Home>>Signaling Pathways>> DNA Damage/DNA Repair>> Checkpoint Kinase (Chk)>>LY2606368

LY2606368 Sale

(Synonyms: 5-[[5-[2-(3-氨基丙氧基)-6-甲氧基苯基]-1H-吡唑-3-基]氨基]-2-吡嗪甲腈,LY2606368) 目录号 : GC15663

A Chk1 inhibitor

LY2606368 Chemical Structure

Cas No.:1234015-52-1

规格 价格 库存 购买数量
5mg
¥1,596.00
现货
25mg
¥5,166.00
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

Kinase experiment [1]:

Preparation Method

Purified CHK1 was incubated with LY2606368 and substrate was added to detect whether LY2606368 was inhibited.

Reaction Conditions

10-6-10nM LY2606368

Applications

LY2606368 is an ATP-competitive protein kinase inhibitor with a Ki of 0.9 nmol/L against purified CHK1.

Cell experiment [2]:

Cell lines

HeLa cells

Preparation Method

Cells were treated with LY2606368 for 7 hours and stained for evidence of DNA DSB using both TUNEL and an antibody for H2AX phosphorylated on serine 139.

Reaction Conditions

33 nmol/L LY2606368 for 7 hours

Applications

LY2606368 triggered S-phase DNA damage, such as pH2AX (S139) and TUNEL positive staining cells significantly increased in S-phase cells. LY2606368 also requires CDC25A and CDK2 to trigger DNA damage. In addition, LY2606368 causes replication catastrophe.

Animal experiment [3]:

Animal models

Female CD-1nu-/nu mice carrying CALU-6 tumors (26-28 g)

Preparation Method

Vehicle consisting of 20% Captisol (CyDex Inc) pH4 or LY2606368 was administered by subcutaneous injection in a volume of 200 ml(10 mmol/L). 4,8, 12, 24, and 48 hours after drug administration, blood for plasma drug exposure was extracted

Dosage form

1, 3.3, or 10 mg/kg LY2606368 twice daily for 2 days

Applications

Up to 72.3% inhibition of tumor growth was observed in the three doses of LY2606368 tested, and mice lost no more than 3% of their body weight, indicating that LY2606368 was well tolerated in any treatment group. In addition, tumor regrowth was slow in the highest dose group during the 28-day recovery period, indicating a durable tumor response to LY2606368.

References:

[1]. King C, Diaz HB, et,al. LY2606368 Causes Replication Catastrophe and Antitumor Effects through CHK1-Dependent Mechanisms. Mol Cancer Ther. 2015 Sep;14(9):2004-13. doi: 10.1158/1535-7163.MCT-14-1037. Epub 2015 Jul 3. PMID: 26141948.

产品描述

LY2606368, an ATP-competitive CHK1 inhibitor, is currently in the clinical stage with a Ki of 0.9 nM and an IC50 of <1 nM. LY2606368 inhibits CHK2 (IC50=8 nM) and RSK1 (IC50=9 nM)[2].

Treatment of cells with LY2606368 results in the rapid appearance of TUNEL and pH2AX-positive double-stranded DNA breaks in the S-phase cell population. Inhibition of apoptosis by the caspase inhibitor Z-VAD-FMK had no effect on chromosome fragmentation, indicating that LY2606368 causes replication catastrophe[1].LY2606368 triggered S-phase DNA damage, such as pH2AX (S139) and TUNEL positive staining cells significantly increased in S-phase cells. LY2606368 also requires CDC25A and CDK2 to trigger DNA damage. In addition, LY2606368 causes replication catastrophe[1]. In primary patient-derived osteosarcoma cells, LY2606368 alone results in strongly reduced clonogenic survival at low nanomolar concentrations and acts by affecting cell cycle progression, induction of apoptosis and induction of double-stranded DNA breakage at concentrations that are well below clinically tolerable and safe plasma concentrations[3]. To investigate the combined effect of the Chk1 inhibitor LY2606368and antitumor drugs (GEM and S 1) on pancreatic cancer cell line SUIT 2. Acombination of LY2606368 and GS showed effective induction of apoptosis[4]. HGSOC cell lines were also sensitive to LY2606368, associated with induction of DNA damage and replication stress. LY2606368 also sensitized these cell lines to PARP inhibition and compromised both HR repair and replication fork stability[5]. BRCAwt HGSOC develops resistance to LY2606368 monotherapy via a prolonged G2 delay induced by lower CDK1/CyclinB1 activity, thus preventing cells from mitotic catastrophe and cell death[6]. PLK1 or CHEK1 inhibitors (BI-2536 or LY2606368) were found to exert a superior anticancer effect against cell lines at low nanomolar concentrations and induce cell-cycle arrest[7].

