Anti-angiogenic agent

抗血管生成剂
  • 文章类型: Journal Article
    新型血管网络的发展是肿瘤生长和进展的基本要求。在过去的十年里,生物标志物和血管生成的潜在分子途径已经被深入研究以破坏肿瘤血管生成的开始和进展。然而,由于毒副作用,抗血管生成剂的临床应用受到限制,获得性耐药性和无法获得有效的生物标志物。
    这篇综述讨论了树枝状纳米载体的发展,这可能是一个有前途的领域,可以探索根除与基于血管生成的癌症治疗相关的当前挑战。具有微妙读数和对分子机制的更好理解的新型药物递送方法已经揭示了树枝状聚合物包含先天的抗血管生成活性,并且掺入抗血管生成剂或基因沉默RNA可以导致协同的抗血管生成和抗癌作用,同时副作用减少。
    Dendrimer介导的靶向血管生成生物标志物有效地导致血管正常化,树枝状聚合物与抗血管生成剂或siRNA或两者的合理连接可能是消除当前基于血管生成的癌症治疗挑战的潜在领域。然而,与树枝状聚合物介导的血管生成生物标志物靶向相关的缺点,如稳定性差或这些生物标志物在正常细胞上的小表达,限制了它在市场规模上的应用。
    UNASSIGNED: Development of novel vascular networks is a fundamental requirement for tumor growth and progression. In the last decade, biomarkers and underlying molecular pathways of angiogenesis have been intensely investigated to disrupt the initiation and progression of tumor angiogenesis. However, the clinical applications of anti-angiogenic agents are constrained due to toxic side effects, acquired drug resistance, and unavailability of validated biomarkers.
    UNASSIGNED: This review discusses the development of dendrimeric nanocarriers that could be a promising domain to explore for the eradication of current challenges associated with angiogenesis-based cancer therapy. Novel drug-delivery approaches with subtle readouts and better understanding of molecular mechanisms have revealed that dendrimers comprise innate anti-angiogenic activity and incorporation of anti-angiogenic agents or gene-silencing RNA could lead to synergistic anti-angiogenic and anticancer effects with reduced side effects.
    UNASSIGNED: Dendrimer-mediated targeting of angiogenic biomarkers has efficiently led to the vascular normalization, and rational linking of dendrimers with anti-angiogenic agent or siRNA or both might be a potential area to eradicate the current challenges of angiogenesis-based cancer therapy. However, drawbacks associated with the dendrimers-mediated targeting of angiogenic biomarkers, such as poor stability or small expression of these biomarkers on the normal cells, limit their application at market scale.
    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: Editorial
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 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: Journal Article
    大多数被批准用于临床使用的预测性生物标志物测量单一分析物,例如遗传改变或蛋白质过表达。我们开发并验证了一种新型生物标志物,旨在实现广泛的临床应用。Xerna™TME面板是一个泛肿瘤,基于RNA表达的分类器,旨在预测对多种肿瘤微环境(TME)靶向治疗的反应,包括免疫疗法和抗血管生成剂。
    Panel算法是一种人工神经网络(ANN),使用124个基因的输入签名进行训练,并在各种实体肿瘤中进行了优化。从298名患者的培训数据中,学会区分四种TME亚型的模型:血管生成(A),免疫活性物质(IA),免疫沙漠(ID),和免疫抑制(IS)。在四个独立的临床队列中评估最终分类器,以测试TME亚型是否可以预测整个胃,卵巢,和黑色素瘤数据集。
    TME亚型代表由血管生成和免疫生物学轴定义的基质表型。该模型在生物标志物阳性和阴性之间产生清晰的界限,并且对于多个治疗假设显示1.6至7倍的临床益处富集。与胃和卵巢抗血管生成数据集的空模型相比,小组在所有标准中的表现都更好。它的准确性也优于PD-L1联合阳性评分(>1),特异性,和阳性预测值(PPV),和微卫星不稳定性高(MSI-H)的敏感性和阴性预测值(NPV)的胃免疫治疗队列。
    TME小组在不同数据集上的强大表现表明,它可能适合用作各种癌症类型和治疗方式的临床诊断。
    UNASSIGNED: Most predictive biomarkers approved for clinical use measure single analytes such as genetic alteration or protein overexpression. We developed and validated a novel biomarker with the aim of achieving broad clinical utility. The Xerna™ TME Panel is a pan-tumor, RNA expression-based classifier, designed to predict response to multiple tumor microenvironment (TME)-targeted therapies, including immunotherapies and anti-angiogenic agents.
