Vaccine Efficacy

疫苗功效
  • 文章类型: Journal Article
    在新兴的SARS-CoV-2变体或谱系以及新疫苗的背景下,准确监测COVID-19疫苗有效性(CVE)是为疫苗接种活动提供信息的关键。
    为了评估2022年秋季和冬季至2023年施用的COVID-19疫苗对有症状的SARS-CoV-2感染(尤其是所有循环病毒和XBB谱系)的有效性在欧洲60岁或以上的人群中,并比较所使用的暴露组和参考组的不同CVE方法。
    这项病例对照研究从VEBIS(疫苗有效性,负担和影响研究),一项多中心研究,从11个欧洲地点收集COVID-19和流感数据:克罗地亚;法国;德国;匈牙利;爱尔兰;葡萄牙;荷兰;罗马尼亚;西班牙,国家;西班牙,纳瓦拉地区;和瑞典。参与者是年龄在60岁或以上,有急性呼吸道感染症状的初级保健患者,他们是在2022年9月至2023年8月COVID-19疫苗接种活动开始后在11个地点招募的。病例和对照被定义为阳性和阴性的患者,分别,逆转录-聚合酶链反应(RT-PCR)检测结果。
    接触的是COVID-19疫苗。暴露组由在2022年秋季和冬季至2023年疫苗接种运动期间以及症状发作前14天或更长时间接受COVID-19疫苗的患者组成。参考组包括在2022年至2023年运动(季节性CVE)之前或之前6个月未接种疫苗的患者,那些从未接种过疫苗的人(绝对CVE),以及在运动前至少6个月或更长时间接种了主要系列疫苗的人(相对CVE)。对于第二助推器的相对CVE,将在运动期间接受第二次加强治疗的患者与运动前6个月或更长时间接受一次加强治疗的患者进行比较.
    结果经RT-PCR确认,医疗护理,有症状的SARS-CoV-2感染。生成了四个CVE估计值:季节性,绝对,相对,和第二助推器的亲戚。CVE使用逻辑回归估计,调整研究地点,症状发作日期,年龄,慢性疾病,和性爱。
    共包括9308名初级保健患者,1687例(1035名女性;中位[IQR]年龄,71[65-79]岁)和7621名对照(4619名女性[61%];中位[IQR]年龄,71[65-78]年)。接种疫苗后14至89天内,季节性CVE为29%(95%CI,14%-42%),绝对CVE为39%(95%CI,6%-60%),相对CVE为31%(95%CI,15%至44%),与所有SARS-CoV-2变体相比,第二助推器的相对CVE为34%(95%CI,18%-47%)。在相同的时间间隔内,季节性CVE为44%(95%CI,-10%至75%),绝对CVE为52%(95%CI,-23%至82%),相对CVE为47%(95%CI,-8%至77%),在高XBB循环期间,第二助推器的相对CVE为46%(95%CI,-13%至77%)。自接种疫苗以来,估计随着时间的推移而下降,疫苗接种后180天没有保护。
    在这项针对年长欧洲人的病例对照研究中,所有CVE方法都表明,2022年至2023年秋季和冬季施用的COVID-19疫苗可提供至少3个月的预防症状,医疗护理,实验室确诊的SARS-CoV-2感染。应使用CVE季节性方法持续监测新的COVID-19疫苗对新出现的SARS-CoV-2变体的有效性。
    UNASSIGNED: In the context of emerging SARS-CoV-2 variants or lineages and new vaccines, it is key to accurately monitor COVID-19 vaccine effectiveness (CVE) to inform vaccination campaigns.
    UNASSIGNED: To estimate the effectiveness of COVID-19 vaccines administered in autumn and winter 2022 to 2023 against symptomatic SARS-CoV-2 infection (with all circulating viruses and XBB lineage in particular) among people aged 60 years or older in Europe, and to compare different CVE approaches across the exposed and reference groups used.
