Fevipiprant

  • 文章类型: Journal Article
    基于生理的药代动力学和吸收建模已越来越多地用于生物制药应用,以定义药物产品质量属性(如溶解)的安全空间。对于feviprant/QAW039,进行了模拟以评估在开发和扩大到商业规模期间,体外溶出对速释薄膜包衣片剂的体内性能的影响。使用观察到的来自生物等效和非生物等效制剂的临床静脉内和口服PK数据建立了feviprant溶出安全空间。使用片剂的质量控制溶出曲线作为GastroPlus™模型输入以估计胃肠道中的体内溶出并模拟人暴露。该模型用于评估剂型的管腔内性能并预测450mg剂量的吸收率极限。各种口服剂型(150-500毫克)的预测模型性能得到了证明,包括禁食健康成年人的非生物等效批次。为了定义450毫克的安全空间,使用理论溶出曲线进行模拟。立即释放口服固体剂型的Q=80%在60分钟或更短时间内溶解的规格反映了安全空间的边界。450mg商业规模批次的溶出曲线在预期生物等效性的溶出区域内。不接近溶解失败的边缘,为拟议的验收标准提供额外的信心。因此,生物等效批次的安全空间允许大于10%的溶出度差异,取代f2相似性分析。
    Physiologically based pharmacokinetic and absorption modeling has increasingly been implemented for biopharmaceutics applications to define the safe space for drug product quality attributes such as dissolution. For fevipiprant/QAW039, simulations were performed to assess the impact of in vitro dissolution on the in vivo performance of immediate-release film-coated tablets during development and scaling up to commercial scale. A fevipiprant dissolution safe space was established using observed clinical intravenous and oral PK data from bioequivalent and non-bioequivalent formulations. Quality control dissolution profiles with tablets were used as GastroPlus™ model inputs to estimate the in vivo dissolution in the gastrointestinal tract and to simulate human exposure. The model was used to evaluate the intraluminal performance of the dosage forms and to predict the absorption rate limits for the 450 mg dose. The predictive model performance was demonstrated for various oral dosage forms (150‒500 mg), including the non-bioequivalent batches in fasted healthy adults. To define the safe space at 450 mg, simulations were performed using theoretical dissolution profiles. A specification of Q = 80% dissolved in 60 min or less for an immediate-release oral solid dosage form reflected the boundaries of the safe space. The dissolution profile of the 450 mg commercial scale batch was within a dissolution region where bioequivalence is anticipated, not near an edge of failure for dissolution, providing additional confidence to the proposed acceptance criteria. Thus, the safe space allowed for a wider than 10% dissolution difference for bioequivalent batches, superseding f2 similarity analyses.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Fevipiprant是一种非甾体类口服前列腺素D2(PGD2)受体2拮抗剂,可减少支气管壁炎症,可能改善哮喘人群的临床结局。在PubMed上进行了系统的审查搜索,Embase,和中央Cochrane登记处.与安慰剂相比,Fevipiprant作为干预组纳入了随机临床试验。对于连续变量,标准化的平均差,对于离散变量,使用Mantel-Haenszel风险比(MH风险比)进行分析。95%和p值<0.05的置信区间被认为是显著的。分析是使用随机效应模型进行的,不考虑异质性。使用I2统计量评估异质性。共五篇文章,包括七项试验,包括在分析中。支气管扩张剂后一秒钟用力呼气量(FEV1)显著增加0.249(0.157-0.341),p<0.001和支气管扩张剂前FEV10.115(0.043至0.188),p=0.002。哮喘控制问卷(ACQ)评分降低-0.124(-0.187至-0.062),p<0.001。在每日剂量为450mg0.77相对风险(RR)(0.61-0.97)的高嗜酸性粒细胞计数人群中,有统计学意义的哮喘恶化减少。在整个人群中,Fevipiprant450mg剂量对哮喘的减少有正偏差,但没有统计学意义。在高嗜酸性粒细胞计数人群中,Feviprant在哮喘恶化方面产生了轻微的统计学显着降低,在总体人群中出现了有利的偏差。在接受治疗的患者中,它显着增加了支气管扩张剂前和后FEV1,并改善了ACQ评分。的好处,尽管具有统计学意义,未能转化为临床重要性。
