mRNA vaccines

mRNA 疫苗
  • 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
    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
    背景:由疫苗接种和自然感染引起的对SARS-CoV-2的混合免疫,在儿科人群中仍然没有得到足够的理解,尽管接种疫苗的儿童中突破性感染率上升。
    方法:我们进行了一项前瞻性纵向研究,特异性,在一组5-11岁的mRNA疫苗接种儿童(n=29)中,突破性SARS-CoV-2感染引起的抗原特异性T细胞反应的细胞因子谱。这项纵向分析涉及疫苗接种后16个月的六个不同时间点,在此期间,我们共分析了159份血液样本。所有随访至少12个月的儿童(n=26)经历了突破性感染。我们使用最少的血液样本进行了细胞因子释放测定,我们通过细胞内细胞因子染色验证了这些反应的细胞起源。
    结果:突破性感染后,接受mRNA疫苗治疗的儿童表现出增强的Spike肽特异性Th1应答.此外,它们的Spike特异性T细胞表现出独特的CD4+IFN-γ+IL10+细胞富集,类似于具有混合免疫的成年人的特征。重要的是,疫苗接种不会阻碍靶向膜的多特异性T细胞反应的发展,核蛋白,和ORF3a/7/8抗原。
    结论:儿童,先前使用基于Spike的mRNA疫苗并经历有症状或无症状的突破性感染,保留了增强和多样化Th1/IL-10抗原特异性T细胞对多种SARS-CoV-2蛋白的反应的能力。这些发现反映了与成人混合细胞免疫相关的特征,已知赋予对严重COVID-19的抗性。
    背景:本研究由新加坡国家医学研究委员会(NMRC)资助(COVID19RF-0019,MOH-000019,MOH-000535,OFLCG19May-0034和MOH-OFYIRG19nov-0002)。
    BACKGROUND: Hybrid immunity to SARS-CoV-2, resulting from both vaccination and natural infection, remains insufficiently understood in paediatric populations, despite increasing rates of breakthrough infections among vaccinated children.
    METHODS: We conducted a prospective longitudinal study to investigate the magnitude, specificity, and cytokine profile of antigen-specific T cell responses elicited by breakthrough SARS-CoV-2 infection in a cohort of mRNA-vaccinated children (n = 29) aged 5-11. This longitudinal analysis involved six distinct time points spanning a 16-month period post-vaccination, during which we analysed a total of 159 blood samples. All children who were followed for at least 12 months (n = 26) experienced a breakthrough infection. We conducted cytokine release assays using minimal blood samples, and we verified the cellular origin of these responses through intracellular cytokine staining.
    RESULTS: After breakthrough infection, children who had received mRNA vaccines showed enhanced Th1 responses specific to Spike peptides. Additionally, their Spike-specific T cells exhibited a distinctive enrichment of CD4+ IFN-γ+IL10+ cells, a characteristic akin to adults with hybrid immunity. Importantly, vaccination did not impede the development of multi-specific T cell responses targeting Membrane, Nucleoprotein, and ORF3a/7/8 antigens.
    CONCLUSIONS: Children, previously primed with a Spike-based mRNA vaccine and experiencing either symptomatic or asymptomatic breakthrough infection, retained the ability to enhance and diversify Th1/IL-10 antigen-specific T cell responses against multiple SARS-CoV-2 proteins. These findings mirror characteristics associated with hybrid cellular immunity in adults, known to confer resistance against severe COVID-19.
    BACKGROUND: This study was funded by the National Medical Research Council (NMRC) Singapore (COVID19RF-0019, MOH-000019, MOH-000535, OFLCG19May-0034 and MOH-OFYIRG19nov-0002).
