Axitinib

阿西替尼
  • 文章类型: Journal Article
    目的:探讨血管内皮生长因子(VEGF)在糖尿病足溃疡(DFU)创面愈合中的作用及其调控机制。
    方法:链脲佐菌素诱导的糖尿病大鼠建立DFU动物模型。VEGF和阿西替尼(VEGFR的特异性抑制剂)用于体内治疗。对不同时间点的伤口进行成像,并通过苏木精和伊红(H&E)染色和Masson三色染色对伤口进行组织学分析。进行免疫组织化学染色以检查伤口中CD31和eNOS的表达。免疫荧光法和实时定量PCR检测巨噬细胞标志物。此外,THP-1分化为巨噬细胞,然后用白细胞介素(IL)-4诱导M2巨噬细胞,其次是VEGF治疗。收集来自VEGF介导的巨噬细胞的条件培养基(CM)以培养人真皮成纤维细胞(HDF)。通过细胞计数试剂盒(CCK)-8,伤口愈合和Transwell测定来测量细胞活力和迁移,分别。
    结果:VEGF处理显著加速DFU大鼠的伤口愈合。VEGF促进胶原沉积,CD31和eNOS表达升高,证实了大鼠糖尿病伤口周围VEGF的促血管生成。同时,VEGF限制促炎细胞因子和增加F4/80和CD206表达,在DFU大鼠的糖尿病伤口中,VEGF治疗后,突出了活化的巨噬细胞和增强的M2巨噬细胞。然而,阿西替尼在DFU大鼠中发挥与VEGF相反的功能。此外,VEGF在体外直接促进巨噬细胞向M2表型极化,来自VEGF介导的M2巨噬细胞的CM显著促进HDFs的增殖,迁移和胶原沉积。
    结论:VEGF可能通过促进M2巨噬细胞极化和成纤维细胞迁移促进DFU创面愈合。
    OBJECTIVE: The objective was to investigate the specific role and the regulatory mechanism of vascular endothelial growth factor (VEGF) during wound healing in diabetic foot ulcer (DFU).
    METHODS: Streptozotocin-induced diabetic rats were used to establish a DFU animal model. VEGF and Axitinib (a specific inhibitor of VEGFR) were used for treatment in vivo. The wounds at different time points were imaged and histological analysis of the wounds were performed by haematoxylin and eosin (H&E) staining and Masson\'s trichrome staining. Immunohistochemical staining was conducted to examine CD31 and eNOS expression in the wounds. Immunofluorescence assay and quantitative real-time PCR were performed to examine macrophage markers. In addition, THP-1 was differentiated to macrophages, and then treated with interleukin (IL)-4 to induce M2 macrophages, followed by VEGF treatment. The conditional medium (CM) from VEGF-mediated macrophages were collected to culture human dermal fibroblasts (HDFs). Cell viability and migration were measured by Cell Counting Kit (CCK)-8, wound-healing and Transwell assays, respectively.
    RESULTS: VEGF treatment remarkably accelerated wound healing of DFU rats. VEGF promoted collagen deposition and elevated CD31 and eNOS expression, confirming the pro-angiogenesis of VEGF around diabetic wound in rats. Meanwhile, VEGF restricted pro-inflammatory cytokines and increased F4/80 and CD206 expression, highlighting the activated macrophages and enhanced M2 macrophages following VEGF treatment in diabetic wounds of DFU rats. However, Axitinib exerted an opposite function to VEGF in DFU rats. Moreover, VEGF directly promoted macrophage polarization toward M2 phenotype in vitro, and the CM from VEGF-mediated M2 macrophages markedly promoted HDFs proliferation, migration and collagen deposition.
    CONCLUSIONS: VEGF might accelerate the wound healing of DFU through promoting M2 macrophage polarization and fibroblast migration.