Up to 72.3% inhibition of tumor growth was observed in the three doses of LY2606368 tested, and mice lost no more than 3% of their body weight, indicating that LY2606368 was well tolerated in any treatment group. In addition, tumor regrowth was slow in the highest dose group during the 28-day recovery period, indicating a durable tumor response to LY2606368[1].

References:
[1]: King C, Diaz HB,et,al. Barda D, Marshall MS. LY2606368 Causes Replication Catastrophe and Antitumor Effects through CHK1-Dependent Mechanisms. Mol Cancer Ther. 2015 Sep;14(9):2004-13. doi: 10.1158/1535-7163.MCT-14-1037. Epub 2015 Jul 3. PMID: 26141948.
[2]: Yin Y, Shen Q, et,al. Chk1 inhibition potentiates the therapeutic efficacy of PARP inhibitor BMN673 in gastric cancer. Am J Cancer Res. 2017 Mar 1;7(3):473-483. PMID: 28401005; PMCID: PMC5385637.
[3]: Heidler CL, Roth EK, et,al. Prexasertib (LY2606368) reduces clonogenic survival by inducing apoptosis in primary patient-derived osteosarcoma cells and synergizes with cisplatin and talazoparib. Int J Cancer. 2020 Aug 15;147(4):1059-1070. doi: 10.1002/ijc.32814. Epub 2019 Dec 19. PMID: 31782150; PMCID: PMC7384073.
[4]: Morimoto Y, Takada K, et,al. Prexasertib increases the sensitivity of pancreatic cancer cells to gemcitabine and S?1. Oncol Rep. 2020 Feb;43(2):689-699. doi: 10.3892/or.2019.7421. Epub 2019 Nov 28. PMID: 31789403.
[5]: Parmar K, Kochupurakkal BS, et,al. The CHK1 Inhibitor Prexasertib Exhibits Monotherapy Activity in High-Grade Serous Ovarian Cancer Models and Sensitizes to PARP Inhibition. Clin Cancer Res. 2019 Oct 15;25(20):6127-6140. doi: 10.1158/1078-0432.CCR-19-0448. Epub 2019 Aug 13. PMID: 31409614; PMCID: PMC6801076.
[6]: Nair J, Huang TT, et,al. Resistance to the CHK1 inhibitor prexasertib involves functionally distinct CHK1 activities in BRCA wild-type ovarian cancer. Oncogene. 2020 Aug;39(33):5520-5535. doi: 10.1038/s41388-020-1383-4. Epub 2020 Jul 9. PMID: 32647134; PMCID: PMC7426265.
[7]: Yoshida K, Yokoi A, et,al. Aberrant Activation of Cell-Cycle-Related Kinases and the Potential Therapeutic Impact of PLK1 or CHEK1 Inhibition in Uterine Leiomyosarcoma. Clin Cancer Res. 2022 May 13;28(10):2147-2159. doi: 10.1158/1078-0432.CCR-22-0100. PMID: 35302600.

