nanovaccines

纳米疫苗
  • 文章类型: Journal Article
    耐药性是癌症化疗中的重大挑战,并且是导致癌症患者恢复不良的主要因素。尽管载药纳米颗粒在克服化疗耐药性方面显示出了希望,他们经常携带药物组合,需要先进的设计和制造工艺。此外,从免疫疗法的角度来看,他们很少接近化疗耐药的肿瘤。在这项研究中,我们开发了一种仅由化疗诱导的耐药肿瘤抗原(CIRTAs)和免疫佐剂Toll样受体(TLR)7/8激动剂R848(CIRTAs@R848)组成的治疗性纳米疫苗.这种纳米疫苗不需要额外的载体并且具有简单的生产过程。它有效地同时向树突状细胞(DCs)递送抗原和免疫刺激剂,促进DC成熟。CIRTAs@R848表现出显著的肿瘤抑制,特别是与免疫检查点阻断(ICB)抗PD-1(αPD-1)联合使用时。联合治疗增加了T细胞向肿瘤的浸润,同时降低了调节性T细胞(Tregs)的比例并调节了肿瘤微环境,导致长期免疫记忆。总的来说,这项研究从一个新的角度介绍了一种治疗化疗耐药肿瘤的创新策略,具有在个性化免疫疗法和精准医学中的潜在应用。
    Drug resistance is a significant challenge in cancer chemotherapy and is a primary factor contributing to poor recovery for cancer patients. Although drug-loaded nanoparticles have shown promise in overcoming chemotherapy resistance, they often carry a combination of drugs and require advanced design and manufacturing processes. Furthermore, they seldom approach chemotherapy-resistant tumors from an immunotherapy perspective. In this study, we developed a therapeutic nanovaccine composed solely of chemotherapy-induced resistant tumor antigens (CIRTAs) and the immune adjuvant Toll-like receptor (TLR) 7/8 agonist R848 (CIRTAs@R848). This nanovaccine does not require additional carriers and has a simple production process. It efficiently delivers antigens and immune stimulants to dendritic cells (DCs) simultaneously, promoting DCs maturation. CIRTAs@R848 demonstrated significant tumor suppression, particularly when used in combination with the immune checkpoint blockade (ICB) anti-PD-1 (αPD-1). The combined therapy increased the infiltration of T cells into the tumor while decreasing the proportion of regulatory T cells (Tregs) and modulating the tumor microenvironment, resulting in long-term immune memory. Overall, this study introduces an innovative strategy for treating chemotherapy-resistant tumors from a novel perspective, with potential applications in personalized immunotherapy and precision medicine.
    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
    硒(Se)是对人体健康至关重要的元素,以其抗癌特性而闻名。尽管硒纳米颗粒(SeNPs)显示出比其他硒化合物更低的毒性和更高的生物相容性,裸露的SeNP在水溶液中不稳定。在这项研究中,几种材料,包括牛血清白蛋白(BSA),壳聚糖,聚甲基乙烯基醚-alt-马来酸酐,和生育酚聚乙二醇琥珀酸酯,探索开发稳定的SeNPs,并进一步评估其作为癌症治疗候选者的潜力。所有优化的SeNP都是球形的,<100nm,尺寸分布窄。在酸性条件下产生的BSA稳定的SeNPs在培养基中表现出最高的稳定性,等离子体,在生理pH下,长时间保持其尺寸≈50-60nm。SeNP在癌细胞系中表现出增强的毒性,同时保留原代人真皮成纤维细胞,强调它们作为有效抗癌剂的潜力。此外,BSA-SeNPs与纳米疫苗的组合在EO771乳腺癌小鼠模型中导致强烈的肿瘤生长减少,显示肿瘤大小减少了三倍。这种协同抗癌作用不仅突出了SeNPs作为有效抗癌剂的作用,而且为开发使用SeNPs改善癌症免疫治疗结果的创新组合方法提供了有价值的见解。
    Selenium (Se) is an element crucial for human health, known for its anticancer properties. Although selenium nanoparticles (SeNPs) have shown lower toxicity and higher biocompatibility than other Se compounds, bare SeNPs are unstable in aqueous solutions. In this study, several materials, including bovine serum albumin (BSA), chitosan, polymethyl vinyl ether-alt-maleic anhydride, and tocopherol polyethylene glycol succinate, are explored to develop stable SeNPs and further evaluate their potential as candidates for cancer treatment. All optimized SeNP are spherical, <100 nm, and with a narrow size distribution. BSA-stabilized SeNPs produced under acidic