Pitavastatin

匹伐他汀
  • 文章类型: Journal Article
    宫颈癌是第四种最普遍的癌症形式,在全球范围内是女性死亡率的重要因素。匹伐他汀是一种抗高脂血症药物,已被证明具有抗癌和抗炎作用。因此,本研究的目的是评价匹伐他汀对宫颈癌的抗癌作用及其分子机制。结果表明,匹伐他汀通过靶向CaSki的细胞周期阻滞和凋亡来显著抑制细胞活力,HeLa和C-33A细胞。匹伐他汀在CaSki和HeLa细胞中引起亚G1-和G0/G1期停滞,在C-33A细胞中引起亚G1-和G2/M期停滞。此外,匹伐他汀通过激活聚ADP-核糖聚合酶(PARP)诱导细胞凋亡,Bax和裂解的caspase3;灭活Bcl-2的表达;并增加线粒体膜去极化。此外,匹伐他汀诱导细胞凋亡并减缓所有三种宫颈细胞系的迁移,由PI3K/AKT和MAPK(JNK,p38和ERK1/2)途径。匹伐他汀在源自癌细胞的异种移植小鼠模型中体内显着抑制肿瘤生长。总的来说,我们的结果确定匹伐他汀是宫颈癌的抗癌药物,将来可能会扩展到临床使用。
    Cervical cancer ranks as the fourth most prevalent form of cancer and is a significant contributor to female mortality on a global scale. Pitavastatin is an anti-hyperlipidemic medication and has been demonstrated to exert anticancer and anti-inflammatory effects. Thus, the purpose of this study was to evaluate the anticancer effect of pitavastatin on cervical cancer and the underlying molecular mechanisms involved. The results showed that pitavastatin significantly inhibited cell viability by targeting cell-cycle arrest and apoptosis in Ca Ski, HeLa and C-33 A cells. Pitavastatin caused sub-G1- and G0/G1-phase arrest in Ca Ski and HeLa cells and sub-G1- and G2/M-phase arrest in C-33 A cells. Moreover, pitavastatin induced apoptosis via the activation of poly-ADP-ribose polymerase (PARP), Bax and cleaved caspase 3; inactivated the expression of Bcl-2; and increased mitochondrial membrane depolarization. Furthermore, pitavastatin induced apoptosis and slowed the migration of all three cervical cell lines, mediated by the PI3K/AKT and MAPK (JNK, p38 and ERK1/2) pathways. Pitavastatin markedly inhibited tumor growth in vivo in a cancer cell-originated xenograft mouse model. Overall, our results identified pitavastatin as an anticancer agent for cervical cancer, which might be expanded to clinical use in the future.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:尽管有有效的策略,EGFR突变肺癌的耐药性仍是一个挑战.代谢重编程是肿瘤耐药的主要机制之一。一类被称为“他汀类药物”的药物抑制脂质胆固醇代谢,广泛用于心血管疾病患者。先前的研究也证明了其在接受EGFR-TKI治疗的肺癌患者中改善治疗效果的能力。因此,他汀类药物对肺癌靶向耐药的影响仍有待研究。
    方法:长时间接触吉非替尼导致从亲本敏感细胞系(PC9)中出现耐药肺癌细胞系(PC9GR),表现出传统的EGFR突变。CCK-8测定用于评估各种浓度的匹伐他汀对细胞增殖的影响。进行RNA测序以检测差异表达的基因及其相关途径。为了检测蛋白质的表达,进行蛋白质印迹。通过异种移植小鼠模型在体内评估匹伐他汀的抗肿瘤活性。
    结果:低剂量维持诱导PC9吉非替尼耐药菌株。细胞培养和动物相关研究验证了匹伐他汀对肺癌细胞增殖的抑制作用,促进细胞凋亡,抑制了EGFR-TKIs的获得性耐药。KEGG通路分析表明,相对于PC9细胞,在PC9GR细胞中hippo/YAP信号通路被激活,匹伐他汀对YAP的表达有抑制作用。用YAPRNA干扰,pAKT,pBAD和BCL-2表达降低,而BAX表达则增加。因此,YAP下调显著增加吉非替尼耐药肺癌细胞的凋亡并降低其存活率。SC79增加pAKT后,吉非替尼诱导的YAP下调细胞凋亡减少,细胞存活率提高。机械上,匹伐他汀的这些作用与YAP途径有关,从而抑制下游AKT/BAD-BCL-2信号通路。
    结论:本研究为临床应用降脂药物匹伐他汀增强肺癌患者对EGFR-TKI药物的敏感性、缓解耐药提供了分子依据。
    BACKGROUND: Despite effective strategies, resistance in EGFR mutated lung cancer remains a challenge. Metabolic reprogramming is one of the main mechanisms of tumor drug resistance. A class of drugs known as \"statins\" inhibit lipid cholesterol metabolism and are widely used in patients with cardiovascular diseases. Previous studies have also documented its ability to improve the therapeutic impact in lung cancer patients who receive EGFR-TKI therapy. Therefore, the effect of statins on targeted drug resistance to lung cancer remains to be investigated.