    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
    目的:急性缺血性卒中(AIS)的病理生理特征常涉及血脑屏障(BBB)功能异常,其特征在于紧密连接蛋白(Tjs)的降解导致渗透性增加。这种功能障碍会加剧脑损伤并导致严重的并发症。BBB的渗透率在AIS的不同阶段波动,并受多种因素的影响。开发恢复BBB功能的有效疗法仍然是AIS治疗中的重大挑战。在AIS的早期阶段,高水平的血管内皮生长因子(VEGF)已被证明会加剧BBB破坏和中风进展。我们的研究旨在探讨VEGF受体抑制剂阿西替尼对BBB功能障碍和脑缺血/再灌注损伤的保护作用。
    方法:构建暴露于氧-葡萄糖剥夺(OGD)的BEnd3细胞模型,以评估阿西替尼(400ng/ml)抗凋亡和病理屏障功能恢复的药理活性。在体内,对大鼠进行1h短暂大脑中动脉闭塞和23h再灌注(tMCAO/R),以研究BBB的通透性和脑组织损伤。在再灌注开始时通过尾静脉施用阿西替尼。通过Evans蓝渗漏和Tjsclaudin-5和occludin的表达水平评估BBB完整性。
    结果:我们的研究表明,与阿西替尼共同孵育可增强OGD损伤的bEnd3细胞的细胞活力,降低LDH泄漏率,并抑制凋亡相关蛋白细胞色素C和Bax的表达。阿西替尼还减轻了对Tjs的损害,并促进了OGD损伤的bEnd.3细胞中跨上皮电阻的恢复。在体内,阿西替尼给药减少了tMCAO/R大鼠脑内伊文思蓝渗漏并上调了Tjs在半暗带脑组织中的表达。值得注意的是,10mg/kg阿西替尼通过减少脑梗死体积和脑水肿体积发挥显著的抗缺血作用。改善神经功能,并减少脑中的促炎细胞因子IL-6和TNF-α。
    结论:我们的研究强调了阿西替尼作为血脑屏障功能的有效保护剂,能够通过抑制VEGF和增加AIS中紧密连接蛋白的表达来促进病理性血脑屏障的恢复。这表明卒中后最初24小时内的VEGF拮抗作用可能是一种新的治疗方法,可以增强血脑屏障功能并减轻缺血再灌注损伤。
    OBJECTIVE: The pathophysiological features of acute ischemic stroke (AIS) often involve dysfunction of the blood-brain barrier (BBB), characterized by the degradation of tight junction proteins (Tjs) leading to increased permeability. This dysfunction can exacerbate cerebral injury and contribute to severe complications. The permeability of the BBB fluctuates during different stages of AIS and is influenced by various factors. Developing effective therapies to restore BBB function remains a significant challenge in AIS treatment. High levels of vascular endothelial growth factor (VEGF) in the early stages of AIS have been shown to worsen BBB breakdown and stroke progression. Our study aimed to investigate the protective effects of the VEGF receptor inhibitor Axitinib on BBB dysfunction and cerebral ischemia/reperfusion-induced injury.
    METHODS: BEnd3 cell exposed to oxygen-glucose deprivation (OGD) model was constructed to estimate pharmacological activity of Axitinib (400 ng/ml) on anti-apoptosis and pathological barrier function recovery. In vivo, rats were subjected to a 1 h transient middle cerebral artery occlusion and 23 h reperfusion (tMCAO/R) to investigate the permeability of BBB and cerebral tissue damage. Axitinib was administered through the tail vein at the beginning of reperfusion. BBB integrity was assessed by Evans blue leakage and the expression levels of Tjs claudin-5 and occludin.
    RESULTS: Our research revealed that co-incubation with Axitinib enhanced the cell viability of OGD-insulted bEnd3 cells, decreased LDH leakage rate, and suppressed the expression of apoptosis-related proteins cytochrome C and Bax. Axitinib also mitigated the damage to Tjs and facilitated the restoration of transepithelial electrical resistance in OGD-insulted bEnd.3 cells. In vivo, Axitinib administration reduced intracerebral Evans blue leakage and up-regulated the expression of Tjs in the penumbra brain tissue in tMCAO/R rats. Notably, 10 mg/kg Axitinib exerted a significant anti-ischemic effect by decreasing cerebral infarct volume and brain edema volume, improving neurological function, and reducing pro-inflammatory cytokines IL-6 and TNF-α in the brain.
    CONCLUSIONS: Our study highlights Axitinib as a potent protectant of blood-brain barrier function, capable of promoting pathological blood-brain barrier recovery through VEGF inhibition and increased expression of tight junction proteins in AIS. This suggests that VEGF antagonism within the first 24 h post-stroke could be a novel therapeutic approach to enhance blood-brain barrier function and mitigate ischemia-reperfusion injury.