    UNASSIGNED: This case-control study obtained data from VEBIS (Vaccine Effectiveness, Burden and Impact Studies), a multicenter study that collects COVID-19 and influenza data from 11 European sites: Croatia; France; Germany; Hungary; Ireland; Portugal; the Netherlands; Romania; Spain, national; Spain, Navarre region; and Sweden. Participants were primary care patients aged 60 years or older with acute respiratory infection symptoms who were recruited at the 11 sites after the start of the COVID-19 vaccination campaign from September 2022 to August 2023. Cases and controls were defined as patients with positive and negative, respectively, reverse transcription-polymerase chain reaction (RT-PCR) test results.
    UNASSIGNED: The exposure was COVID-19 vaccination. The exposure group consisted of patients who received a COVID-19 vaccine during the autumn and winter 2022 to 2023 vaccination campaign and 14 days or more before symptom onset. Reference group included patients who were not vaccinated during or in the 6 months before the 2022 to 2023 campaign (seasonal CVE), those who were never vaccinated (absolute CVE), and those who were vaccinated with at least the primary series 6 months or more before the campaign (relative CVE). For relative CVE of second boosters, patients receiving their second booster during the campaign were compared with those receiving 1 booster 6 months or more before the campaign.
    UNASSIGNED: The outcome was RT-PCR-confirmed, medically attended, symptomatic SARS-CoV-2 infection. Four CVE estimates were generated: seasonal, absolute, relative, and relative of second boosters. CVE was estimated using logistic regression, adjusting for study site, symptom onset date, age, chronic condition, and sex.
    UNASSIGNED: A total of 9308 primary care patients were included, with 1687 cases (1035 females; median [IQR] age, 71 [65-79] years) and 7621 controls (4619 females [61%]; median [IQR] age, 71 [65-78] years). Within 14 to 89 days after vaccination, seasonal CVE was 29% (95% CI, 14%-42%), absolute CVE was 39% (95% CI, 6%-60%), relative CVE was 31% (95% CI, 15% to 44%), and relative CVE of second boosters was 34% (95% CI, 18%-47%) against all SARS-CoV-2 variants. In the same interval, seasonal CVE was 44% (95% CI, -10% to 75%), absolute CVE was 52% (95% CI, -23% to 82%), relative CVE was 47% (95% CI, -8% to 77%), and relative CVE of second boosters was 46% (95% CI, -13% to 77%) during a period of high XBB circulation. Estimates decreased with time since vaccination, with no protection from 180 days after vaccination.
    UNASSIGNED: In this case-control study among older Europeans, all CVE approaches suggested that COVID-19 vaccines administered in autumn and winter 2022 to 2023 offered at least 3 months of protection against symptomatic, medically attended, laboratory-confirmed SARS-CoV-2 infection. The effectiveness of new COVID-19 vaccines against emerging SARS-CoV-2 variants should be continually monitored using CVE seasonal approaches.
    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
    分位数回归已成为分析竞争风险数据的广泛使用的工具。然而,具有连续标记的竞争风险数据的分位数回归仍然很少。标记变量是经典竞争风险模型中失败原因的扩展,其中失败原因被仅在未经审查的失败时间观察到的连续标记代替。连续标记变量的实例是测量感染病毒与疫苗构建体中包含的病毒之间的不相似性的遗传距离。在这篇文章中,我们提出了一种新的标记特定分位数回归模型。所提出的估计方法从标记附近的数据中借用强度,并基于诱导的平滑估计方程,这与现有的具有离散原因的竞争风险数据的方法有很大不同。所得估计量的渐近性质是在标记和分位数连续体中建立的。此外,提出了一种标记特异性分位数型疫苗效力,并开发了其统计推断程序。进行了模拟研究,以评估所提出的估计和假设检验程序的有限样本性能。提供了第一个HIV疫苗效力试验的应用。
    Quantile regression has become a widely used tool for analysing competing risk data. However, quantile regression for competing risk data with a continuous mark is still scarce. The mark variable is an extension of cause of failure in a classical competing risk model where cause of failure is replaced by a continuous mark only observed at uncensored failure times. An example of the continuous mark variable is the genetic distance that measures dissimilarity between the infecting virus and the virus contained in the vaccine construct. In this article, we propose a novel mark-specific quantile regression model. The proposed estimation method borrows strength from data in a neighbourhood of a mark and is based on an induced smoothed estimation equation, which is very different from the existing methods for competing risk data with discrete causes. The asymptotic properties of the resulting estimators are established across mark and quantile continuums. In addition, a mark-specific quantile-type vaccine efficacy is proposed and its statistical inference procedures are developed. Simulation studies are conducted to evaluate the finite sample performances of the proposed estimation and hypothesis testing procedures. An application to the first HIV vaccine efficacy trial is provided.