    METHODS: Prolonged exposure to gefitinib resulted in the emergence of a resistant lung cancer cell line (PC9GR) from the parental sensitive cell line (PC9), which exhibited a traditional EGFR mutation. The CCK-8 assay was employed to assess the impact of various concentrations of pitavastatin on cellular proliferation. RNA sequencing was conducted to detect differentially expressed genes and their correlated pathways. For the detection of protein expression, Western blot was performed. The antitumor activity of pitavastatin was evaluated in vivo via a xenograft mouse model.
    RESULTS: PC9 gefitinib resistant strains were induced by low-dose maintenance. Cell culture and animal-related studies validated that the application of pitavastatin inhibited the proliferation of lung cancer cells, promoted cell apoptosis, and restrained the acquired resistance to EGFR-TKIs. KEGG pathway analysis showed that the hippo/YAP signaling pathway was activated in PC9GR cells relative to PC9 cells, and the YAP expression was inhibited by pitavastatin administration. With YAP RNA interference, pAKT, pBAD and BCL-2 expression was decreased, while BAX expression as increased. Accordingly, YAP down-regulated significantly increased apoptosis and decreased the survival rate of gefitinib-resistant lung cancer cells. After pAKT was increased by SC79, apoptosis of YAP down-regulated cells induced by gefitinib was decreased, and the cell survival rate was increased. Mechanistically, these effects of pitavastatin are associated with the YAP pathway, thereby inhibiting the downstream AKT/BAD-BCL-2 signaling pathway.
    CONCLUSIONS: Our study provides a molecular basis for the clinical application of the lipid-lowering drug pitavastatin enhances the susceptibility of lung cancer to EGFR-TKI drugs and alleviates drug resistance.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    他汀类药物可降低慢性肾脏病(CKD)患者心血管事件的风险。尽管糖尿病(DM)是他汀类药物治疗的副作用,一些研究表明,匹伐他汀不会引起DM。本研究调查了匹伐他汀对红细胞膜脂肪酸(FA)含量的影响,影响DM和心血管疾病的发生。此外,评估匹伐他汀治疗后脂联素和糖化血红蛋白(HbA1c)水平的变化.
    共纳入45名患者,其中28人完成了研究。超过24周,16例患者接受2mg匹伐他汀,12例患者接受10mg阿托伐他汀。如果需要额外的脂质对照,则在12周后调整剂量。匹伐他汀和阿托伐他汀组分别有10例和9例DM患者,分别。红细胞膜FAs和脂联素水平测定采用气相色谱法和酶联免疫吸附试验,分别。
    在两组中,饱和FAs,棕榈酸,反式油酸,总胆固醇,低密度脂蛋白胆固醇水平显著低于基线水平.匹伐他汀组红细胞膜中花生四烯酸(AA)含量明显增加,但脂联素水平未受影响。用匹伐他汀治疗的患者HbA1c水平降低。没有与他汀类药物治疗相关的不良反应。
    匹伐他汀治疗CKD患者可能通过改变红细胞膜AA水平来改善糖代谢。此外,匹伐他汀对CKD和DM患者的血糖控制无不良影响.