    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
    不断出现严重急性呼吸综合征冠状病毒2(SARS-CoV-2)的新变种,增强的可传播性,显著的免疫逃逸,免疫力下降需要加强疫苗接种。我们评估了安全性,免疫原性,SARS-CoV-2mRNA疫苗SYS6006与活性对照疫苗的异源性加强剂在随机,开放标签,在中国接受过两剂或三剂SARS-CoV-2灭活疫苗的18岁或以上健康成年人中进行的积极对照3期试验。该试验于2022年12月开始,持续了6个月。参与者被随机分配(总体比例:3:1)接受一剂SYS6006(N=2999)或基于祖先受体结合区的剂量,明矾佐剂重组蛋白SARS-CoV-2疫苗(N=1000),包括520名免疫原性亚组参与者。SYS6006加强显示出良好的安全性特征,其中大多数AE为1级或2级,并且在第14天和第28天诱导出稳健的野生型和OmicronBA.5中和抗体应答,显示出相对于对照疫苗的免疫原性优势并满足主要目标。针对任何严重程度的COVID-19,任何变体的相对疫苗效力为51.6%(95%CI,35.5-63.7),BA.5为66.8%(48.6-78.5),XBB为37.7%(2.4-60.3),从第7天到第6个月。在接种疫苗和感染的混合免疫参与者中,对于由二次感染引起的任何严重程度的COVID-19,相对疫苗有效率为68.4%(31.1-85.5)。所有COVID-19病例均为轻度。SYS6006异源加强显示良好的安全性,对BA.5相关COVID-19具有优异的免疫原性和高效力,并对XBB相关COVID-19具有保护作用,特别是在杂交免疫群体中。试验注册:中国临床试验注册中心:ChiCTR2200066941。
    Continuous emergence of new variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), enhanced transmissibility, significant immune escape, and waning immunity call for booster vaccination. We evaluated the safety, immunogenicity, and efficacy of heterologous booster with a SARS-CoV-2 mRNA vaccine SYS6006 versus an active control vaccine in a randomized, open-label, active-controlled phase 3 trial in healthy adults aged 18 years or more who had received two or three doses of SARS-CoV-2 inactivated vaccine in China. The trial started in December 2022 and lasted for 6 months. The participants were randomized (overall ratio: 3:1) to receive one dose of SYS6006 (N = 2999) or an ancestral receptor binding region-based, alum-adjuvanted recombinant protein SARS-CoV-2 vaccine (N = 1000), including 520 participants in an immunogenicity subgroup. SYS6006 boosting showed good safety profiles with most AEs being grade 1 or 2, and induced robust wild-type and Omicron BA.5 neutralizing antibody response on Days 14 and 28, demonstrating immunogenicity superiority versus the control vaccine and meeting the primary objective. The relative vaccine efficacy against COVID-19 of any severity was 51.6% (95% CI, 35.5-63.7) for any variant, 66.8% (48.6-78.5) for BA.5, and 37.7% (2.4-60.3) for XBB, from Day 7 through Month 6. In the vaccinated and infected hybrid immune participants, the relative vaccine efficacy was 68.4% (31.1-85.5) against COVID-19 of any severity caused by a second infection. All COVID-19 cases were mild. SYS6006 heterologous boosting demonstrated good safety, superior immunogenicity and high efficacy against BA.5-associated COVID-19, and protected against XBB-associated COVID-19, particularly in the hybrid immune population.Trial registration: Chinese Clinical Trial Registry: ChiCTR2200066941.
    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:近年来,像COVID-19这样的传染病对全球社会经济产生了深远的影响。mRNA疫苗由于其快速发展而获得了突出的地位,工业适应性,简单,以及对新变种的反应能力。值得注意的是,2023年诺贝尔生理学或医学奖承认对mRNA疫苗研究的重大贡献。
    方法:我们的研究使用了WebofScienceCoreCollection(WoSCC)数据库进行了全面的文献计量分析,涵盖2003年至2023年关于mRNA疫苗的5512篇论文。我们制作了合作地图,共同引文分析,和关键词聚类来评估该领域的发展历史和成就。
    结果:分析产生了突出国家/机构的知识图谱,有影响力的作家,经常出版和被高度引用的期刊,和开创性的参考文献。正在进行的研究热点包括免疫反应,稳定性增强,在癌症预防和治疗中的应用,并利用mRNA技术防治传染病。
    结论:mRNA疫苗代表了传染病预防的变革发展。这项研究提供了对该领域发展的见解,并确定了关键的研究重点,促进疫苗技术的进步和应对未来的挑战。
    BACKGROUND: In recent years, infectious diseases like COVID-19 have had profound global socio-economic impacts. mRNA vaccines have gained prominence due to their rapid development, industrial adaptability, simplicity, and responsiveness to new variants. Notably, the 2023 Nobel Prize in Physiology or Medicine recognized significant contributions to mRNA vaccine research.
