Nano-immunotherapy

纳米免疫疗法
  • 文章类型: Journal Article
    以“冷肿瘤”为特征的乳腺癌表现出低水平的免疫细胞浸润,这限制了常规免疫疗法的疗效。最近的研究集中在利用纳米技术结合肿瘤微环境调节将“冷肿瘤”转化为“热肿瘤”的策略上。该方法涉及使用靶向和修饰肿瘤微环境的功能化纳米颗粒,以促进抗肿瘤免疫细胞的浸润和活化。通过传递免疫激活剂或阻断免疫抑制信号,这些纳米粒子激活了休眠的免疫反应,增强肿瘤免疫原性和治疗反应。这些策略不仅有望提高乳腺癌患者对现有免疫疗法的反应率,还可能为新的治疗途径铺平道路。为乳腺癌的免疫治疗提供了新的方向。
    Breast cancer characterized as \"cold tumors\" exhibit low levels of immune cell infiltration, which limits the efficacy of conventional immunotherapy. Recent studies have focused on strategies using nanotechnology combined with tumor microenvironment modulation to transform \"cold tumors\" into \"hot tumors\". This approach involves the use of functionalized nanoparticles that target and modify the tumor microenvironment to promote the infiltration and activation of antitumor immune cells. By delivering immune activators or blocking immunosuppressive signals, these nanoparticles activate otherwise dormant immune responses, enhancing tumor immunogenicity and the therapeutic response. These strategies not only promise to increase the response rate of breast cancer patients to existing immunotherapies but also may pave new therapeutic avenues, providing a new direction for the immunotherapy of breast cancer.
    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
    血液恶性肿瘤(HMs)包括具有显著发病率和死亡率的不同组的血液肿瘤。免疫疗法已成为HMs患者的一种有效和关键的治疗方式。尽管在过去十年中,在理解和实施HMs的免疫疗法方面取得了显著进步,几个挑战依然存在。这些挑战包括免疫相关的不良反应,治疗性抗原在体内的精确生物分布和消除,肿瘤的免疫耐受,和肿瘤细胞在肿瘤微环境(TME)内的免疫逃避。纳米技术,具有在纳米尺度上操纵材料特性的能力,有可能通过改善药物靶向和稳定性等各个方面来解决这些障碍并彻底改变治疗结果。纳米技术和免疫治疗的融合催生了纳米免疫治疗,抗肿瘤治疗的一个专门分支。纳米技术已经在嵌合抗原受体T细胞(CAR-T)治疗中找到了应用,癌症疫苗,免疫检查点抑制剂,以及其他针对HMs的免疫治疗策略。在这篇综述中,我们描述了最近的发展,并讨论了目前在纳米免疫治疗领域的挑战,为这些疾病的基于纳米技术的治疗方法的潜力提供了新的见解。
    Hematological malignancies (HMs) encompass a diverse group of blood neoplasms with significant morbidity and mortality. Immunotherapy has emerged as a validated and crucial treatment modality for patients with HMs. Despite notable advancements having been made in understanding and implementing immunotherapy for HMs over the past decade, several challenges persist. These challenges include immune-related adverse effects, the precise biodistribution and elimination of therapeutic antigens in vivo, immune tolerance of tumors, and immune evasion by tumor cells within the tumor microenvironment (TME). Nanotechnology, with its capacity to manipulate material properties at the nanometer scale, has the potential to tackle these obstacles and revolutionize treatment outcomes by improving various aspects such as drug targeting and stability. The convergence of nanotechnology and immunotherapy has given rise to nano-immunotherapy, a specialized branch of anti-tumor therapy. Nanotechnology has found applications in chimeric antigen receptor T cell (CAR-T) therapy, cancer vaccines, immune checkpoint inhibitors, and other immunotherapeutic strategies for HMs. In this review, we delineate recent developments and discuss current challenges in the field of nano-immunotherapy for HMs, offering novel insights into the potential of nanotechnology-based therapeutic approaches for these diseases.