    BACKGROUND: Statins reduce the risk of cardiovascular events in patients with chronic kidney disease (CKD). Although diabetes mellitus (DM) is a reported side effect of statin treatment, some studies have indicated that pitavastatin does not cause DM. The present study investigated the effect of pitavastatin on the fatty acid (FA) content of erythrocyte membranes, which affects the occurrence of DM and cardiovascular diseases. In addition, changes in adiponectin and glycated hemoglobin (HbA1c) levels were evaluated after pitavastatin treatment.
    METHODS: A total of 45 patients were enrolled, 28 of whom completed the study. Over 24 weeks, 16 patients received 2 mg pitavastatin and 12 patients received 10 mg atorvastatin. Dosages were adjusted after 12 weeks if additional lipid control was required. There were 10 and nine patients with DM in the pitavastatin and atorvastatin groups, respectively. Erythrocyte membrane FAs and adiponectin levels were measured using gas chromatography and enzyme-linked immunosorbent assay, respectively.
    RESULTS: In both groups, saturated FAs, palmitic acid, trans-oleic acid, total cholesterol, and low-density lipoprotein cholesterol levels were significantly lower than those at baseline. The arachidonic acid (AA) content in the erythrocyte membrane increased significantly in the pitavastatin group, but adiponectin levels were unaffected. HbA1c levels decreased in patients treated with pitavastatin. No adverse effects were associated with statin treatment.
    CONCLUSIONS: Pitavastatin treatment in patients with CKD may improve glucose metabolism by altering erythrocyte membrane AA levels. In addition, pitavastatin did not adversely affect glucose control in patients with CKD and DM.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    鉴于由于固有的抗癌性机制而导致的针对各种癌症类型的当前治疗方式的有限功效,发现有效的抗癌剂提出了巨大的挑战。癌症免疫化疗是乳腺癌治疗和克服癌症耐药性的替代策略。人吲哚胺2,3-双加氧酶(hIDO1)和人色氨酸2,3-双加氧酶2(hTDO2)在色氨酸代谢中起关键作用,导致犬尿氨酸和其他生物活性代谢产物的产生。此过程促进烟酰胺二核苷酸(NAD)的从头合成,促进抗癌性。这项研究使用FDA批准的药物的药物再利用策略确定了一种新的双重hIDO1/hTDO2抑制剂。在这里,我们描述了基于配体的药效团模型的发展,该模型基于12种已报道hIDO1/hTDO2抑制活性的化合物的训练集。我们进行了药效团搜索,然后对2568种FDA批准的针对这两种酶的药物进行了高通量虚拟筛选。导致十次点击,其中四个具有双重抑制活性的高潜力。为了进一步进行计算机模拟和体外生物学研究,抗高胆固醇血症药物匹伐他汀被认为是本研究的首选药物。分子动力学(MD)模拟表明,匹伐他汀与hIDO1和hTDO2受体形成稳定的复合物,为其潜在的治疗功效提供结构基础。在纳摩尔(nM)浓度下,它对两种检查的酶都表现出显着的体外酶抑制活性。此外,匹伐他汀对BT-549,MCF-7和HepG2细胞系表现出有效的细胞毒性活性(IC50=16.82,9.52和1.84µM,分别)。其抗癌活性主要是由于通过HepG2癌细胞的细胞周期分析发现的G1/S期停滞的诱导。最终,用匹伐他汀治疗HepG2癌细胞影响caspase-3的显著激活,伴随着细胞凋亡生物标志物如IDO的下调,TDO,STAT3、P21、P27、IL-6和AhR。
    Discovering effective anti-cancer agents poses a formidable challenge given the limited efficacy of current therapeutic modalities against various cancer types due to intrinsic resistance mechanisms. Cancer immunochemotherapy is an alternative strategy for breast cancer treatment and overcoming cancer resistance. Human Indoleamine 2,3-dioxygenase (hIDO1) and human Tryptophan 2,3-dioxygenase 2 (hTDO2) play pivotal roles in tryptophan metabolism, leading to the generation of kynurenine and other bioactive metabolites. This process facilitates the de novo synthesis of Nicotinamide Dinucleotide (NAD), promoting cancer resistance. This study identified a new dual hIDO1/hTDO2 inhibitor using a drug repurposing strategy of FDA-approved drugs. Herein, we delineate the development of a ligand-based pharmacophore model based on a training set of 12 compounds with reported hIDO1/hTDO2 inhibitory activity. We conducted a pharmacophore search followed by