    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:大多数疫苗有效性下降的证据仅限于疫苗接种后相对较短的时间。
    方法:本研究使用了从日本关东地区某市的医疗管理索赔和疫苗接种记录的链接数据库获得的数据。研究期从2020年4月1日延长至2022年12月31日。使用时间依赖性分段Cox比例风险模型分析了COVID-19疫苗的有效性持续时间,性别和癌症史,糖尿病,慢性阻塞性肺疾病,哮喘,慢性肾病,和心血管疾病作为协变量。
    结果:在174,757名符合条件的个人中,14,416人(8.3%)被诊断为COVID-19,936人(0.54%)被诊断为COVID-19。基于时间依赖性Cox回归模型并参考非疫苗组的多变量分析显示,单剂量组的COVID-19发病率较低(风险比,0.76[95%置信区间,0.63-0.91]),两剂(0.89[0.85-0.93]),三剂量(0.80[0.76-0.85]),四剂量(0.93[0.88-1.00]),和五剂量组(0.72[0.62-0.84])。单剂量组的COVID-19相关住院发生率较低(0.42[0.21-0.81]),两剂量(0.44[0.35-0.56]),三剂量(0.38[0.30-0.47]),四剂量(0.20[0.14-0.28]),和五剂量(0.11[0.014-0.86])组。基于时间依赖性分段Cox比例风险模型并参考非疫苗组的多变量分析显示,该疫苗对COVID-19的发病率有4个月的显著预防效果,住院≥6个月的预防效果。
    结论:疫苗接种后,随着时间的推移,疫苗的有效性逐渐减弱;然而,对COVID-19发病率和住院的保护作用维持4个月和≥6个月,分别。这些结果可能有助于在COVID-19大流行后制定常规疫苗接种计划。
    BACKGROUND: Most evidence of the waning of vaccine effectiveness is limited to a relatively short period after vaccination.
    METHODS: Data obtained from a linked database of healthcare administrative claims and vaccination records maintained by the municipality of a city in the Kanto region of Japan were used in this study. The study period extended from April 1, 2020, to December 31, 2022. The duration of the effectiveness of the COVID-19 vaccine was analyzed using a time-dependent piecewise Cox proportional hazard model using the age, sex and history of cancer, diabetes, chronic obstructive pulmonary disease, asthma, chronic kidney disease, and cardiovascular disease as covariates.
    RESULTS: Among the 174,757 eligible individuals, 14,416 (8.3%) were diagnosed with COVID-19 and 936 (0.54%) were hospitalized for COVID-19. Multivariate analysis based on the time-dependent Cox regression model with reference of non-vaccine group revealed a lower incidence of COVID-19 in the one-dose group (hazard ratio, 0.76 [95% confidence interval, 0.63-0.91]), two-dose (0.89 [0.85-0.93]), three-dose (0.80 [0.76-0.85]), four-dose (0.93 [0.88-1.00]), and five-dose (0.72 [0.62-0.84]) groups. A lower incidence of COVID-19-related hospitalization was observed in the one-dose group (0.42 [0.21-0.81]), two-dose (0.44 [0.35-0.56]), three-dose (0.38 [0.30-0.47]), four-dose (0.20 [0.14-0.28]), and five-dose (0.11 [0.014-0.86]) groups. Multivariable analyses based on the time-dependent piecewise Cox proportional hazard model with reference of non-vaccine group revealed significant preventive effects of the vaccine for 4 months for the incidence of COVID-19 and ≥ 6 months for hospitalization.
