solid tumor treatment

  • 文章类型: Journal Article
    聚焦超声(FUS)触发的纳米级药物输送,作为治疗实体瘤的智能刺激响应系统,进行了计算研究,以增强药物的局部递送和治疗功效。热敏脂质体(TSL)的整合,作为负载多柔比星(DOX)的纳米载体,FUS,提供了一种有前途的药物递送系统。完全耦合的偏微分方程组,包括FUS传播的Helmholtz方程,生物传热,间质液流量,药物在组织和细胞空间的运输,首先提出了这种治疗方法的药效学模型。然后通过有限元方法求解方程以计算细胞内药物浓度和治疗功效。这项研究的主要目的是提出一个多物理和多尺度模型来模拟药物释放,运输,并运送到实体瘤,然后分析FUS暴露时间和药物释放速率如何影响这些过程。我们的发现不仅显示了模型复制这种治疗方法的能力,但也证实了这种治疗的好处,改善了肿瘤中的药物聚集和减少了健康组织中的药物递送。例如,治疗后肿瘤细胞的存活率下降到62.4%,因为大量的药物被输送到癌细胞。接下来,三种释放速率的组合(超快,快,和缓慢)和FUS暴露时间(10、30和60分钟)进行检查。曲线下面积(AUC)结果表明,30分钟FUS暴露和快速药物释放的组合导致实际和有效的治疗反应。
    Focused Ultrasound (FUS)-triggered nano-sized drug delivery, as a smart stimuli-responsive system for treating solid tumors, is computationally investigated to enhance localized delivery of drug and treatment efficacy. Integration of thermosensitive liposome (TSL), as a doxorubicin (DOX)-loaded nanocarrier, and FUS, provides a promising drug delivery system. A fully coupled partial differential system of equations, including the Helmholtz equation for FUS propagation, bio-heat transfer, interstitial fluid flow, drug transport in tissue and cellular spaces, and a pharmacodynamic model is first presented for this treatment approach. Equations are then solved by finite element methods to calculate intracellular drug concentration and treatment efficacy. The main objective of this study is to present a multi-physics and multi-scale model to simulate drug release, transport, and delivery to solid tumors, followed by an analysis of how FUS exposure time and drug release rate affect these processes. Our findings not only show the capability of model to replicate this therapeutic approach, but also confirm the benefits of this treatment with an improvement of drug aggregation in tumor and reduction of drug delivery in healthy tissue. For instance, the survival fraction of tumor cells after this treatment dropped to 62.4%, because of a large amount of delivered drugs to cancer cells. Next, a combination of three release rates (ultrafast, fast, and slow) and FUS exposure times (10, 30, and 60 min) was examined. Area under curve (AUC) results show that the combination of 30 min FUS exposure and rapid drug release leads to a practical and effective therapeutic response.
    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
    基于功能性免疫细胞转移的嵌合抗原受体T(CAR-T)细胞疗法正呈现出蓬勃发展的局面。然而,复杂的制造过程,高成本,和令人失望的结果在实体瘤的治疗限制了它的使用。令人鼓舞的是,它促进了融合免疫学的新策略的发展,细胞生物学,和生物材料来克服这些障碍。近年来,通过适当设计的生物材料辅助的CAR-T工程提高了治疗效果并减少了副作用,为改善癌症免疫治疗提供可持续的策略。同时,生物材料的低成本和多样性也为工业生产和商业化提供了可能性。这里,我们总结了生物材料作为基因载体在CAR-T细胞生成中的作用,并强调了体内原位构建的优势.然后,我们重点研究了如何将生物材料与CAR-T细胞结合,以更好地实现实体瘤治疗中的协同免疫疗法.最后,我们描述了生物材料在CAR-T治疗中的潜在挑战和前景。这篇综述旨在提供基于生物材料的CAR-T肿瘤免疫治疗的详细概述,以帮助研究人员参考和定制用于CAR-T治疗的生物材料,以提高免疫治疗的疗效。
    Chimeric antigen receptor-T (CAR-T) cell therapy based on functional immune cell transfer is showing a booming situation. However, complex manufacturing processes, high costs, and disappointing results in the treatment of solid tumors have limited its use. Encouragingly, it has facilitated the development of new strategies that fuse immunology, cell biology, and biomaterials to overcome these obstacles. In recent years, CAR-T engineering assisted by properly designed biomaterials has improved therapeutic efficacy and reduced side effects, providing a