PIK-75_DataSheet_MedChemExpress
Nicka IP 75

氯化鎳過低 7g/l 7ASD以上發濛
普瑞得哈氏實驗教材
IP AD的影響
AD過高 5ml/l 全片光亮
AD過低 0.5ml/l 黑水位線出現 HCD.LCD灰暗
普瑞得哈氏實驗教材
四. 結 論
問題 偏低 偏高 帶出損耗大 分析並調整之 改善對策
從以上哈氏片外觀結果可總結出: NI-IP 75工藝參數作用如下:
PH值的影響
PH過高 4.5 8ASD以上白霧
PH過低 1.0 全片光亮
普瑞得哈氏實驗教材
鎳濃度的影響
鎳濃度過高 120g/l 全片光亮
Hale Waihona Puke 鎳濃度過低 60g/l HCD輕微白霧
普瑞得哈氏實驗教材
硼酸的影響
硼酸過高 50g/l 全片光亮
硼酸過低 30g/l HCD燒焦
普瑞得哈氏實驗教材
氯化鎳的影響
參數 作用
Ni
提供主鹽
高電流區白霧
NiCl2.6H2O
增加鍍液導 電性.促進陽 極溶解 緩衝 PH 提高鍍層亮 度,防變色 維持電流密 度範圍 維持電流密 度範圍
高電流區發濛
脆性增加
H3BO3 IP 75 AD
高電流區燒焦 高低電流區灰暗
鍍液易結晶 影響鍍層品質 分析並調整之 (或加IP-AD,以 1ml/l哈氏槽調整) 以IP-R2(3ml/l降 0.1個PH)調整之 調整之
PH 溫度
鍍液導電性增加 電流密度上限受影響
高電流區白霧燒焦 電流密度上限受影 響
普瑞得哈氏實驗教材
Ram Tech Ni- IP 75工藝
普瑞得哈氏實驗教材
Ni- IP 75工藝
內容 1.簡介 2.組成及操作條件 3.哈氏片外觀 4.總結
L755507_DataSheet_MedChemExpress

Inhibitors, Agonists, Screening Libraries Data SheetBIOLOGICAL ACTIVITY:L755507 is a potent, selective agonist of β3–AR with an IC 50 of 35 nM.IC50 & Target: IC50: 35 nM (β3–AR)[1]In Vitro: L755507 causes a robust concentration–dependent increase in cAMP accumulation (pEC 50 values of 8.5 and 12.3,respectively). Maximal cAMP accumulation with zinterol and L755507 is increased after pretreatment with pertussis toxin. In contrast to cAMP, zinterol, L755507 and L748337 increase phosphorylation of extracellular signal–regulated kinase 1/2 (Erk1/2) with very high potency (pEC 50 values of 10.9, 11.7 and 11.6)[1]. L755507 and Scr7 do not reduce cell viability significantly. Scr7 does not affect cell cycle distribution in a range of 10 to 200 μM. L755507 significantly decreases the proportion of cells in the G2/M phase at 10 μM or 40μM and increases the S–phase cells at 10 μM compare with the DMSO–treated cells [2]. PROTOCOL (Extracted from published papers and Only for reference)Cell Assay:[1]The cytosensor microphysiometer is used to measure β3–AR–mediated increases in ECAR . In brief, CHO β3 cells areseeded into 12–mm Transwell inserts at 5×105 cells/cup and left to adhere overnight. On the day of experiment, cells are equilibrated for 2 h, and cumulative concentration–response curves to L755507, zinterol, or L748337 are constructed in paired sister cells with each concentration of drug exposed to cells for 14 min. Results are expressed as a percentage of the maximal response to L755507. In experiments examining the effect of inhibitors, cells are treated for 30 min before stimulation with appropriate drugs. All drugs are diluted in modified RPMI 1640 medium. These results are expressed as a percentage of the maximal response to L755507, zinterol, or L748337 over basal [1].References:[1]. Sato M, et al. The beta3–adrenoceptor agonist4–[[(Hexylamino)carbonyl]amino]–N–[4–[2–[[(2S)–2–hydroxy–3–(4–hydroxyphenoxy)propyl]amino]ethyl]–phenyl]–benzenesulfonamide (L755507) andantagonist (S)–N–[4–[2–[[3–[3–(acetamidomethyl)phenoxy]–2–hydroxypropyl]amino]–ethyl]phenyl]benzenesulfonamide (L748337) activate different signaling pathways in Chinese hamster ovary–K1 cells stably expressing the human beta3–adrenoceptor. Mol Pharmacol. 