conditions present the highest stability in medium, plasma, and at physiological pH, maintaining their size ≈50-60 nm for an extended period. SeNPs demonstrate enhanced toxicity in cancer cell lines while sparing primary human dermal fibroblasts, underscoring their potential as effective anticancer agents. Moreover, the combination of BSA-SeNPs with a nanovaccine results in a strong tumor growth reduction in an EO771 breast cancer mouse model, demonstrating a three-fold decrease in tumor size. This synergistic anticancer effect not only highlights the role of SeNPs as effective anticancer agents but also offers valuable insights for developing innovative combinatorial approaches using SeNPs to improve the outcomes of cancer immunotherapy.
    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
    慢性乙型肝炎病毒(HBV)感染是一个全球性的健康问题,大大增加了发展肝病的风险。诱导抗HB血清转换并实现针对慢性HBV感染的持久免疫应答的新策略的开发仍然具有挑战性。这里,我们发现,慢性HBV感染影响了STING介导的树突状细胞(DCs)宿主免疫应答诱导所涉及的信号通路,然后产生了淋巴结靶向的纳米疫苗,该疫苗共递送了乙型肝炎表面抗原(HBsAg)和环状单磷酸二鸟苷酸(c-di-GMP)(命名为PP-SG纳米疫苗).在HBV携带者小鼠中评估了PP-SG纳米疫苗用于CHB治疗的可行性和效率。血清样品分析HBsAg,抗HBs,HBVDNA,丙氨酸转氨酶水平,和肝脏样本的HBVDNA和RNA和HBcAg进行评估,伴随着在PP-SG纳米疫苗治疗过程中HBV特异性细胞和体液免疫反应的分析。PP-SG纳米疫苗增加抗原吞噬和DC成熟,有效和安全地消除HBV,实现了针对HBV再注射的持久免疫反应,并破坏慢性HBV感染诱导的免疫耐受,其特征是HBV特异性CD8+T和CD4+T细胞的产生和多功能性以及免疫检查点分子的下调。用PP-SG纳米疫苗免疫的HBV携带者小鼠实现了部分抗HBs血清转换。PP-SG纳米疫苗可以诱导足够和持久的病毒抑制和实现抗HBs血清转化,使其成为临床慢性乙型肝炎治疗的有希望的疫苗候选。
    Chronic hepatitis B virus (HBV) infection is a global health problem that substantially increases the risk of developing liver disease. The development of a novel strategy to induce anti-HB seroconversion and achieve a long-lasting immune response against chronic HBV infection remains challenging. Here, we found that chronic HBV infection affected the signaling pathway involved in STING-mediated induction of host immune responses in dendritic cells (DCs) and then generated a lymph node-targeted nanovaccine that co-delivered hepatitis B surface antigen (HBsAg) and cyclic diguanylate monophosphate (c-di-GMP) (named the PP-SG nanovaccine). The feasibility and efficiency of the PP-SG nanovaccine for CHB treatment were evaluated in HBV-carrier mice. Serum samples were analyzed for HBsAg, anti-HBs, HBV DNA, and alanine aminotransferase levels, and liver samples were evaluated for HBV DNA and RNA and HBcAg, accompanied by an analysis of HBV-specific cellular and humoral immune responses during PP-SG nanovaccine treatment. The PP-SG nanovaccine increased antigen phagocytosis and DC maturation, efficiently and safely eliminated HBV, achieved a long-lasting immune response against HBV reinjection, and disrupted chronic HBV infection-induced immune tolerance, as characterized by the generation and multifunctionality of HBV-specific CD8+ T and CD4+ T cells and the downregulation of immune checkpoint molecules. HBV-carrier mice immunized with the PP-SG nanovaccine achieved partial anti-HBs seroconversion. The PP-SG nanovaccine can induce sufficient and persistent viral suppression and achieve anti-HBs seroconversion, rendering it a promising vaccine candidate for clinical chronic hepatitis B therapy.