    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
    背景:阿西替尼和免疫检查点抑制剂(ICIs)的组合在晚期肾细胞癌(RCC)的治疗中显示出有希望的疗效。本研究旨在前瞻性地评估其安全性,功效,和新辅助托里帕利马联合阿西替尼在非转移性透明细胞肾癌中的生物标志物。
    方法:这是一个单一的机构,单臂II期临床试验。纳入具有非转移性活检证实的透明细胞RCC(T2-T3N0-1M0)的患者。患者将接受阿西替尼5mg,每天两次,每3周(三个周期)联合使用托里帕利马240mg,持续12周。然后,患者将在新辅助治疗后接受部分(PN)或根治性肾切除术(RN)。主要终点是客观反应率(ORR)。次要终点包括无病生存率,安全,围手术期并发症发生率。预测性生物标志物参与探索性分析。
    结果:共有20名患者被纳入研究,其中19人正在接受手术。一名患者拒绝手术。主要终点ORR为45%。πORR的后验分布均值为0.44(95%可信区间:0.24-0.64),满足预定义的主端点,ORR为32%。在肾切除术前,95%的患者观察到肿瘤缩小。此外,4例患者达到病理完全缓解.25%的患者发生≥3级不良事件,包括高血压,高血糖症,谷丙转氨酶/谷草转氨酶(ALT/AST)增加,和蛋白尿。术后,观察到1例4a级和8例1-2级并发症.与病情稳定的患者相比,应答者在精氨酸酶1(ARG1)等免疫因子上表现出显著差异,黑色素瘤抗原(MAGEs),树突状细胞(DC),TNF超家族成员13(TNFSF13),Apelin受体(APLNR),和C-C基序趋化因子配体3如1(CCL3-L1)。该试验的局限性在于样本量小。
    结论:新佐剂托里帕利单抗联合阿西替尼在局部晚期透明细胞RCC中显示出令人鼓舞的活性和可接受的毒性,值得进一步研究。
    背景:clinicaltrials.gov,NCT04118855。
    BACKGROUND: A combination of axitinib and immune checkpoint inhibitors (ICIs) demonstrated promising efficacy in the treatment of advanced renal cell carcinoma (RCC). This study aims to prospectively evaluate the safety, efficacy, and biomarkers of neoadjuvant toripalimab plus axitinib in non-metastatic clear cell RCC.
    METHODS: This is a single-institution, single-arm phase II clinical trial. Patients with non-metastatic biopsy-proven clear cell RCC (T2-T3N0-1M0) are enrolled. Patients will receive axitinib 5 mg twice daily combined with toripalimab 240 mg every 3 weeks (three cycles) for up to 12 weeks. Patients then will receive partial (PN) or radical nephrectomy (RN) after neoadjuvant therapy. The primary endpoint is objective response rate (ORR). Secondary endpoints include disease-free survival, safety, and perioperative complication rate. Predictive biomarkers are involved in exploratory analysis.
    RESULTS: A total of 20 patients were enrolled in the study, with 19 of them undergoing surgery. One patient declined surgery. The primary endpoint ORR was 45%. The posterior distribution of πORR had a mean of 0.44 (95% credible intervals: 0.24-0.64), meeting the predefined primary endpoint with an ORR of 32%. Tumor shrinkage was observed in 95% of patients prior to nephrectomy. Furthermore, four patients achieved a pathological complete response. Grade ≥3 adverse events occurred in 25% of patients, including hypertension, hyperglycemia, glutamic pyruvic transaminase/glutamic oxaloacetic transaminase (ALT/AST) increase, and proteinuria. Postoperatively, one grade 4a and eight grade 1-2 complications were noted. In comparison to patients with stable disease, responders exhibited significant differences in immune factors such as Arginase 1(ARG1), Melanoma antigen (MAGEs), Dendritic Cell (DC), TNF Superfamily Member 13 (TNFSF13), Apelin Receptor (APLNR), and C-C Motif Chemokine Ligand 3 Like 1 (CCL3-L1). The limitation of this trial was the small sample size.