2008 Nov;74(5):1417–28.[2]. Guoling Li, et al. Small molecules enhance CRISPR/Cas9–mediated homology–directed genome editing in primary cells. Sci Rep. 2017; 7: 8943.Product Name:L755507Cat. No.:HY-19334CAS No.:159182-43-1Molecular Formula:C30H40N4O6S Molecular Weight:584.73Target:Adrenergic Receptor Pathway:GPCR/G Protein Solubility:DMSO: 125 mg/mLCaution: Product has not been fully validated for medical applications. For research use only.Tel: 609-228-6898 Fax: 609-228-5909 E-mail: tech@Address: 1 Deer Park Dr, Suite Q, Monmouth Junction, NJ 08852, USA。
grp75分子量

grp75分子量
(原创实用版)
目录
1.GRP75 的概述
2.GRP75 的分子量
3.GRP75 的功能与应用
正文
1.GRP75 的概述
GRP75,全称为葡萄糖调节蛋白 75,是一种在人体内广泛存在的蛋白质。
它主要参与葡萄糖代谢过程,对维持血糖稳定起到关键作用。
作为一种重要的调节因子,GRP75 能够与其他蛋白质相互作用,进而调节它们的活性。
2.GRP75 的分子量
GRP75 的分子量并没有一个固定的数值,因为它可能存在多种同种物,这些同种物的分子量可能略有差异。
然而,通常情况下,GRP75 的分子量约为 75kDa(千道尔顿),这也是它名称的由来。
3.GRP75 的功能与应用
GRP75 具有多种生物学功能,其中包括:
- 调节胰岛素分泌:GRP75 通过与胰岛β细胞相互作用,促进胰岛素的分泌,从而降低血糖水平。
- 调节糖原合成与降解:GRP75 能够影响肝细胞和肌肉细胞中的糖原合成与降解,进而调节血糖水平。
- 参与脂肪酸代谢:GRP75 与脂肪酸结合蛋白相互作用,调节脂肪酸的代谢过程。
由于 GRP75 在葡萄糖代谢中的关键作用,它被认为是治疗糖尿病和相关代谢性疾病的重要靶点。
研究人员正在努力研究 GRP75 的结构和功能,以期开发出更具针对性和有效性的药物。
总之,GRP75 是一种重要的蛋白质,它在葡萄糖代谢中发挥着关键作用。
grp75分子量

grp75分子量1. 什么是grp75grp75是一种热休克蛋白(heat shock protein,HSP),它属于HSP70家族的一员。
HSP70家族是一类高度保守的蛋白质,广泛存在于原核生物和真核生物中。
grp75在真核生物中的分布广泛,包括动物、植物和微生物等。
2. grp75的分子量grp75的分子量约为75 kDa(千道尔顿)。
kDa是蛋白质分子量的常用单位,它表示千道尔顿(kilodalton)的意思。
道尔顿是一种质量单位,用于表示原子、分子或其他微观粒子的质量。
3. grp75的结构和功能grp75是一种具有多个结构域的蛋白质。
它包含一个N末端的ATP结合域(ATP-binding domain)和一个C末端的亚麻酸结合域(substrate-binding domain)。
这些结构域使得grp75具有多种功能。
grp75的主要功能是作为分子伴侣蛋白(molecular chaperone)参与蛋白质的折叠、组装和转运过程。
它能够与其他蛋白质相互作用,帮助它们正确地折叠成功能性的构象。
grp75还能够促进蛋白质的转运,将它们从细胞质转运到细胞器中。
此外,grp75还参与细胞应激反应和细胞凋亡等生物学过程。
在细胞应激反应中,grp75能够通过与其他蛋白质的相互作用,帮助细胞对环境变化做出适应。
在细胞凋亡过程中,grp75参与调控细胞凋亡信号通路,对细胞的生死决策起到重要的调节作用。
4. grp75的研究意义grp75的研究对于理解细胞内蛋白质折叠和转运机制具有重要意义。
通过研究grp75的结构和功能,可以揭示蛋白质折叠过程中的关键步骤和参与者,有助于发现新的药物靶点和治疗策略。
此外,grp75在肿瘤发生和发展过程中也起到重要的作用。
研究发现,grp75在肿瘤细胞中的表达水平通常较高,与肿瘤的侵袭和转移能力密切相关。
因此,grp75有望成为肿瘤治疗的新靶点,为肿瘤治疗提供新的思路和方法。
化学治疗chemotherapy对病原体微生物9955

内酰胺环
内酰胺酶
-内酰胺类抗生素的共性
• 药动学 • 吸收后广泛分布于全身组织,关节
、胸腔、心包液和胆汁中含量较 高,容易达到治疗浓度 • 在前列腺液、脑和眼组织液中浓 度低 • 在体内不被代谢, 以原形迅速由 肾小管分泌排出, t1/2较短
药理作用和机制
• 属于快速杀菌药 • 敏感菌
–大多数G+球菌(链、葡) –G+杆菌(炭、白、破) –G-球菌(脑、淋) –G-杆菌(伤寒,副伤寒,百,肠,痢) –螺旋体(梅,幽),放线菌
LD50/ED50或LD5/ED95 值越大表明药物疗效毒性 比越大
临床应用价值可能较高
抗微生物药物作用机制
1. 抑制细胞壁合成 –-LAs类
2. 增加胞浆膜通透性 – 氨基糖苷类通过离子吸附 – 制霉菌素结合真菌胞膜麦角固 醇 – 多粘菌素结合胞浆磷脂
-LAs类 转肽酶
3. 抑制细菌蛋白合成 –氯霉素、林可霉素和大环内酯 类作用于50S亚单位 –四环素和氨基糖苷类作用于30S 亚单位
机体、药物及病原体相互作用关系
抗菌药物发展简史
• Pasteur 和 Joubert 最先认识到 微生物的产物有可能抗菌
• 1928年,Fleming发现青霉素 • 1939年Florey置备成功青霉素,
1941年治疗感染, 开辟了抗生素 化疗的新纪元
• 1935年,Domagk合成磺胺药
• 抗生素(antibiotics): 由各种微 生物(包括细菌真菌放线菌)产生 的, 能抑制其它微生物的物质
3. 螺旋体所致感染 钩端螺旋体、梅毒(大剂量) 、 回归热等
4. G+杆菌所致感染 破伤风、白喉、炭疽病,应与相 应的抗毒素合用
药动学
• 易被胃酸及消化酶破坏,不宜 口服
PIK-75 Hydrochloride_PI3K_CAS号372196-77-5说明书_AbMole中国

分子量488.74溶解性(25°C)DMSO 3 mg/mL分子式C H BrN O S.HCl Water <1 mg/mLCAS号372196-77-5Ethanol <1 mg/mL储存条件3年 -20°C 粉末状生物活性PIK-75是一种p110α抑制剂,IC50为5.8 nM (比作用于p110β效果强200倍),亚型特异性的突变体在Ser773,也有效抑制DNA-PK,IC50为2 nM。