    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
    癌症仍然是全球主要的死亡原因,要求早期诊断和有效治疗。传统的治疗方法由于其需要更多的特异性和全身毒性而常常达不到。在这个充满挑战的景观中,纳米金刚石(ND)作为一种潜在的解决方案出现,减轻传统方法的局限性。ND是微小的碳颗粒,可以模仿传统钻石的化学稳定性和硬度,并利用纳米材料的优势。ND因其独特的特性而脱颖而出,这些特性使它们成为有前途的nanotheranosics候选人,在一个平台上结合治疗和成像能力。许多这些应用依赖于粒子表面的设计,作为表面的作用是至关重要的运输生物活性分子,防止聚集,和建筑复合材料。这篇评论深入探讨了ND的独特特征,结构和光学特性,以及它们在推进癌症诊断和治疗方法方面的深远意义。该报告深入研究了这些特殊的ND特性如何推动精确肿瘤靶向技术的发展,提高化疗作为化学增敏剂的有效性,利用免疫治疗策略,促进精准医学,并创建用于靶向治疗的局部缩微胶片设备。
    Cancer remains a leading global cause of mortality, demanding early diagnosis and effective treatment. Traditional therapeutic methods often fall short due to their need for more specificity and systemic toxicity. In this challenging landscape, nanodiamonds (ND) emerge as a potential solution, mitigating the limitations of conventional approaches. ND are tiny carbon particles that mimic traditional diamonds chemical stability and hardness and harness nanomaterials\' advantages. ND stands out for the unique properties that make them promising nanotheranostics candidates, combining therapeutic and imaging capabilities in one platform. Many of these applications depend on the design of the particle\'s surface, as the surface\'s role is crucial in transporting bioactive molecules, preventing aggregation, and building composite materials. This review delves into ND\'s distinctive features, structural and optical characteristics, and their profound relevance in advancing cancer diagnosis and treatment methods. The report delves into how these exceptional ND properties drive the development of state-of-the-art techniques for precise tumor targeting, boosting the effectiveness of chemotherapy as a chemosensitizer, harnessing immunotherapy strategies, facilitating precision medicine, and creating localized microfilm devices for targeted therapies.
    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
    纳米技术的进步为多发性骨髓瘤(MM)的诊断和治疗提供了新的途径,以骨髓中浆细胞的克隆性增殖为特征的血液恶性肿瘤。这篇综述阐明了纳米技术彻底改变骨髓瘤治疗的潜力,专注于基于纳米颗粒的药物递送系统,纳米成像技术,和纳米免疫疗法。基于纳米颗粒的药物递送系统提供增强的药物靶向,减少全身毒性,并提高治疗效果。我们讨论了纳米载体的最新发展,如脂质体,聚合物纳米颗粒,和无机纳米粒子,用于化疗药物的输送,siRNA和miRNA在MM治疗中的应用。我们深入研究了提供空间多维数据的纳米级成像技术,提供肿瘤微环境的整体视图。这种空间分辨率可以帮助破译癌细胞与其周围环境之间复杂的相互作用,促进高度靶向治疗的发展。最后,我们探索纳米免疫疗法的新兴领域,它使用纳米粒子来调节免疫系统以治疗骨髓瘤。具体来说,我们考虑如何使用纳米粒子将肿瘤抗原传递给抗原呈递细胞,从而增强机体对骨髓瘤细胞的免疫反应。总之,纳米技术在改善MM患者的预后和生活质量方面具有广阔的前景。然而,仍然存在一些挑战,包括需要进一步的临床前和临床试验来评估这些新兴策略的安全性和有效性.未来的研究还应该集中在开发个性化的纳米医学方法上,这可以根据个体患者的遗传和分子特征为他们量身定制治疗方法。
    Advances in nanotechnology have provided novel avenues for the diagnosis and treatment of multiple myeloma (MM), a hematological malignancy characterized by the clonal proliferation of plasma cells in the bone marrow. This review elucidates the potential of nanotechnology to revolutionize myeloma therapy, focusing on nanoparticle-based drug delivery systems, nanoscale imaging techniques, and nano-immunotherapy. Nanoparticle-based drug delivery systems offer enhanced drug targeting, reduced systemic toxicity, and improved therapeutic efficacy. We discuss the latest developments in nanocarriers, such as liposomes, polymeric nanoparticles, and inorganic nanoparticles, used for the delivery of chemotherapeutic agents, siRNA, and miRNA in MM treatment. We delve into nanoscale imaging techniques which provide spatial multi-omic data, offering a holistic view of the tumor microenvironment. This spatial resolution can help decipher the complex interplay between cancer cells and their surrounding environment, facilitating the development of highly targeted therapies. Lastly, we explore the burgeoning field of nano-immunotherapy, which employs nanoparticles to modulate the immune system for myeloma treatment. Specifically, we consider how nanoparticles can be used to deliver tumor antigens to antigen-presenting cells, thus enhancing the body\'s immune response against myeloma cells. In conclusion, nanotechnology holds great promise for improving the prognosis and quality of life of MM patients. However, several challenges remain, including the need for further preclinical and clinical trials to assess the safety and efficacy of these emerging strategies. Future research should also focus on developing personalized nanomedicine approaches, which could tailor treatments to individual patients based on their genetic and molecular profiles.