    CONCLUSIONS: Neoadjuvant toripalimab combined with axitinib shows encouraging activity and acceptable toxicity in locally advanced clear cell RCC and warrants further study.
    BACKGROUND: clinicaltrials.gov, NCT04118855.
    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
    一种商业抗癌药物,阿西替尼,表现出非常稳定的双重排放的辨别人血清白蛋白。
    A commercial anti-cancer drug, axitinib, exhibits very stable dual emissions for discrimination of human serum albumin.
    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
    目的:免疫检查点抑制剂(ICIs)加酪氨酸激酶抑制剂(TKIs)已成为转移性肾细胞癌患者的一线治疗方法。本研究旨在探讨肿瘤浸润B淋巴细胞(TIBs)对联合治疗的影响。
    方法:对2020年3月至2023年6月期间接受抗PD-1抗体联合阿西替尼治疗的115例转移性透明细胞肾细胞癌(mccRCC)患者的临床记录进行回顾性分析。观察目标:客观反应率(ORR),和总生存率(OS),无进展生存期(PFS)和免疫谱。
    结果:高TIB患者预示联合治疗的ORR较低(p=0.033)。TIB是联合治疗mccRCC患者OS(p=0.013)和PFS(p=0.021)较差的独立预测因子。TIBs浸润与更多的CD4+T相关(p<0.001),CD8+T(p<0.001),M2巨噬细胞(p=0.020)和调节性T细胞(Tregs)(p=0.004)。在TIBs高患者中,CD4+T细胞(p=0.038、0.029和0.002)和CD8+T细胞(p=0.006、0.026和<0.001)中PD-1、CTLA-4和TIM-3阳性率显著升高。
    结论:我们的研究显示TIBs浸润预测了抗PD-1抗体联合阿西替尼治疗mccRCC患者的不良结局。作为一个推论,TIB与M2巨噬细胞和Tregs呈正相关,导致随后的多个免疫检查点相关的T细胞耗尽。因此,仅PD-1阻断不足以有效逆转高TIBsmccRCC患者的T细胞耗竭。
    OBJECTIVE: Immune checkpoint inhibitors (ICIs) plus tyrosine kinase inhibitors (TKIs) has become first-line therapy for metastatic renal cell carcinoma patients. This study aims to investigate the effect of tumor infiltrating B lymphocytes (TIBs) on the combination therapy.
    METHODS: The retrospective analysis was conducted on the clinical records of 115 metastatic clear cell renal cell carcinoma (mccRCC) patients treated with anti-PD-1 antibody plus Axitinib between March 2020 and June 2023. Observation target: objective response rate (ORR), and overall survival (OS), progression-free survival (PFS) and immune profile.
    RESULTS: Patients with high TIBs portended lower ORR of the combination therapy (p = 0.033). TIBs was an independent predictor for poorer OS (p = 0.013) and PFS (p = 0.021) in mccRCC patients with combination treatment. TIBs infiltration was associated with more CD4+T (p < 0.001), CD8+T (p < 0.001), M2 macrophages (p = 0.020) and regulatory T cells (Tregs) (p = 0.004). In TIBs high patients, the percentages of PD-1, CTLA-4 and TIM-3 positive rate were significantly increased in CD4+T (p = 0.038, 0.029 and 0.002 respectively) and CD8+T cells (p = 0.006, 0.026 and < 0.001 respectively).
    CONCLUSIONS: Our study revealed TIBs infiltration predicted adverse outcomes in mccRCC patients treated with anti-PD-1 antibody plus Axitinib. As a corollary, TIBs positively associated with M2 macrophages and Tregs, leading to subsequent multiple immune checkpoints related exhaustion of T cells. Thus, only PD-1 blockade are inadequate to reverse T cells exhaustion effectively in high TIBs mccRCC patients.
