vaccine platform

  • 文章类型: Journal Article
    幽门螺杆菌引起全球流行的感染,与慢性胃炎甚至胃癌的发展高度相关。随着抗生素耐药性的增加,科学家们已经开始寻找更好的疫苗设计策略来根除幽门螺杆菌定植。然而,虽然目前的策略更喜欢用单一的幽门螺杆菌抗原配制疫苗,他们的潜力尚未充分发挥。外膜囊泡(OMV)是潜在的平台,因为它们可以递送多种抗原。在这项研究中,我们设计了三种关键的幽门螺杆菌抗原蛋白(UreB,卡加,和VacA)到源自鼠伤寒沙门氏菌的OMV表面(S.使用血红蛋白蛋白酶(Hbp)自转运蛋白系统的鼠伤寒)突变菌株。在各种淘汰赛策略中,我们发现,从ΔrfbPΔfliCΔfljBΔompA突变体中分离出的OMV可以引起免疫球蛋白G(IgG)和A(IgA)水平的明显增加,并有效地触发T辅助细胞1-和17-偏向的细胞免疫反应,在预防幽门螺杆菌方面发挥着至关重要的作用。接下来,衍生自ΔrfbPΔfliCΔfljBΔompA突变体的OMV用作载体以递送幽门螺杆菌抗原的不同组合。小鼠模型中的抗体和细胞因子水平以及攻击实验表明,共同递送UreB和CagA可以保护免受幽门螺杆菌和抗原特异性T细胞应答。总之,从鼠伤寒沙门氏菌ΔrfbPΔfliCΔfljBΔompA突变株作为载体,同时使用Hbp自转运蛋白系统导入幽门螺杆菌UreB和CagA作为抗原蛋白,将大大有利于控制幽门螺杆菌感染。
    外膜囊泡(OMV),作为一种新型的抗原递送平台,已用于各种病原体甚至肿瘤的疫苗设计。肠病沙门氏菌(S.鼠伤寒),作为一种易于工程化并具有佐剂功效和免疫刺激能力的细菌,已成为继大肠杆菌之后纯化OMV的首选细菌载体。本研究的重点是幽门螺杆菌的设计;(H.幽门螺杆菌)疫苗,利用基因修饰的沙门氏菌OMV呈递幽门螺杆菌的几种主要抗原,包括UreB,VacA和CagA.筛选并鉴定了最佳的沙门氏菌OMV递送载体和抗原组合,为H.pylori疫苗的开发提供了新的思路,并为其他难以开发的细菌疫苗提供了一个集成的抗原递送平台,病毒,甚至肿瘤。
    Helicobacter pylori causes globally prevalent infections that are highly related to chronic gastritis and even development of gastric carcinomas. With the increase of antibiotic resistance, scientists have begun to search for better vaccine design strategies to eradicate H. pylori colonization. However, while current strategies prefer to formulate vaccines with a single H. pylori antigen, their potential has not yet been fully realized. Outer membrane vesicles (OMVs) are a potential platform since they could deliver multiple antigens. In this study, we engineered three crucial H. pylori antigen proteins (UreB, CagA, and VacA) onto the surface of OMVs derived from Salmonella enterica serovar Typhimurium (S. Typhimurium) mutant strains using the hemoglobin protease (Hbp) autotransporter system. In various knockout strategies, we found that OMVs isolated from the ΔrfbP ΔfliC ΔfljB ΔompA mutants could cause distinct increases in immunoglobulin G (IgG) and A (IgA) levels and effectively trigger T helper 1- and 17-biased cellular immune responses, which perform a vital role in protecting against H. pylori. Next, OMVs derived from ΔrfbP ΔfliC ΔfljB ΔompA mutants were used as a vector to deliver different combinations of H. pylori antigens. The antibody and cytokine levels and challenge experiments in mice model indicated that co-delivering UreB and CagA could protect against H. pylori and antigen-specific T cell responses. In summary, OMVs derived from the S. Typhimurium ΔrfbP ΔfliC ΔfljB ΔompA mutant strain as the vector while importing H. pylori UreB and CagA as antigenic proteins using the Hbp autotransporter system would greatly benefit controlling H. pylori infection.