    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
    背景:肿瘤免疫治疗不仅可以消除原发病灶,还会产生长期免疫记忆,有效抑制肿瘤的转移和复发。然而,免疫疗法在临床实践中也显示出许多局限性。近年来,纳米材料和免疫疗法的结合为完全消除肿瘤带来了新的光,其出色的抗肿瘤作用和可忽略的副作用。
    方法:利用WebofScience核心论集(WOSCC)检索并获取WOSCC成立以来抗肿瘤纳米免疫治疗的相关文献。Bibliometrix,VOSviewer,CiteSpace,GraphPad棱镜,采用Excel进行统计分析和可视化。年产量,积极的机构,核心期刊,主要作者,关键词,主要国家,关键文档,并对纳入期刊的影响因子进行评价。
    结果:2004年至2022年共纳入443项相关研究,文章的年增长率达到惊人的16.85%。就出版物数量而言,领先的国家是中国和美国。控释杂志,生物材料,ActaBiomaterialia,Theranostics,先进材料,ACSNano是核心期刊,发表有关该领域最新进展的高质量文献。集中于树突状细胞和药物递送的文章在该领域占很大比例。关键词如调节性T细胞,肿瘤微环境,免疫检查点封锁,药物输送,光动力疗法,光热疗法,肿瘤相关巨噬细胞是成熟度较高的最热主题之一.树突状细胞,疫苗,T细胞将成为未来研究的热点和新兴课题。
    结论:纳米材料与抗肿瘤免疫治疗的联合治疗,即抗肿瘤纳米免疫疗法日益受到重视。抗肿瘤纳米免疫疗法正在经历从简单到复杂的转变,从表型到机制。
    BACKGROUND: Tumor immunotherapy can not only eliminate the primary lesion, but also produce long-term immune memory, effectively inhibiting tumor metastasis and recurrence. However, immunotherapy also showed plenty of limitations in clinical practice. In recent years, the combination of nanomaterials and immunotherapy has brought new light for completely eliminating tumors with its fabulous anti-tumor effects and negligible side effects.
    METHODS: The Core Collection of Web of Science (WOSCC) was used to retrieve and obtain relevant literatures on antitumor nano-immunotherapy since the establishment of the WOSCC. Bibliometrix, VOSviewer, CiteSpace, GraphPad Prism, and Excel were adopted to perform statistical analysis and visualization. The annual output, active institutions, core journals, main authors, keywords, major countries, key documents, and impact factor of the included journals were evaluated.
    RESULTS: A total of 443 related studies were enrolled from 2004 to 2022, and the annual growth rate of articles reached an astonishing 16.85%. The leading countries in terms of number of publications were China and the United States. Journal of Controlled Release, Biomaterials, Acta Biomaterialia, Theranostics, Advanced Materials, and ACS Nano were core journals publishing high-quality literature on the latest advances in the field. Articles focused on dendritic cells and drug delivery accounted for a large percentage in this field. Key words such as regulatory T cells, tumor microenvironment, immune checkpoint blockade, drug delivery, photodynamic therapy, photothermal therapy, tumor-associated macrophages were among the hottest themes with high maturity. Dendritic cells, vaccine, and T cells tend to become the popular and emerging research topics in the future.
    CONCLUSIONS: The combined treatment of nanomaterials and antitumor immunotherapy, namely antitumor nano-immunotherapy has been paid increasing attention. Antitumor nano-immunotherapy is undergoing a transition from simple to complex, from phenotype to mechanism.