    CONCLUSIONS: Vaccine effectiveness showed gradual attenuation with time after vaccination; however, protective effects against the incidence of COVID-19 and hospitalization were maintained for 4 months and ≥ 6 months, respectively. These results may aid in formulating routine vaccination plans after the COVID-19 pandemic.
    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:支持区域疫苗政策和实践的COVID-19疫苗有效性数据在非洲有限。因此,本综述旨在评估在非洲使用的COVID-19疫苗的疗效和有效性.
    方法:我们系统地检索了同行评审的随机对照试验(RCT),前瞻性和回顾性队列研究,以及报道非洲VE的病例对照研究。我们进行了偏见风险评估,这篇综述的结果被综合并以叙述的形式呈现,包括表格和数字。合成的重点是COVID-19VE对各种水平的疾病状况和结果(感染,住院治疗或危重,和死亡),时间点,和关注的变体。
    结果:共13项研究,总样本量为913,285名参与者,包括在这次审查中。大多数研究(8/13)来自南非,38.5%(5/13)是随机临床试验。研究报告说,全剂量的辉瑞-BioNTech疫苗对β(B.1.351)和Delta变体的COVID-19感染的VE为100%,对Delta变体的住院治疗的VE为96.7%。约翰逊和约翰逊疫苗的VE范围从38.1%-62.0%针对住院治疗和51.9%-86%针对β(B1.351)变体的危重疾病。Oxford-AstraZeneca疫苗对Omicron变体住院的VE为89.4%,但对B.1.351变体无效(10.4%)。Sinopharm疫苗对感染的VE为67%,对Delta变体的住院治疗的VE为46%。
    结论:在非洲使用的COVID-19疫苗可有效预防感染,住院治疗,和死亡。这些审查结果强调,所有利益攸关方需要齐心协力,加强COVID-19疫苗的获取和供应,并加强公众对接触高风险人群的认识,非洲人口中未接种疫苗的群体。
    BACKGROUND: Data on COVID-19 vaccine effectiveness to support regional vaccine policy and practice are limited in Africa. Thus, this review aimed to evaluate the efficacy and effectiveness of COVID-19 vaccines administered in Africa.
    METHODS: We systematically searched peer-reviewed randomized controlled trials (RCTs), prospective and retrospective cohort studies, and case-control studies that reported on VE in Africa. We carried out a risk of bias assessment, and the findings of this review were synthesized and presented in a narrative form, including tables and figures. The synthesis was focused on COVID-19 VE against various levels of the disease condition and outcomes (infection, hospitalization or critical, and death), time points, and variants of concern.
    RESULTS: A total of 13 studies, with a total sample size of 913,285 participants, were included in this review. The majority (8/13) of studies were from South Africa and 38.5% (5/13) were randomized clinical trials. The studies reported that a full dose of Pfizer-BioNTech vaccine had a VE of 100% against COVID-19 infection by Beta (B.1.351) and Delta variants and 96.7% against hospitalization by Delta variant. The Johnson and Johnson vaccine had VE ranging from 38.1%-62.0% against hospitalization and 51.9%- 86% against critical disease by Beta (B 1.351) variant. The Oxford-AstraZeneca vaccine had a VE of 89.4% against hospitalization by the Omicron variant but was not effective against the B.1.351 variant (10.4%). The Sinopharm vaccine had a VE of 67% against infection and 46% against hospitalization by Delta variant.