与纯化的p110α结合, PIK-75是非竞争性抑制剂,K为 36 nM。
PIK-75也有效抑制DNA-PK,IC50为2 nM。
PIK-75 (1 μM) 通过显著降低无刺激的非哮喘气道平滑肌细胞(ASM),哮喘 ASM细胞 , 和肺成纤维细胞的线粒体活性,降低细胞寿命。
实验操作来自于公开的文献,仅供参考细胞实验细胞系LN229:EGFR (PTENwt) and U87:EGFR (PTENmt) cells方法Cell proliferation assay and flow cytometry. For viability, 105 cells were seeded in 12-well plates in the presence of erlotinib, PI-103, PIK-90, rapamycin, erlotinib plus PIK-90, erlotinib plus rapamycin, erlotinib plus PIK-90, and rapamycin or erlotinib plus PI-103 for 72h. Cell viability was determined using a WST-1 assay (Roche Molecular Biochemicals).浓度0, 0.1, 0.5, 1µM处理时间72h动物实验动物模型Human primary GBM43 cells female BALB/c nu/nu mice bearing xenografts model配制DMSO:H20剂量40 mg/kg给药处理daily i.p.不同实验动物依据体表面积的等效剂量转换表(数据来源于FDA指南)小鼠大鼠兔豚鼠仓鼠狗重量 (kg)0.020.15 1.80.40.0810体表面积 (m)0.0070.0250.150.050.020.5K系数36128520动物 A (mg/kg) = 动物 B (mg/kg) ×动物 B的K系数动物 A的K系数例如,依据体表面积折算法,将白藜芦醇用于小鼠的剂量22.4 mg/kg 换算成大鼠的剂量,需要将22.4 mg/kg 乘以小鼠的K系数(3),再除以大鼠的K系数(6),得到白藜芦醇用于大鼠的等效剂量为11.2 mg/kg。
Bevirimat_174022-42-5_DataSheet_MedChemExpress

Caution: Not fully tested. For research purposes only Medchemexpress LLC
www.medch r p x e m e h c d e m . w w w : b e AW Sm Uo ,c 0 4. 5s 8s 0e r p Jx Ne ,m n e oh t c e cd ne i rm P @ ,o y f an Wi : l ni oa sm n iE k l i W 8 1
References: [1]. Smith PF, et al. Phase I and II study of the safety, virologic effect, and pharmacokinetics/pharmacodynamics of single-dose 3-o-(3',3'-dimethylsuccinyl)betulinic acid (bevirimat) against human immunodeficiency virus infection. Antimicrob Agents Chemother. 2007 Oct;51(10):3574-81. ( ) [2]. Salzwedel K, et al. Maturation inhibitors: a new therapeutic class targets the virus structure. AIDS Rev. 2007 Jul-Sep;9(3):162-72. [3]. Martin DE, et al. Bevirimat: a novel maturation inhibitor for the treatment of HIV-1 infection. Antivir Chem Chemother. 2008;19(3):107-13.
b-AP15_DataSheet_MedChemExpress

Inhibitors, Agonists, Screening Libraries Data SheetBIOLOGICAL ACTIVITY:b–AP15 is a specific inhibitor of the deubiquitinating enzymes UCHL5 and Usp14.IC50 & Target: UCHL5/Usp14[1]In Vitro: Purified 19S proteasomes (5 nM) are treated with indicated concentrations of b–AP15 and DUB activity is determined by detectionof Ub–AMC cleavage. The IC 50 value (2.1±0.411 μM) is determined from log concentration curves in Graph Pad Prism using non linear regression analysis. b–AP15 as a previously unidentified class of proteasome inhibitor that abrogates thedeubiquitinating activity of the 19S regulatory particle. b–AP15 inhibited the activity of two 19S regulatory–particle–associated deubiquitinases, ubiquitin C–terminal hydrolase 5 (UCHL5) and ubiquitin–specific peptidase 14 (USP14), resulting in accumulation of polyubiquitin. b–AP15 induced tumor cell apoptosis that is insensitive to TP53 status and overexpression of the apoptosisinhibitor BCL2[1]. The ability of b–AP15 is determined to inhibit proteasome deubiquitinase activity using Ub–AMC as the substrate.An IC 50 of 16.8±2.8 μM is observed [2]. b–AP15 is a specific