mRNA vaccines

mRNA 疫苗
  • 文章类型: Journal Article
    胰腺腺癌(PAAD)是最致命的恶性肿瘤之一。和信使核糖核酸疫苗,它们构成了最新一代的疫苗技术,有望为胰腺癌的治疗带来新的思路。合并并分析癌症基因组图谱-PAAD和基因型-组织表达数据。使用加权基因共表达网络分析来鉴定与免疫和氧化应激相关的基因中与肿瘤突变负荷相关的基因模块。通过单因素Cox回归分析筛选差异表达的免疫相关氧化应激基因,这些基因通过非负矩阵分解进行分析。免疫浸润分析后,采用最小绝对收缩和选择算子回归结合Cox回归构建模型,并根据模型构建后的受试者工作特性曲线和决策曲线分析曲线预测模型的有用性。最后,使用基因集富集分析结合京都基因和基因组百科全书和基因本体论生物学过程分析进行代谢途径富集分析。该模型由ERAP2,间充质上皮转化因子(MET),与现有模型相比,CXCL9和血管紧张素原(AGT)基因可用于帮助更准确地预测胰腺癌患者的预后。ERAP2参与免疫激活,在癌症免疫逃避中起重要作用。MET与肝细胞生长因子结合,导致c-MET的二聚化和磷酸化。这激活了各种信号通路,包括MAPK和PI3K,为了调节增殖,入侵,和癌细胞的迁移。CXCL9过表达与不良患者预后相关,并减少PAAD肿瘤微环境中CD8细胞毒性T淋巴细胞的数量。AGT被肾素酶裂解以产生血管紧张素1,并且AGT转换酶裂解血管紧张素1以产生血管紧张素2。胰腺癌诊断后暴露于AGT转换酶抑制剂与提高生存率相关本研究中确定的4个基因-ERAP2,MET,CXCL9和AGT有望成为信使核糖核酸疫苗开发的靶标,需要进一步深入研究。
    Adenocarcinoma of the pancreas (PAAD) is one of the deadliest malignant tumors, and messenger ribonucleic acid vaccines, which constitute the latest generation of vaccine technology, are expected to lead to new ideas for the treatment of pancreatic cancer. The Cancer Genome Atlas-PAAD and Genotype-Tissue Expression data were merged and analyzed. Weighted gene coexpression network analysis was used to identify gene modules associated with tumor mutational burden among the genes related to both immunity and oxidative stress. Differentially expressed immune-related oxidative stress genes were screened via univariate Cox regression analysis, and these genes were analyzed via nonnegative matrix factorization. After immune infiltration analysis, least absolute shrinkage and selection operator regression combined with Cox regression was used to construct the model, and the usefulness of the model was predicted based on the receiver operating characteristic curve and decision curve analysis curves after model construction. Finally, metabolic pathway enrichment was analyzed using gene set enrichment analysis combined with Kyoto Encyclopedia of Genes and Genomes and gene ontology biological process analyses. This model consisting of the ERAP2, mesenchymal-epithelial transition factor (MET), CXCL9, and angiotensinogen (AGT) genes can be used to help predict the prognosis of pancreatic cancer patients more accurately than existing models. ERAP2 is involved in immune activation and is important in cancer immune evasion. MET binds to hepatocyte growth factor, leading to the dimerization and phosphorylation of c-MET. This activates various signaling pathways, including MAPK and PI3K, to regulate the proliferation, invasion, and migration of cancer cells. CXCL9 overexpression is associated with a poor patient prognosis and reduces the number of CD8 + cytotoxic T lymphocytes in the PAAD tumor microenvironment. AGT is cleaved by the renin enzyme to produce angiotensin 1, and AGT-converting enzyme cleaves angiotensin 1 to produce angiotensin 2. Exposure to AGT-converting enzyme inhibitors after pancreatic cancer diagnosis is associated with improved survival. The 4 genes identified in the present study - ERAP2, MET, CXCL9, and AGT - are expected to serve as targets for messenger ribonucleic acid vaccine development and need to be further investigated in depth.