    CONCLUSIONS: COVID-19 vaccines administered in Africa were effective in preventing infections, hospitalization, and death. These review findings underscore the need for concerted efforts of all stakeholders to enhance the access and availability of COVID-19 vaccines and reinforce public awareness to reach the high-risk, unvaccinated group of the African 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
    世界卫生组织于2023年5月5日宣布2019年冠状病毒病(COVID-19)大流行结束。开发了几种疫苗,并公布了有关其有效性的新数据。然而,疫苗的临床试验是在Omicron变体出现之前进行的,有些人群仍需要对疫苗的有效性进行测试.本研究的总体目标是分析COVID-19疫苗在Omicron变种之前和之后在NuevoLeon人群中考虑合并症的患者中接种的效果,墨西哥。
    墨西哥社会保障研究所的流行病学COVID-19数据来自墨西哥东北部的67家医院,从2020年7月到2023年5月,共收集了669,393个案例,255,819报道了SARS-CoV-2阳性逆转录定量聚合酶链反应(RT-qPCR)测试或COVID-19抗原快速测试阳性。
    在Omicron之前(BO,2020-2021年),两剂COVID-19疫苗接种14天后,BNT162b2和ChAdOx1疫苗在非共病和所有共病亚组中对感染有效,而在Omicron(AO,2022年至2023年),没有任何疫苗对感染没有显著的效果。关于住院BO,BNT162b2,ChAdOx1,CoronaVac和mRNA-1273显着保护非合并症患者,而BNT162b2,ChAdOx1和mRNA-1273则保护所有合并症亚组免于住院。AO,BNT162b2,ChAdOx1,CoronaVac和mRNA-1273对非合并症患者的住院有效,而对于大多数合并症患者,BNT162b2,ChAdOx1和CoronaVac对住院有效。在BO期间,使用大多数疫苗可以保护非共病患者免受COVID-19的死亡,而在高血压患者中,使用mRNA-1273疫苗的AO效果降低,和糖尿病。
    BO,COVID-19疫苗对感染有效,住院治疗,和死亡,而AO,COVID-19疫苗未能保护人群免受COVID-19感染。观察到AO对住院和死亡的不同有效性。
    UNASSIGNED: The end of the coronavirus disease 2019 (COVID-19) pandemic has been declared by the World Health Organization on May 5, 2023. Several vaccines were developed, and new data is being published about their effectiveness. However, the clinical trials for the vaccines were performed before the Omicron variant appeared and there are population groups where vaccine effectiveness still needs to be tested. The overarching goal of the present study was to analyze the effects of COVID-19 vaccination before and after the Omicron variant in patients considering comorbidities in a population from Nuevo Leon, Mexico.
    UNASSIGNED: Epidemiological COVID-19 data from the Mexican Social Security Institute were collected from 67 hospitals located in northeastern Mexico, from July 2020 to May 2023, and a total of 669,393 cases were compiled, 255,819 reported a SARS-CoV-2 positive reverse transcription quantitative polymerase chain reaction (RT-qPCR) test or a positive COVID-19 antigen rapid test.
    UNASSIGNED: Before Omicron (BO, 2020-2021), after 14 days of two doses of COVID-19 vaccine, BNT162b2 and ChAdOx1 vaccines were effective against infection in non-comorbid and all comorbid subgroups, whereas after Omicron (AO, 2022- 2023) there was no significant effectiveness against infection with none of the vaccines. Regarding hospitalization BO, BNT162b2, ChAdOx1, CoronaVac and mRNA-1273 significantly protected non-comorbid patients whereas BNT162b2, ChAdOx1, and mRNA-1273, protected all comorbid subgroups against hospitalization. AO, BNT162b2, ChAdOx1, CoronaVac and mRNA-1273 were effective against hospitalization in non-comorbid patients whereas for most comorbid subgroups BNT162b2, ChAdOx1 and CoronaVac were effective against hospitalization. Non-comorbid patients were protected against death as an outcome of COVID-19 during the BO period with most vaccines whereas a reduction in effectiveness was observed AO with mRNA-1273 vaccines in patients with hypertension, and diabetes mellitus.
    UNASSIGNED: BO, COVID-19 vaccines were effective against infection, hospitalization, and death whereas AO, COVID-19 vaccines failed to protect the population from COVID-19 infection. A varying effectiveness against hospitalization and death is observed AO.
    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:2022-23年美国流感季节在2022年秋季初达到顶峰。
    方法:晚期流感疫苗对门诊患者的有效性(VE),在美国流感VE网络参与者中使用检测阴性设计计算实验室确诊的流感.