    UNASSIGNED: The Panel algorithm is an artificial neural network (ANN) trained with an input signature of 124 genes that was optimized across various solid tumors. From the 298-patient training data, the model learned to discriminate four TME subtypes: Angiogenic (A), Immune Active (IA), Immune Desert (ID), and Immune Suppressed (IS). The final classifier was evaluated in four independent clinical cohorts to test whether TME subtype could predict response to anti-angiogenic agents and immunotherapies across gastric, ovarian, and melanoma datasets.
    UNASSIGNED: The TME subtypes represent stromal phenotypes defined by angiogenesis and immune biological axes. The model yields clear boundaries between biomarker-positive and -negative and showed 1.6-to-7-fold enrichment of clinical benefit for multiple therapeutic hypotheses. The Panel performed better across all criteria compared to a null model for gastric and ovarian anti-angiogenic datasets. It also outperformed PD-L1 combined positive score (>1) in accuracy, specificity, and positive predictive value (PPV), and microsatellite-instability high (MSI-H) in sensitivity and negative predictive value (NPV) for the gastric immunotherapy cohort.
    UNASSIGNED: The TME Panel\'s strong performance on diverse datasets suggests it may be amenable for use as a clinical diagnostic for varied cancer types and therapeutic modalities.
    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
    目的:中枢神经系统(CNS)转移,包括脑转移(BM)和软脑膜转移(LM)在表皮生长因子受体(EGFR)突变的非小细胞肺癌(NSCLC)中很常见,并与不良结果相关。在这项研究中,我们评估了在既往酪氨酸激酶抑制剂(TKI)治疗后出现BM/LM进展的NSCLC患者中,单独使用furmonertinib160mg或联合使用抗血管生成药物的疗效.
    方法:纳入发展为BM(BM队列)或LM进展(LM队列)的EGFR突变NSCLC患者,作为二线或后续治疗,每天接受furmonertinib160mg,有或没有抗血管生成剂。通过颅内无进展生存期(iPFS)评估颅内疗效。
    结果:纳入了BM队列中的12例患者和LM队列中的16例患者。BM队列中几乎一半的患者和LM队列中的大多数患者的身体状况较差,东部肿瘤协作组表现状态(ECOG-PS)≥2。在BM队列中,给予单一药物furmonertinib或联合治疗的iPFS中位数为3.6个月(95CI1.435-5.705),LM队列中的4.3个月(95CI2.094-6.486)。亚组和单变量分析表明,在BM队列中,良好的ECOG-PS与furmonertinib的良好疗效相关(iPFS中位数=2.1,ECOG-PS≥2与14.6个月ECOG-PS<2,P<0.05)。总的来说,任何级别的不良事件(AE)发生在46.4%的患者中(13/28).其中,14.3%的患者(28人中有4人)出现3级或更高的不良事件,一切都在控制之中,导致没有剂量减少或暂停。
    结论:对于先前EGFR-TKI治疗后出现BM/LM进展的晚期NSCLC患者,单药furmonertinib160mg或联合抗血管生成药物是一种可选的挽救疗法。具有有希望的疗效和可接受的安全性,值得进一步探索。
    OBJECTIVE: Central nervous system (CNS) metastases including brain metastases (BM) and leptomeningeal metastases (LM) are frequent in epidermal growth factor receptor (EGFR)-mutated non-small cell lung cancer (NSCLC), and are correlated with poor outcomes. In this study, we evaluated the efficacy of single-agent furmonertinib 160 mg or combining with anti-angiogenic agent in NSCLC patients who had developed BM/LM progression from previous tyrosine kinase inhibior (TKI) treatment.
    METHODS: EGFR-mutated NSCLC patients who developed BM (the BM cohort) or LM progression (the LM cohort) were included, having received furmonertinib 160 mg daily as second-line or later treatment, with or without anti-angiogenic agents. The intracranial efficacy was evaluated by intracranial progression-free survival (iPFS).
