investigated the specificities of these inhibitors in targeting the associated pathways/molecules using western blot analyses.
Results: Co-treatment of JQ1 or OTX-015 with temsirolimus synergistically suppressed NB cell growth/survival by inducing G1 cell cycle arrest and apoptosis with greatest efficacy in MYCN-amplified NB cells. Mechanistically, the co-treatment of JQ1 or OTX-015 with temsirolimus significantly downregulated the expression levels of phosphorylated 4EBP1/p70-S6K/eIF4E (mTOR components) and BRD4 (BET protein)/MYCN proteins. Further, this combination significantly inhibited global protein synthesis, compared to single agents. Our findings also demonstrated that both JQ1 and temsirolimus chemosensitized NB cells when tested in combination with cisplatin chemotherapy.
Conclusions: Together, our findings demonstrate synergistic efficacy of JQ1 or OTX-015 and temsirolimus against MYCN-driven NB, by dual-inhibition of MYCN (targeting transcription) and mTOR (targeting translation). Additional preclinical evaluation is warranted to determine the clinical utility of targeted therapy for high-risk NB patients.

BET inhibitor suppresses migration of human hepatocellular carcinoma by inhibiting SMARCA4

Sci Rep2021 Jun 3;11(1):11799.PMID: 34083693DOI: 10.1038/s41598-021-91284-2

Hepatocellular carcinoma (HCC) is one of the most prevalent and poorly responsive cancers worldwide. Bromodomain and extraterminal (BET) inhibitors, such as JQ1 and OTX-015, inhibit BET protein binding to acetylated residues in histones. However, the physiological mechanisms and regulatory processes of BET inhibition in HCC remain unclear. To explore BET inhibitors' potential role in the molecular mechanisms underlying their anticancer effects in HCC, we analyzed BET inhibitor-treated HCC cells' gene expression profiles with RNA-seq and bioinformatics analysis. BET inhibitor treatment significantly downregulated genes related to bromodomain-containing proteins 4 (BRD4), such as ACSL5, SLC38A5, and ICAM2. Importantly, some cell migration-related genes, including AOC3, CCR6, SSTR5, and SCL7A11, were significantly downregulated. Additionally, bioinformatics analysis using Ingenuity Knowledge Base Ingenuity Pathway Analysis (IPA) revealed that SMARCA4 regulated migration response molecules. Furthermore, knockdown of SMARCA4 gene expression by siRNA treatment significantly reduced cell migration and the expression of migration-related genes. In summary, our results indicated that BET inhibitor treatment in HCC cell lines reduces cell migration through the downregulation of SMARCA4.

Safety and Efficacy of Bromodomain and Extra-Terminal Inhibitors for the Treatment of Hematological Malignancies and Solid Tumors: A Systematic Study of Clinical Trials

Front Pharmacol2021 Jan 26;11:621093.PMID: 33574760DOI: 10.3389/fphar.2020.621093

Background: The upregulated expression of BET proteins is closely associated with the occurrence and development of hematological malignancies and solid tumors. Several BET inhibitors have been developed, and some have been in phase I/II of clinical trials. Here, the safety, efficacy, and pharmacodynamics of ten BET inhibitors currently in clinical trials were evaluated. Methods: We retrieved and reviewed published reports on the clinical trials of twelve BET inhibitors including AZD5153, ABBV-075, BMS-986158, CPI-0610, GSK525762, OTX-015, PLX51107, INCB054329, INCB057643, FT-1101, CC-90010, and ODM-207 for patients with hematological malignancies and solid tumors and summarized their published target genes. Results: In the monotherapy of BET inhibitors, the most common and severe (grade ≥3) hematological adverse events (AEs) are thrombocytopenia, anemia, and neutropenia. The most common non-hematological syndromes are diarrhea, nausea, fatigue, dysgeusia, and decreased appetite, while the most severe AE is pneumonia. Additionally, T max of these BET inhibitors was between 0.5-6 h, but the range for T 1/2 varied significantly. According to published data, the rates of SD, PD, CR and PR were 27.4%, 37.6%, 3.5%, and 5.7%, respectively, which is not very satisfactory. In addition to BRD4, oncogene MYC is another common target gene of these BET inhibitors. Ninety-seven signaling pathways may be regulated by BET inhibitors. Conclusion: All BET inhibitors reviewed in our study exhibited exposure-dependent thrombocytopenia, which may limit their clinical application. Moreover, further efforts are necessary to explore the optimal dosing schemes and combinations to maximize the efficacy of BET inhibitors.

Dual Inhibition of Histone Deacetylases and the Mechanistic Target of Rapamycin Promotes Apoptosis in Cell Line Models of Uveal Melanoma

Invest Ophthalmol Vis Sci2021 Sep 2;62(12):16.PMID: 34533562DOI: 10.1167/iovs.62.12.16

Purpose: Over 90% of uveal melanomas harbor pathogenic variants of the GNAQ or GNA11 genes that activate survival pathways. As previous studies found that Ras-mutated cell lines were vulnerable to a combination of survival pathway inhibitors and the histone-deacetylase inhibitor romidepsin, we investigated whether this combination would be effective in models of uveal melanoma.
Methods: A small-scale screen of inhibitors of bromodomain-containing protein 4 (BRD4; OTX-015), extracellular signal-related kinase (ERK; ulixertinib), mechanistic target of rapamycin (mTOR; AZD-8055), or phosphoinositide 3-kinase (PI3K; GDC-0941) combined with a clinically relevant administration of romidepsin was performed on a panel of uveal melanoma cell lines (92.1, Mel202, MP38, and MP41) and apoptosis was quantified by flow cytometry after 48 hours. RNA sequencing analysis was performed on Mel202 cells treated with romidepsin alone, AZD-8055 alone, or the combination, and protein changes were validated by immunoblot.
Results: AZD-8055 with romidepsin was the most effective combination in inducing apoptosis in the cell lines. Increased caspase-3 and PARP cleavage were noted in the cell lines when they were treated with romidepsin and mTOR inhibitors. RNA sequencing analysis of Mel202 cells revealed that apoptosis was the most affected pathway in the romidepsin/AZD-8055-treated cells. Increases in pro-apoptotic BCL2L11 and decreases in anti-apoptotic BIRC5 and BCL2L1 transcripts noted in the sequencing analysis were confirmed at the protein level in Mel202 cells.
Conclusions: Our data suggest that romidepsin in combination with mTOR inhibition could be an effective treatment strategy against uveal melanoma due in part to changes in apoptotic proteins.