    结果:在2022年12月12日至2023年4月30日的2561名参与者中,有91例实验室确诊的流感病例主要患有A(H1N1)pdm09(6B.1A.5a.2a.1)或A(H3N2)(3C.2a1b.2a2b)。总的来说,VE为30%(95%置信区间-9%,54%);低后期活动排除了大多数亚组的估计。
    结论:2022-23年晚期门诊流感VE无统计学意义。基因组表征可以改善对流行流感后高峰的流感病毒的鉴定。
    BACKGROUND: The 2022-23 US influenza season peaked early in fall 2022.
    METHODS: Late-season influenza vaccine effectiveness (VE) against outpatient, laboratory-confirmed influenza was calculated among participants of the US Influenza VE Network using a test-negative design.
    RESULTS: Of 2561 participants enrolled from December 12, 2022 to April 30, 2023, 91 laboratory-confirmed influenza cases primarily had A(H1N1)pdm09 (6B.1A.5a.2a.1) or A(H3N2) (3C.2a1b.2a.2b). Overall, VE was 30% (95% confidence interval -9%, 54%); low late-season activity precluded estimation for most subgroups.
    CONCLUSIONS: 2022-23 late-season outpatient influenza VE was not statistically significant. Genomic characterization may improve the identification of influenza viruses that circulate postinfluenza peak.
    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
    我们计算了日本SARS-CoV-2OmicronBA.2主导时期COVID-19患者家庭接触者的攻击率。在近期(<3个月)未接种疫苗的家庭接触者中,完全接种疫苗的接触者的攻击率低于未完全接种疫苗的接触者。证明间接疫苗的有效性。
    We calculated attack rates for household contacts of COVID-19 patients during the SARS-CoV-2 Omicron BA.2-dominant period in Japan. Attack rates among household contacts without recent (<3 months) vaccination was lower for contacts of index patients with complete vaccination than for contacts of index patients without complete vaccination, demonstrating indirect vaccine 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
    接种流感疫苗可将流感的风险降低40%至60%,它们依赖于流感血凝素(HA)特异性中和抗体的产生,而忽略了神经氨酸酶(NA)作为重要的表面靶标.使用标准化NA浓度的疫苗接种可以提供更广泛和更持久的针对流感感染的保护。在这方面,我们旨在比较显示在VLP表面的NA与可溶性NA的效力。杆状病毒表达系统(BEVS)和新型无病毒Tnms42昆虫细胞系用于在基于gag的VLP上表达N2NA。为了生产高纯度和高浓度的VLP免疫原,使用两步色谱纯化工艺与超速离心相结合。在主要/加强疫苗接种计划中,接种1µgN2-VLP的小鼠免于死亡,而接受相同剂量的可溶性无佐剂NA的小鼠死于致死性感染。此外,加强前和攻击前血清的NA抑制测定和NA-ELISA证实VLP制剂诱导的NA特异性抗体水平高于可溶性无佐剂化NA。
    Vaccination against influenza virus can reduce the risk of influenza by 40% to 60%, they rely on the production of neutralizing antibodies specific to influenza hemagglutinin (HA) ignoring the neuraminidase (NA) as an important surface target. Vaccination with standardized NA concentration may offer broader and longer-lasting protection against influenza infection. In this regard, we aimed to compare the potency of a NA displayed on the surface of a VLP with a soluble NA. The baculovirus expression system (BEVS) and the novel virus-free Tnms42 insect cell line were used to express N2 NA on gag-based VLPs. To produce VLP immunogens with high levels of purity and concentration, a two-step chromatography purification process combined with ultracentrifugation was used. In a prime/boost vaccination scheme, mice vaccinated with 1 µg of the N2-VLPs were protected from mortality, while mice receiving the same dose of unadjuvanted NA in soluble form succumbed to the lethal infection. Moreover, NA inhibition assays and NA-ELISAs of pre-boost and pre-challenge sera confirm that the VLP preparation induced higher levels of NA-specific antibodies outperforming the soluble unadjuvanted NA.