    RESULTS: Totally 12 patients in the BM cohort and 16 patients in the LM cohort were included. Almost one half of patients in the BM cohort and a majority in the LM cohort had a poor physical status, with a Eastern Cooperative Oncology Group performance status (ECOG-PS) ≥2. The administration of single-agent furmonertinib or combination treatment achieved a median iPFS of 3.6 months (95%CI 1.435-5.705) in the BM cohort, and 4.3 months (95%CI 2.094-6.486) in the LM cohort. Subgroup and univariate analysis has shown that a good ECOG-PS correlated with a favorable efficacy of furmonertinib in the BM cohort (median iPFS = 2.1 with ECOG-PS ≥ 2 vs. 14.6 months with ECOG-PS < 2, P < 0.05). Overall, any grade of adverse events (AEs) occured in 46.4% of patients (13/28). Among them, 14.3% of patients (4 of 28) had grade 3 or higher AEs, and were all under control, led to no dose reductions or suspension.
    CONCLUSIONS: Single-agent furmonertinib 160 mg or in combination of anti-angiogenic agent is an optional salvage therapy for advanced NSCLC patients who developed BM/LM progression from prior EGFR-TKI treatment, with a promising efficacy and an acceptable safety profile, and is worth of further exploration.
    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: Multicenter Study
    目的:Ramucirumab(RAM)和多西他赛(DOC)是晚期非小细胞肺癌(NSCLC)一线治疗后常用的药物。因此,我们旨在阐明先前治疗后RAM和DOC的测序策略,包括免疫检查点抑制剂(ICI),单独的细胞毒性剂(CTx),贝伐单抗(BEV),和酪氨酸激酶抑制剂(TKI)。
    方法:我们招募了接受了RAM和DOC的NSCLC患者,并比较了有和没有ICI,单纯CTx的组,BEV,还有TKI,分别。通过肿瘤对这种治疗的反应,患者进一步分为“完全缓解(CR)+部分缓解(PR),“稳定的疾病,\"和\"进行性疾病\"组,分别。我们比较了这些组的RAM和DOC疗效。
    结果:总计,237名患者登记。在先前有ICI的组中,客观缓解率和疾病控制率显着高于没有先前ICI的组(分别为p=0.012和0.028),中位无进展生存期(PFS)也显著延长(p=0.027).单独使用和不使用CTx的组之间的PFS没有显着差异,BEV,还有TKI.多因素分析显示,既往ICI是改善PFS的独立因素。此外,与之前没有ICI的组相比,之前有CR+PR的ICI组显著延长PFS(p=0.013).
    结论:RAM和DOC可以优选在ICI后给药,而不是单独在CTx后给药,BEV,或者TKI,and,此外,如果先前的ICI具有良好的肿瘤反应,则增强。
    OBJECTIVE: Ramucirumab (RAM) and docetaxel (DOC) are commonly used after first-line therapy for advanced non-small cell lung cancer (NSCLC). Therefore, we aimed to elucidate sequencing strategies of RAM and DOC following prior treatments, including immune checkpoint inhibitor (ICI), cytotoxic agent (CTx) alone, bevacizumab (BEV), and tyrosine kinase inhibitor (TKI).
    METHODS: We recruited patients with NSCLC who received RAM and DOC and compared the groups with and without prior ICI, CTx alone, BEV, and TKI, respectively. By tumor response to such treatments, the patients were further classified into \"complete response (CR) + partial response (PR),\" \"stable disease,\" and \"progressive disease\" groups, respectively. We compared RAM and DOC efficacy among these groups.
    RESULTS: In total, 237 patients were registered. In the group with prior ICI, the objective response rate and disease control rate were significantly higher than those without prior ICI (p = 0.012 and 0.028, respectively), and the median progression-free survival (PFS) was also significantly longer (p = 0.027). There were no significant differences in PFS between the groups with and without CTx alone, BEV, and TKI. Multivariate analysis revealed that prior ICI was an independent factor associated with better PFS. Furthermore, the prior ICI group with CR + PR significantly prolonged PFS compared to the group without prior ICI (p = 0.013).
    CONCLUSIONS: RAM and DOC may be preferably administered after ICI, rather than after CTx alone, BEV, or TKI, and, furthermore, enhanced if the prior ICI has a favorable tumor response.