high-throughput virtual screening of 2568 FDA-approved drugs against both enzymes, resulting in ten hits, four of them with high potential of dual inhibitory activity. For further in silico and in vitro biological investigation, the anti-hypercholesterolemic drug Pitavastatin deemed the drug of choice in this study. Molecular dynamics (MD) simulations demonstrated that Pitavastatin forms stable complexes with both hIDO1 and hTDO2 receptors, providing a structural basis for its potential therapeutic efficacy. At nanomolar (nM) concentration, it exhibited remarkable in vitro enzyme inhibitory activity against both examined enzymes. Additionally, Pitavastatin demonstrated potent cytotoxic activity against BT-549, MCF-7, and HepG2 cell lines (IC50 = 16.82, 9.52, and 1.84 µM, respectively). Its anticancer activity was primarily due to the induction of G1/S phase arrest as discovered through cell cycle analysis of HepG2 cancer cells. Ultimately, treating HepG2 cancer cells with Pitavastatin affected significant activation of caspase-3 accompanied by down-regulation of cellular apoptotic biomarkers such as IDO, TDO, STAT3, P21, P27, IL-6, and AhR.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    脂质紊乱,主要是高胆固醇血症,是波兰最常见的心血管(CV)危险因素(这甚至适用于3/4的人)。低密度脂蛋白胆固醇(LDL-C)血清水平是确定CV风险并确定降脂治疗(LLT)目标的基本脂质参数。降脂治疗可改善心血管预后,并在一级和二级心血管预防中延长寿命。尽管有有效的降脂药物和有关其有益作用的可靠数据,LDL-C控制水平严重不足.这是相关的,除其他外,医生的惰性和患者对副作用的恐惧。血脂学的发展使药物具有良好的安全性并使治疗个性化。匹伐他汀,第三有效的降脂他汀类药物,其特点是肌肉并发症和新的糖尿病病例的风险较低,因为它的代谢不同。因此,匹伐他汀对于糖尿病高危患者或现有糖尿病患者是非常好的治疗选择,以及有心血管风险的患者。这份专家意见文件试图就使用匹伐他汀治疗脂质紊乱的地方和可能性提出建议。
    Lipid disorders, primarily hypercholesterolemia, are the most common cardiovascular (CV) risk factor in Poland (this applies even 3/4 of people). The low-density lipoprotein cholesterol (LDL-C) serum level is the basic lipid parameter that should be measured to determine CV risk and determines the aim and target of lipid-lowering treatment (LLT). Lipid-lowering treatment improves cardiovascular prognosis and prolongs life in both primary and secondary cardiovascular prevention. Despite the availability of effective lipid-lowering drugs and solid data on their beneficial effects, the level of LDL-C control is highly insufficient. This is related, among other things, to physician inertia and patients\' fear of side effects. The development of lipidology has made drugs available with a good safety profile and enabling personalisation of therapy. Pitavastatin, the third most potent lipid-lowering statin, is characterised by a lower risk of muscle complications and new cases of diabetes due to its being metabolised differently. Thus, pitavastatin is a very good therapeutic option in patients at high risk of diabetes or with existing diabetes, and in patients at cardiovascular risk. This expert opinion paper attempts at recommendation on the place and possibility of using pitavastatin in the treatment of lipid disorders.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    肺癌是全球癌症相关死亡的最常见原因,由多种因素引起,包括高脂肪饮食(HFD)。CD36,一种脂肪酸受体,与代谢相关的疾病密切相关,包括心血管疾病和癌症。然而,CD36在HFD加速非小细胞肺癌(NSCLC)中的作用尚不清楚.在体内,我们饲喂C57BL/6J野生型(WT)和CD36敲除(CD36-/-)小鼠正常食物或HFD在有或没有匹伐他汀的2周前皮下注射LLC1细胞。体外,用游离脂肪酸(FFA)处理A549和NCI-H520细胞以模拟HFD情况以探索潜在机制。我们发现HFD在体内促进LLC1肿瘤生长,FFA增加A549和NCI-H520细胞的细胞增殖和迁移。降脂药匹伐他汀抑制了增强的细胞或肿瘤生长,减少脂质积累。更重要的是,我们发现,NSCLC患者的血浆可溶性CD36(sCD36)水平高于健康患者.与WT小鼠相比,在CD36-/-小鼠中LLC1细胞的增殖被大大抑制,在HFD组中被匹伐他汀进一步抑制。在分子水平上,我们发现CD36抑制,用匹伐他汀或质粒,通过AKT/mTOR途径降低增殖和迁移相关蛋白的表达。一起来看,我们证明匹伐他汀或其他抑制剂抑制CD36表达可能是NSCLC治疗的可行策略.