    METHODS: Our study employed a comprehensive bibliometric analysis using the Web of Science Core Collection (WoSCC) database, encompassing 5,512 papers on mRNA vaccines from 2003 to 2023. We generated cooperation maps, co-citation analyses, and keyword clustering to evaluate the field\'s developmental history and achievements.
    RESULTS: The analysis yielded knowledge maps highlighting countries/institutions, influential authors, frequently published and highly cited journals, and seminal references. Ongoing research hotspots encompass immune responses, stability enhancement, applications in cancer prevention and treatment, and combating infectious diseases using mRNA technology.
    CONCLUSIONS: mRNA vaccines represent a transformative development in infectious disease prevention. This study provides insights into the field\'s growth and identifies key research priorities, facilitating advancements in vaccine technology and addressing future challenges.
    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
    肾移植受者(KTRs),像其他实体器官移植受者一样,对mRNA疫苗表现出次优的反应,只有大约一半在两次剂量后实现血清转化。然而,加强剂量的有效性,特别是在产生中和抗体(NAb),仍然知之甚少,因为大多数研究主要集中在非中和抗体上。这里,我们纵向评估了一年中40个KTRs对SARS-CoV-2mRNA疫苗的体液反应,在第二次给药后约5个月给予加强剂量后,检查抗尖峰IgG和NAb的变化。我们发现加强后5个月体液反应显著增加,与第二剂量后观察到的减弱反应形成鲜明对比。值得注意的是,近四分之一的参与者即使在加强剂量后仍未达到保护性血浆水平.我们还发现,较高的估计肾小球滤过率(eGFR)与疫苗接种后更强大的体液反应相关。总之,这些发现强调了加强剂量在增强KTRs持久体液免疫中的有效性,如在升压后5个月的患者中发现的NAb的保护水平所证明的那样,尤其是那些具有较高的eGFR率。
    Kidney transplant recipients (KTRs), like other solid organ transplant recipients display a suboptimal response to mRNA vaccines, with only about half achieving seroconversion after two doses. However, the effectiveness of a booster dose, particularly in generating neutralizing antibodies (NAbs), remains poorly understood, as most studies have mainly focused on non-neutralizing antibodies. Here, we have longitudinally assessed the humoral response to the SARS-CoV-2 mRNA vaccine in 40 KTRs over a year, examining changes in both anti-spike IgG and NAbs following a booster dose administered about 5 months post-second dose. We found a significant humoral response increase 5 months post-booster, a stark contrast to the attenuated response observed after the second dose. Of note, nearly a quarter of participants did not achieve protective plasma levels even after the booster dose. We also found that the higher estimated glomerular filtration rate (eGFR) correlated with a more robust humoral response postvaccination. Altogether, these findings underscore the effectiveness of the booster dose in enhancing durable humoral immunity in KTRs, as evidenced by the protective level of NAbs found in 65% of the patients 5 months post- booster, especially those with higher eGFR rates.
    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
    复发性COVID-19感染,尽管全球疫苗接种,强调了加强剂量的需要。已经提出了一种异源加强剂,以增强对SARS-CoV-2病毒的新出现的变体的免疫力和保护作用。在这份报告中,我们的目的是评估安全性,和CORENAPCIN的免疫原性,作为三剂灭活疫苗后的第四剂加强剂量。
    方法:该研究是以双盲,随机化,基于mRNA的候选疫苗的安慰剂对照1期临床试验,CORENAPCIN.该疫苗作为异源加强剂注射到18-50岁的健康伊朗成年人中,这些成年人以前曾接受过三剂灭活的SARS-CoV-2疫苗。在研究中,30名参与者被随机分配接受两种不同剂量(25µg和50µg)的COReNAPCIN或安慰剂。候选疫苗包含编码SARS-CoV-2的融合前稳定的Spike蛋白的完整序列的mRNA,其配制在脂质纳米颗粒内。主要终点是安全性,次要目标是直到接种后6个月的体液免疫原性。追求细胞免疫原性作为探索性结果。
    结果:COReNAPCIN在两种剂量的接种个体中均具有良好的耐受性,没有危及生命或其他严重不良事件。最明显的不良事件是注射部位疼痛,疲劳和肌痛。关于免疫原性,尽管SARS-CoV-2抗体的血清阳性率是由于所有的疫苗接种史和一些参与者以前的SARS-CoV-2感染,25和50µgCORENAPCIN的接受者,接种疫苗两周后,显示抗RBD水平增加6·6和8·1倍,抗刺突抗体水平增加11·5和21·7倍,分别。几何平均病毒中和滴度在25µg组中达到10.2倍,在50µg预加强水平组中达到8.4倍。六个月后,与25和50µg组的基线水平相比,测得的抗尖峰抗体浓度仍保持2.8和6.3的几何平均倍数上升,分别。此外,疫苗接种后,特异性IFN-γT细胞应答的显著增加强调了细胞免疫的激活.