    Outer membrane vesicles (OMVs), as a novel antigen delivery platform, has been used in vaccine design for various pathogens and even tumors. Salmonella enterica serovar Typhimurium (S. Typhimurium), as a bacterium that is easy to engineer and has both adjuvant efficacy and immune stimulation capacity, has become the preferred bacterial vector for purifying OMVs after Escherichia coli. This study focuses on the design of Helicobacter pylori ;(H. pylori) vaccines, utilizing genetically modified Salmonella OMVs to present several major antigens of H. pylori, including UreB, VacA and CagA. The optimal Salmonella OMV delivery vector and antigen combinations are screened and identified, providing new ideas for the development of H. pylori vaccines and an integrated antigen delivery platform for other difficult to develop vaccines for bacteria, viruses, and even 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: Journal Article
    目前的抗原递送平台,如明矾和纳米粒子,不容易可调,因此可能不会产生最佳的适应性免疫反应。我们通过用含有靶抗原的水溶液装载冻干的微孔退火颗粒(MAP)来创建抗原递送平台。在施用抗原负载的MAP(VaxMAP)后,生物材料重建形成具有持续释放曲线的即时抗原负载多孔支架区域以最大化体液免疫。与明矾相似,VaxMAP诱导淋巴结中的CD4T滤泡辅助(Tfh)细胞和生发中心(GC)B细胞反应。负载SARS-CoV-2刺突蛋白的VaxMAP提高了幅度,中和,和抗受体结合结构域抗体与明矾疫苗接种小鼠相比的持续时间。与HA1负载明矾相比,单次注射流感特异性HA1负载VaxMAP增强了中和抗体,并引发了针对流感病毒攻击的更大保护。因此,VaxMAP是一个平台,可用于促进适应性免疫细胞反应,以产生更强大的中和抗体,以及更好的病原体攻击保护。
    Current antigen delivery platforms, such as alum and nanoparticles, are not readily tunable, thus may not generate optimal adaptive immune responses. We created an antigen delivery platform by loading lyophilized Microporous Annealed Particle (MAP) with aqueous solution containing target antigens. Upon administration of antigen loaded MAP (VaxMAP), the biomaterial reconstitution forms an instant antigen-loaded porous scaffold area with a sustained release profile to maximize humoral immunity. VaxMAP induced CD4+ T follicular helper (Tfh) cells and germinal center (GC) B cell responses in the lymph nodes similar to Alum. VaxMAP loaded with SARS-CoV-2 spike protein improved the magnitude, neutralization, and duration of anti-receptor binding domain antibodies compared to Alum vaccinated mice. A single injection of Influenza specific HA1-loaded-VaxMAP enhanced neutralizing antibodies and elicited greater protection against influenza virus challenge than HA1-loaded-Alum. Thus, VaxMAP is a platform that can be used to promote adaptive immune cell responses to generate more robust neutralizing antibodies, and better protection upon pathogen challenge.
    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
    各种疫苗平台,包括新兴平台,已应用于COVID-19疫苗的开发。生物技术初创公司经常引领新医疗技术的发展,而大型制药公司和公共机构长期以来一直为疫苗开发做出贡献。在这项研究中,对参与COVID-19疫苗开发的疫苗平台和开发者进行了分析,阐明使用的疫苗平台的趋势,开发商的国家分布,以及疫苗平台技术和国家的开发人员概况的差异。分析表明,传统的,已建立,和新兴的疫苗平台已被广泛使用,较旧的平台在临床开发中更先进。它也展示了中国的崛起,除了美国,虽然许多国家一直在发展。初创公司为病毒载体和基于RNA的疫苗的开发做出了重大贡献,表明它们在新技术应用中的重要作用。主要开发商因国家和地区而异。联盟,包括国际合作,在晚期临床开发中取得了进展。基于这些结果,大流行疫苗开发的未来前景以及对政策和公司战略的影响进行了讨论。
    Various vaccine platforms, including emerging platforms, have been applied in the development of COVID-19 vaccines. Biotechnology startups often lead the development of new medical technologies, whereas major pharmaceutical companies and public institutions have long contributed to vaccine development. In this study, vaccine platforms and developers involved in COVID-19 vaccine development were analyzed, elucidating the trends of vaccine platforms used, the country distribution of the developers, and differences in the profiles of developers by vaccine platform technologies and country. The analysis revealed that conventional, established, and emerging vaccine platforms have been widely used and that older platforms are more advanced in clinical development. It also demonstrated the emergence of China, in addition to the U.S., while many pharmerging countries have been engaged in development. Startups have significantly contributed to the development of viral vector and RNA-based vaccines, suggesting their important role in the application of novel technologies. The major developers differ by country and region. Alliances, including international collaborations, have progressed in late clinical development. Based on these results, future perspectives of pandemic vaccine development and implications for policy and corporate strategies are discussed.