sustainable strategy for improving cancer immunotherapy. At the same time, the low cost and diversity of biomaterials also offer the possibility of industrial production and commercialization. Here, we summarize the role of biomaterials as gene delivery vehicles in the generation of CAR-T cells and highlight the advantages of in-situ construction in vivo. Then, we focused on how biomaterials can be combined with CAR-T cells to better enable synergistic immunotherapy in the treatment of solid tumors. Finally, we describe biomaterials\' potential challenges and prospects in CAR-T therapy. This review aims to provide a detailed overview of biomaterial-based CAR-T tumor immunotherapy to help investigators reference and customize biomaterials for CAR-T therapy to improve the efficacy of 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: Journal Article
    嵌合抗原受体T细胞(CART)疗法是治疗复发和难治性B细胞恶性肿瘤的里程碑。然而,在实体瘤中尚未获得CART细胞的有益作用。在这里,我们实施了一种多孔微针贴片,该贴片可容纳CART细胞,并允许在植入瘤床或手术后切除腔中时原位渗透介导的CART细胞接种.加载在微针尖端的孔中的CART细胞容易地以均匀分散的方式被护送到肿瘤而不失去其活性。与直接肿瘤内注射相比,这种微针介导的局部递送增强了CART细胞的浸润和免疫刺激。这个可剪裁的补丁提供了一个变革性的平台,用于治疗各种肿瘤的活细胞的分散接种。
    Chimeric antigen receptor T cell (CAR T) therapy was a milestone in the treatment of relapsed and refractory B cell malignancies. However, beneficial effects of CAR T cells have not been obtained in solid tumors yet. Herein, we implement a porous microneedle patch that accommodates CAR T cells and allows in situ penetration-mediated seeding of CAR T cells when implanted in the tumor bed or in the post-surgical resection cavity. CAR T cells loaded in the pores of the microneedle tips were readily escorted to the tumor in an evenly scattered manner without losing their activity. Such microneedle-mediated local delivery enhanced infiltration and immunostimulation of CAR T cells as compared to direct intratumoral injection. This tailorable patch offers a transformative platform for scattered seeding of living cells for treating a variety of tumors.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Comparative Study
    OBJECTIVE: The present meta-analysis study was performed to identify the potential cardiotoxicity risks when using Vascular Endothelial Growth Factor Receptor Tyrosine kinase inhibitors (VEGFR-TKIs) as anticancer drugs in patients with solid tumors.
    METHODS: Pubmed, Embase, the Cochrane Central Register of Controlled Trials, and ClinicalTrials.gov databases were searched for the randomized controlled trials. We have included 45 randomized controlled trials (RCTs) associated with nine VEGFR-TKIs Food and Drug Administration (FDA)-approved drugs used to treat patients with solid tumors. To evaluate the trials\' risk of bias, Cochrane Risk of Bias Tool was assessed. A direct comparison was assessed by RevMan5.3 software, calculating the odds ratio (OR) and 95% confidence interval (CI). Heterogeneity was tested by the I2 statistic and Chi-square test for P value. Bayesian network meta-analysis was performed using Stata 15.0 and GeMTC 0.14.3 software, calculated OR along with corresponding 95% credible interval (CrI). The model\'s convergence was evaluated by the potential scale reduced factor (PSRF). Consistency between direct and indirect comparisons was assessed by the \"node-splitting\" method.
