Viral Fusion Proteins

病毒融合蛋白
  • 文章类型: Journal Article
    病毒与细胞膜的融合是包膜病毒生命周期中的关键步骤。病毒融合蛋白促进了这一过程,其中许多是构象pH敏感的。pH变化如何引发这种融合的细节在很大程度上仍然难以捉摸。这项研究提出了原型III类融合蛋白的冷冻电子显微镜(cryo-EM)结构,GP64,在其融合前和早期中间状态下,揭示了伴随膜融合过程的结构中间体。这些结构确定了pH敏感开关的参与,包括H23、H245和H304,在感测触发膜融合的初始步骤的低pH时。该开关的pH传感作用通过细胞-细胞合胞体形成和双重染料标记的测定得到证实。研究结果表明,多个组氨酸残基之间的配位充当pH传感器和激活剂。多组氨酸开关在病毒融合中的参与适用于感染人的流感病毒和其他病毒的融合剂,这可能导致开发抗病毒疗法和疫苗的策略。
    The fusion of viruses with cellular membranes is a critical step in the life cycle of enveloped viruses. This process is facilitated by viral fusion proteins, many of which are conformationally pH-sensitive. The specifics of how changes in pH initiate this fusion have remained largely elusive. This study presents the cryo-electron microscopy (cryo-EM) structures of a prototype class III fusion protein, GP64, in its prefusion and early intermediate states, revealing the structural intermediates accompanying the membrane fusion process. The structures identify the involvement of a pH-sensitive switch, comprising H23, H245, and H304, in sensing the low pH that triggers the initial step of membrane fusion. The pH sensing role of this switch is corroborated by assays of cell-cell syncytium formation and dual dye-labeling. The findings demonstrate that coordination between multiple histidine residues acts as a pH sensor and activator. The involvement of a multi-histidine switch in viral fusion is applicable to fusogens of human-infecting thogotoviruses and other viruses, which could lead to strategies for developing anti-viral therapies and 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
    人类呼吸道合胞病毒(RSV)是一个重要的健康问题,特别是对于婴儿,年幼的孩子,和老人。已知这种病毒由于环境因素和群体免疫而不断进化。鉴于此,我们的研究旨在分析2020-2022年科威特RSV-A和RSV-B基因型中G蛋白的遗传变异性.在2020年1月至2022年9月之间,我们从急性呼吸道感染的住院患者中收集了490个呼吸道样本。使用多重实时PCR测试并证实这些样品对RSV呈阳性。随后,使用先进的纳米孔测序技术对样品进行核酸测序,以分析全长G基因。序列分析表明,64株(76%)为RSV-A,20株(24%)为RSV-B。RSV-A的G基因属于GA2.3.5基因型,而所有RSV-B基因型均属于GB5.0.5a。检测到RSV-A和RSV-B的新谱系和亚谱系,表明新菌株在科威特的传播。许多独特和新的氨基酸变化,包括插入,在科威特分离株的G蛋白中发现,第二高变区的变异性最高。在G蛋白中也发现了数量增加的N和O连接的糖基化位点。这可以推测改变RSV的抗原性。已确定的RSV-A和RSV-B基因型的G蛋白变化可能是由免疫压力引起的,并可能影响科威特循环菌株的抗原特性。这可能潜在地导致可以逃避免疫应答的新的RSV变体。我们对RSV-A和RSV-B的G蛋白的深入分析可以帮助开发更有效的治疗和疫苗。
    The human respiratory syncytial virus (RSV) is a significant health concern, particularly for infants, young children, and the elderly. This virus is known to evolve continuously due to environmental factors and herd immunity. In light of this, our study aimed to analyze the genetic variability of the G protein in RSV-A and RSV-B genotypes in Kuwait from 2020 to 2022. Between January 2020 and September 2022, we collected 490 respiratory samples from hospitalized patients with acute respiratory tract infections. These samples were tested and confirmed positive for RSV using multiplex Real-Time PCR. Subsequently, the samples underwent nucleic acid sequencing using the advanced Nanopore sequencing technology to analyze the full-length G gene. Sequence analysis showed that 64 isolates (76%) were RSV-A, and 20 isolates (24%) were RSV-B. The G genes of RSV-A belonged to genotype GA2.3.5, while all the RSV-B genotypes belonged to GB5.0.5a. New lineages and sub-lineages of RSV-A and RSV-B were detected, indicating the circulation of new strains in Kuwait. Many unique and new amino acid changes, including insertions, were found in the G proteins of Kuwaiti isolates, with the highest variability in the second hypervariable region. An increased number of