    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
    正在进行的研究正在积极探索使用免疫检查点抑制剂通过抑制PD-1/PD-L1轴和重新激活细胞毒性T效应细胞的功能来治疗实体瘤。许多类型的实体瘤,然而,其特征是致密而坚硬的基质,难以治疗。机械治疗学已经形成了最近一类药物,旨在恢复肿瘤微环境的生物力学异常,与增加的刚度和低灌注有关。这里,我们开发了一种含有吡非尼酮的聚合物制剂,已成功恢复乳腺肿瘤和肉瘤的肿瘤微环境。我们发现胶束制剂可以诱导与4T1和E0771三阴性乳腺肿瘤和MCA205纤维肉瘤的小鼠模型相似的机械治疗效果,但剂量比游离吡非尼酮低100倍。重要的是,吡非尼酮胶束与免疫检查点抑制的组合显着延迟原发性肿瘤的生长,导致E0771肿瘤模型的总生存期和完全治愈的显着改善。此外,联合治疗增加CD4+和CD8+T细胞浸润和抑制髓源性抑制细胞,创造良好的免疫刺激条件,这导致了免疫记忆。超声剪切波弹性成像(SWE)能够监测治疗期间肿瘤硬度的变化,建议最佳治疗条件。胶束封装是机械治疗学的一个有前途的策略,和成像方法,比如SWE,可以帮助他们的临床翻译。
    Ongoing research is actively exploring the use of immune checkpoint inhibitors to treat solid tumors by inhibiting the PD-1/PD-L1 axis and reactivating the function of cytotoxic T effector cells. Many types of solid tumors, however, are characterized by a dense and stiff stroma and are difficult to treat. Mechanotherapeutics have formed a recent class of drugs that aim to restore biomechanical abnormalities of the tumor microenvironment, related to increased stiffness and hypo-perfusion. Here, we have developed a polymeric formulation containing pirfenidone, which has been successful in restoring the tumor microenvironment in breast tumors and sarcomas. We found that the micellar formulation can induce similar mechanotherapeutic effects to mouse models of 4T1 and E0771 triple negative breast tumors and MCA205 fibrosarcoma tumors but with a dose 100-fold lower than that of the free pirfenidone. Importantly, a combination of pirfenidone-loaded micelles with immune checkpoint inhibition significantly delayed primary tumor growth, leading to a significant improvement in overall survival and in a complete cure for the E0771 tumor model. Furthermore, the combination treatment increased CD4+ and CD8+ T cell infiltration and suppressed myeloid-derived suppressor cells, creating favorable immunostimulatory conditions, which led to immunological memory. Ultrasound shear wave elastography (SWE) was able to monitor changes in tumor stiffness during treatment, suggesting optimal treatment conditions. Micellar encapsulation is a promising strategy for mechanotherapeutics, and imaging methods, such as SWE, can assist their clinical translation.
    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
    了解纳米粒子的特性如何影响它们的细胞相互作用是改进载体设计的瓶颈。巨噬细胞极化控制其在解决感染或组织修复中的积极作用。为了揭示碳水化合物靶向甘露糖受体对巨噬细胞表面的影响,使用甘露糖(M)和甘露聚糖(Mn)对无药物的岩藻依聚糖/壳聚糖纳米颗粒进行官能化。通过使用岩藻依聚糖的壳聚糖自组装获得聚电解质复合物纳米颗粒。功能化的纳米粒子根据其物理化学特性进行了表征,化学简介,和碳水化合物取向。纳米粒子的大小从200到400nm不等,是单分散的,并且具有稳定的负ζ电位,具有低聚集趋势。非官能化和官能化的纳米颗粒保持其性质长达12周。在THP-1单核细胞和THP-1分化的巨噬细胞中对所有设计的纳米颗粒进行细胞活力和内化研究。在两种免疫细胞中均证实了甘露糖受体的表达。碳水化合物功能化的纳米颗粒导致它们的活化和促炎细胞因子白细胞介素(IL)-1β的产生,IL-6和肿瘤坏死因子(TNF)-α。M-和Mn-包被的纳米颗粒都将巨噬细胞朝向M1-极化状态调节。这些发现证明了这些纳米平台的定制以在体外相互作用和改变巨噬细胞表型,并代表其单独或与负载的药物组合用于未来研究的治疗潜力。