    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
    新的预防性疫苗平台对于对抗呼吸道感染至关重要。T和B记忆细胞介导的保护的功效,通过腺病毒载体产生,进行了测试,以评估新的基于腺病毒的传染病平台的有效性。腺病毒AdV1(佐剂)的组合,配备共刺激配体(ICOSL和CD40L),rRBD(抗原:重组非糖基化刺突蛋白rRBD)用于促进T和B淋巴细胞的分化。腺病毒AdV2(佐剂),没有配体,结合rRBD,作为一个控制。体外T细胞对AdV1+rRBD组合的反应显示CD8+平台特异性T细胞增加(37.2±0.7%vs.23.1±2.1%),和T细胞通过CD8TEMRA对SARS-CoV-2起作用(50.0±1.3%vs.36.0±3.2%)。用AdV1rRBD处理后诱导记忆B细胞(84.1±0.8%vs.82.3±0.4%)或rRBD(94.6±0.3%vs.82.3±0.4%)。观察到用rRBD+Ab诱导后从IgM和IgD向同种型IgG的类别转换。RNA-seq谱分析鉴定了与抗病原体的T辅助细胞分化相关的基因表达模式。分析确定了控制保护性免疫诱导的信号通路,包括MAPK级联,脂肪细胞因子,cAMP,TNF,和Toll样受体信号通路。AdV1+rRBD制剂诱导IL-6、IL-8和TNF。VEROE6细胞系的RNA-seq显示了平台与平台刺激的凋亡基因表达的差异。嘲笑.总之,AdV1+rRBD有效地产生T和B记忆细胞介导的保护,在产生CD8+平台特异性T细胞和同种型转换的IgG记忆B细胞方面呈现有希望的结果。该平台通过控制Th1、Th2和Th17细胞分化基因表达模式来诱导保护性免疫。需要进一步研究以确认其有效性。
    New prophylactic vaccine platforms are imperative to combat respiratory infections. The efficacy of T and B memory cell-mediated protection, generated through the adenoviral vector, was tested to assess the effectiveness of the new adenoviral-based platforms for infectious diseases. A combination of adenovirus AdV1 (adjuvant), armed with costimulatory ligands (ICOSL and CD40L), and rRBD (antigen: recombinant nonglycosylated spike protein rRBD) was used to promote the differentiation of T and B lymphocytes. Adenovirus AdV2 (adjuvant), without ligands, in combination with rRBD, served as a control. In vitro T-cell responses to the AdV1+rRBD combination revealed that CD8+ platform-specific T-cells increased (37.2 ± 0.7% vs. 23.1 ± 2.1%), and T-cells acted against SARS-CoV-2 via CD8+TEMRA (50.0 ± 1.3% vs. 36.0 ± 3.2%). Memory B cells were induced after treatment with either AdV1+rRBD (84.1 ± 0.8% vs. 82.3 ± 0.4%) or rRBD (94.6 ± 0.3% vs. 82.3 ± 0.4%). Class-switching from IgM and IgD to isotype IgG following induction with rRBD+Ab was observed. RNA-seq profiling identified gene expression patterns related to T helper cell differentiation that protect against pathogens. The analysis determined signaling pathways controlling the induction of protective immunity, including the MAPK cascade, adipocytokine, cAMP, TNF, and Toll-like receptor signaling pathway. The AdV1+rRBD formulation induced IL-6, IL-8, and TNF. RNA-seq of the VERO E6 cell line showed differences in the apoptosis gene expression stimulated with the platforms vs. mock. In conclusion, AdV1+rRBD effectively generates T and B memory cell-mediated protection, presenting promising results in producing CD8+ platform-specific T cells and isotype-switched IgG memory B cells. The platform induces protective immunity by controlling the Th1, Th2, and Th17 cell differentiation gene expression patterns. Further studies are required to confirm its effectiveness.