N and O-linked glycosylation sites were also identified in the G protein, which could speculate to alter the antigenicity of RSV. The identified changes in the G protein of RSV-A and RSV-B genotypes might result from immune pressure and could affect the antigenic characteristics of circulating strains in Kuwait. This could potentially lead to new RSV variants that can evade the immune response. Our in-depth analysis of the G proteins of both RSV-A and RSV-B could aid in the development of more potent treatments and 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
    新城疫(ND)是由1型禽副粘病毒的毒株,也称为新城疫病毒(NDV)。尽管接种了疫苗,报道的埃塞俄比亚疫情的频率有所增加。2022年1月至6月,在埃塞俄比亚中部地区的六个商业养鸡场进行了积极的疫情调查,以确定正在传播的NDV菌株。从怀疑感染了NDV的鸡中收集了30个合并的组织样本。还对农场主和兽医进行了问卷调查,以收集有关农场和爆发状况的信息。使用无特异性病原体(SPF)胚胎鸡蛋分离NDV,并使用血凝和逆转录酶聚合酶链反应(RT-PCR)进行检测。使用SPF胚胎鸡蛋中融合(F)蛋白基因序列的系统发育分析和平均死亡时间(MDT)测试,确定了田间NDV分离株的基因型和毒力。问卷调查结果显示,ND导致发病率(23.1%),死亡率(16.3%),病死率(70.8%),和重大的经济损失。使用血凝和RT-PCR测试了30个组织样本中的11个NDV阳性。MDT测试和序列分析显示,存在被分类为II类病毒致病型的基因型VII的毒性NDV,与本地使用的疫苗株(基因型II)不同。当前的强毒NDV融合蛋白切割位点基序的氨基酸序列揭示了112RRQKR→F117,与当地使用的无毒疫苗株(112GRQGR→L117)不同。流行病学数据,MDT结果,切割位点序列,系统发育分析表明,目前的NDV分离株具有强毒力。四个NDV序列保藏在GenBank中,登录号为F基因(PP726912-15)和M基因(PP726916-19)。无毒疫苗株和循环毒力NDV之间的遗传差异可以解释本地使用的疫苗提供的低水平保护。需要进一步的研究来更好地了解不同生产系统中的循环NDV基因型。
    Newcastle disease (ND) is caused by virulent strains of avian paramyxovirus type 1, also known as Newcastle disease virus (NDV). Despite vaccination, the frequency of reported outbreaks in Ethiopia has increased. From January to June 2022, an active outbreak investigation was conducted in six commercial chicken farms across areas of central Ethiopia to identify the circulating NDV strains. Thirty pooled tissue specimens were collected from chickens suspected of being infected with NDV. A questionnaire survey of farm owners and veterinarians was also carried out to collect information on the farms and the outbreak status. NDV was isolated using specific-pathogen-free (SPF)-embryonated chicken eggs and detected using haemagglutination and the reverse transcriptase-polymerase chain reaction (RT-PCR). The genotype and virulence of field NDV isolates were determined using phylogenetic analysis of fusion (F) protein gene sequences and the mean death time (MDT) test in SPF-embryonated chicken eggs. The questionnaire results revealed that ND caused morbidity (23.1%), mortality (16.3%), case fatality (70.8%), and significant economic losses. Eleven of thirty tissue specimens tested positive for NDV using haemagglutination and RT-PCR. The MDT testing and sequence analysis revealed the presence of virulent NDV classified as genotype VII of class II velogenic pathotype and distinct from locally used vaccine strains (genotype II). The amino acid sequences of the current virulent NDV fusion protein cleavage site motif revealed 112RRQKR↓F117, unlike the locally used avirulent vaccine strains (112GRQGR↓L117). The epidemiological data, MDT results, cleavage site sequence, and phylogenetic analysis all indicated that the present NDV isolates were virulent. The four NDV sequences were deposited in GenBank with accession numbers F gene (PP726912-15) and M gene (PP726916-19). The genetic difference between avirulent vaccine strains and circulating virulent NDV could explain the low level of protection provided by locally used vaccines. Further studies are needed to better understand the circulating NDV genotypes in different production systems.