USP14 and UCHL5 inhibitor, which blocks growth and induces apoptosis in MM cells [3].In Vivo: b–AP15 (2.5 mg/kg) inhibits tumor growth in syngenic mice models with less frequent administration schedules. We administered b–AP15 to C57BL/6J mice with Lewis lung carcinomas (LLCs) using a 2–d–on, 2–d–off schedule and to BALB/c mice with orthotopic breast carcinoma (4T1) using a 1–d–on, 3–d–off schedule. b–AP15 significantly inhibited tumor growth in both models, with T/C=0.16 (P≤0.01) for the C57BL/6J mice and T/C=0.25 (P≤0.001) for the BALB/c mice. A reduction in the number of pulmonary metastases also is observed in the group of mice with 4T1 breast carcinomas treated with b–AP15[1].PROTOCOL (Extracted from published papers and Only for reference)Kinase Assay:[1]For deubiquitinase inhibition assays, 19S regulatory particle (5 nM), 26S (5 nM) UCH–L1 (5 nM), UCH–L3 (0.3 nM),USP2CD (5 nM) USP7CD (5 nM) USP8CD (5 nM) or BAP1 (5 nM) is incubated with DMSO or b–AP15 and monitored the cleavage of ubiquitin–AMC (1,000 nM) using a Wallac VICTOR Multilabel counter or a Tecan Infinite M1000 equipped with 380 nm excitation and 460 nm emission filters [1].Cell Assay: b–AP15 is dissolved in DMSO and stored, and then diluted with appropriate medium before use [2]. [2]Cell viability is monitored by either the fluorometric microculture cytotoxicity assay or the MTT assay. For the MTT assay, cells are seeded into 96–well flat–bottomed plates overnight and exposed to drugs, using DMSO as the control. At the end of incubations, 10 μl of a stock solution of 5 mg/mL MTT is added into each well, and the plates are incubated 4 hours at 37°C. Formazan crystals are dissolved with 100 μL 10% SDS/10 mM HCl solution overnight at 37°C. Absorbance is measured using an enzyme–linkedimmunosorbent assay (ELISA) plate reader at 590 nm [2].Animal Administration: b–AP15 is dissolved it in Cremophor EL and polyethylene glycol 400 (1:1) by heating to reach aworking concentration of 2 mg/mL. Working stock is 1:10 diluted in 0.9% saline immediately before injection (Mice)[1].[1]Mice [1]For the squamous carcinoma model, 1×106 FaDu cells are subcutaneously injected into the right rear flank of female SCIDProduct Name:b–AP15Cat. No.:HY-13989CAS No.:1009817-63-3Molecular Formula:C 22H 17N 3O 6Molecular Weight:419.39Target:Deubiquitinase Pathway:Cell Cycle/DNA Damage Solubility:DMSO: ≥ 44 mg/mLmice. Tumor growth is measured by the formula length×width2×0.44. When tumors have grown to a size of approximately 200 mm3 (defined as day 0), mice are randomized to receive either vehicle (n=10) or b–AP15 (n=15) at 5 mg per kg of body weight by daily subcutaneous injection. For the colon carcinoma model, we subcutaneously injected 2.5 × 106 HCT–116 colon carcinoma cells overexpressing Bcl2 into the right flank of female nude mice. We treated mice with 5 mg of b–AP15 per kg of body weight by intraperitoneal injection. For the lung carcinoma model, we subcutaneously injected 2×105 LLC cells into the right rear