    Fevipiprant is a non-steroidal oral prostaglandin D2 (PGD2) receptor 2 antagonist that reduces bronchial wall inflammation, possibly improving clinical outcomes in the asthmatic population. A systemic review search was conducted on PubMed, Embase, and Central Cochrane Registry. Randomized clinical trials were included with Fevipiprant as an intervention arm compared to placebo. For continuous variables, the standardized mean difference, and for discrete variables, Mantel-Haenszel Risk Ratio (MH Risk ratio) was used for analysis. Confidence interval of 95% and p-value < 0.05 was considered significant. The analysis was done using a random-effects model irrespective of heterogeneity. Heterogeneity was evaluated using the I2 statistic. A total of five articles, including seven trials, were included in the analysis. There was significant increase in post-bronchodilator forced expiratory volume in one second (FEV1) 0.249 (0.157-0.341), p<0.001 and pre-bronchodilator FEV1 0.115 (0.043 to 0.188), p=0.002. A decrease in asthma control questionnaire (ACQ) score of -0.124 (-0.187 to -0.062), p<0.001, was reported. Statistically significant asthma exacerbation reduction was reported in the high eosinophil count population with a daily dose of 450mg 0.77 relative risks (RR) (0.61-0.97). There was a positive deviation toward Fevipiprant 450mg dose for asthma reduction in the overall population, but it was not statistically significant. Fevipiprant produced a slight statistically significant reduction in asthma exacerbations in the high eosinophil count population with favorable deviation in the overall population. It significantly increased pre-and post-bronchodilator FEV1 and improved ACQ scores in treated patients. The benefits, though statistically significant, failed to translate into clinical importance.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    从免疫细胞或其他细胞类型释放的前列腺素D2(PGD2)激活其受体,D类前列腺素受体(DP)1和2(DP1和DP2),促进过敏性和肺部疾病的炎症反应。前列腺素介导的炎症也可能导致血管疾病,例如腹主动脉瘤(AAA)。然而,DP受体在AAA发病机制中的作用尚未得到系统研究。在本研究中,在两种不同的AAA形成小鼠模型中测试了DP1缺陷小鼠和DP1或DP2的药理学抑制剂:血管紧张素II(AngII)输注和氯化钙(CaCl2)应用。DP1缺陷小鼠[杂合(DP1+/-)和纯合(DP1-/-)]均被保护免受CaCl2诱导的AAA形成,与基质金属肽酶(MMP)活性降低和外膜炎症细胞浸润有关。在AngII输液模型中,DP1+/-小鼠,但不是DP1-/-小鼠,表现出减少的AAA形成。有趣的是,在DP1-/-小鼠中检测到对AngII输注的DP2受体的代偿性上调,提示DP2受体在AAA中的潜在作用。用DP1(laropiprant)或DP2(feviprant)的选择性拮抗剂治疗,防止AAA形成,与减少的弹性蛋白降解和主动脉炎症反应。总之,PGD2信号有助于小鼠的AAA形成,表明DP受体拮抗剂,已经在过敏性和肺部疾病中进行了广泛的测试,可能是改善AAA的有希望的候选人。
    Prostaglandin D2 (PGD2) released from immune cells or other cell types activates its receptors, D prostanoid receptor (DP)1 and 2 (DP1 and DP2), to promote inflammatory responses in allergic and lung diseases. Prostaglandin-mediated inflammation may also contribute to vascular diseases such as abdominal aortic aneurysm (AAA). However, the role of DP receptors in the pathogenesis of AAA has not been systematically investigated. In the present study, DP1-deficient mice and pharmacological inhibitors of either DP1 or DP2 were tested in two distinct mouse models of AAA formation: angiotensin II (AngII) infusion and calcium chloride (CaCl2) application. DP1-deficient mice [both heterozygous (DP1+/-) and homozygous (DP1-/-)] were protected against CaCl2-induced AAA formation, in conjunction with decreased matrix metallopeptidase (MMP) activity and adventitial inflammatory cell infiltration. In the AngII infusion model, DP1+/- mice, but not DP1-/- mice, exhibited reduced AAA formation. Interestingly, compensatory up-regulation of the DP2 receptor was detected in DP1-/- mice in response to AngII infusion, suggesting a potential role for DP2 receptors in AAA. Treatment with selective antagonists of DP1 (laropiprant) or DP2 (fevipiprant) protected against AAA formation, in conjunction with reduced elastin degradation and aortic inflammatory responses. In conclusion, PGD2 signaling contributes to AAA formation in mice, suggesting that antagonists of DP receptors, which have been extensively tested in allergic and lung diseases, may be promising candidates to ameliorate AAA.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Clinical Trial, Phase III