    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
    尼帕病毒(NiV)是一种高致病性人畜共患病毒,它被列入世界卫生组织研究与发展蓝图清单,死亡率高达70%。由于其高致病性和爆发效力,迫切需要针对NiV的治疗对策。由于NiV需要在生物安全等级(BSL)4设施内处理,我们利用杆状病毒表达载体系统(BEVS)开发了一个安全的药物筛选平台,该系统基于NiV诱导的合胞体形成,可在BSL-1设施内处理.为了重建NiV诱导的BEVS合胞体形成,产生了两种杆状病毒来表达负责诱导合胞体形成的重组蛋白,包括一种表现出共表达的NiV融合蛋白(NiV-F)和NiV附着糖蛋白(NiV-G)的杆状病毒和另一种表现出人EphrinB2蛋白的杆状病毒。有趣的是,当培养基被修饰为具有较低的pH水平并补充胆固醇时,在感染的昆虫细胞中观察到合胞体形成。几种化合物的融合抑制性能,如植物化学物质和多磺化萘胺化合物,使用这个平台进行评估。在这些化合物中,suramin在杆状病毒表达系统中对NiV诱导的合胞体显示出最高的融合抑制活性。此外,我们的计算机模拟结果提供了苏拉明与NiV-G的中心孔和EphrinB2的G-H环相互作用的分子水平一瞥,这可能是其融合抑制活性的可能原因。
    Nipah virus (NiV) is known to be a highly pathogenic zoonotic virus, which is included in the World Health Organization Research & Development Blueprint list of priority diseases with up to 70% mortality rate. Due to its high pathogenicity and outbreak potency, a therapeutic countermeasure against NiV is urgently needed. As NiV needs to be handled within a Biological Safety Level (BSL) 4 facility, we had developed a safe drug screening platform utilizing a baculovirus expression vector system (BEVS) based on a NiV-induced syncytium formation that could be handled within a BSL-1 facility. To reconstruct the NiV-induced syncytium formation in BEVS, two baculoviruses were generated to express recombinant proteins that are responsible for inducing the syncytium formation, including one baculovirus exhibiting co-expressed NiV fusion protein (NiV-F) and NiV attachment glycoprotein (NiV-G) and another exhibiting human EphrinB2 protein. Interestingly, syncytium formation was observed in infected insect cells when the medium was modified to have a lower pH level and supplemented with cholesterol. Fusion inhibitory properties of several compounds, such as phytochemicals and a polysulfonated naphthylamine compound, were evaluated using this platform. Among these compounds, suramin showed the highest fusion inhibitory activity against NiV-induced syncytium in the baculovirus expression system. Moreover, our in silico results provide a molecular-level glimpse of suramin\'s interaction with NiV-G\'s central hole and EphrinB2\'s G-H loop, which could be the possible reason for its fusion inhibitory activity.