    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
    前列腺癌(PCa)的特征是免疫反应有限的“冷肿瘤”,使肿瘤对免疫检查点抑制剂(ICI)具有抗性。治疗性信使RNA(mRNA)疫苗已成为一种有希望的策略,通过增强免疫反应性和显着增强抗肿瘤功效来克服这一挑战。在我们的研究中,我们合成了利乐,混合多种肿瘤相关抗原的mRNA疫苗,免疫,一种免疫增强佐剂,旨在诱导有效的抗肿瘤免疫。ImmunER表现出促进树突状细胞(DC)成熟的能力,加强DC迁移,并在细胞和动物水平上改善抗原呈递。此外,利乐,结合免疫,诱导骨髓来源的树突状细胞(BMDCs)转化为cDC1-CCL22并上调JAK-STAT1途径,促进IL-12,TNF-α的释放,和其他细胞因子。这种级联导致T细胞的增殖和活化增强,从而有效杀死肿瘤细胞。体内实验进一步揭示了Tetra+ImmunER增加了RM-1-PSMA肿瘤组织中的CD8+T细胞浸润和活化。总之,我们的发现强调了Tetra和ImmunERmRNA-LNP整合治疗在PCa中具有强大的抗肿瘤免疫的潜力.
    Prostate cancer (PCa) is characterized as a \"cold tumor\" with limited immune responses, rendering the tumor resistant to immune checkpoint inhibitors (ICI). Therapeutic messenger RNA (mRNA) vaccines have emerged as a promising strategy to overcome this challenge by enhancing immune reactivity and significantly boosting anti-tumor efficacy. In our study, we synthesized Tetra, an mRNA vaccine mixed with multiple tumor-associated antigens, and ImmunER, an immune-enhancing adjuvant, aiming to induce potent anti-tumor immunity. ImmunER exhibited the capacity to promote dendritic cells (DCs) maturation, enhance DCs migration, and improve antigen presentation at both cellular and animal levels. Moreover, Tetra, in combination with ImmunER, induced a transformation of bone marrow-derived dendritic cells (BMDCs) to cDC1-CCL22 and up-regulated the JAK-STAT1 pathway, promoting the release of IL-12, TNF-α, and other cytokines. This cascade led to enhanced proliferation and activation of T cells, resulting in effective killing of tumor cells. In vivo experiments further revealed that Tetra + ImmunER increased CD8+T cell infiltration and activation in RM-1-PSMA tumor tissues. In summary, our findings underscore the promising potential of the integrated Tetra and ImmunER mRNA-LNP therapy for robust anti-tumor immunity in PCa.
    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
    一个健康的方法,它整合了人类的健康,动物,植物,和不同层次的生态系统,对于解决相互关联的健康威胁至关重要。mRNA疫苗的出现补充了这一点,彻底改变了疾病预防。它们提供广谱的有效性,可以快速定制以靶向特定的病原体。它们的用途超越了人类医学,在兽医实践中显示出控制疾病和降低人畜共患传播风险的潜力。这篇综述将mRNA疫苗和一个健康放在蜱传疾病的背景下。这些疫苗赋予跨物种免疫力的潜力是巨大的,可能破坏人畜共患疾病的传播周期并保护人类和动物的健康,在减少蜱种群的同时,病原体的感染和循环。蜱和蜱传病原体mRNA疫苗的开发和应用代表了全球卫生的综合战略,在我们相互联系的世界中,为所有物种培育更健康的生态系统。
    The One Health approach, which integrates the health of humans, animals, plants, and ecosystems at various levels, is crucial for addressing interconnected health threats. This is complemented by the advent of mRNA vaccines, which have revolutionized disease prevention. They offer broad-spectrum effectiveness and can be rapidly customized to target specific pathogens. Their utility extends beyond human medicine, showing potential in veterinary practices to control diseases and reduce the risk of zoonotic transmissions. This review place mRNA vaccines and One Health in the context of tick-borne diseases. The potential of these vaccines to confer cross-species immunity is significant, potentially disrupting zoonotic disease transmission cycles and protecting the health of both humans and animals, while reducing tick populations, infestations and circulation of pathogens. The development and application of mRNA vaccines for tick and tick-borne pathogens represent a comprehensive strategy in global health, fostering a healthier ecosystem for all species in our interconnected world.