    Lung cancer is the most common cause of cancer-related deaths worldwide and is caused by multiple factors, including high-fat diet (HFD). CD36, a fatty acid receptor, is closely associated with metabolism-related diseases, including cardiovascular disease and cancer. However, the role of CD36 in HFD-accelerated non-small-cell lung cancer (NSCLC) is unclear. In vivo, we fed C57BL/6J wild-type (WT) and CD36 knockout (CD36-/-) mice normal chow or HFD in the presence or absence of pitavastatin 2 weeks before subcutaneous injection of LLC1 cells. In vitro, A549 and NCI-H520 cells were treated with free fatty acids (FFAs) to mimic HFD situation for exploration the underlying mechanisms. We found that HFD promoted LLC1 tumor growth in vivo and that FFAs increased cell proliferation and migration in A549 and NCI-H520 cells. The enhanced cell or tumor growth was inhibited by the lipid-lowering agent pitavastatin, which reduced lipid accumulation. More importantly, we found that plasma soluble CD36 (sCD36) levels were higher in NSCLC patients than those in healthy ones. Compared to that in WT mice, the proliferation of LLC1 cells in CD36-/- mice was largely suppressed, which was further repressed by pitavastatin in HFD group. At the molecular level, we found that CD36 inhibition, either with pitavastatin or plasmid, reduced proliferation- and migration-related protein expression through the AKT/mTOR pathway. Taken together, we demonstrate that inhibition of CD36 expression by pitavastatin or other inhibitors may be a viable strategy for NSCLC treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景和目的:我们最近报道了氟伐他汀,阿托伐他汀,辛伐他汀和瑞舒伐他汀使用家兔肠道制剂具有钙通道拮抗活性。目前的研究集中在匹伐他汀和洛伐他汀可能抑制血管L型钙通道的作用。可能具有血管松弛作用。还测试了在氨氯地平存在下匹伐他汀和洛伐他汀的联合作用的血管舒张作用。材料和方法:匹伐他汀和洛伐他汀对80mM氯化钾(KCL)诱导的收缩和1µM去甲肾上腺素(N。在分离的兔主动脉条制剂中研究了E)诱导的收缩。通过构建钙浓度响应曲线(CCRC)进一步验证了对80mMKCL诱导的血管收缩的放松作用。在不存在和存在三种不同浓度的匹伐他汀和洛伐他汀的情况下,使用CCRC作为阴性对照。维拉帕米用作具有L-型钙通道结合活性的标准药物。在其他一系列实验中,我们研究了匹伐他汀之间的药物相互作用,洛伐他汀,和氨氯地平.结果:这项研究的结果表明,洛伐他汀比匹伐他汀更有效,因为在完整的主动脉中具有相对较低的EC50(7.44×10-5±0.16M)和(4.55×10-5±0.10M)用于KCL诱导的收缩。对于KCL诱导的收缩,完整和裸露的主动脉中的洛伐他汀振幅为,分别,24%和35.5%;而匹伐他汀在完整和裸露的主动脉中用于KCL诱导的收缩的幅度分别为34%和40%,分别。当我们在EC50(LogCa++M)中添加氨氯地平时,观察到他汀类药物的EC50值的左移。CCRC的右移说明匹伐他汀和洛伐他汀具有钙通道拮抗作用。与匹伐他汀相比,测试浓度(6.74×10-7M)的洛伐他汀产生了相对较低的EC50(-2.5±0.10)LogCaM的右移,这进一步证实洛伐他汀相对更有效。EC50的右移类似于维拉帕米的右移。在氨氯地平存在下观察到匹伐他汀和洛伐他汀的加成效应(p<0.05)。结论:匹伐他汀和洛伐他汀通过对L型电压门控钙通道的抑制作用来缓解KCL(80mM)诱导的血管收缩。洛伐他汀和匹伐他汀还放松了去甲肾上腺素(1µM)诱导的收缩,从而深入了解匹伐他汀和洛伐他汀的双重作用模式。