    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
    纳米支架的应用已成为疫苗设计中一个很有前景的策略,基于蛋白质的纳米颗粒为生物相容性和有效递送抗原提供了理想的途径。这里,我们提出了一种新的内源性衣壳形成蛋白,活化调节细胞骨架相关蛋白(ARC),可以通过即插即用策略(SpyCatcher3/SpyTag3)进行工程改造,用于多价展示抗原。结合ARC的自组装能力和灵活的模块化,基于ARC的疫苗引发了针对Mpox或SARS-CoV-2的强大免疫应答,与基于铁蛋白的疫苗诱导的免疫应答相当。此外,基于ARC的纳米粒子用作免疫刺激剂,有效刺激树突状细胞并促进生发中心反应。即使没有佐剂,基于ARC的疫苗在致命攻击模型中产生保护性免疫应答。因此,这项研究显示了ARC作为一种新型的基于蛋白质的纳米载体用于多价表面展示病原抗原的可行性,并证明了利用重组哺乳动物逆转录病毒样蛋白作为生物活性分子递送载体的潜力.
    The application of nanoscale scaffolds has become a promising strategy in vaccine design, with protein-based nanoparticles offering desirable avenues for the biocompatible and efficient delivery of antigens. Here, we presented a novel endogenous capsid-forming protein, activated-regulated cytoskeleton-associated protein (ARC), which could be engineered through the plug-and-play strategy (SpyCatcher3/SpyTag3) for multivalent display of antigens. Combined with the self-assembly capacity and flexible modularity of ARC, ARC-based vaccines elicited robust immune responses against Mpox or SARS-CoV-2, comparable to those induced by ferritin-based vaccines. Additionally, ARC-based nanoparticles functioned as immunostimulants, efficiently stimulating dendritic cells and facilitating germinal center responses. Even without adjuvants, ARC-based vaccines generated protective immune responses in a lethal challenge model. Hence, this study showed the feasibility of ARC as a novel protein-based nanocarrier for multivalent surface display of pathogenic antigens and demonstrated the potential of exploiting recombinant mammalian retrovirus-like protein as a delivery vehicle for bioactive molecules.