    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
    在我们最近的研究中,我们探讨了三维(3D)测量肿瘤体积在预测肾细胞癌(RCC)患者生活质量(QoL)改善中的功效,接受阿西替尼和抗PD-L1抗体治疗的患者。这项研究包括18例RCC患者,包括10名男性和8名女性,平均年龄56.83±9.94岁。通过利用3D切片器软件,我们分析了治疗前后的CT扫描,以评估肿瘤体积的变化.通过FKSI-DRS问卷评估患者的QoL。我们的研究结果表明,所有患者的3D模型都被成功创建,基于RECIST1.1标准的治疗应答分类与体积分析之间存在中度一致性(kappa=0.556,p=0.001).值得注意的是,9例患者报告治疗后QoL有临床意义的改善.有趣的是,与CT测量的直径变化相比,3D模型显示的肿瘤体积变化在预测QoL改善方面显示出更高的曲线下面积,尽管这种差异没有统计学意义(z=0.593,p=0.553).此外,一项多变量分析将基于3D模型的肿瘤体积变化确定为QoL改善的独立预测因子(比值比=1.073,95%CI1.002-1.149,p=0.045).总之,我们的研究表明,与传统的基于CT的直径测量相比,通过3D模型测量的肿瘤体积变化可能更有效地预测RCC患者的症状改善.这提供了一种评估治疗反应和患者健康状况的新方法,在RCC治疗领域取得了重大进展。
    In our recent study, we explored the efficacy of three-dimensional (3D) measurement of tumor volume in predicting the improvement of quality of life (QoL) in patients suffering from renal cell cancer (RCC), who were treated with axitinib and anti-PD-L1 antibodies. This study encompassed 18 RCC patients, including 10 men and 8 women, with an average age of 56.83 ± 9.94 years. By utilizing 3D Slicer software, we analyzed pre- and post-treatment CT scans to assess changes in tumor volume. Patients\' QoL was evaluated through the FKSI-DRS questionnaire. Our findings revealed that 3D models for all patients were successfully created, and there was a moderate agreement between treatment response classifications based on RECIST 1.1 criteria and volumetric analysis (kappa = 0.556, p = 0.001). Notably, nine patients reported a clinically meaningful improvement in QoL following the treatment. Interestingly, the change in tumor volume as indicated by the 3D model showed a higher area under the curve in predicting QoL improvement compared to the change in diameter measured by CT, although this difference was not statistically significant (z = 0.593, p = 0.553). Furthermore, a multivariable analysis identified the change in tumor volume based on the 3D model as an independent predictor of QoL improvement (odds ratio = 1.073, 95% CI 1.002-1.149, p = 0.045).In conclusion, our study suggests that the change in tumor volume measured by a 3D model may be a more effective predictor of symptom improvement in RCC patients than traditional CT-based diameter measurements. This offers a novel approach for assessing treatment response and patient well-being, presenting a significant advancement in the field of RCC 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
    儿科神经母细胞瘤以及许多其他癌症的治疗策略受到不利的肿瘤微环境(TME)的限制。在这项研究中,神经母细胞瘤的TMEs根据其遗传特征分为四个不同的亚型:免疫富集,免疫沙漠,非增生性和纤维化。根据亚型的分子特征构建免疫评分和增殖评分,以量化神经母细胞瘤中癌细胞的免疫微环境或恶性程度。分别。免疫评分与患者对免疫疗法的反应相关;增殖评分是神经母细胞瘤的独立预后生物标志物,并且被证明比现有的临床预测因子更准确。进一步验证了该双重评分系统,并证实了与免疫景观和恶性程度相关的保守分子模式。通过双重评分系统,阿西替尼和BI-2536被确认为神经母细胞瘤的候选药物。体内和体外实验均表明,阿西替尼诱导的神经母细胞瘤细胞的焦亡激活了抗肿瘤免疫力并抑制了肿瘤生长;BI-2536在神经母细胞瘤细胞中诱导细胞周期停滞在S期。神经母细胞瘤的综合双重评分系统可以预测预后并筛选治疗策略,从而提供个性化治疗。
    Treatment strategies for paediatric neuroblastoma as well as many other cancers are limited by the unfavourable tumour microenvironment (TME). In this study, the TMEs of neuroblastoma were grouped by their genetic signatures into four distinct subtypes: immune enriched, immune desert, non-proliferative and fibrotic. An Immune Score and a Proliferation Score were constructed based on the molecular features of the subtypes to quantify the immune microenvironment or malignancy degree of cancer cells in neuroblastoma, respectively. The Immune Score correlated with a patient\'s response to immunotherapy; the Proliferation Score was an independent prognostic biomarker for neuroblastoma and proved to be more accurate than the existing clinical predictors. This double scoring system was further validated and the conserved molecular pattern associated with immune landscape and malignancy degree was confirmed. Axitinib and BI-2536 were confirmed as candidate drugs for neuroblastoma by the double scoring system. Both in vivo and in vitro experiments demonstrated that axitinib-induced pyroptosis of neuroblastoma cells activated anti-tumour immunity and inhibited tumour growth; BI-2536 induced cell cycle arrest at the S phase in neuroblastoma cells. The comprehensive double scoring system of neuroblastoma may predict prognosis and screen for therapeutic strategies which could provide personalized treatments.