    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
    疾病修饰疗法(DMT)会影响多发性硬化症(pwMS)患者对严重急性呼吸综合征冠状病毒2型(SARS-CoV-2)疫苗的细胞免疫反应。在这项研究中,我们旨在通过测量各种DMT上pwMS中的广泛细胞因子谱来阐明所涉及的抗原特异性T细胞的特征。我们检查了全血培养物中SARS-CoV-2特异性T细胞反应,其特征是释放白介素(IL)-2,IL-4,IL-5,IL-10,IL-13,IL-17A,干扰素-γ(IFN-γ),和肿瘤坏死因子-α(TNF-α),以及在两个或三个剂量的mRNA或病毒载体疫苗(VVV)后,在pwMS中靶向SARS-CoV-2刺突蛋白的抗体(AB)。对于mRNA疫苗接种无应答者,给予NVX-CoV2373蛋白质疫苗,和免疫反应进行了评估。我们的发现表明,pwMS中对SARS-CoV-2疫苗的免疫反应偏向Th1表型,以IL-2和IFN-γ为特征。此外,观察到以IL-5为特征的Th2应答,并且在较小程度上观察到IL-4、IL-10和IL-13。因此,IL-2和IL-5水平的测定可以补充传统的IFN-γ测定,从而更全面地表征SARS-CoV-2疫苗的细胞应答.我们的结果为接受不同DMT的pwMS提供了全面的细胞因子谱,并为在该患者人群中设计疫苗接种策略提供了有价值的见解。
    Disease-modifying therapies (DMTs) impact the cellular immune response to severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) vaccines in patients with multiple sclerosis (pwMS). In this study, we aim to elucidate the characteristics of the involved antigen-specific T cells via the measurement of broad cytokine profiles in pwMS on various DMTs. We examined SARS-CoV-2-specific T cell responses in whole blood cultures characterized by the release of interleukin (IL)-2, IL-4, IL-5, IL-10, IL-13, IL-17A, interferon-gamma (IFN-γ), and tumor necrosis factor-alpha (TNF-α), as well as antibodies (AB) targeting the SARS-CoV-2 spike protein in pwMS following either two or three doses of mRNA or viral vector vaccines (VVV). For mRNA vaccination non-responders, the NVX-CoV2373 protein-based vaccine was administered, and immune responses were evaluated. Our findings indicate that immune responses to SARS-CoV-2 vaccines in pwMS are skewed towards a Th1 phenotype, characterized by IL-2 and IFN-γ. Additionally, a Th2 response characterized by IL-5, and to a lesser extent IL-4, IL-10, and IL-13, is observed. Therefore, the measurement of IL-2 and IL-5 levels could complement traditional IFN-γ assays to more comprehensively characterize the cellular responses to SARS-CoV-2 vaccines. Our results provide a comprehensive cytokine profile for pwMS receiving different DMTs and offer valuable insights for designing vaccination strategies in this patient population.
    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mRNA疫苗主要通过肌肉注射给药,诱导良好的全身免疫,但粘膜免疫有限。通过mRNA疫苗接种实现粘膜免疫可以减少病原体在进入位点的复制并减少人与人之间的传播。然而,向粘膜递送mRNA疫苗面临着mRNA降解等挑战,进入细胞不良,和反应原性。将mRNA封装在细胞外囊泡中可以保护mRNA并降低反应原性,使粘膜mRNA疫苗成为可能。已经研究了来自可食用水果的植物来源的细胞外囊泡作为mRNA载体。动物研究表明,通过口服给药时,橙色衍生的细胞外囊泡中的mRNA可以引起全身和粘膜免疫反应,鼻部,或肌内途径。一旦冻干,这些产品表现出显著的稳定性。优化mRNA,提高翻译效率,免疫原性,反应原性,可以通过调整5'上限区域来获得稳定性,poly-A尾巴,密码子选择,以及核苷类似物的使用。最近的研究还提出了含有RNA聚合酶以及环状mRNA构建体的自扩增RNA疫苗。来自肠胃外灌注动物的数据证明了用非佐剂蛋白进行鼻免疫的功效,在人类中的研究表明,肠胃外疫苗与粘膜自然暴露于相同抗原的组合提供了保护并减少了传播。因此,粘膜mRNA疫苗接种至少在用肠胃外疫苗预处理的生物体中是有益的。这种做法可以广泛应用于传染病的治疗。
    Current mRNA vaccines are mainly administered via intramuscular injection, which induces good systemic immunity but limited mucosal immunity. Achieving mucosal immunity through mRNA vaccination could diminish pathogen replication at the entry site and reduce interhuman transmission. However, delivering mRNA vaccines to mucosae faces challenges like mRNA degradation, poor entry into cells, and reactogenicity. Encapsulating mRNA in extracellular vesicles may protect the mRNA and reduce reactogenicity, making mucosal mRNA vaccines possible. Plant-derived