flank of female C57/B6 mice. When tumors had grown to a size of approximately 50 mm3 (defined as day 0), we randomized mice to receive either vehicle (n=4) or b–AP15 (n=4) at 5 mg per kg of body weight intraperitoneally, with a treatment cycle consisting of 2 d of treatment followed by 2 d of rest (2 d on, 2 d off) for 2 weeks.References:[1]. D'Arcy P, et al. Inhibition of proteasome deubiquitinating activity as a new cancer therapy. Nat Med. 2011 Nov 6;17(12):1636–40.[2]. Wang X, et al. The 19S Deubiquitinase Inhibitor b–AP15 is Enriched in Cells and Elicits Rapid Commitment to Cell Death. Mol Pharmacol. 2014 Jun;85(6):932–45.[3]. Tian Z, et al. A novel small molecule inhibitor of deubiquitylating enzyme USP14 and UCHL5 induces apoptosis in multiple myeloma and overcomes bortezomib resistance. Blood. 2014 Jan 30;123(5):706–16.Caution: Product has not been fully validated for medical applications. For research use only.Tel: 609-228-6898 Fax: 609-228-5909 E-mail: tech@Address: 1 Deer Park Dr, Suite Q, Monmouth Junction, NJ 08852, USA。
- 1、下载文档前请自行甄别文档内容的完整性,平台不提供额外的编辑、内容补充、找答案等附加服务。
- 2、"仅部分预览"的文档,不可在线预览部分如存在完整性等问题,可反馈申请退款(可完整预览的文档不适用该条件!)。
- 3、如文档侵犯您的权益,请联系客服反馈,我们会尽快为您处理(人工客服工作时间:9:00-18:30)。
Inhibitors, Agonists, Screening Libraries Data SheetBIOLOGICAL ACTIVITY:PIK–75 is a p110α inhibitor with IC50 of 5.8 nM (200–fold more potently than p110β), isoform–specific mutants at Ser773, and also potently inhibits DNA–PK with IC50 of 2 nM.IC50 value: 5.8/2 nM(p110α/DNA–PK) [1] [2]Target: p110α; DNA–PKin vitro: PIK–75 shows the impressive potency and isoform selectivity at p110α while the corresponding IC50 values are 1300 nM, 76nM and 510 nM for other PI3K isoforms, p110β, –γ, and –δ, respectively. Furthermore, when binding to purified p110α, PIK–75 is a noncompetitive inhibitor with respect to ATP with Ki of 36 nM and competitive with respect to the substrate PI with Ki of 2.3 nM [1].PIK–75 also shows potent inhibition of DNA–PK [2]. PIK–75 (1 μM) reduces cell survival by significantly decreasing mitochondrial activity in unstimulated nonasthmatic airway smooth muscle (ASM) cells, asthmatic ASM cells, and lung fibroblasts. While inTGFβ–stimulated ASM cells, PIK75 only decreases mitochondrial activity in asthmatic cells without effects in nonasthmatic cells [3].in vivo: In the ErbB3WT tumor model, PIK–75 reduces in vitro chemotactic response to HRGβ1 and lowers pAkt levels by 40%. Besides,PIK–75 significantly reduces tumor cell motility and in vivo invasion in ErbB3WT primary tumors [4]. In the CD1 male mice, PIK–75leads to serious impairments in the insulin tolerance test (ITT) and glucose tolerance test (GTT), and an increase in glucose production during a pyruvate tolerance test (PTT) [5].PROTOCOL (Extracted from published papers and Only for reference)Cell assay [6]A total of 2,000 human pancreatic cancer cells (MIA PaCa–2 or AsPC–1) per well were plated in 96–well flat–bottom plates and then treated with either gemcitabine, PIK–75 alone or in combination