    Background and Objectives: We have recently reported that Fluvastatin, Atorvastatin, Simvastatin and Rosuvastatin have calcium channel antagonistic activities using rabbits\' intestinal preparations. The current study is focused on the effects of Pitavastatin and Lovastatin for possible inhibition of vascular L-Type calcium channels, which may have vasorelaxant effect(s). Combined effects of Pitavastatin and Lovastatin in the presence of Amlodipine were also tested for vasorelaxation. Materials and Methods: Possible relaxing effects of Pitavastatin and Lovastatin on 80 mM Potassium chloride (KCL)-induced contractions and on 1 µM norepinephrine (N.E)-induced contractions were studied in isolated rabbit\'s aortic strips preparations. Relaxing effects on 80 mM KCL-induced vascular contractions were further verified by constructing Calcium Concentration Response Curves (CCRCs), in the absence and presence of three different concentrations of Pitavastatin and Lovastatin using CCRCs as negative control. Verapamil was used as a standard drug that has L-Type calcium channel binding activity. In other series of experiments, we studied drug interaction(s) among Pitavastatin, Lovastatin, and amlodipine. Results: The results of this study imply that Lovastatin is more potent than Pitavastatin for having comparatively lower EC50 (7.44 × 10-5 ± 0.16 M) in intact and (4.55 × 10-5 ± 0.10 M) in denuded aortae for KCL-induced contractions. Lovastatin amplitudes in intact and denuded aortae for KCL-induced contractions were, respectively, 24% and 35.5%; whereas amplitudes for Pitavastatin in intact and denuded aortae for KCL-induced contractions were 34% and 40%, respectively. A left shift in the EC50 values for the statins was seen when we added amlodipine in EC50 (Log Ca++ M). Right shift for CCRCs state that Pitavastatin and Lovastatin have calcium channel antagonistic effects. Lovastatin in test concentration (6.74 × 10-7 M) produced a right shift in relatively lower EC50 (-2.5 ± 0.10) Log Ca++ M as compared to Pitavastatin, which further confirms that lovastatin is relatively more potent. The right shift in EC50 resembles the right shift of Verapamil. Additive effect of Pitavastatin and Lovastatin was noted in presence of amlodipine (p < 0.05). Conclusions: KCL (80 mM)-induced vascular contractions were relaxed by Pitavastatin and Lovastatin via inhibitory effects on L-Type voltage-gated calcium channels. Lovastatin and Pitavastatin also relaxed Norepinephrine (1 µM)-induced contractions giving an insight for involvement of dual mode of action of Pitavastatin and Lovastatin.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Clinical Trial, Phase III
    目的:依泽替米贝联合他汀类药物治疗是进一步降低低密度脂蛋白胆固醇(LDL-C)水平的有效选择。因此,我们研究了固定剂量的匹伐他汀/依泽替米贝联合用药的长期疗效和安全性(K-924LD:2mg/10mg;K-924HD:4mg/10mg).