    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
    抗血管生成治疗是破坏营养供应和饥饿肿瘤的有效方法,但它受到疗效差和负反馈诱导的肿瘤复发的限制。在这项研究中,据报道,神经纤毛蛋白-1(NRP-1)靶向纳米药物(命名为FPPT@Axi)通过结合光动力疗法(PDT)和抗血管生成来抑制时空肿瘤。简而言之,FPPT@Axi通过利用靶向NRP-1的嵌合肽(Fmoc-K(PpIX)-PEG8-TKPRR)包封抗血管生成药物阿西替尼(Axitinib)来制备。重要的是,NRP-1介导的靶向特性使FPPT@Axi能够选择性地集中在血管内皮细胞和乳腺癌细胞,促进原位活性氧(ROS)的产生,以进行特定的血管破坏,并在光刺激下增强细胞凋亡。此外,联合传递的Axi可以进一步抑制血管内皮生长因子受体(VEGFR),从而削弱PDT诱导的肿瘤新生血管形成的负反馈。因此,FPPT@Axi通过阻断血管生成时空抑制肿瘤生长,破坏肿瘤血管,诱导肿瘤细胞凋亡。这种NRP-1介导的靶向共递送系统揭示了通过使用临床批准的PDT和化学疗法构建具有翻译潜力的吸引人的候选物。
    Antiangiogenic therapy is an effective way to disrupt nutrient supply and starve tumors, but it is restricted by poor efficacy and negative feedback-induced tumor relapse. In this study, a neuropilin-1 (NRP-1)-targeted nanomedicine (designated as FPPT@Axi) is reported for spatiotemporal tumor suppression by combining photodynamic therapy (PDT) with antiangiogenesis. In brief, FPPT@Axi is prepared by utilizing an NRP-1-targeting chimeric peptide (Fmoc-K(PpIX)-PEG8-TKPRR) to encapsulate the antiangiogenic drug Axitinib (Axi). Importantly, the NRP-1-mediated targeting property enables FPPT@Axi to selectively concentrate at vascular endothelial and breast cancer cells, facilitating the production of reactive oxygen species (ROS) in situ for specific vascular disruption and enhanced cell apoptosis under light stimulation. Moreover, the codelivered Axi can further inhibit vascular endothelial growth factor receptor (VEGFR) to impair the negative feedback of PDT-induced tumor neovascularization. Consequently, FPPT@Axi spatiotemporally restrains the tumor growth through blocking angiogenesis, destroying tumor vessels, and inducing tumor apoptosis. Such an NRP-1-mediated targeting codelivery system sheds light on constructing an appealing candidate with translational potential by using clinically approved PDT and chemotherapy.
    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
    当前的分析旨在从中国医疗保健系统的角度评估托里帕利马联合阿西替尼对先前未经治疗的RCC患者的经济效益。
    开发分区生存模型以模拟20年时间范围内3周患者的过渡,以评估与舒尼替尼相比,托里帕利马联合阿西替尼治疗晚期肾癌的成本效益。生存数据来自RENOTORCH试验,从数据库和已发表的文献中获得成本和效用投入。总成本,寿命年(LYs),质量调整寿命年(QALYs),增量成本效益比(ICER)是模型输出。进行了亚组分析和敏感性分析,以提高模型结果的全面性和鲁棒性。
    在基本情况分析中,与舒尼替尼相比,托里帕利马加阿西替尼可以带来额外的1.19LYs和0.65QALYs,边际成本为41,499.23美元,导致ICER为64,337.49美元/QALY,高于WTP阈值。ICER总是超过所有亚组的WTP阈值。敏感性分析表明模型结果是稳健的。
    从中国医疗系统的角度来看,托里帕利玛联合阿西替尼不太可能成为未经治疗的晚期肾癌患者的具有成本效益的一线治疗方案。
    UNASSIGNED: The current analysis aimed to evaluate the economic benefit of toripalimab plus axitinib for previously untreated RCC patients from the Chinese healthcare system perspective.