    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
    胶质母细胞瘤(GBM)是成人最致命的中枢神经系统(CNS)肿瘤之一。作为护理标准(SOC)的补充,各种免疫疗法可改善其他癌症的治疗效果。其中,肿瘤疫苗可以作为补充单一疗法,或与其他免疫疗法一起提高临床疗效,如免疫检查点阻断(ICB)和嵌合抗原受体T细胞(CAR-T)治疗。先前对GBM治疗性疫苗的研究表明,由于低突变负担,很少有新抗原可以在GBM中靶向。和单肽治疗性疫苗在肿瘤控制中作为单一疗法的功效有限。结合不同的抗原,包括新抗原,肿瘤相关抗原(TAA),和病原体来源的抗原,优化疫苗设计或疫苗接种策略可能有助于临床疗效的提高。在这次审查中,我们讨论了当前的GBM治疗性疫苗平台,评估和潜在的抗原靶标,当前的挑战,和疗效改善的视角机会。
    Glioblastoma (GBM) is one of the most lethal central nervous systems (CNS) tumours in adults. As supplements to standard of care (SOC), various immunotherapies improve the therapeutic effect in other cancers. Among them, tumour vaccines can serve as complementary monotherapy or boost the clinical efficacy with other immunotherapies, such as immune checkpoint blockade (ICB) and chimeric antigen receptor T cells (CAR-T) therapy. Previous studies in GBM therapeutic vaccines have suggested that few neoantigens could be targeted in GBM due to low mutation burden, and single-peptide therapeutic vaccination had limited efficacy in tumour control as monotherapy. Combining diverse antigens, including neoantigens, tumour-associated antigens (TAAs), and pathogen-derived antigens, and optimizing vaccine design or vaccination strategy may help with clinical efficacy improvement. In this review, we discussed current GBM therapeutic vaccine platforms, evaluated and potential antigenic targets, current challenges, and perspective opportunities for efficacy improvement.
    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
    Pichinde病毒(PICV)可以感染多种动物,已被开发为安全有效的疫苗载体。我们先前的研究表明,用表达猪H3N2甲型流感病毒(IAV-S)的血凝素(HA)基因的重组PICV载体疫苗接种的猪产生了病毒中和抗体,并受到同源H3N2株的感染保护。本研究的目的是评估表达来自三种共同循环的IAV-S亚型:H1N1、H1N2和H3N2的HA抗原的三价PICV载体疫苗的免疫原性和保护效力。用三价PICV疫苗免疫的猪开发了针对所有三种匹配的IAV-S的病毒中和(VN)和血凝抑制(HI)抗体。在用H1N1毒株攻击感染后,接种三价疫苗的六只猪中的五只在鼻拭子和支气管肺泡灌洗液中没有IAV-SRNA基因组的证据,而所有未接种疫苗的对照猪在这两种类型的样品中显示高拷贝数的IAV-S基因组RNA。总的来说,我们的结果表明,三价PICV载体疫苗可引发针对3种靶向IAV-S毒株的抗体应答,并在猪中提供针对同源病毒攻击的保护.因此,PICV显示出作为在猪中递送多种疫苗抗原的病毒载体被探索的潜力。
    Pichinde virus (PICV) can infect several animal species and has been developed as a safe and effective vaccine vector. Our previous study showed that pigs vaccinated with a recombinant PICV-vectored vaccine expressing the hemagglutinin (HA) gene of an H3N2 influenza A virus of swine (IAV-S) developed virus-neutralizing antibodies and were protected against infection by the homologous H3N2 strain. The objective of the current study was to evaluate the immunogenicity and protective efficacy of a trivalent PICV-vectored vaccine expressing HA antigens from the three co-circulating IAV-S subtypes: H1N1, H1N2, and H3N2. Pigs immunized with the trivalent PICV vaccine developed virus-neutralizing (VN) and hemagglutination inhibition (HI) antibodies against all three matching IAV-S. Following challenge infection with the H1N1 strain, five of the six pigs vaccinated with the trivalent vaccine had no evidence of IAV-S RNA genomes in nasal swabs and bronchoalveolar lavage fluid, while all non-vaccinated control pigs showed high number of copies of IAV-S genomic RNA in these two types of samples. Overall, our results demonstrate that the trivalent PICV-vectored vaccine elicits antibody responses against the three targeted IAV-S strains and provides protection against homologous virus challenges in pigs. Therefore, PICV exhibits the potential to be explored as a viral vector for delivering multiple vaccine antigens in swine.