    Understanding how nanoparticles\' properties influence their cellular interactions is a bottleneck for improving the design of carriers. Macrophage polarization governs their active role in solving infections or tissue repair. To unravel the effect of carbohydrate-targeting mannose receptors on the macrophage surface, drug-free fucoidan/chitosan nanoparticles were functionalized using mannose (M) and mannan (Mn). Polyelectrolyte complex nanoparticles were obtained upon chitosan self-assembly using fucoidan. The functionalized nanoparticles were characterized in terms of their physicochemical characteristics, chemical profile, and carbohydrate orientation. The nanoparticles varied in size from 200 to 400 nm, were monodisperse, and had a stable negative zeta potential with a low aggregation tendency. The nonfunctionalized and functionalized nanoparticles retained their properties for up to 12 weeks. Cell viability and internalization studies were performed for all the designed nanoparticles in the THP-1 monocytes and THP-1-differentiated macrophages. The expression of the mannose receptor was verified in both immune cells. The carbohydrate-functionalized nanoparticles led to their activation and the production of pro-inflammatory cytokines interleukin (IL)-1β, IL-6, and tumour necrosis factor (TNF)-α. Both M- and Mn-coated nanoparticles modulate macrophages toward an M1-polarized state. These findings demonstrate the tailoring of these nanoplatforms to interact and alter the macrophage phenotype in vitro and represent their therapeutic potential either alone or in combination with a loaded drug for future studies.
    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
    血管生成在黑色素瘤的进展和转移中起关键作用,巨噬细胞的促血管生成作用是当前抗血管生成疗法失败的一个主要原因。这里,已经开发了结合阿魏酚和聚(I:C)(阿魏酚/聚(I:C))的纳米免疫疗法来增强巨噬细胞的抗血管生成活性以抑制黑色素瘤。我们的发现表明,阿魏酚/聚(I:C)是一种高效的抗肿瘤疗法,毒性有限。体内和体外实验均表明该组合在阻止血管生成方面是成功的。Ferumoxytol/poly(I:C)被证明可以降低内皮细胞的活力,从而阻碍了管道的形成。特别是,阿魏酚/聚(I:C)能够使巨噬细胞极化为M1表型并降低血管内皮生长因子的表达,这又放大了阿魏酚/聚(I:C)的抗血管生成特性。阿魏酚/聚(I:C)纳米免疫疗法的这种组合丰富了阿魏酚的抗血管生成治疗性,并将为黑色素瘤的治疗提供新的思路。
    Angiogenesis plays a key role in the progression and metastasis of melanoma, and the pro-angiogenic effect of macrophages is one major reason for the failure of current anti-angiogenic therapies. Here, a nano-immunotherapy combining ferumoxytol and poly(I:C) (ferumoxytol/poly(I:C)) has been developed to boost the anti-angiogenic activities of macrophages to inhibit melanoma. Our findings demonstrated that ferumoxytol/poly(I:C) was a highly efficacious anti-tumor therapy with limited toxicity. Both in vivo and in vitro experiments indicated that this combination was successful in impeding angiogenesis. Ferumoxytol/poly(I:C) was demonstrated to reduce the viability of endothelial cells, thus hindering tube formation. Particularly, ferumoxytol/poly(I:C) was able to polarize macrophages to the M1 phenotype and decrease the expression of vascular endothelial growth factor, which in turn amplified the anti-angiogenic properties of ferumoxytol/poly(I:C). This combination of ferumoxytol/poly(I:C) nano-immunotherapy enriches the anti-angiogenic therapeutic nature of ferumoxytol and will shed new light on the treatment of melanoma.