    UNASSIGNED: The partitioned survival model was developed to simulate 3-week patients\' transition in 20-year time horizon to evaluate the cost-effectiveness of toripalimab plus axitinib compared with sunitinib for advanced RCC. Survival data were gathered from the RENOTORCH trial, and cost and utility inputs were obtained from the database and published literature. Total cost, life-years (LYs), quality-adjusted life-years (QALYs), and incremental cost-effectiveness ratio (ICER) were the model outputs. Subgroup analyses and sensitivity analyses were conducted to increase the comprehensiveness and estimate the robustness of the model results.
    UNASSIGNED: In the base-case analysis, compared with sunitinib, toripalimab plus axitinib could bring additional 1.19 LYs and 0.65 QALYs, with the marginal cost of $41,499.23, resulting in the ICER of $64,337.49/QALY, which is higher than the WTP threshold. And ICERs were always beyond the WTP threshold of all subgroups. Sensitivity analyses demonstrated the model results were robust.
    UNASSIGNED: Toripalimab plus axitinib was unlikely to be the cost-effective first-line therapy for patients with previously untreated advanced RCC compared with sunitinib from the Chinese healthcare system perspective.
    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
    血管内皮生长因子B(VEGFB)已被充分证明通过与VEGF受体(VEGFR)结合在调节血管功能中起关键作用。然而,VEGFB和VEGFRs在青春期乳腺发育中的具体作用尚不清楚.在这项研究中,我们观察到,用阿西替尼阻断VEGF受体抑制青春期乳腺发育.同时,通过用阿西替尼阻断VEGF受体,抑制了乳腺上皮细胞(HC11)的增殖.此外,VEGFR1而不是VEGFR2和NRP1的敲低引起HC11增殖的抑制,提示VEGFR1在此过程中的重要作用。此外,阿西替尼或VEGFR1敲低导致PI3K/Akt途径的抑制。然而,Akt激活剂SC79消除了阿西替尼和/或VEGFR1敲低诱导的HC11增殖抑制,表明PI3K/Akt途径参与.最后,VEGFB和VEGFR1的敲减通过抑制PI3K/Akt通路抑制小鼠乳腺的青春期发育。总之,结果表明,VEGFB/VEGFR1信号的敲除通过抑制PI3K/Akt通路抑制小鼠青春期乳腺发育,为青春期乳腺发育的调控提供了新的靶点。
    Vascular endothelial growth factor B (VEGFB) has been well demonstrated to play a crucial role in regulating vascular function by binding to the VEGF receptors (VEGFRs). However, the specific role of VEGFB and VEGFRs in pubertal mammary gland development remains unclear. In this study, we observed that blocking the VEGF receptors with Axitinib suppressed the pubertal mammary gland development. Meanwhile, the proliferation of mammary epithelial cells (HC11) was repressed by blocking the VEGF receptors with Axitinib. Additionally, knockdown of VEGFR1 rather than VEGFR2 and NRP1 elicited the inhibition of HC11 proliferation, suggesting the essential role of VEGFR1 during this process. Furthermore, Axitinib or VEGFR1 knockdown led to the inhibition of the PI3K/Akt pathway. However, the inhibition of HC11 proliferation induced by Axitinib and or VEGFR1 knockdown was eliminated by the Akt activator SC79, indicating the involvement of the PI3K/Akt pathway. Finally, the knockdown of VEGFB and VEGFR1 suppressed the pubertal development of mice mammary gland with the inhibition of the PI3K/Akt pathway. In summary, the results showed that knockdown of the VEGFB/VEGFR1 signaling suppresses pubertal mammary gland development of mice via the inhibition of the PI3K/Akt pathway, which provides a new target for the regulation of pubertal mammary gland development.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号