extracellular vesicles from edible fruits have been investigated as mRNA carriers. Studies in animals show that mRNA vehiculated in orange-derived extracellular vesicles can elicit both systemic and mucosal immune responses when administered by the oral, nasal, or intramuscular routes. Once lyophilized, these products show remarkable stability. The optimization of mRNA to improve translation efficiency, immunogenicity, reactogenicity, and stability can be obtained through adjustments of the 5\'cap region, poly-A tail, codons selection, and the use of nucleoside analogues. Recent studies have also proposed self-amplifying RNA vaccines containing an RNA polymerase as well as circular mRNA constructs. Data from parenterally primed animals demonstrate the efficacy of nasal immunization with non-adjuvanted protein, and studies in humans indicate that the combination of a parenteral vaccine with the natural exposure of mucosae to the same antigen provides protection and reduces transmission. Hence, mucosal mRNA vaccination would be beneficial at least in organisms pre-treated with parenteral vaccines. This practice could have wide applications for the treatment of infectious 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
    增强我们对mRNA疫苗的理解可能有助于将来设计新型疫苗,旨在增强免疫保护,同时最大程度地减少反应原反应。在进行此设计之前,确定适应性免疫是否与疫苗的反应原性相关是很重要的.我们研究了一个用mRNA疫苗接种的大型队列来回答这个问题。这是一项针对真实世界数据的观察性研究。反应性数据来自VigilVacCOVID研究。从健康记录中检索免疫原性(体液和细胞)数据。定义了一个主要种群(n=215)和两个亚群(亚群1,n=3563;亚群2,n=597)。对亚群1和2进行敏感性分析,以探索结果的一致性。我们分析了不良反应的强度和类型与引发的抗体滴度的发展和数量之间的关系。作为亚群1的探索性分析,我们评估了反应原性与细胞免疫原性之间的关联。发烧的发病率较高,萎靡不振,包括严重病例在内的肌痛与抗体滴度阳性的发展和数量显着相关。细胞免疫没有观察到显著的发现。我们观察到免疫原性和反应原性之间的正相关。这些发现可能与我们对mRNA疫苗功能的理解的未来发展有关。
    Enhancing our comprehension of mRNA vaccines may facilitate the future design of novel vaccines aimed at augmenting immune protection while minimising reactogenic responses. Before this design is carried out, it is important to determine whether adaptive immunity correlates with the reactogenicity profile of vaccines. We studied a large cohort that was vaccinated with mRNA vaccines to answer this question. This was an observational study with real-world data. Reactogenicity data were obtained from the VigilVacCOVID study. Immunogenicity (humoral and cellular) data were retrieved from health records. One main population (n = 215) and two subpopulations were defined (subpopulation 1, n = 3563; subpopulation 2, n = 597). Sensitivity analyses were performed with subpopulations 1 and 2 to explore the consistency of results. We analysed the association of the intensity and types of adverse reactions with the development and quantity of elicited antibody titres. As an exploratory analysis in subpopulation 1, we assessed the association between reactogenicity and cellular immunogenicity. A higher incidence of fever, malaise, and myalgia including severe cases was significantly associated with the development and quantity of positive antibody titres. No significant findings were observed with cellular immunity. We observed a positive association between immunogenicity and reactogenicity. These findings can be relevant for the future development of our understanding of how mRNA vaccines function.