    RESULTS: In this network meta-analysis, a total of 20,027 patients from 45 randomized controlled trials and associated with nine FDA-approved VEGFR-TKIs (axitinib, cabozantinib, lenvatinib, nintedanib, pazopanib, regorafenib, sorafenib, sunitinib, vandetanib), were enrolled. Findings indicated that lenvatinib had the most significant probability of provoking all grades cardiovascular incident and hypertension, followed by vandetanib, cabozantinib, axitinib, pazopanib, sorafenib, sunitinib, regorafenib and nintedanib. The nine agent\'s severe cardiovascular and severe hypertension risk was probably similar. The ranking probability of cardiac toxicity shows that vandetanib ranked most likely to have the highest risk for cardiotoxicity among all the VEGFR-TKIs reviewed, followed by pazopanib, axitinib, sorafenib, sunitinib. In contrast, regorafenib and nintedanib did not exhibit an increased risk of cardiac damage.
    CONCLUSIONS: The association between the nine VEGFR-TKIs with potential cardiotoxicity occurrence was reviewed. Both the regorafenib and nintedanib did not display detectable signs of cardiotoxic damage. In contrast, lenvatinib and vandetanib are ranked to have the most severe cardiotoxicity side impacts. These results may provide information for clinical practice guidelines, implementing strategies in selecting the adequate VEGFR-TKIs, and understanding the cardiovascular toxicity inflicted by the VEGFR-TKIs.
    UNASSIGNED: CRD 42,020,167,307.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Chimeric antigen receptor T-cell (CAR-T) therapy has achieved good therapeutic efficacy in the treatment of hematological malignancies. In August 2017, Novartis Kymriah (CAR-T cells targeting CD19) was approved by the FDA, indicating the real entry of CAR-T cell therapy into clinical applications and making CAR-T cell therapy the most attractive technology in the field of tumor treatment. In October 2017, the FDA approved the world\'s second CAR-T cell therapy-Yescarta. The launch of these products has attracted wide attention to CAR-T cell therapy. CAR-T cell therapy has achieved significant effect in the treatment of tumors, however, CAR-T therapy also faces clinical problems, such as cytokine release syndrome (CRS), poor therapeutic efficacy in solid tumors, and high rates of tumor recurrence. At present, the side effects of CAR-T therapy have attracted a large amount of attention, which has resulted in investigations into strategy establishment. With a deepening understanding of CAR-T therapy and the continuous optimization of therapeutic regimens, its toxicity and side effects have been partially controlled. This study set out to analyze the problems in the clinical application of CAR-T therapy encountered in recent years and to introduce corresponding strategies, with the aim of providing a basis of reference for clinicians and scientists in the management of CAR-T therapy in clinical practice and in the CAR-T therapy research.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Herein, the biodegradable micelle-forming amphiphilic N-(2-hydroxypropyl) methacrylamide (HPMA)-based polymer conjugates with the anticancer drug doxorubicin (Dox) designed for enhanced tumor accumulation were investigated, and the influence of their stability in the bloodstream on biodistribution, namely, tumor uptake, and in vivo antitumor efficacy were evaluated in detail. Dox was attached to the polymer carrier by a hydrazone bond enabling pH-controlled drug release. While the polymer-drug conjugates were stable in a buffer at pH 7.4 (mimicking bloodstream environment), Dox was released in a buffer under mild acidic conditions modeling the tumor microenvironment or cells. The amphiphilic polymer carriers differed in the structure of hydrophobic cholesterol derivative moieties bound to the HPMA copolymers via a hydrolyzable hydrazone bond, exhibiting different rates of micellar structure disintegration at various pH values. Considerable dependence of the studied polymer-drug conjugate biodistribution on the stability of the micellar structure was observed in neutral, bloodstream-mimicking, environment, showing that a faster rate of the micelle disintegration in pH 7.4 increased the conjugate blood clearance, decreased tumor accumulation, and significantly reduced the tumor:blood and tumor:muscle ratios. Similarly, the final therapeutic outcome was strongly affected by the stability of the micellar structure because the most stable micellar conjugate showed an almost similar therapeutic outcome as the water-soluble, nondegradable, high-molecular-weight starlike HPMA copolymer-Dox conjugate, which was highly efficient in the treatment of solid tumors in mice. Based on the results, we conclude that the bloodstream stability of micellar polymer-anticancer drug conjugates, in addition to their low side toxicity, is a crucial parameter for their efficient solid tumor accumulation and high in vivo antitumor activity.
    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