LY2606368 是一种 ATP 竞争性 CHK1 抑制剂,目前处于临床阶段,Ki 为 0.9 nM,IC50 <1 nM。 LY2606368 抑制 CHK2 (IC50=8 nM) 和 RSK1 (IC50=9 nM)[2]

用 LY2606368 处理细胞导致 TUNEL 和 pH2AX- 快速出现S 期细胞群中的阳性双链 DNA 断裂。半胱天冬酶抑制剂 Z-VAD-FMK 对细胞凋亡的抑制对染色体断裂没有影响,表明 LY2606368 导致复制灾难[1]。LY2606368 引发 S 期 DNA 损伤,如 pH2AX (S139) S期细胞中TUNEL阳性染色细胞明显增多。 LY2606368 还需要 CDC25A 和 CDK2 来触发 DNA 损伤。此外,LY2606368 会导致复制灾难[1]。在原发性患者来源的骨肉瘤细胞中,LY2606368 单独导致低纳摩尔浓度下的克隆形成存活率大大降低,并通过影响细胞周期进程、诱导细胞凋亡和诱导双链 DNA 断裂而起作用,浓度远低于临床可耐受和安全的血浆浓度[3]。研究Chk1抑制剂LY2606368和抗肿瘤药物(GEM和S 1)联合作用对胰腺癌细胞系SUIT 2的影响。LY2606368和GS联用显示出有效诱导细胞凋亡[4]。 HGSOC 细胞系也对 LY2606368 敏感,与 DNA 损伤和复制应激的诱导有关。 LY2606368 还使这些细胞系对 PARP 抑制敏感,并损害 HR 修复和复制叉稳定性[5]。 BRCAwt HGSOC 通过较低的 CDK1/CyclinB1 活性诱导的延长的 G2 延迟对 LY2606368 单一疗法产生耐药性,从而防止细胞发生有丝分裂灾难和细胞死亡[6]。 PLK1 或 CHEK1 抑制剂(BI-2536 或 LY2606368)被发现在低纳摩尔浓度下对细胞系发挥优异的抗癌作用,并诱导细胞周期停滞[7]

在所测试的三个剂量的 LY2606368 中观察到高达 72.3% 的肿瘤生长抑制,小鼠体重减轻不超过 3%,表明 LY2606368 在任何治疗组中都具有良好的耐受性。此外,在 28 天的恢复期内,最高剂量组的肿瘤再生缓慢,表明对 LY2606368[1] 的持久肿瘤反应。

Chemical Properties

Cas No. 1234015-52-1 SDF
别名 5-[[5-[2-(3-氨基丙氧基)-6-甲氧基苯基]-1H-吡唑-3-基]氨基]-2-吡嗪甲腈,LY2606368
化学名 (Z)-5-((5-(2-(3-aminopropoxy)-6-methoxyphenyl)-1H-pyrazol-3(2H)-ylidene)amino)pyrazine-2-carbonitrile
分子式 C18H19N7O2 分子量 365.39
溶解度 <0.73mg/mL 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 2.7368 mL 13.684 mL 27.368 mL
5 mM 0.5474 mL 2.7368 mL 5.4736 mL
10 mM 0.2737 mL 1.3684 mL 2.7368 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

Targeting DNA Damage Response Promotes Antitumor Immunity through STING-Mediated T-cell Activation in Small Cell Lung Cancer

Cancer Discov2019 May;9(5):646-661.PMID: 30777870DOI: 10.1158/2159-8290.CD-18-1020