    方法:我们进行了第三阶段,多中心,一项开放标签试验,纳入接受匹伐他汀(2或4mg)但未达到LDL-C管理目标的高胆固醇血症患者.根据患者是否接受了匹伐他汀2和4mg,将患者纳入K-924LD和HD组。分别,治疗52周。K-924每天口服一次。主要目的是在第52周检查LDL-C相对于基线的百分比变化,并在所有患者中进行最后观察(LOCF)。
    结果:在评估的109例患者中,62和47被分配到K-924LD和HD组,分别。在所有患者中,LDL-C较基线(134.4±37.9mg/dL)下降-30.3±14.3%(p<0.001)。因此,91.8%和37.5%的一级和二级预防患者达到了LDL-C管理目标,分别。这些结果在K-924LD组和HD组中是一致的。在安全分析中,K-924HD组患者发生单一药物不良反应.
    结论:替换匹伐他汀单药治疗后,发现K-924在52周内有效且耐受性良好。因此,K-924可以在不增加药物数量的情况下加强LDL-C降低治疗。
    OBJECTIVE: Ezetimibe administration with ongoing statin therapy is an effective option for further lowering low-density lipoprotein cholesterol (LDL-C) levels. Thus, we investigated the long-term efficacy and safety of fixed-dose combination of pitavastatin/ezetimibe (K-924 LD: 2 mg/10 mg; K-924 HD: 4 mg/10 mg).
    METHODS: We conducted a phase III, multicenter, open-label trial involving patients with hypercholesterolemia receiving pitavastatin (2 or 4 mg) who had not achieved their LDL-C management target. Patients were enrolled into the K-924 LD and HD groups based on whether they had received pitavastatin 2 and 4 mg, respectively, and treated for 52 weeks. K-924 was administered orally once daily. The primary objective was to examine the percent change in LDL-C from baseline at week 52 with last observation carried forward imputation (LOCF) in all patients.
    RESULTS: Of the 109 patients evaluated, 62 and 47 were assigned to the K-924 LD and HD groups, respectively. In all patients, LDL-C decreased by -30.3±14.3% (p<0.001) from baseline (134.4±37.9 mg/dL). Consequently, 91.8% and 37.5% of the patients for primary and secondary prevention reached their LDL-C management target, respectively. These results were consistent in both the K-924 LD and HD groups. In the safety analysis, a single adverse drug reaction occurred in a patient in the K-924 HD group.
    CONCLUSIONS: After replacing pitavastatin monotherapy, K-924 was found to be effective and well-tolerated over 52 weeks. Thus, K-924 can contribute to intensifying LDL-C-lowering therapy without increasing the number of medications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    本研究旨在研究电生理,闪烁显像术,匹伐他汀对坐骨神经损伤大鼠的组织病理学影响及对功能状态的影响。
    将30只Wistar白化病大鼠分为3组,每组10只:假手术组(无损伤),对照组(神经损伤),和匹伐他汀组(引起神经损伤,口服匹伐他汀2mg/kg,每天1次,共21天)。在干预之前和结束时,采用CatWalk系统进行定量步态分析和坐骨神经传导研究.干预之后,对腓肠肌进行了闪烁扫描评估,并对坐骨神经进行了组织病理学检查。
    干预前和第21天的坐骨神经传导在各组之间没有显着差异(p>0.05)。根据定量步态分析,对照组在个体方面有显着差异,静态,动态,和协调参数(p<0.05)。组织病理学检查显示,两组之间的总髓鞘轴突计数和平均轴突直径存在显着差异(p<0.001)。
    匹伐他汀对坐骨神经损伤大鼠的神经再生和运动功能恢复有效。
    UNASSIGNED: This study aims to investigate the electrophysiological, scintigraphic, and histopathological effects of pitavastatin and its impact on functional status in rats with sciatic nerve injury.