    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
    TNX-1800是一种合成来源的活重组嵌合马痘病毒(rcHPXV)候选疫苗,表达武汉SARS-CoV-2刺突(S)蛋白。这项研究的主要目的是评估TNX-1800在受到USA-WA1/2020SARS-CoV-2攻击的两种非人灵长类动物中的免疫原性和功效。TNX-1800疫苗接种耐受性良好,无严重不良事件或临床参数显著变化。单剂量的TNX-1800在非洲绿猴和食蟹猴中产生体液反应,通过抗SARS-CoV-2SIgG的总结合和针对USA-WA1/2020菌株的中和抗体滴度进行测量。此外,单剂量的TNX-1800在食蟹猴中诱导干扰素-γ(IFN-γ)介导的T细胞应答。在受到SARS-CoV-2的攻击后,非洲绿猴和食蟹猴在上呼吸道和下呼吸道表现出病毒的快速清除。未来的研究将评估TNX-1800对新出现的变体的功效,并证明其在人类中的安全性。
    TNX-1800 is a synthetically derived live recombinant chimeric horsepox virus (rcHPXV) vaccine candidate expressing Wuhan SARS-CoV-2 spike (S) protein. The primary objective of this study was to evaluate the immunogenicity and efficacy of TNX-1800 in two nonhuman primate species challenged with USA-WA1/2020 SARS-CoV-2. TNX-1800 vaccination was well tolerated with no serious adverse events or significant changes in clinical parameters. A single dose of TNX-1800 generated humoral responses in African Green Monkeys and Cynomolgus Macaques, as measured by the total binding of anti-SARS-CoV-2 S IgG and neutralizing antibody titers against the USA-WA1/2020 strain. In addition, a single dose of TNX-1800 induced an interferon-gamma (IFN-γ)-mediated T-cell response in Cynomolgus Macaques. Following challenge with SARS-CoV-2, African Green and Cynomolgus Macaques exhibited rapid clearance of virus in the upper and lower respiratory tract. Future studies will assess the efficacy of TNX-1800 against newly emerging variants and demonstrate its safety in humans.