Despite recent advances in the use of immunotherapy, only a minority of patients with small cell lung cancer (SCLC) respond to immune checkpoint blockade (ICB). Here, we show that targeting the DNA damage response (DDR) proteins PARP and checkpoint kinase 1 (CHK1) significantly increased protein and surface expression of PD-L1. PARP or CHK1 inhibition remarkably potentiated the antitumor effect of PD-L1 blockade and augmented cytotoxic T-cell infiltration in multiple immunocompetent SCLC in vivo models. CD8+ T-cell depletion reversed the antitumor effect, demonstrating the role of CD8+ T cells in combined DDR-PD-L1 blockade in SCLC. We further demonstrate that DDR inhibition activated the STING/TBK1/IRF3 innate immune pathway, leading to increased levels of chemokines such as CXCL10 and CCL5 that induced activation and function of cytotoxic T lymphocytes. Knockdown of cGAS and STING successfully reversed the antitumor effect of combined inhibition of DDR and PD-L1. Our results define previously unrecognized innate immune pathway-mediated immunomodulatory functions of DDR proteins and provide a rationale for combining PARP/CHK1 inhibitors and immunotherapies in SCLC. SIGNIFICANCE: Our results define previously unrecognized immunomodulatory functions of DDR inhibitors and suggest that adding PARP or CHK1 inhibitors to ICB may enhance treatment efficacy in patients with SCLC. Furthermore, our study supports a role of innate immune STING pathway in DDR-mediated antitumor immunity in SCLC.See related commentary by Hiatt and MacPherson, p. 584.This article is highlighted in the In This Issue feature, p. 565.

Prediction of DNA Repair Inhibitor Response in Short-Term Patient-Derived Ovarian Cancer Organoids

Cancer Discov2018 Nov;8(11):1404-1421.PMID: 30213835DOI: 10.1158/2159-8290.CD-18-0474

Based on genomic analysis, 50% of high-grade serous ovarian cancers (HGSC) are predicted to have DNA repair defects. Whether this substantial subset of HGSCs actually have functional repair defects remains unknown. Here, we devise a platform for functional profiling of DNA repair in short-term patient-derived HGSC organoids. We tested 33 organoid cultures derived from 22 patients with HGSC for defects in homologous recombination (HR) and replication fork protection. Regardless of DNA repair gene mutational status, a functional defect in HR in the organoids correlated with PARP inhibitor sensitivity. A functional defect in replication fork protection correlated with carboplatin and CHK1 and ATR inhibitor sensitivity. Our results indicate that a combination of genomic analysis and functional testing of organoids allows for the identification of targetable DNA damage repair defects. Larger numbers of patient-derived organoids must be analyzed to determine whether these assays can reproducibly predict patient response in the clinic.Significance: Patient-derived ovarian tumor organoids grow rapidly and match the tumors from which they are derived, both genetically and functionally. These organoids can be used for DNA repair profiling and therapeutic sensitivity testing and provide a rapid means of assessing targetable defects in the parent tumor, offering more suitable treatment options. Cancer Discov; 8(11); 1404-21. ?2018 AACR. This article is highlighted in the In This Issue feature, p. 1333.

CHK1 Inhibitor Blocks Phosphorylation of FAM122A and Promotes Replication Stress

Mol Cell2020 Nov 5;80(3):410-422.e6.PMID: 33108758DOI: 10.1016/j.molcel.2020.10.008

While effective anti-cancer drugs targeting the CHK1 kinase are advancing in the clinic, drug resistance is rapidly emerging. Here, we demonstrate that CRISPR-mediated knockout of the little-known gene FAM122A/PABIR1 confers cellular resistance to CHK1 inhibitors (CHK1is) and cross-resistance to ATR inhibitors. Knockout of FAM122A results in activation of PP2A-B55¦Á, a phosphatase that dephosphorylates the WEE1 protein and rescues WEE1 from ubiquitin-mediated degradation. The resulting increase in WEE1 protein expression reduces replication stress, activates the G2/M checkpoint, and confers cellular resistance to CHK1is. Interestingly, in tumor cells with oncogene-driven replication stress, CHK1 can directly phosphorylate FAM122A, leading to activation of the PP2A-B55¦Á phosphatase and increased WEE1 expression. A combination of a CHK1i plus a WEE1 inhibitor can overcome CHK1i resistance of these tumor cells, thereby enhancing anti-cancer activity. The FAM122A expression level in a tumor cell can serve as a useful biomarker for predicting CHK1i sensitivity or resistance.