    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
    近年来,转移性肾细胞癌(mRCC)的治疗性医疗设备一直在扩展,随着VEGF/VEGFR(血管内皮生长因子/血管内皮生长因子受体)抑制剂的引入,mTOR(哺乳动物雷帕霉素靶蛋白)抑制剂和免疫检查点(IC)抑制剂。目前,对于mRCC的一线治疗,可以选择VEGFR-TKI(VEGFR-酪氨酸激酶抑制剂)单一疗法,ICI-ICI(免疫检查点抑制剂)组合和ICI-VEGFRTKI组合。然而,一致的一部分患者不能从ICIs的一线治疗中获益;此外,联合治疗方案的使用使患者面临显著的毒性.因此,迫切需要开发对VEGFR-TKIs和ICIs反应的预后和预测性生物标志物,和血清IL-8的测量正在成为该领域的潜在候选者。最近对大型II期和III期试验的回顾性分析发现,升高的基线血清IL-8与较高水平的肿瘤和循环免疫抑制性骨髓细胞相关。降低T细胞活化和对治疗的反应差。这些发现必须在前瞻性临床试验中得到证实;然而,它们为血清IL-8作为VEGFR-TKIs和ICIs耐药的生物标志物的潜在用途提供了证据.考虑到正在改变转移性肾细胞癌管理的新药和治疗方案的数量,血清IL-8可能成为为每个患者定制最佳治疗的宝贵资源。
    The therapeutic armamentarium of metastatic Renal Cell Carcinoma (mRCC) has consistently expanded in recent years, with the introduction of VEGF/VEGFR (Vascular Endothelial Growth Factor/Vascular Endothelial Growth Factor Receptor) inhibitors, mTOR (mammalian Target Of Rapamycin) inhibitors and Immune Checkpoint (IC) inhibitors. Currently, for the first-tline treatment of mRCC it is possible to choose between a VEGFR-TKI (VEGFR-Tyrosine Kinase Inhibitor) monotherapy, an ICI-ICI (Immune Checkpoint Inhibitor) combination and an ICI-VEGFRTKI combination. However, a consistent part of patients does not derive benefit from first-line therapy with ICIs; moreover, the use of combination regimens exposes patients to significant toxicities. Therefore, there is a critical need to develop prognostic and predictive biomarkers of response to VEGFR-TKIs and ICIs, and measurement of serum IL-8 is emerging as a potential candidate in this field. Recent retrospective analyses of large phase II and phase III trials found that elevated baseline serum IL-8 correlated with higher levels of tumor and circulating immunosuppressive myeloid cells, decreased T cell activation and poor response to treatment. These findings must be confirmed in prospective clinical trials; however, they provide evidence for a potential use of serum IL-8 as biomarker of resistance to VEGFR-TKIs and ICIs. Considering the amount of new agents and treatment regimens which are transforming the management of metastatic renal cell carcinoma, serum IL-8 could become a precious resource in tailoring the best therapy for each individual patient with the disease.
    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
    表皮生长因子受体(EGFR)突变的非小细胞肺癌(NSCLC)患者的类型可以从EGFR/血管EGFR(VEGFR)途径的双重抑制中获得显着的临床益处仍不清楚。关于吸烟习惯在EGFR-TKI加抗血管生成剂的临床获益中的重要性尚未达成共识。
    PubMed,EMBASE,系统检索了所有II/III期随机临床试验(RCTs)的Cochrane数据库,这些试验调查了EGFR-TKI联合抗血管生成药的疗效,并按吸烟习惯分层(2021年10月更新).主要结果是吸烟者和非吸烟者PFS/OS的合并HR,EGFR-TKI联合抗血管生成治疗在吸烟者和非吸烟者之间的疗效差异,通过PFS和OS的差异来衡量。
    确定了7个II/III期RCTs,涉及1452名患者。汇总分析表明,EGFR-TKI加抗血管生成剂可将进展风险降低40%(HR,与单独使用EGFR-TKI相比,吸烟者为0.60;95CI0.48-0.75),但非吸烟者(HR,0.92;95CI0.68-1.25)。比较分析进一步表明,吸烟的EGFR突变的NSCLC患者从EGFR-TKI加抗血管生成剂治疗中获得了更高的无进展生存期(PFS)(HR,0.68;95CI0.51-0.91)。与PFS的结果一致,接受EGFR-TKI加抗血管生成剂的吸烟者似乎比非吸烟者表现出更好的总体生存率(OS),但没有达到统计学上的显着程度(HR,0.60;95CI0.23-1.52)。Meta回归分析显示治疗线无显著影响(P=0.52),试验阶段(P=0.52),EGFR-TKI型(P=0.13),或抗血管生成剂类型(P=0.50)对多变量模型下PFS的影响大小。
    综合分析表明,EGFR-TKI加抗血管生成药物导致吸烟EGFR突变患者的PFS良好,与不吸烟者相比,这可能为临床医生提供有用的指导。
    The types of epidermal growth factor receptor (EGFR)-mutant non-small-cell lung cancer (NSCLC) patients who could obtain significant clinical benefit from the dual inhibition of EGFR/vascular EGFR (VEGFR) pathways remain unclear. No consensus has been reached on the significance of smoking habits in clinical benefit obtained from EGFR-TKI plus anti-angiogenic agents.