    慢性鼻窦炎伴鼻息肉(CRSwNP)与哮喘有关,尤其是晚期发作。目前CRSwNP的治疗方案有局限性,对其他安全有效的疗法的需求尚未得到满足。
    THUNDER研究的目的是确定前列腺素D2受体2(DP2)拮抗剂feviprant在CRSwNP和合并哮喘患者中的疗效和安全性。通过改善鼻息肉评分(主要终点)来衡量,鼻塞评分,鼻窦结果测试22分,和宾夕法尼亚大学气味鉴定测试成绩。
    雷声是3b阶段,随机化,多中心,双盲,安慰剂对照,平行组,feviprant150mg或450mg每日一次与安慰剂的16周研究。所有患者每天200μg鼻内接受糠酸莫米松。
    98例患者被随机分配到feviprant150mg(n=32),fevipiprant450毫克(n=34),或安慰剂(n=32)。对于feviprant150mg,第16周鼻息肉评分相对于基线的平均(SE)变化为0.20(0.224),对于feviprant450mg,-0.10(0.216),安慰剂为0.14(0.233)。平均治疗差异为0.05(95%置信区间,-0.59,0.70;调整后的P=.979)对于feviprant150mg与安慰剂和-0.25(95%置信区间,-0.88,0.39;调整后的P=.656)对于feviprant450mg与安慰剂。feviprant与安慰剂的次要终点没有有意义的差异。
    THUNDER没有提供任何证据证明fevipiprant在治疗CRSwNP和哮喘患者中的作用;未来的研究可能会确立其他DP2拮抗剂的作用,特别是阿司匹林加重的呼吸系统疾病患者。
    Chronic rhinosinusitis with nasal polyps (CRSwNP) is associated with asthma, particularly of late onset. Current treatment options for CRSwNP have limitations, and there is an unmet need for other safe and effective therapies.
    The aim of the THUNDER study was to determine the efficacy and safety of the prostaglandin D2 receptor 2 (DP2) antagonist fevipiprant in patients with CRSwNP and concomitant asthma, measured by improvement in nasal polyp score (primary end point), nasal congestion score, Sinonasal Outcome Test 22 score, and University of Pennsylvania Smell Identification Test score.
    THUNDER was a phase 3b, randomized, multicenter, double-blind, placebo-controlled, parallel-group, 16-week study of fevipiprant 150 mg or 450 mg once daily versus placebo. All patients received intranasal mometasone furoate 200 μg daily.
    Ninety-eight patients were randomly assigned to fevipiprant 150 mg (n = 32), fevipiprant 450 mg (n = 34), or placebo (n = 32). Mean (SE) change from baseline in nasal polyp score at week 16 was 0.20 (0.224) for fevipiprant 150 mg, -0.10 (0.216) for fevipiprant 450 mg, and 0.14 (0.233) for placebo. Mean treatment difference was 0.05 (95% confidence interval, -0.59, 0.70; adjusted P = .979) for fevipiprant 150 mg versus placebo and -0.25 (95% confidence interval, -0.88, 0.39; adjusted P = .656) for fevipiprant 450 mg versus placebo. There was no meaningful difference in the secondary end points for fevipiprant versus placebo.
    THUNDER provided no evidence of a role for fevipiprant in the treatment of patients with CRSwNP and asthma; future studies may establish a role for other DP2 antagonists, specifically in patients with aspirin-exacerbated respiratory disease.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Clinical Trial, Phase III
    BACKGROUND: The prostaglandin D2 (PGD2) receptor 2 (DP2 receptor) pathway is an important regulator of the inflammatory cascade in asthma, which can be stimulated by allergic or non-allergic triggers. Fevipiprant is an oral, non-steroidal, highly selective, reversible antagonist of the DP2 receptor that inhibits the binding of PGD2 and its metabolites.