    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
    牛β冠状病毒(BoCoV)是一种与人类冠状病毒OC43密切相关的牛的肠道致病菌。接种疫苗是为了预防BoCoV引起的疾病,但是在保护和免疫逃避对驱动进化的影响方面存在知识差距。在这项研究中,将免疫表位定位到BoCoV结构蛋白上,包括穗和血凝素酯酶(HE),然后支持爱尔兰临床分离株的靶向基因测序和选择性压力分析。在一些定位的免疫表位中多样化选择和氨基酸变化的普遍性增加表明免疫逃逸正在选择在这些区域中出现的非同义突变。与其他相比,选择分析和测序为中和抗体(nAb)表位提供了更多的支持,表明nAbs是BoCoV免疫反应的重要分支。刺突和HE序列的系统发育分析表明,这项研究中的爱尔兰分离株位于欧洲进化枝,除了一个位于亚洲/美国进化枝的HE序列,而这个样本的刺突基因在欧洲进化枝。欧洲和亚洲/美国分离株之间的重组将产生这样的序列。这项研究收集了证据,表明逃避nAb反应的压力有助于BoCoV进化。
    Bovine betacoronavirus (BoCoV) is a pneumoenteric pathogen of cattle that is closely related to human coronavirus OC43. Vaccines are administered to protect against diseases caused by BoCoV, but knowledge gaps exist with regard to correlates of protection and the effect of immune evasion on driving evolution. In this study, immune epitopes were mapped onto BoCoV structural proteins, including spike and haemagglutinin esterase (HE), and then supported with targeted gene sequencing of Irish clinical isolates and selective pressure analysis. Increased prevalence of diversifying selection and amino acid changes in some mapped immune epitopes suggests that immune escape is selecting for non-synonymous mutations arising in these regions. Selection analysis and sequencing provided increased support for neutralising antibody (nAb) epitopes compared to others, suggesting that nAbs are an important arm of the immune response to BoCoV. Phylogenetic analysis of spike and HE sequences showed that Irish isolates from this study were in the European clade, except for one HE sequence that sat in the Asian/American clade, while the spike gene of this sample was in the European clade. Recombination between a European and an Asian/American isolate would give rise to such a sequence. This study has gathered evidence suggesting that pressure to evade the nAb response is contributing to BoCoV evolution.
    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
    许多包膜病毒,比如冠状病毒,流感,和呼吸道合胞病毒(RSV),利用I类融合蛋白进入细胞。在这个过程中,蛋白质从融合前过渡到融合后状态,经历实质性和不可逆的构象变化。融合前构象在疫苗开发中反复显示出巨大的潜力。然而,这种状态的不稳定性对其在疫苗中的实际应用提出了挑战。虽然非天然二硫化物在维持预融合结构方面是有效的,鉴定稳定二硫键仍然是一项复杂的任务。这里,我们提出了一种通用的计算方法来系统地识别融合前稳定的二硫化物。我们的方法评估了二硫键的几何约束,并引入了一个排名系统来估计它们在稳定融合前构象方面的潜力。我们假设,限制构象转换的初始阶段的二硫化物可以比阻止后期展开的二硫化物提供更高的融合前状态稳定性。我们的算法在RSVF蛋白上的实施导致发现了支持我们假设的融合前稳定的二硫化物。此外,我们的顶级设计在棉鼠模型中作为疫苗候选物的评估证明了对RSV感染的强大保护作用,强调了我们疫苗开发方法的潜力。
    Numerous enveloped viruses, such as coronaviruses, influenza, and respiratory syncytial virus (RSV), utilize class I fusion proteins for cell entry. During this process, the proteins transition from a prefusion to a postfusion state, undergoing substantial and irreversible conformational changes. The prefusion conformation has repeatedly shown significant potential in vaccine development. However, the instability of this state poses challenges for its practical application in vaccines. While non-native disulfides have been effective in maintaining the prefusion structure, identifying stabilizing disulfide bonds remains an intricate task. Here, we present a general computational approach to systematically identify prefusion-stabilizing disulfides. Our method assesses the geometric constraints of disulfide bonds and introduces a ranking system to estimate their potential in stabilizing the prefusion conformation. We hypothesized that disulfides restricting the initial stages of the conformational switch could offer higher stability to the prefusion state than those preventing unfolding at a later stage. The implementation of our algorithm on the RSV F protein led to the discovery of prefusion-stabilizing disulfides that supported our hypothesis. Furthermore, the evaluation of our top design as a vaccine candidate in a cotton rat model demonstrated robust protection against RSV infection, highlighting the potential of our approach for vaccine development.