    PubMed, EMBASE, and Cochrane databases for all phase II/III randomized clinical trials (RCTs) investigating the efficacy of EGFR-TKI combined with anti-angiogenic agents stratified by smoking habits (updated October 2021) were searched systematically. The primary outcomes were the pooled HRs for PFS/OS in smokers and non-smokers, and differences in efficacy of EGFR-TKI plus anti-angiogenic treatment between smokers and non-smokers, measured by difference in PFS and OS.
    Seven phase II/III RCTs involving 1452 patients were identified. The pooled analysis demonstrated that EGFR-TKI plus anti-angiogenic agent could decrease the risk of progression by 40% (HR, 0.60; 95%CI 0.48-0.75) in smokers when compared with EGFR-TKI alone, but not in non-smokers (HR, 0.92; 95%CI 0.68-1.25). The comparison analysis further demonstrated that EGFR-mutated NSCLC patients who smoked obtained greater progression-free survival (PFS) benefit from treatment with EGFR-TKI plus anti-angiogenic agents (HR, 0.68; 95%CI 0.51-0.91). Consistent with the results for PFS, smokers receiving EGFR-TKI plus anti-angiogenic agents appeared to exhibit better overall survival (OS) than non-smokers but not to a statistically significant degree (HR, 0.60; 95%CI 0.23-1.52). Meta-regression analysis revealed no significant effect of the line of treatment (P = 0.52), trial phase (P = 0.52), EGFR-TKI type (P = 0.13), or anti-angiogenic agent type (P = 0.50) on PFS effect sizes under multivariate models.
    Comprehensive analysis suggested that EGFR-TKI plus anti-angiogenic agents led to favorable PFS among smoking EGFR-mutant patients, comparable to nonsmokers, which might provide a useful guide for clinicians.
    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: Case Reports
    PD-1/PD-L1 inhibitor immunotherapy has showed impressive activity in various cancers, especially relapsed/refractory (r/r) classical Hodgkin lymphoma (cHL). However, acquired resistance is inevitable for most patients. Sometimes severe side effects also lead to treatment termination. When immunotherapy failed, alternative treatment options are limited. In the past few years, we have used the anti-angiogenic agent apatinib and PD-1 inhibitor camrelizumab to treat cHL patients who failed prior immunotherapy. In this study, we analyzed the data of these patients.
    Patients with r/r cHL who had failed immunotherapy and subsequently received apatinib-camrelizumab (AC) combination therapy were included in this study. Patient data were collected from medical records and follow-up system. The efficacy and safety of AC therapy were analyzed.
    Seven patients who failed immunotherapy were identified in our database, of which five patients acquired immunotherapy resistance and two patients experienced severe side effects. They received a combination of camrelizumab (200 mg every four weeks) and apatinib (425 mg or 250 mg per day). As of the cut-off date, these patients had received a median of 4 cycles (range, 2 - 31) of treatment. Two (2/7) patients achieved complete response, four (4/7) partial response, and one (1/7) stable disease. The median progression-free survival was 10.0 months (range, 2.0 - 27.8). Low-dose apatinib (250 mg) plus camrelizumab was well tolerated and had no unexpected side effects. Besides, no reactive cutaneous capillary endothelial proliferation was observed in AC-treated patients.
    Low dose apatinib plus camrelizumab might be a promising treatment option for r/r cHL patients who have failed immunotherapy. This combination treatment is worthy of further investigation in more patients including solid cancer patients who have failed immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    Aim: Advanced esophageal squamous cell carcinoma (ESCC) is a lethal disease with poor response to conventional chemotherapy. Immunotherapy showed better activity than chemotherapy in late-line treatment. However, the rate and duration of response are far from satisfactory. The efficacy of an anti-angiogenic agent combined with immunotherapy for ESCC is unknown. Results: A patient with ESCC experienced disease relapse after chemo-radiotherapy. The disease progressed after combined chemotherapy. A combination regimen of the PD-1 inhibitor camrelizumab and the anti-angiogenic agent apatinib was administered. The patient achieved a PET/CT-confirmed durable complete response with mild toxicity. Conclusion: The PD-1 inhibitor combined with the anti-angiogenic agent is effective and safe for the treatment of ESCC. This regimen is worth investigation in clinical trials.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号