    METHODS: SPIRIT, a 2-treatment period (52-week, double-blind and optional 104-week single-blind), randomised, placebo-controlled, multicentre, parallel-group study, assessed the long-term safety of fevipiprant (150 mg and 450 mg o.d.) added to standard of care in patients ≥ 12 years with uncontrolled asthma. Stratified block randomisation was used. Patients were randomised in an approximate ratio of 3:3:1 (fevipiprant 150 mg, fevipiprant 450 mg or placebo). Patients were either newly enrolled or had participated in a previous fevipiprant Phase 3 trial. Primary endpoints were: time-to-first treatment emergent adverse event (AE); serious AE; and AE leading to discontinuation from study treatment. Data from both treatment periods were combined for analyses. Data were collected during study site visits.
    RESULTS: In total, 1093 patients were randomised to receive fevipiprant 150 mg, 1085 to fevipiprant 450 mg, and 360 to placebo. Overall, 1184 patients had ≥ 52 weeks\' treatment, while 163 received ≥ 104 weeks\' treatment. Both doses were well tolerated, with a safety profile similar to placebo both in new patients and in those enrolled from previous studies. In exploratory analyses, reduced rates of moderate-to-severe asthma exacerbations, increased time-to-first moderate-to-severe asthma exacerbation and improved FEV1 were observed for both doses of fevipiprant versus placebo; these were without multiplicity adjustment and should be interpreted with caution. SPIRIT was terminated early, on 16 December 2019, by the Sponsor.
    CONCLUSIONS: In patients with uncontrolled asthma, the addition of fevipiprant had a favourable long-term safety profile.
    BACKGROUND: Clinicaltrials.gov, NCT03052517, prospectively registered 23 January 2017, https://clinicaltrials.gov/ct2/show/NCT03052517 .
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Letter
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Comparative Study
    背景:通过前列腺素D2受体2(DP2)的前列腺素D2(PGD2)信号传导有助于特应性和非特应性哮喘。抑制DP2在某些哮喘患者亚群中显示出治疗益处,改善嗜酸性粒细胞气道炎症。PGD2代谢物通过DP2信号延长哮喘患者的炎症反应。PGD2代谢物对嗜酸性粒细胞和ILC2活性的作用尚未完全理解。
    方法:从特应性哮喘患者外周血中分离嗜酸性粒细胞和ILC2s。嗜酸性粒细胞形状改变,在存在或不存在选择性DP2拮抗剂feviprant下用7种PGD2代谢物刺激后,测量ILC2迁移和IL-5/IL-13细胞因子分泌。
    结果:除9α外,选定的代谢物诱导的嗜酸性粒细胞形状改变具有相似的纳摩尔效力,11β-PGF2。代谢物之间嗜酸性粒细胞前向散射的最大值相当。除9α外,ILC2在选定代谢物存在下剂量依赖性迁移,11β-PGF2,EC50值为17.4至91.7nM。与PGD2相比,在Δ12-PGD2,15-脱氧-Δ12,14-PGD2,PGJ2,Δ12-PGJ2和15-脱氧-Δ12,14-PGJ2的存在下,绝对细胞迁移得到增强。ILC2细胞因子的产生也是剂量依赖性的,但与细胞迁移相比,其平均效力降低了六倍(IL-5范围为108.1至526.9nM,IL-13范围:125.2至788.3nM)。与PGD2相比,在大多数代谢物存在下,细胞因子的绝对分泌减少。Fevipiprant剂量依赖性地抑制嗜酸性粒细胞形状改变,具有(亚)纳摩尔效力的ILC2迁移和ILC2细胞因子分泌。
    结论:前列腺素D2代谢物以DP2依赖性方式启动ILC2迁移和IL-5和IL-13细胞因子分泌。我们的数据表明,代谢物对于体内嗜酸性粒细胞活化和ILC2迁移可能很重要,而对于ILC2细胞因子分泌的程度较小。
    BACKGROUND: Prostaglandin D2 (PGD2) signaling via prostaglandin D2 receptor 2 (DP2) contributes to atopic and non-atopic asthma. Inhibiting DP2 has shown therapeutic benefit in certain subsets of asthma patients, improving eosinophilic airway inflammation. PGD2 metabolites prolong the inflammatory response in asthmatic patients via DP2 signaling. The role of PGD2 metabolites on eosinophil and ILC2 activity is not fully understood.
    METHODS: Eosinophils and ILC2s were isolated from peripheral blood of atopic asthmatic patients. Eosinophil shape change, ILC2 migration and IL-5/IL-13 cytokine secretion were measured after stimulation with seven PGD2 metabolites in presence or absence of the selective DP2 antagonist fevipiprant.