of both drugs with indicated concentrations. At the indicated times,20 μl of 1 mg/ml MTT in PBS was added to each well and further incubated for 4 h. After centrifugation and removal of the medium,150 μl of DMSO was added to each well to dissolve the formazan crystals. The absorbance was measured at 562 nm using an ELx808absorbance microplate reader. Absorbance of untreated cells was designated as 100%, and the relative viable cells were expressed as a percentage of this value. The drug interaction was evaluated by using the combination index (CI) according to the method of Chou and Talalay using CompuSyn software.Animal administration [6]Animal use procedures were approved by the Institutional Animal Care and Use Committee of Georgetown University Medical Center.MIA PaCa–2 cells (1.7×106 cells/mouse) mixed with Matrigel were injected subcutaneously into the flank of male athymic nude(Foxn1nu) mice aged 6–weeks. Gemcitabine (50 mg/ml) was dissolved in PBS and PIK–75 (20 mg/ml) was dissolved in DMSO. Injection solution was made as 10% of Cremophor EL and 3% of poly(ethylene glycol) 400 in sterile water. Before administration of compounds,gemcitabine was further diluted in PBS and DMSO or PIK–75 was further diluted in the injection solution and sterilized by 0.2 μm filterProduct Name:PIK–75Cat. No.:HY-13281CAS No.:372196-77-5Molecular Formula:C 16H 15BrClN 5O 4S Molecular Weight:488.74Target:DNA–PK; DNA–PK Pathway:PI3K/Akt/mTOR; Cell Cycle/DNA Damage Solubility:10 mM in DMSOunit. These diluents were mixed with 1:1 ratio and administered into peritoneal cavity of the mouse. Gemcitabine (20 mg/kg) or gemcitabine (20 mg/kg)/PIK–75 (2 mg/kg) combination was administered twice per week and vehicle control and PIK–75 (2 mg/kg) were administered 5 times per week. The body weights and tumor sizes were measured 3 times per week. Tumor volumes were calculated as width (mm) × length (mm) × height (mm)/2.References:[1]. Zheng Z, et al. Isoform–selective inhibition of phosphoinositide 3–kinase: identification of a new region of nonconserved amino acids critical for p110αinhibition. Mol Pharmacol. 2011, 80(4), 657–664.[2]. WO/2003/072557, 09/04/2003[3]. Moir LM, et al. Phosphatidylinositol 3–kinase isoform–specific effects in airway mesenchymal cell function. J Pharmacol Exp Ther. 2011, 337(2), 557–566.[4]. Smirnova T, et al. Phosphoinositide 3–kinase signaling is critical for ErbB3–driven breast cancer cell motility and metastasis. Oncogene. 2012, 31(6), 706–715.[5]. Smith GC, et al. Effects of acutely inhibiting PI3K isoforms and mTOR on regulation of glucose metabolism in vivo. Biochem J. 2012, 442(1), 161–169.[6]. Duong HQ, et al. Inhibition of NRF2 by PIK–75 augments sensitivity of pancreatic cancer cells to gemcitabine. Int J Oncol. 2014 Mar;44(3):959–69.Caution: Product has not been fully validated for medical applications. For research use only.Tel: 609-228-6898 Fax: 609-228-5909 E-mail: tech@Address: 1 Deer Park Dr, Suite Q, Monmouth Junction, NJ 08852, USA。