    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
    mRNA疫苗是一种诱导免疫应答的疫苗。编码抗原的mRNA通过疫苗载体传递到免疫细胞中,它们是由于编码抗原的mRNA翻译而产生的,一种蛋白质。例如,COVID-19mRNA疫苗产生COVID-19病毒的刺突蛋白,而对于流感病毒,mRNA疫苗靶向血凝素蛋白来治疗流感,它需要根据大流行或季节性病毒进行修改,因为它能够适应免疫反应,这使得疫苗的开发艰巨。蛋白质分子促进适应性免疫应答,其消除和终止相应的病毒或病原体。将mRNA疫苗递送到体内有许多挑战;因此,mRNA的封装(通常在脂质纳米颗粒中)对于保护mRNA免受人体周围环境的影响是必要的。在这篇评论文章中,我们主要专注于生产,配方,和一般的mRNA疫苗的稳定性,更多地阐述和关注SARS-CoV-2或COVID-19和流感病毒,这已经成为一个主要的问题,因为这些病毒已经变成了危及生命的疾病。
    A mRNA vaccine is a type of vaccine that induces an immune response. Antigen-encoding mRNA is delivered via vaccine carriers into the immune cells, which are produced because of antigen-encoding mRNA translation, a protein. For example, COVID-19 mRNA vaccines produce the spike protein of the COVID-19 virus, whereas for influenza virus, mRNA vaccines target the haemagglutinin protein to treat the flu, and it requires modifications depending on the pandemic or seasonal viruses as it is capable of adapting the immune response, which makes the development of vaccines arduous. The protein molecule promotes an adaptive immune response that eliminates and terminates the corresponding virus or pathogen. There are many challenges to delivering an mRNA vaccine into the body; hence, the encapsulation of the mRNA (usually within lipid nanoparticles) is necessary to protect the mRNA from the body\'s surrounding environment. In this review article, we focus mainly on the production, formulation, and stabilization of mRNA vaccines in general, elaborating more on and focusing more on SARS-CoV-2, or COVID-19, and influenza viruses, which have become a major concern as these viruses have turned into life-threatening 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
    我们的目的是评估疗效,安全,和SARS-CoV-2mRNA疫苗(OmicronBA.5)LVRNA012的免疫原性在中国已免疫但无SARS-CoV-2感染的成年人中作为加强剂。
    这是一个单中心,随机化,双盲,安慰剂对照的3期临床试验招募健康的成年参与者(≥18岁),这些参与者至少在6个月前完成了两剂或三剂灭活的COVID-19疫苗,在蚌埠,安徽省,中国。符合条件的参与者被随机分配(1:1)接受LVRNA012疫苗(100ug)或安慰剂的加强肌内疫苗接种。主要终点是疫苗接种后14天,加强剂量的LVRNA012疫苗或安慰剂对任何严重程度的症状性COVID-19的保护功效。干预后14天至180天,实验室确认的COVID-19感染被确定,在干预后7至90天之间,每月8次积极监测有症状的疾病,在90至180天之间每月至少一次。
    招募了2615名参与者,并以1:1的比例随机分配到疫苗组(1308)或安慰剂组(1307)。在加强免疫后14天,共有141人(LVRNA012组46人,安慰剂组95人)出现有症状的COVID-19感染,显示疫苗效力为51.9%(95%CI,31.3%至66.4%)。在XBB在社区流行期间,大多数感染是在干预后90天检测到的。在LVRNA012疫苗接种后,64%的参与者报告了不良反应,但大多数是轻度或中度的。与安慰剂组(GMT12.5[8.4,18.7])相比,在第14天,用LVRNA012mRNA疫苗的加强疫苗接种可显著增强针对Omicron变体XBB.1.5(GMT132.3[99.8,175.4])的中和抗体滴度(先前免疫的个体)。
    LVRNA012mRNA疫苗具有免疫原性,在omicron占主导地位的时期,在预防COVID-19方面表现出强劲的疗效。
    ClinicalTrials.gov,标识符NCT05745545。
    UNASSIGNED: We aimed to evaluate the efficacy, safety, and immunogenicity of a SARS-CoV-2 mRNA vaccine (Omicron BA.5) LVRNA012 given as the booster in immunized but SARS-CoV-2 infection-free adults in China.
    UNASSIGNED: This is a single-center, randomized, double-blind, placebo-controlled phase 3 clinical trial enrolling healthy adult participants (≥18 years) who had completed two or three doses of inactivated COVID-19 vaccines at least 6 months before, in Bengbu, Anhui province, China. Eligible participants were randomly assigned (1:1) to receive a booster intramuscular vaccination with an LVRNA012 vaccine (100ug) or placebo. The primary endpoint was the protective efficacy of a booster dose of the LVRNA012 vaccine or placebo against symptomatic COVID-19 of any severity 14 days after vaccination. Laboratory-confirmed COVID-19 infections were identified from 14 days to 180 days after intervention, with active surveillance for symptomatic illness 8 times per month between 7 to 90 days and at least once per month between 90 to 180 days after intervention.
    UNASSIGNED: 2615 participants were recruited and randomly assigned in a 1:1 ratio to either the vaccine group (1308) or the placebo group (1307). A total of 141 individuals (46 in the LVRNA012 group and 95 in the placebo group) developed symptomatic COVID-19 infection 14 days after the booster immunization, showing a vaccine efficacy of 51.9% (95% CI, 31.3% to 66.4%). Most infections were detected 90 days after intervention during a period when XBB was prevalent in the community. Adverse reactions were reported by 64% of participants after the LVRNA012 vaccination, but most of them were mild or moderate. The booster vaccination with the LVRNA012 mRNA vaccine could significantly enhance neutralizing antibody titers against the Omicron variant XBB.1.5 (GMT 132.3 [99.8, 175.4]) than did those in the placebo group (GMT 12.5 [8.4, 18.7]) at day 14 for the previously immunized individuals.