Prexasertib (LY2606368) reduces clonogenic survival by inducing apoptosis in primary patient-derived osteosarcoma cells and synergizes with cisplatin and talazoparib

Int J Cancer2020 Aug 15;147(4):1059-1070.PMID: 31782150DOI: 10.1002/ijc.32814

Progress in the systemic control of osteosarcoma has been limited over the past decades thus indicating the urgent clinical need for the development of novel treatment strategies. Therefore, we have recently developed new preclinical models to study promising novel agents for the treatment of pediatric osteosarcoma. The checkpoint kinase (chk) inhibitor prexasertib (LY2606368) and its salt form (LSN2940930) have recently been shown to be active in adult and pediatric malignancies, including sarcoma. We have now tested the potency of prexasertib in clonogenic survival assays in two new lines of primary patient-derived osteosarcoma cells and in two established osteosarcoma cell lines as a single agent and in combination with cisplatin and the poly ADP-ribose polymerase (PARP) inhibitor talazoparib. Prexasertib alone results in strongly reduced clonogenic survival at low nanomolar concentrations and acts by affecting cell cycle progression, induction of apoptosis and induction of double-stranded DNA breakage at concentrations that are well below clinically tolerable and safe plasma concentrations. In combination with cisplatin and talazoparib, prexasertib acts in a synergistic fashion. Chk1 inhibition by prexasertib and its combination with the DNA damaging agent cisplatin and the PARP-inhibitor talazoparib thus emerges as a potential new treatment option for pediatric osteosarcoma which will now have to be tested in preclinical primary patient derived in vivo models and clinical studies.

Epigenetic and Immune-Cell Infiltration Changes in the Tumor Microenvironment in Hepatocellular Carcinoma

Front Immunol2021 Dec 2;12:793343.PMID: 34925377DOI: 10.3389/fimmu.2021.793343

Background: Epigenetics regulate gene expression without altering the DNA sequence. Epigenetics targeted chemotherapeutic approach can be used to overcome treatment resistance and low response rate in HCC. However, a comprehensive review of genomic data was carried out to determine the role of epigenesis in the tumor microenvironment (TME), immune cell-infiltration characteristics in HCC is still insufficient.
Methods: The association between epigenetic-related genes (ERGs), inflammatory response-related genes (IRRGs) and CRISPR genes was determined by merging genomic and CRISPR data. Further, characteristics of immune-cell infiltration in the tumor microenvironment was evaluated.
Results: Nine differentially expressed genes (ANP32B, ASF1A, BCORL1, BMI1, BUB1, CBX2, CBX3, CDK1, and CDK5) were shown to be independent prognostic factors based on lasso regression in the TCGA-LIHC and ICGC databases. In addition, the results showed significant differences in expression of PDCD-1 (PD-1) and CTLA4 between the high- and low-epigenetic score groups. The CTRP and PRISM-derived drug response data yielded four CTRP-derived compounds (SB-743921, GSK461364, gemcitabine, and paclitaxel) and two PRISM-derived compounds (dolastatin-10 and LY2606368). Patients with high ERGs benefited more from immune checkpoint inhibitor (ICI) therapy than patients with low ERGs. In addition, the high ERGs subgroup had a higher T cell exclusion score, while the low ERGs subgroup had a higher T cell dysfunction. However, there was no difference in microsatellite instability (MSI) score among the two subgroups. Further, genome-wide CRISPR-based loss-of function screening derived from DepMap was conducted to determine key genes leading to HCC development and progression. In total, 640 genes were identified to be essential for survival in HCC cell lines. The protein-protein interaction (PPI) network demonstrated that IRRGs PSEN1 was linked to most ERGs and CRISPR genes such as CDK1, TOP2A, CBX2 and CBX3.
Conclusion: Epigenetic alterations of cancer-related genes in the tumor microenvironment play a major role in carcinogenesis. This study showed that epigenetic-related novel biomarkers could be useful in predicting prognosis, clinical diagnosis, and management in HCC.