    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
    纤维肉瘤,恶性间质瘤,具有侵袭性和高复发率,导致预后不良。蒽环类药物,如阿霉素(DOX),代表纤维肉瘤的一线化疗,但往往表现出次优的疗效。最近,利用干扰素基因刺激因子(STING)介导的先天免疫已成为癌症治疗的希望策略。在化学免疫疗法中整合化学疗法与免疫调节剂已显示出增强治疗结果的潜力。在这里,我们引入了先进的树突状细胞(DC)纳米疫苗,cGAMP@PLGA@CRTM(GP@CRTM),联合低剂量DOX增强纤维肉瘤化疗免疫疗法。纳米疫苗由包裹STING激动剂2,3-cGAMP(cGAMP@PLGA,GP)作为其核心,以钙网蛋白(CRT)高表达的纤维肉瘤细胞膜(CRTM)作为外壳。将CRT暴露在疫苗表面有助于招募DC和刺激摄取,促进STING激动剂和肿瘤抗原向DC的有效同时递送。这种双重递送方法有效激活DCs中的STING途径,引发持续的免疫刺激。同时,低剂量DOX减少化疗相关的副作用,直接杀死一部分肿瘤细胞,并增加肿瘤的免疫原性,从而进一步放大免疫治疗性能。因此,这些发现证明了DC纳米疫苗GP@CRTM作为化疗助推器的潜力.低剂量DOX与DC纳米疫苗的协同结合成为一种强大的化学免疫治疗策略,优化全身纤维肉瘤治疗。
    Fibrosarcoma, a malignant mesenchymal tumor, is characterized by aggressive invasiveness and a high recurrence rate, leading to poor prognosis. Anthracycline drugs, such as doxorubicin (DOX), represent the frontline chemotherapy for fibrosarcoma, but often exhibit suboptimal efficacy. Recently, exploiting the stimulator of interferon genes (STING)-mediated innate immunity has emerged as a hopeful strategy for cancer treatment. Integrating chemotherapy with immunomodulators in chemo-immunotherapy has shown potential for enhancing treatment outcomes. Herein, we introduce an advanced dendritic cell (DC) nanovaccine, cGAMP@PLGA@CRTM (GP@CRTM), combined with low-dose DOX to enhance fibrosarcoma chemo-immunotherapy. The nanovaccine consists of poly(lactic-co-glycolic acid) (PLGA) nanoparticles encapsulating the STING agonist 2,3-cGAMP (cGAMP@PLGA, GP) as its core, and a calreticulin (CRT) high-expressing fibrosarcoma cell membrane (CRTM) as the shell. Exposing CRT on the vaccine surface aids in recruiting DCs and stimulating uptake, facilitating efficient simultaneous delivery of STING agonists and tumor antigens to DCs. This dual delivery method effectively activates the STING pathway in DCs, triggering sustained immune stimulation. Simultaneously, low-dose DOX reduces chemotherapy-related side effects, directly kills a subset of tumor cells, and increases tumor immunogenicity, thus further amplifying immune therapeutic performance. Hence, these findings demonstrate the potential of DC nanovaccine GP@CRTM as a booster for chemotherapy. Synergistically combining low-dose DOX with the DC nanovaccine emerges as a powerful chemo-immunotherapy strategy, optimizing systemic fibrosarcoma therapy.
    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
    已经设计了纳米疫苗以克服与常规疫苗相关的限制。有效的递送方法如工程载体或智能纳米颗粒(NPs)是诱导自我耐受性和优化疫苗免疫原性且副作用最小的关键必要条件。NP可以用作佐剂,免疫原,或纳米载体以开发用于有效抗原递送的纳米疫苗。携带多种肿瘤抗原和免疫刺激剂的多载纳米疫苗可以有效地增加对肿瘤细胞的免疫力。它们可以被生物工程化以增强与树突细胞的相互作用并允许逐渐和恒定的抗原释放。修改NP表面属性,使用高密度脂蛋白模拟纳米盘,以及开发基于纳米的人工抗原呈递细胞,如树突状细胞衍生的外泌体,是增强针对肿瘤细胞的抗原呈递和免疫反应的新开发技术之一。本综述概述了不同的观点,改进,以及当前癌症治疗和疫苗接种方案成功临床应用的障碍。描述了不同类型的纳米疫苗和掺入其结构中的纳米颗粒的免疫调节作用。使用纳米疫苗预防和治疗艾滋病等常见疾病的优势,疟疾,讨论了癌症和结核病。Further,描述了开发最佳癌症疫苗的潜在途径。鉴于癌细胞和肿瘤微环境的免疫抑制特性,免疫调节剂和免疫检查点抑制剂与其他常规抗癌疗法联合应用对于提高免疫反应的有效性是必要的.