    结论:COReNAPCIN加强剂显示良好的安全性,耐受性,和免疫原性概况,支持其进一步的临床开发(试验注册:IRCT20230131057293N1)。
    The recurrent COVID-19 infection, despite global vaccination, highlights the need for booster doses. A heterologous booster has been suggested to enhance immunity and protection against emerging variants of concern of the SARS-CoV-2 virus. In this report, we aimed to assess the safety, and immunogenicity of COReNAPCIN, as a fourth booster dose after three doses of inactivated vaccines.
    METHODS: The study was conducted as a double-blind, randomized, placebo-controlled phase 1 clinical trial of the mRNA-based vaccine candidate, COReNAPCIN. The vaccine was injected as a heterologous booster in healthy Iranian adults aged 18-50 who had previously received three doses of inactivated SARS-CoV-2 vaccines. In the study, 30 participants were randomly assigned to receive either COReNAPCIN in two different doses (25 µg and 50 µg) or placebo. The vaccine candidate contained mRNA encoding the complete sequence of the pre-fusion stabilized Spike protein of SARS-CoV-2, formulated within lipid nanoparticles. The primary endpoint was safety and the secondary objective was humoral immunogenicity until 6 months post-vaccination. The cellular immunogenicity was pursued as an exploratory outcome.
    RESULTS: COReNAPCIN was well tolerated in vaccinated individuals in both doses with no life-threatening or other serious adverse events. The most noticeable solicited adverse events were pain at the site of injection, fatigue and myalgia. Regarding the immunogenicity, despite the seroprevalence of SARS-CoV-2 antibodies due to the vaccination history for all and previous SARS-CoV-2 infection for some participants, the recipients of 25 and 50 µg COReNAPCIN, two weeks post-vaccination, showed 6·6 and 8·1 fold increase in the level of anti-RBD, and 11·5 and 21·7 fold increase in the level of anti-spike antibody, respectively. The geometric mean virus neutralizing titers reached 10.2 fold in the 25 µg group and 8.4 fold in 50 µg group of pre-boost levels. After 6 months, the measured anti-spike antibody concentration still maintains a geometric mean fold rise of 2.8 and 6.3, comparing the baseline levels in 25 and 50 µg groups, respectively. Additionally, the significant increase in the spike-specific IFN-ϒ T-cell response upon vaccination underscores the activation of cellular immunity.
    CONCLUSIONS: COReNAPCIN booster showed favorable safety, tolerability, and immunogenicity profile, supporting its further clinical development (Trial registration: IRCT20230131057293N1).