    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: Review
    宿主免疫系统具有以特异性和适应性方式靶向和杀死癌细胞的内在能力,可以通过癌症免疫疗法进一步增强。旨在训练免疫系统以增强抗肿瘤免疫反应。几种不同类别的癌症免疫疗法已成为临床上新的标准癌症疗法。包括癌症疫苗,免疫检查点抑制剂,过继性T细胞疗法,和溶瘤病毒治疗。尽管对一部分患者有显著的生存益处,低应答率和免疫毒性仍然是当前癌症免疫治疗的主要挑战.在过去的几十年里,纳米医学以极大的热情被深入研究,导致纳米粒子平台和纳米工程技术的显著进步。纳米医学和免疫疗法的进展也导致了纳米免疫疗法一个新生的研究领域的出现,其目的是在纳米医学的帮助下实现免疫治疗的全部治疗潜力。特别是,纳米载体在免疫肿瘤学中提供了一个令人兴奋的机会来增强活性,增加特异性,降低毒性,并通过增强免疫刺激活性和有利地调节药理学性质来维持免疫剂的抗肿瘤功效。这篇综述讨论了纳米载体用于癌症免疫治疗的潜力,并介绍了旨在使用基于纳米载体的工程方法改善临床癌症免疫治疗方式的临床前研究。它还讨论了纳米载体解决免疫肿瘤学当前面临的挑战以及将其转化为临床应用的挑战的潜力。
    The host immune system possesses an intrinsic ability to target and kill cancer cells in a specific and adaptable manner that can be further enhanced by cancer immunotherapy, which aims to train the immune system to boost the antitumor immune response. Several different categories of cancer immunotherapy have emerged as new standard cancer therapies in the clinic, including cancer vaccines, immune checkpoint inhibitors, adoptive T cell therapy, and oncolytic virus therapy. Despite the remarkable survival benefit for a subset of patients, the low response rate and immunotoxicity remain the major challenges for current cancer immunotherapy. Over the last few decades, nanomedicine has been intensively investigated with great enthusiasm, leading to marked advancements in nanoparticle platforms and nanoengineering technology. Advances in nanomedicine and immunotherapy have also led to the emergence of a nascent research field of nano-immunotherapy, which aims to realize the full therapeutic potential of immunotherapy with the aid of nanomedicine. In particular, nanocarriers present an exciting opportunity in immuno-oncology to boost the activity, increase specificity, decrease toxicity, and sustain the antitumor efficacy of immunological agents by potentiating immunostimulatory activity and favorably modulating pharmacological properties. This review discusses the potential of nanocarriers for cancer immunotherapy and introduces preclinical studies designed to improve clinical cancer immunotherapy modalities using nanocarrier-based engineering approaches. It also discusses the potential of nanocarriers to address the challenges currently faced by immuno-oncology as well as the challenges for their translation to clinical applications.
    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
    纳米人工抗原呈递细胞(aAPC),旨在体外或体内激活T细胞的合成免疫细胞模拟物,提供细胞免疫疗法的有效替代方案。然而,描述纳米-aAPC拓扑效应的综合研究,包括纳米粒子形态和配体密度,缺乏。这里,我们系统地研究了基于聚合物小体的aAPC对T细胞活化的拓扑效应。我们采用了由可生物降解的聚(乙二醇)-嵌段-聚(d,具有球形或管状形状和可变尺寸的l-丙交酯(PEG-PDLLA)聚合物囊泡,用控制密度的αCD3和αCD28抗体官能化。我们的结果表明,高配体密度导致T细胞活化增强,可以通过使用较大尺寸的聚合物囊泡来进一步增强。在低配体密度下,聚合物体形状和大小的影响更明显,显示与球形或较小的对应物相比,大的细长聚合物囊泡更好地激活T细胞。这项研究证明了聚合物囊泡作为aAPC的能力,并强调了拓扑结构在其合理设计中的作用。
    Nanosized artificial antigen-presenting cells (aAPCs), synthetic immune cell mimics that aim to activate T cells ex or in vivo, offer an effective alternative to cellular immunotherapies. However, comprehensive studies that delineate the effect of nano-aAPC topology, including nanoparticle morphology and ligand density, are lacking. Here, we systematically studied the topological effects of polymersome-based aAPCs on T cell activation. We employed an aAPC library created from biodegradable poly(ethylene glycol)-block-poly(d,l-lactide) (PEG-PDLLA) polymersomes with spherical or tubular shape and variable sizes, which were functionalized with αCD3 and αCD28 antibodies at controlled densities. Our results indicate that high ligand density leads to enhancement in T cell activation, which can be further augmented by employing polymersomes with larger size. At low ligand density, the effect of both polymersome shape and size was more pronounced, showing that large elongated polymersomes better activate T cells compared to their spherical or smaller counterparts. This study demonstrates the capacity of polymersomes as aAPCs and highlights the role of topology for their rational design.
    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