    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:为了抗击2019年冠状病毒病(COVID-19),加强疫苗接种策略很重要。然而,加强疫苗平台的最佳给药方式尚不清楚.在这里,我们旨在评估3种或4种异源与同源加强方案的利弊.
    方法:从2022年11月3日至2023年12月21日,我们搜索了五个数据库中的随机临床试验(RCT)。审稿人筛选,提取的数据,并使用Cochrane偏差风险2工具独立评估偏差风险。我们对我们的主要结局(全因死亡率;实验室确认的有症状和严重COVID-19;严重不良事件[SAE])和次要结局(生活质量[QoL];不良事件[AE]视为非严重)进行了荟萃分析和试验序贯分析(TSA)。我们用分级方法评估了证据。对2023年前后的试验进行了亚组分析,三个或四个助推器,免疫受损状态,后续行动,偏见的风险,异源加强疫苗平台,和助推器的效价。
    结果:我们纳入了29项RCT和43项比较(12,538名参与者)。异源加强方案可能不会降低全因死亡率的相对风险(RR)(11项试验;RR0.86;95%CI0.33至2.26;I20%;非常低的确定性证据);实验室确认的有症状的COVID-19(14项试验;RR0.95;95%CI0.72至1.25;I20%;非常低的确定性);或严重的COVID-19(10项;IRR0.51;33%至2非常低的对于安全结果,异源加强方案可能对SAE没有影响(27项试验;RR1.15;95%CI0.68~1.95;I20%;非常低的确定性),但可能会增加被认为非严重的AE(20项试验;RR1.19;95%CI1.08~1.32;I264.4%;非常低的确定性)。没有可用的QoL数据。我们的TSA表明,累积Z曲线对于任何结果都没有达到徒劳。
    结论:根据我们目前的样本量,我们无法推断任何结果的影响差异,但是异源加强方案似乎会导致更多的非严重AE。此外,更可靠的数据有助于更新这篇评论。
    BACKGROUND: To combat coronavirus disease 2019 (COVID-19), booster vaccination strategies are important. However, the optimal administration of booster vaccine platforms remains unclear. Herein, we aimed to assess the benefits and harms of three or four heterologous versus homologous booster regimens.
    METHODS: From November 3 2022 to December 21, 2023, we searched five databases for randomised clinical trials (RCT). Reviewers screened, extracted data, and assessed bias risks independently with the Cochrane risk-of-bias 2 tool. We conducted meta-analyses and trial sequential analyses (TSA) on our primary (all-cause mortality; laboratory confirmed symptomatic and severe COVID-19; serious adverse events [SAE]) and secondary outcomes (quality of life [QoL]; adverse events [AE] considered non-serious). We assessed the evidence with the GRADE approach. Subgroup analyses were stratified for trials before and after 2023, three or four boosters, immunocompromised status, follow-up, risk of bias, heterologous booster vaccine platforms, and valency of booster.
    RESULTS: We included 29 RCTs with 43 comparisons (12,538 participants). Heterologous booster regimens may not reduce the relative risk (RR) of all-cause mortality (11 trials; RR 0.86; 95% CI 0.33 to 2.26; I2 0%; very low certainty evidence); laboratory-confirmed symptomatic COVID-19 (14 trials; RR 0.95; 95% CI 0.72 to 1.25; I2 0%; very low certainty); or severe COVID-19 (10 trials; RR 0.51; 95% CI 0.20 to 1.33; I2 0%; very low certainty). For safety outcomes, heterologous booster regimens may have no effect on SAE (27 trials; RR 1.15; 95% CI 0.68 to 1.95; I2 0%; very low certainty) but may raise AE considered non-serious (20 trials; RR 1.19; 95% CI 1.08 to 1.32; I2 64.4%; very low certainty). No data on QoL was available. Our TSAs showed that the cumulative Z curves did not reach futility for any outcome.
    CONCLUSIONS: With our current sample sizes, we were not able to infer differences of effects for any outcomes, but heterologous booster regimens seem to cause more non-serious AE. Furthermore, more robust data are instrumental to update this review.