    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
    尽管之前的研究表明,在主要系列COVID-19mRNA疫苗接种后,死亡风险没有增加,缺乏关于加强剂量的报告。这项研究旨在评估除主要系列外的mRNA疫苗增强剂之后的死亡风险。这项嵌套病例对照研究包括两个城市的两个年龄特定队列(截至2021年2月1日18-64岁和≥65岁)。确定所有死亡病例,并在每个死亡日期(索引日期)对每个病例进行5个对照,并根据市政当局进行风险集抽样,年龄,和性爱。mRNA疫苗(第一至第五剂量)的调整比值比(aOR)和95%置信区间(CIs)是通过使用条件逻辑回归模型在指标日期前21和42天内与未接种疫苗进行比较来估计的。18-64岁的队列包括431例(平均年龄,57.0岁;男人,58.2%)和2,155名对照(平均年龄,56.0;男子,58.2%),而≥65年队列包括12,166例(84.0;50.2%)和60,830例对照(84.0,50.2%)。在18-64年队列中,第三次和第四次给药后0-21天的aOR(95%CI)分别为0.62(0.24,1.62)和0.38(0.08,1.84),分别。≥65年队列中第三至第五剂量后的aOR(95%CI)为0.36(0.31,0.43),0.30(0.25,0.37),和0.26(0.20,0.33),分别。总之,mRNA疫苗的加强剂量不会增加死亡风险.这些发现可以帮助后续的疫苗运动和缓解疫苗的犹豫。
    Although previous studies have shown no increased mortality risk after the primary series of COVID-19 mRNA vaccines, reports on booster doses are lacking. This study aimed to evaluate mortality risk after the mRNA vaccine boosters in addition to the primary series. This nested case-control study included two age-specific cohorts (18-64 and ≥65 years as of February 1, 2021) in two municipalities. All deaths were identified and matched five controls for each case at each date of death (index date) with risk set sampling according to municipality, age, and sex. The adjusted odds ratios (aORs) and 95% confidence intervals (CIs) for mRNA vaccines (first to fifth doses) were estimated by comparing with no vaccination within 21 and 42 days before the index date using a conditional logistic regression model. The 18-64-years cohort comprised 431 cases (mean age, 57.0 years; men, 58.2%) and 2,155 controls (mean age, 56.0; men, 58.2%), whereas the ≥65-years cohort comprised 12,166 cases (84.0; 50.2%) and 60,830 controls (84.0, 50.2%). The aORs (95% CI) in 0-21 days after the third and fourth doses in the 18-64-years cohort were 0.62 (0.24, 1.62) and 0.38 (0.08, 1.84), respectively. The aORs (95% CI) after the third to fifth doses in the ≥65 years cohort were 0.36 (0.31, 0.43), 0.30 (0.25, 0.37), and 0.26 (0.20, 0.33), respectively. In conclusion, booster doses of mRNA vaccines do not increase mortality risk. These findings could help subsequent vaccine campaigns and alleviate vaccine hesitancy.
    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:尽管有一些关于COVID-19疫苗和体外受精(IVF)治疗的数据,其对血清免疫球蛋白G(IgG)水平的潜在影响尚未进行前瞻性评估.这项研究旨在评估COVID-19疫苗和IgG水平对IVF结局的影响。
    方法:这种观察,队列研究在转诊IVF病房进行.从2021年3月至4月招募了在COVID-19疫苗接种期间接受IVF治疗的夫妇。该研究比较了38名接受过辉瑞mRNACOVID-19疫苗接种的女性和10名没有感染过这种病毒的女性。我们还比较了24名女性接种前和接种后的IVF治疗方法。还评估了血清学滴度与IVF治疗结果之间的关系。
    结果:在主要结果测量方面,在接种和未接种/未感染组之间没有发现显着差异。然而,未接种疫苗组有更高的妊娠率趋势(57%vs.23%,p=0.078),但交货率没有差异(p=0.236),孕周(p=0.537)或出生率(p=0.671)。
    结论:我们谨慎声明,COVID-19mRNA疫苗不会影响生育结局,包括受精,妊娠率和分娩率,产科结果,和精液参数,无论测量的IgG水平。
    BACKGROUND: Although there are some data regarding the COVID-19 vaccine and in in vitro fertilization (IVF) treatments, its potential impact in terms of serum immunoglobulin G (IgG) levels has not been evaluated prospectively. This study aimed to assess the effect of COVID-19 vaccine and IgG levels on IVF outcomes.
    METHODS: This observational, cohort study was conducted at a referral IVF unit. Couples undergoing IVF treatment during the COVID-19 vaccination period were recruited from March-April 2021. The study compared 38 women who had received the Pfizer mRNA COVID-19 vaccination to 10 women who had not and were not infected by the virus. We also compared pre- and post-vaccination IVF treatments for 24 women. The relation between serologic titers and IVF treatment outcomes was also assessed.
    RESULTS: No significant difference was found between the vaccinated and unvaccinated/uninfected groups regarding the main outcome measures. However, there was a trend toward a higher pregnancy rate for the unvaccinated group (57% vs. 23%, p = 0.078) but no difference in delivery rate (p = 0.236), gestational week (p = 0.537) or birth rate (p = 0.671).