    Nanovaccines have been designed to overcome the limitations associated with conventional vaccines. Effective delivery methods such as engineered carriers or smart nanoparticles (NPs) are critical requisites for inducing self-tolerance and optimizing vaccine immunogenicity with minimum side effects. NPs can be used as adjuvants, immunogens, or nanocarriers to develop nanovaccines for efficient antigen delivery. Multiloaded nanovaccines carrying multiple tumor antigens along with immunostimulants can effectively increase immunity against tumor cells. They can be biologically engineered to boost interactions with dendritic cells and to allow a gradual and constant antigen release. Modifying NPs surface properties, using high-density lipoprotein-mimicking nanodiscs, and developing nano-based artificial antigen-presenting cells such as dendritic cell-derived-exosomes are amongst the new developed technologies to enhance antigen-presentation and immune reactions against tumor cells. The present review provides an overview on the different perspectives, improvements, and barriers of successful clinical application of current cancer therapeutic and vaccination options. The immunomodulatory effects of different types of nanovaccines and the nanoparticles incorporated into their structure are described. The advantages of using nanovaccines to prevent and treat common illnesses such as AIDS, malaria, cancer and tuberculosis are discussed. Further, potential paths to develop optimal cancer vaccines are described. Given the immunosuppressive characteristics of both cancer cells and the tumor microenvironment, applying immunomodulators and immune checkpoint inhibitors in combination with other conventional anticancer therapies are necessary to boost the effectiveness of the immune 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
    长期以来,卵巢癌(OC)的死亡率一直是妇科恶性肿瘤中最高的。虽然OC被认为是一种免疫原性肿瘤,免疫治疗效果不理想。免疫抑制微环境是其中一个原因,另一个原因是缺乏公认的疫苗有效抗原。化疗,作为OC最常用的治疗方法之一,在治疗过程中可以产生化疗相关抗原(CAAs),并显示出原位疫苗的效果。在这里,我们设计了一种抗原捕获纳米疫苗NP-TP1@M-M,其肿瘤靶向肽TMTP1和树突状细胞(DC)受体甘露糖组装在表面,佐剂单磷酰脂质A(MPLA)包裹在聚(D,L-丙交酯-共-乙交酯)(PLGA)纳米颗粒。PLGA本身具有抗原捕获能力。TMTP1是由我们的研究小组筛选的肿瘤归巢肽,具有广泛而优异的肿瘤靶向能力。经过这些修改,NP-TP1@M-M可以在化疗后捕获和富集更多的肿瘤特异性抗原,刺激DC成熟,激活适应性免疫,并结合免疫检查点封锁,以最大限度地释放身体的免疫潜能,为OC的治疗提供了一种良好的治疗策略。
    The mortality of ovarian cancer (OC) has long been the highest among gynecological malignancies. Although OC is considered to be an immunogenic tumor, the effect of immunotherapy is not satisfactory. The immunosuppressive microenvironment is one reason for this, and the absence of recognized effective antigens for vaccines is another. Chemotherapy, as one of the most commonly used treatment for OC, can produce chemotherapy-associated antigens (CAAs) during treatment and show the effect of in situ vaccine. Herein, we designed an antigen capture nano-vaccine NP-TP1@M-M with tumor targeting peptide TMTP1 and dendritic cell (DC) receptor mannose assembled on the surface and adjuvant monophosphoryl lipid A (MPLA) encapsulated in the core of poly (D, L-lactide-co-glycolide) (PLGA) nanoparticles. PLGA itself possessed the ability of antigen capture. TMTP1 was a tumor-homing peptide screened by our research team, which held extensive and excellent tumor targeting ability. After these modifications, NP-TP1@M-M could capture and enrich more tumor-specific antigens after chemotherapy, stimulate DC maturation, activate the adaptive immunity and combined with immune checkpoint blockade to maximize the release of the body\'s immune potential, providing an eutherapeutic strategy for the treatment of OC.