    RESULTS: Selected metabolites induced eosinophil shape change with similar nanomolar potencies except for 9α,11β-PGF2. Maximal values in forward scatter of eosinophils were comparable between metabolites. ILC2s migrated dose-dependently in the presence of selected metabolites except for 9α,11β-PGF2 with EC50 values ranging from 17.4 to 91.7 nM. Compared to PGD2, the absolute cell migration was enhanced in the presence of Δ12-PGD2, 15-deoxy-Δ12,14-PGD2, PGJ2, Δ12-PGJ2 and 15-deoxy-Δ12,14-PGJ2. ILC2 cytokine production was dose dependent as well but with an average sixfold reduced potency compared to cell migration (IL-5 range 108.1 to 526.9 nM, IL-13 range: 125.2 to 788.3 nM). Compared to PGD2, the absolute cytokine secretion was reduced in the presence of most metabolites. Fevipiprant dose-dependently inhibited eosinophil shape change, ILC2 migration and ILC2 cytokine secretion with (sub)-nanomolar potencies.
    CONCLUSIONS: Prostaglandin D2 metabolites initiate ILC2 migration and IL-5 and IL-13 cytokine secretion in a DP2 dependent manner. Our data indicate that metabolites may be important for in vivo eosinophil activation and ILC2 migration and to a lesser extent for ILC2 cytokine secretion.
    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
    Accumulating evidence has shown that prostaglandin D2 (PGD2)-chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2) pathway plays an important role in promoting eosinophilic airway inflammation in asthma. We aimed to assess the efficacy and safety of CRTH2 antagonist fevipiprant in patients with persistent asthma compared with placebo.
    We identified eligible studies by searching PubMed, EMBASE, the Cochrane Central Register of Controlled Trials and ClinicalTrials.gov. The study was registered as CRD 42020221714 ( http://www.crd.york.ac.uk/PROSPERO ). Ten randomized controlled trials with 7902 patients met our inclusion criteria. A statistically significant benefit of fevipiprant compared with placebo was shown in improving forced expiratory volume in 1 s (MD 0.05 L, 95% CI: 0.02 to 0.07; p < 0.0001), Asthma Control Questionnaire score (MD -0.10, 95% CI: -0.16 to -0.04; p = 0.001), and Asthma Quality of Life Questionnaire score (MD 0.08, 95% CI: 0.03 to 0.13; p = 0.003). Fevipiprant decreased number of patients with at least one asthma exacerbation requiring administration of systemic corticosteroids for 3 days or more (RR 0.86, 95% CI: 0.77 to 0.97; p = 0.01). Some benefits were a little more pronounced in the high eosinophil population (with an elevated blood eosinophil count or sputum eosinophil percentage) and in the 450 mg dose group. Fevipiprant was well tolerated with no safety issues compared with placebo. Fevipiprant could safely improve asthma outcomes compared to placebo. However, most of the differences didn\'t reach the minimal clinically important difference (MCID), thus the clinical benefits remained to be confirmed.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Fevipiprant is an oral, non-steroidal, highly selective, reversible antagonist of the prostaglandin D2 (DP2) receptor. The DP2 receptor is a mediator of inflammation expressed on the membrane of key inflammatory cells, including eosinophils, Th2 cells, type 2 innate lymphoid cells, CD8+ cytotoxic T cells, basophils and monocytes, as well as airway smooth muscle and epithelial cells. The DP2 receptor pathway regulates the allergic and non-allergic asthma inflammatory cascade and is activated by the binding of prostaglandin D2. Fevipiprant is metabolised by several uridine 5\'-diphospho glucuronosyltransferase enzymes to an inactive acyl-glucuronide (AG) metabolite, the only major human metabolite. Both fevipiprant and its AG metabolite are eliminated by urinary excretion; fevipiprant is also possibly cleared by biliary excretion. These parallel elimination pathways suggested a low risk of major drug-drug interactions (DDI), pharmacogenetic or ethnic variability for fevipiprant, which was supported by DDI and clinical studies of fevipiprant. Phase II clinical trials of fevipiprant showed reduction in sputum eosinophilia, as well as improvement in lung function, symptoms and quality of life in patients with asthma. While fevipiprant reached the most advanced state of development to date of an oral DP2 receptor antagonist in a worldwide Phase III clinical trial programme, the demonstrated efficacy did not support further clinical development in asthma.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号