AI总结:根据提供的论文列表,这些研究主要集中在以下几个医学领域:1. 纳米技术和药物递送系统:多篇论文探讨了纳米载体、纳米催化和纳米技术在药物设计和癌症治疗中的应用。例如,使用纳米催化剂和纳米载体提高药物的靶向性和疗效,以及开发基于纳米技术的新型抗癌疗法。2. 肿瘤微环境与抗肿瘤治疗:研究涉及肿瘤微环境中细胞间的相互作用及其对抗癌治疗的影响。特别是对肿瘤酸性环境、生物化学特性及化疗药物在不同氧合状态下的反应进行了深入分析,旨在优化抗癌治疗方案。3. 化学动力学与合成方法:一些论文关注于通过特定化学反应(如光诱导自由基硫醇-烯点击化学)来构建具有特殊功能的分子结构,为药物设计提供了新的思路和技术手段。4. 蛋白质降解与靶向治疗:研究提出创新策略以促进目标蛋白的降解,作为克服癌症耐药性的潜在途径,并介绍了一种全新的治疗性方法——“蛋白降解靶向嵌合体”(PROTACs),用于解决难治性癌症问题。5. 分子结构与生物活性关系:利用先进的计算模型研究分子结构如何影响其生物活性,特别是在低氧或正常氧条件下药物沉积物的行为,这对于理解药物作用机制至关重要。6. 多肽药物与生物制药创新:强调了多肽药物在未来生物制药领域的地位,讨论了它们在心脏疾病、神经退行性疾病等多个方面的应用前景,并指出这一领域正处于快速发展阶段,带来了许多新机遇。综上所述,这些论文反映了当前医学研究中对先进材料科学、精准医疗及个性化治疗的高度关注,特别是在癌症治疗和其他重大疾病的防治方面取得了显著进展。
Scan me!
共3篇 平均IF=26.6 (6.4-40.4)更多分析
  • 2区Q1影响因子: 6.4
    打开PDF
    1. Targeting PD-L1 in non-small cell lung cancer using CAR T cells.
    1. 使用CAR T细胞在非小细胞肺癌中靶向PD-L1。
    作者:Liu Ming , Wang Xu , Li Wei , Yu Xinfang , Flores-Villanueva Pedro , Xu-Monette Zijun Y , Li Ling , Zhang Mingzhi , Young Ken H , Ma Xiaodong , Li Yong
    期刊:Oncogenesis
    日期:2020-08-13
    DOI :10.1038/s41389-020-00257-z
    Antibodies against programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have dramatically changed the landscape of therapies for non-small cell lung carcinoma (NSCLC); however, the majority of patients do not respond to these agents. In addition, hyperprogressive disease (HPD) develops in a larger portion of NSCLC patients treated with PD-1/PD-L1 inhibitors than in patients treated with standard chemotherapy. The use of chimeric antigen receptor (CAR) T cells has been successful to treat blood cancers but not for solid tumors like NSCLC. In this work, we constructed CAR T cells that target PD-L1 and evaluated their efficacy in NSCLC with either high or low PD-L1 expression. PD-L1-CAR T cells exhibited antigen-specific activation, cytokine production, and cytotoxic activity against PD-L1 NSCLC cells and xenograft tumors. Furthermore, the addition of a subtherapeutic dose of local radiotherapy improved the efficacy of PD-L1-CAR T cells against PD-L1 NSCLC cells and tumors. Our findings indicate that PD-L1-CAR T cells represent a novel therapeutic strategy for patients with PD-L1-positive NSCLC, particularly for those who are susceptible to HPD.
  • 1区Q1影响因子: 40.4
    打开PDF
    2. Next-generation immuno-oncology agents: current momentum shifts in cancer immunotherapy.
    2. 下一代免疫肿瘤学代理:目前癌症免疫疗法的动量。
    期刊:Journal of hematology & oncology
    日期:2020-04-03
    DOI :10.1186/s13045-020-00862-w
    Cancer immunotherapy has reached a critical point, now that immune checkpoint inhibitors and two CAR-T products have received market approval in treating 16 types of cancers and 1 tissue-agnostic cancer indication. Accompanying these advances, the 2018 Nobel Prize was awarded for the discovery of immune checkpoint pathways, which has led to the revolution of anti-cancer treatments. However, expanding the indications of immuno-oncology agents and overcoming treatment resistance face mounting challenges. Although combination immunotherapy is an obvious strategy to pursue, the fact that there have been more failures than successes in this effort has served as a wake-up call, placing emphasis on the importance of building a solid scientific foundation for the development of next-generation immuno-oncology (IO) agents. The 2019 China Cancer Immunotherapy Workshop was held to discuss the current challenges and opportunities in IO. At this conference, emerging concepts and strategies for IO development were proposed, focusing squarely on correcting the immunological defects in the tumor microenvironment. New targets such as Siglec-15 and new directions including neoantigens, cancer vaccines, oncolytic viruses, and cytokines were reviewed. Emerging immunotherapies were discussed in the areas of overcoming primary and secondary resistance to existing immune checkpoint inhibitors, activating effector cells, and targeting immunosuppressive mechanisms in the tumor microenvironment. In this article, we highlight old and new waves of IO therapy development, and provide perspectives on the latest momentum shifts in cancer immunotherapy.
  • 1区Q1影响因子: 26.6
    打开PDF
    3. Programming CAR-T cells to kill cancer.
    3. 对CAR-T细胞编程以杀死癌症。
    作者:Labanieh Louai , Majzner Robbie G , Mackall Crystal L
    期刊:Nature biomedical engineering
    日期:2018-06-11
    DOI :10.1038/s41551-018-0235-9
    T cells engineered to express chimeric antigen receptors (CARs) that are specific for tumour antigens have led to high complete response rates in patients with haematologic malignancies. Despite this early success, major challenges to the broad application of CAR-T cells as cancer therapies remain, including treatment-associated toxicities and cancer relapse with antigen-negative tumours. Targeting solid tumours with CAR-T cells poses additional obstacles because of the paucity of tumour-specific antigens and the immunosuppressive effects of the tumour microenvironment. To overcome these challenges, T cells can be programmed with genetic modules that increase their therapeutic potency and specificity. In this Review Article, we survey major advances in the engineering of next-generation CAR-T therapies for haematologic cancers and solid cancers, with particular emphasis on strategies for the control of CAR specificity and activity and on approaches for improving CAR-T-cell persistence and overcoming immunosuppression. We also lay out a roadmap for the development of off-the-shelf CAR-T cells.
logo logo
$!{favoriteKeywords}