    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:英国于2021年9月交付了第一支“加强”COVID-19疫苗,最初是针对严重疾病高危人群,然后是所有成年人。BNT162b2Pfizer-BioNTech疫苗最初使用,然后还有ModernamRNA-1273。
    方法:经英国国家卫生局批准,我们使用常规临床数据来评估BNT162b2或mRNA-1273强化治疗的有效性,而在接受两次主要疗程疫苗剂量的符合条件的成年人中,BNT162b2或mRNA-1273强化治疗的有效性.在与加强优先状态和先前的COVID-19免疫相关的因素上,我们将每个加强接受者与未加强对照进行了匹配。我们调整了Cox模型中的其他因素,估计增加剂量后182天(6个月)的危险比。我们估计了总体和以下时间段内的危险比:1-14、15-42、43-69、70-97、98-126、127-152和155-182天。结果包括SARS-CoV-2测试阳性,COVID-19住院,COVID-19死亡,非COVID-19死亡,和骨折。
    结果:我们将8,198,643名加强接受者与未加强对照进行了匹配。6个月随访期间调整后的危险比为:SARS-CoV-2测试阳性0.75(0.74,0.75);COVID-19住院0.30(0.29,0.31);COVID-19死亡0.11(0.10,0.14);非COVID-19死亡0.22(0.21,0.23);骨折0.77(0.75,0.78)。加强疫苗对严重COVID-19相关结果的估计有效性在加强剂量后的前3个月达到峰值。到6个月,SARS-CoV-2试验阳性的累积发生率在加强者中高于未加强者.
    结论:我们估计COVID-19加强疫苗接种,与没有加强疫苗接种相比,对COVID-19住院和COVID-19死亡提供实质性保护,但对SARS-CoV-2检测阳性仅提供有限保护。与未增强个体相比,增强个体的骨折发生率较低,这可能表明无法测量的混杂因素。观察性研究应报告针对非目标和阴性对照结果的估计疫苗有效性。
    BACKGROUND: The UK delivered its first \"booster\" COVID-19 vaccine doses in September 2021, initially to individuals at high risk of severe disease, then to all adults. The BNT162b2 Pfizer-BioNTech vaccine was used initially, then also Moderna mRNA-1273.
    METHODS: With the approval of the National Health Service England, we used routine clinical data to estimate the effectiveness of boosting with BNT162b2 or mRNA-1273 compared with no boosting in eligible adults who had received two primary course vaccine doses. We matched each booster recipient with an unboosted control on factors relating to booster priority status and prior COVID-19 immunization. We adjusted for additional factors in Cox models, estimating hazard ratios up to 182 days (6 months) following booster dose. We estimated hazard ratios overall and within the following periods: 1-14, 15-42, 43-69, 70-97, 98-126, 127-152, and 155-182 days. Outcomes included a positive SARS-CoV-2 test, COVID-19 hospitalization, COVID-19 death, non-COVID-19 death, and fracture.
    RESULTS: We matched 8,198,643 booster recipients with unboosted controls. Adjusted hazard ratios over 6-month follow-up were: positive SARS-CoV-2 test 0.75 (0.74, 0.75); COVID-19 hospitalization 0.30 (0.29, 0.31); COVID-19 death 0.11 (0.10, 0.14); non-COVID-19 death 0.22 (0.21, 0.23); and fracture 0.77 (0.75, 0.78). Estimated effectiveness of booster vaccines against severe COVID-19-related outcomes peaked during the first 3 months following the booster dose. By 6 months, the cumulative incidence of positive SARS-CoV-2 test was higher in boosted than unboosted individuals.
    CONCLUSIONS: We estimate that COVID-19 booster vaccination, compared with no booster vaccination, provided substantial protection against COVID-19 hospitalization and COVID-19 death but only limited protection against positive SARS-CoV-2 test. Lower rates of fracture in boosted than unboosted individuals may suggest unmeasured confounding. Observational studies should report estimated vaccine effectiveness against nontarget and negative control outcomes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号