    UNASSIGNED: The LVRNA012 mRNA vaccine is immunogenic, and shows robust efficacy in preventing COVID-19 during the omicron-predominate period.
    UNASSIGNED: ClinicalTrials.gov, identifier NCT05745545.
    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
    这项纵向前瞻性对照多中心研究旨在监测COVID-19疫苗接种后突破性感染(BTI)引起的三种暴露所产生的免疫力,考虑到预先存在的对普通冠状病毒(CoV)的细胞介导免疫力,这可能会影响细胞对SARS-CoV-2的反应性。在完全接种疫苗的(F)个体中确定了抗SARS-CoV-2-尖峰IgG抗体(抗S-IgG)和针对尖峰蛋白(S)-和核衣壳蛋白(N)蛋白的细胞反应性PCR确认感染后1至24周,与部分接种疫苗(PBTI)和未接种疫苗(U)相比,经历了BTI(FBTI)或加强疫苗接种(FBooster)。与U相比,在F+BTI中发现了高亲和力抗S-IgG,后者表现出增加的持久促炎细胞因子对S-刺激。CoV在U中与较高的细胞反应性相关,而在F中没有发现相关性。该研究表明,在FBTI中,通过三种暴露可诱导显着的S特异性细胞反应,从而建立基本免疫。只有U似乎受益于预先存在的CoV免疫,但与在BTI后从增强的体液和细胞免疫免疫中免疫受益的F+BTI相比,显示了炎性免疫应答。这项研究表明,具有来自COVID-19疫苗接种和BTI的混合免疫的个体获得了稳定的体液和细胞免疫应答,并维持了至少6个月。我们的发现证实了卫生当局通过三种S蛋白暴露来建立基本免疫力的建议。
    This longitudinal prospective controlled multicenter study aimed to monitor immunity generated by three exposures caused by breakthrough infections (BTI) after COVID-19-vaccination considering pre-existing cell-mediated immunity to common-corona-viruses (CoV) which may impact cellular reactivity against SARS-CoV-2. Anti-SARS-CoV-2-spike-IgG antibodies (anti-S-IgG) and cellular reactivity against Spike-(S)- and nucleocapsid-(N)-proteins were determined in fully-vaccinated (F) individuals who either experienced BTI (F+BTI) or had booster vaccination (F+Booster) compared to partially vaccinated (P+BTI) and unvaccinated (U) from 1 to 24 weeks post PCR-confirmed infection. High avidity anti-S-IgG were found in F+BTI compared to U, the latter exhibiting increased long-lasting pro-inflammatory cytokines to S-stimulation. CoV was associated with higher cellular reactivity in U, whereas no association was seen in F. The study illustrates the induction of significant S-specific cellular responses in F+BTI building-up basic immunity by three exposures. Only U seem to benefit from pre-existing CoV immunity but demonstrated inflammatory immune responses compared to F+BTI who immunologically benefit from enhanced humoral and cellular immunity after BTI. This study demonstrates that individuals with hybrid immunity from COVID-19-vaccination and BTI acquire a stable humoral and cellular immune response that is maintained for at least 6 months. Our findings corroborate recommendations by health authorities to build on basic immunity by three S-protein exposures.
    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
    我们研究了编码SARS-CoV-2S蛋白(mRNA-RBD)受体结合域的实验性mRNA疫苗的无针喷射注射递送。通过无针喷射注射器用mRNA-RBD免疫BALB/c小鼠诱导高水平的具有病毒中和活性和病毒特异性T细胞应答的抗体。在接受mRNA-RBD肌内注射的小鼠组中,免疫应答较低。已经证明了这种简单而安全的mRNA递送方法的有效性。因此,喷射注射mRNA疫苗可以很好地替代脂质纳米粒。
    We studied a needle-free jet injection delivery of an experimental mRNA vaccine encoding the receptor-binding domain of the SARS-CoV-2 S protein (mRNA-RBD). Immunization of BALB/c mice with mRNA-RBD by a needle-free jet injector induced high levels of antibodies with virus-neutralizing activity and a virus-specific T-cell response. The immune response was low in the group of mice that received intramuscular injection of mRNA-RBD. The effectiveness of this simple and safe method of mRNA delivering has been demonstrated. Thus, jet injection of mRNA vaccine can be a good alternative to lipid nanoparticles.
    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