    CONCLUSIONS: We cautiously state that the COVID-19 mRNA vaccine does not affect fertility outcomes, including fertilization, pregnancy and delivery rates, obstetric outcomes, and semen parameters, regardless of measured IgG levels.
    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
    在这里,我们进行了多中心开放标签,随机2期和3期研究,以评估严重急性呼吸道综合征冠状病毒2(SARS-CoV-2)Omicron特异性(BA.1/B.1.1.529)的安全性和免疫原性,单价,热稳定,自扩增mRNA疫苗,GEMCOVAC-OM,当在接受两剂BBV152或ChAdOx1nCoV-19的健康成人中作为增强剂进行皮内给药时。GEMCOVAC-OM在2期和3期均具有良好的耐受性,没有相关的严重不良事件。在第2阶段,在140名参与者中,将GEMCOVAC-OM的安全性和免疫原性与我们的原型mRNA疫苗GEMCOVAC-19(D614G变体特异性)进行了比较。接种疫苗后第29天,使用GEMCOVAC-OM(P<0.0001)和GEMCOVAC-19(P<0.0001)的抗标(BA.1)IgG抗体显着升高。然而,GEMCOVAC-OM组的IgG滴度(主要终点)和血清转化率较高(P<0.0001).在第3阶段,在3,140名参与者(安全性队列)中,将GEMCOVAC-OM与ChAdOx1nCoV-19进行了比较,其中包括420名参与者的免疫原性队列。在第29天,在GEMCOVAC-OM臂中,针对SARS-CoV-2的BA.1变体的中和抗体滴度显着高于基线(P<0.0001),但在ChAdOx1nCoV-19臂中没有(P=0.1490)。GEMCOVAC-OM在中和抗体滴度和血清转换率方面不劣质(主要终点),并且优于ChAdOx1nCoV-19(最小二乘几何平均比率的下限95%置信区间>1,血清转换差异>0%为优势)。在第29天,GEMCOVAC-OM的抗尖峰IgG抗体和血清转化(次要终点)显著更高(P<0.0001)。这些结果证明GEMCOVAC-OM是安全的并且增强针对B.1.1.29变体的免疫应答。印度临床试验注册标识符:CTRI/2022/10/046475。
    Here we conducted a multicenter open-label, randomized phase 2 and 3 study to assess the safety and immunogenicity of a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron-specific (BA.1/B.1.1.529), monovalent, thermostable, self-amplifying mRNA vaccine, GEMCOVAC-OM, when administered intradermally as a booster in healthy adults who had received two doses of BBV152 or ChAdOx1 nCoV-19. GEMCOVAC-OM was well tolerated with no related serious adverse events in both phase 2 and phase 3. In phase 2, the safety and immunogenicity of GEMCOVAC-OM was compared with our prototype mRNA vaccine GEMCOVAC-19 (D614G variant-specific) in 140 participants. At day 29 after vaccination, there was a significant rise in anti-spike (BA.1) IgG antibodies with GEMCOVAC-OM (P < 0.0001) and GEMCOVAC-19 (P < 0.0001). However, the IgG titers (primary endpoint) and seroconversion were higher with GEMCOVAC-OM (P < 0.0001). In phase 3, GEMCOVAC-OM was compared with ChAdOx1 nCoV-19 in 3,140 participants (safety cohort), which included an immunogenicity cohort of 420 participants. At day 29, neutralizing antibody titers against the BA.1 variant of SARS-CoV-2 were significantly higher than baseline in the GEMCOVAC-OM arm (P < 0.0001), but not in the ChAdOx1 nCoV-19 arm (P = 0.1490). GEMCOVAC-OM was noninferior (primary endpoint) and superior to ChAdOx1 nCoV-19 in terms of neutralizing antibody titers and seroconversion rate (lower bound 95% confidence interval of least square geometric mean ratio >1 and difference in seroconversion >0% for superiority). At day 29, anti-spike IgG antibodies and seroconversion (secondary endpoints) were significantly higher with GEMCOVAC-OM (P < 0.0001). These results demonstrate that GEMCOVAC-OM is safe and boosts immune responses against the B.1.1.529 variant. Clinical Trial Registry India identifier: CTRI/2022/10/046475 .
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号