    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
    纳米疫苗治疗是免疫学和个性化医学研究的一个令人兴奋的领域,在增强免疫反应和针对特定疾病方面有着巨大的希望。它们的小尺寸允许免疫细胞有效吸收,导致强大的免疫激活。它们可以掺入免疫刺激分子以提高疫苗效力。因此,纳米疫苗可以个性化靶向肿瘤特异性抗原,激活免疫系统对抗癌细胞。目前,有充分的证据表明纳米疫苗作为癌症治疗的有效性和潜力。然而,有罕见的癌症纳米疫苗的文献计量学分析。在这里,我们对已发表的与纳米疫苗治疗癌症相关的研究进行了文献计量和视觉分析,未来纳米疫苗的发展趋势。
    我们收集了基于WebofScienceCoreCollectionSCI扩展数据库的文献。文献计量分析是通过利用可视化分析工具VOSviewer进行的,共同发生(COOC),城市空间,Bibliometrix(R-ToolofR-Studio),还有Hitcite.
    本研究共纳入517篇文献。中国是出版物最多的国家,也是当地引文总分(TLCS)最高的国家。中国科学院拥有该领域最大的研究数量,最多产的作者是南开大学的德令孔。该领域最著名的期刊是生物材料。研究主要集中在肿瘤纳米疫苗的治疗过程,纳米疫苗的颗粒组成和应用,提示纳米疫苗的潜在热点和趋势。
    在这项研究中,我们总结了涉及纳米疫苗的出版物的特征和变化趋势,并对最具影响力的国家进行了分类,机构,作者,期刊,关于癌症纳米疫苗的热点和趋势。随着纳米材料和肿瘤免疫治疗技术的不断发展,纳米疫苗为癌症提供了一个具有重要临床价值和潜在应用的研究领域。
    UNASSIGNED: Nanovaccine treatment is an exciting area of research in immunology and personalized medicine, holding great promise for enhancing immune responses and targeting specific diseases. Their small size allows efficient uptake by immune cells, leading to robust immune activation. They can incorporate immune-stimulating molecules to boost vaccine efficacy. Therefore, nanovaccine can be personalized to target tumor-specific antigens, activating the immune system against cancer cells. Currently, there have been ample evidence showing the effectiveness and potential of nanovaccine as a treatment for cancer. However, there was rare bibliometric analysis of nanovaccine for cancer. Here we performed a bibliometric and visual analysis of published studies related to nanovaccine treatment for cancer, providing the trend of future development of nanovaccine.
    UNASSIGNED: We collected the literatures based on the Web of Science Core Collection SCI-Expanded database. The bibliometric analysis was performed via utilizing visualization analysis tools VOSviewer, Co-Occurrence (COOC), Citespace, Bibliometrix (R-Tool of R-Studio), and HitCite.
    UNASSIGNED: A total of 517 literatures were included in this study. China is the country with the most publications and the highest total local citation score (TLCS). The Chinese Academy of Sciences holds the largest research count in this field and the most prolific author is Deling Kong from Nankai University. The most prominent journal for publishing in this area is Biomaterials. The researches mainly focus on the therapeutic process of tumor nanovaccines, the particle composition and the application of nanovaccines, suggesting the potential hotspots and trends of nanovaccine.
    UNASSIGNED: In this study, we summarized the characteristics and variation trends of publications involved in nanovaccine, and categorized the most influential countries, institutions, authors, journals, hotspots and trends regarding the nanovaccine for cancer. With the continuous development of nanomaterials and tumor immunotherapy, nanovaccine for cancer provides a research field of significant clinical value and potential application.