    UNASSIGNED: A total of 30 Wistar albino rats were divided into three equal groups including 10 rats in each group: sham group (no injury), control group (nerve injury induced), and pitavastatin group (nerve injury induced and 2 mg/kg of pitavastatin administered orally once a day for 21 days). Before and at the end of intervention, quantitative gait analysis with the CatWalk system and sciatic nerve conduction studies were performed. After the intervention, the gastrocnemius muscle was scintigraphically evaluated, and the sciatic nerve was histopathologically examined.
    UNASSIGNED: There was no significant difference in the sciatic nerve conduction before the intervention and Day 21 among the groups (p>0.05). According to the quantitative gait analysis, there were significant differences in the control group in terms of the individual, static, dynamic, and coordination parameters (p<0.05). The histopathological examination revealed a significant difference in the total myelinated axon count and mean axon diameter among the groups (p<0.001).
    UNASSIGNED: Pitavastatin is effective in nerve regeneration and motor function recovery in rats with sciatic nerve injury.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:匹伐他汀是一种降胆固醇药物,临床上应用广泛。除了这个效果,匹伐他汀已显示出诱导皮肤鳞状细胞癌(SCC)细胞凋亡的潜力。
    目的:本研究旨在探讨匹伐他汀的作用及可能的作用机制。
    方法:用匹伐他汀处理SCC细胞(SCC12和SCC13细胞),通过Westernblot证实诱导细胞凋亡。为了检查匹伐他汀诱导的细胞凋亡是否与胆固醇合成途径中中间介质数量的减少有关,补充甲羟戊酸后,匹伐他汀诱导的细胞凋亡的变化,角鲨烯,研究了香叶基香叶基焦磷酸(G3GPP)和多立康醇。
    结果:匹伐他汀剂量依赖性诱导皮肤SCC细胞凋亡,但相同浓度的匹伐他汀对正常角质形成细胞的活力没有影响。在补充实验中,添加甲羟戊酸或下游代谢物GGMP抑制了匹伐他汀诱导的细胞凋亡.作为检查对细胞内信号传导的影响的结果,匹伐他汀降低了Yes1相关的转录调节因子和Ras同源家族成员A,并增加了Rac家族小GTP酶1和c-JunN末端激酶(JNK)活性。匹伐他汀对信号分子的所有这些作用在补充有甲羟戊酸或GGb时恢复。此外,匹伐他汀诱导的皮肤SCC细胞凋亡被JNK抑制剂抑制。
    结论:这些结果表明,匹伐他汀通过GPB依赖性JNK激活诱导皮肤SCC细胞凋亡。
    BACKGROUND: Pitavastatin is a cholesterol-lowering drug and is widely used clinically. In addition to this effect, pitavastatin has shown the potential to induce apoptosis in cutaneous squamous cell carcinoma (SCC) cells.
    OBJECTIVE: The purpose of this study is to investigate the effects and possible action mechanisms of pitavastatin.
    METHODS: SCC cells (SCC12 and SCC13 cells) were treated with pitavastatin, and induction of apoptosis was confirmed by Western blot. To examine whether pitavastatin-induced apoptosis is related to a decrease in the amount of intermediate mediators in the cholesterol synthesis pathway, the changes in pitavastatin-induced apoptosis after supplementation with mevalonate, squalene, geranylgeranyl pyrophosphate (GGPP) and dolichol were investigated.
    RESULTS: Pitavastatin dose-dependently induced apoptosis of cutaneous SCC cells, but the viability of normal keratinocytes was not affected by pitavastatin at the same concentrations. In supplementation experiments, pitavastatin-induced apoptosis was inhibited by the addition of mevalonate or downstream metabolite GGPP. As a result of examining the effect on intracellular signaling, pitavastatin decreased Yes1 associated transcriptional regulator and Ras homolog family member A and increased Rac family small GTPase 1 and c-Jun N-terminal kinase (JNK) activity. All these effects of pitavastatin on signaling molecules were restored when supplemented with either mevalonate or GGPP. Furthermore, pitavastatin-induced apoptosis of cutaneous SCC cells was inhibited by a JNK inhibitor.
    CONCLUSIONS: These results suggest that pitavastatin induces apoptosis of cutaneous SCC cells through GGPP-dependent JNK activation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号