    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
    猪甲型流感病毒(IAV-S)是高度流行的并且对猪生产者造成重大经济损失。由于病毒的高度可变和快速演变的性质,开发一个安全和通用的疫苗平台至关重要,该平台允许频繁更新疫苗免疫原,以应对新的病毒株的出现。这项研究的主要目的是评估使用脂质纳米颗粒(LNP)作为纳米载体在猪中递送编码病毒血凝素(HA)基因的DNA质粒的可行性。肌肉内施用单剂量的LNP-DNA疫苗在猪中导致强烈的全身和粘膜反应。重要的是,接种疫苗的猪完全免受同源IAV-S株的攻击感染,12只接种疫苗的猪中只有1只在其鼻腔中脱落少量的病毒基因组RNA。在尸检时,在接种疫苗的猪的肺中没有观察到总体或微观损伤。因此,LNP-DNA疫苗在保护猪对抗同源IAV-S毒株方面非常有效,并且可以作为快速开发IAV-S疫苗的有希望的平台。
    The Influenza A virus of swine (IAV-S) is highly prevalent and causes significant economic losses to swine producers. Due to the highly variable and rapidly evolving nature of the virus, it is critical to develop a safe and versatile vaccine platform that allows for frequent updates of the vaccine immunogens to cope with the emergence of new viral strains. The main objective of this study was to assess the feasibility of using lipid nanoparticles (LNPs) as nanocarriers for delivering DNA plasmid encoding the viral hemagglutinin (HA) gene in pigs. The intramuscular administration of a single dose of the LNP-DNA vaccines resulted in robust systemic and mucosal responses in pigs. Importantly, the vaccinated pigs were fully protected against challenge infection with the homologous IAV-S strain, with only 1 out of 12 vaccinated pigs shedding a low amount of viral genomic RNA in its nasal cavity. No gross or microscopic lesions were observed in the lungs of the vaccinated pigs at necropsy. Thus, the LNP-DNA vaccines are highly effective in protecting pigs against the homologous IAV-S strain and can serve as a promising platform for the rapid development of IAV-S vaccines.
    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
    TNX-1800是一种临床前阶段合成衍生的活减毒嵌合马痘病毒疫苗,经工程改造以表达SARS-CoV-2刺突(S)基因。这项研究的目的是评估安全性,耐受性,在叙利亚金仓鼠和新西兰白兔中施用TNX-1800的免疫原性。通过经皮接种以三个剂量接种动物。数据显示,三种TNX-1800疫苗剂量水平的单次经皮施用在仓鼠和兔中都具有良好的耐受性。在所有剂量水平下,与仓鼠相比,兔子在疫苗部位反应方面更令人沮丧。最后,在疫苗接种部位未检测到TNX-1800基因组.接种疫苗后,所有动物均有抗SARS-CoV-2刺突蛋白IgG特异性抗体应答.这些数据表明TNX-1800感染是有限的,无症状,在这项研究结束时,单剂量就能产生免疫反应。
    TNX-1800 is a preclinical stage synthetic-derived live attenuated chimeric horsepox virus vaccine engineered to express the SARS-CoV-2 spike (S) gene. The objectives of this study were to assess the safety, tolerability, and immunogenicity of TNX-1800 administration in Syrian golden hamsters and New Zealand white rabbits. Animals were vaccinated at three doses via percutaneous inoculation. The data showed that the single percutaneous administration of three TNX-1800 vaccine dose levels was well tolerated in both hamsters and rabbits. At all dose levels, rabbits were more decerning regarding vaccine site reaction than hamsters. Lastly, no TNX-1800 genomes could be detected at the site of vaccination. Post-vaccination, all animals had anti-SARS-CoV-2 spike protein IgG specific antibody responses. These data demonstrate that TNX-1800 infection was limited, asymptomatic, and cleared by the end of this study, and a single dose was able to generate immune responses.
    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
    本审查报告了加速/大流行和备灾的关键促成因素的概述,涵盖CMC战略以及疫苗开发中的技术创新。与会者就推动持续加速疫苗开发的实施障碍和机会进行了讨论,并考虑了从COVID大流行中吸取的经验教训和解决未满足医疗需求的直接经验。这些思考集中在(i)从目标抗原识别到发射和生命周期管理的跨学科技术期望框架的重要性;(ii)在类似或产品/疫苗类型中使用现有平台知识;(iii)实施创新和数字工具以实现快速开发和创新控制策略。
    This review reports on an overview of key enablers of acceleration/pandemic and preparedness, covering CMC strategies as well as technical innovations in vaccine development. Considerations are shared on implementation hurdles and opportunities to drive sustained acceleration for vaccine development and considers learnings from the COVID pandemic and direct experience in addressing unmet medical needs. These reflections focus on (i) the importance of a cross-disciplinary framework of technical expectations ranging from target antigen identification to launch and life-cycle management; (ii) the use of prior platform knowledge across similar or products/vaccine types; (iii) the implementation of innovation and digital tools for fast development and innovative control strategies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号