    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
    首批批准的人类使用的针对严重急性呼吸道综合症冠状病毒2(SARS-CoV-2)的疫苗是基于纳米技术的。虽然它们是模块化的,迅速生产,可以减轻疾病的严重程度,目前可用的疫苗在预防感染方面受到限制,强调全球对新型预防性疫苗技术的需求。记住这一点,我们着手开发一种灵活的纳米疫苗平台,用于鼻腔给药以诱导粘膜免疫,这是防止呼吸道病毒感染的最佳保护的基础。下一代多表位纳米疫苗共同递送免疫原性肽,通过免疫信息学工作流程选择,以及PD-L1表达的佐剂和调节剂。作为一个案例研究,我们以SARS-CoV-2肽作为相关抗原来验证该方法。该平台可以引起针对SARS-CoV-2的局部和系统性细胞和体液特异性反应。这导致免疫球蛋白A(IgA)的分泌,能够中和SARS-CoV-2,包括关注的变种,遵循异源免疫策略。考虑到当前基于纳米技术的疫苗所需的冷链分布的局限性,这表明冻干的纳米疫苗在室温下是长期稳定的,并且在重建时保持其体内功效。这使其与发展中国家特别相关,并提供了适应未来病毒威胁的模块化系统。
    The first approved vaccines for human use against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are nanotechnology-based. Although they are modular, rapidly produced, and can reduce disease severity, the currently available vaccines are restricted in preventing infection, stressing the global demand for novel preventive vaccine technologies. Bearing this in mind, we set out to develop a flexible nanovaccine platform for nasal administration to induce mucosal immunity, which is fundamental for optimal protection against respiratory virus infection. The next-generation multiepitope nanovaccines co-deliver immunogenic peptides, selected by an immunoinformatic workflow, along with adjuvants and regulators of the PD-L1 expression. As a case study, we focused on SARS-CoV-2 peptides as relevant antigens to validate the approach. This platform can evoke both local and systemic cellular- and humoral-specific responses against SARS-CoV-2. This led to the secretion of immunoglobulin A (IgA), capable of neutralizing SARS-CoV-2, including variants of concern, following a heterologous immunization strategy. Considering the limitations of the required cold chain distribution for current nanotechnology-based vaccines, it is shown that the lyophilized nanovaccine is stable for long-term at room temperature and retains its in vivo efficacy upon reconstitution. This makes it particularly relevant for developing countries and offers a modular system adaptable to future viral threats.
    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
    靶向特异性新抗原的个性化癌症疫苗已被设想为癌症免疫疗法中最有希望的方法之一。然而,鉴定的新抗原的物理化学变异性限制了它们的功效以及统一格式的疫苗生产.在这里,我们开发了一种基于聚(2-恶唑啉)(POx)的统一纳米疫苗平台,以化学缀合新抗原肽,不管它们的物理化学性质。该疫苗系统可以自组装成具有均匀尺寸(约50nm)的纳米颗粒,并改善抗原积累以及淋巴结中的浸润以增加抗原呈递。与游离肽相比,使用与来自MC38肿瘤细胞系的三种预测的肽新抗原肽缀合的该系统的体内疫苗接种诱导100%稳健的CD8+T细胞应答和优异的肿瘤清除。这种基于POx的疫苗载体代表了提高个性化癌症疫苗的筛选的新抗原肽的可用性和功效的可推广方法。
    Personalized cancer vaccines targeting specific neoantigens have been envisioned as one of the most promising approaches in cancer immunotherapy. However, the physicochemical variability of the identified neoantigens limits their efficacy as well as vaccine manufacturing in a uniform format. Herein, we developed a uniform nanovaccine platform based on poly(2-oxazoline)s (POx) to chemically conjugate neoantigen peptides, regardless of their physicochemical properties. This vaccine system could self-assemble into nanoparticles with uniform size (around 50 nm) and improve antigen accumulation as well as infiltration in the lymph node to increase antigen presentation. In vivo vaccination using this system conjugated with three predicted peptide neoantigen peptides from the MC38 tumor cell line induced 100% robust CD8+ T cell responses and superior tumor clearance compared to free peptides. This POx-based vaccine carrier represents a generalizable approach to increase the availability and efficacy of screened neoantigen peptides for a personalized cancer vaccine.
    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