    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
    RSV感染仍然是全世界儿童的严重威胁,尤其是,在中低收入国家。经由粘膜的疫苗递送具有在呼吸道中诱导局部免疫应答的巨大潜力。以前,我们报道了高免疫原性RSV病毒样颗粒(RSV-VLP)的发展,其基础是构象稳定的前-F蛋白(preFg),糖蛋白和基质蛋白。这里,探讨RSV-VLPs的粘膜递送是否是诱导RSV特异性粘膜和全身免疫的有效策略,RSV-VLP经鼻给药,BALB/c小鼠的舌下和肺部途径。结果表明,通过粘膜途径用VLP免疫诱导最小的粘膜反应,但促进适度水平的血清IgG抗体,增强的T细胞反应和脾细胞上肺归巢标志物CXCR3的表达。通过所有三种粘膜途径用VLP免疫提供针对RSV攻击的保护,而没有RSV诱导的病理学的迹象。
    RSV infection remains a serious threat to the children all over the world, especially, in the low-middle income countries. Vaccine delivery via the mucosa holds great potential for inducing local immune responses in the respiratory tract. Previously, we reported the development of highly immunogenic RSV virus-like-particles (RSV-VLPs) based on the conformationally stable prefusogenic-F protein (preFg), glycoprotein and matrix protein. Here, to explore whether mucosal delivery of RSV-VLPs is an effective strategy to induce RSV-specific mucosal and systemic immunity, RSV-VLPs were administered via the nasal, sublingual and pulmonary routes to BALB/c mice. The results demonstrate that immunization with the VLPs via the mucosal routes induced minimal mucosal response and yet facilitated modest levels of serum IgG antibodies, enhanced T cell responses and the expression of the lung-homing marker CXCR3 on splenocytes. Immunization with VLPs via all three mucosal routes provided protection against RSV challenge with no signs of RSV induced pathology.
    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
    呼吸道合胞病毒(RSV)劫持胆固醇或自噬途径以促进最佳复制。然而,我们对相关分子机制的理解仍然有限。这里,我们显示RSV感染通过下调溶酶体酸性脂肪酶的活性来阻断胆固醇从溶酶体向内质网的转运,激活SREBP2-LDLR轴,并促进溶酶体中外源性胆固醇的摄取和积累。高胆固醇水平会损害ORP1L的VAP-A结合活性,并促进动力蛋白-动力蛋白的募集,PLEKHM1或HOPSVPS39至Rab7-RILP,从而促进自噬体的负端转运和自溶酶体的形成。富含胆固醇的溶酶体的酸化抑制和功能障碍通过抑制自溶酶体降解来损害自噬通量,促进RSV融合蛋白的积累。在胆固醇消耗或LDLR敲低后,RSV-F的储存几乎被废除。最重要的是,LDLR的敲除在体内有效抑制RSV感染。这些发现阐明了RSV如何共同调节溶酶体胆固醇重编程和自噬的分子机制,并揭示了LDLR作为抗RSV药物开发的新靶标。
    Respiratory syncytial virus (RSV) hijacks cholesterol or autophagy pathways to facilitate optimal replication. However, our understanding of the associated molecular mechanisms remains limited. Here, we show that RSV infection blocks cholesterol transport from lysosomes to the endoplasmic reticulum by downregulating the activity of lysosomal acid lipase, activates the SREBP2-LDLR axis, and promotes uptake and accumulation of exogenous cholesterol in lysosomes. High cholesterol levels impair the VAP-A-binding activity of ORP1L and promote the recruitment of dynein-dynactin, PLEKHM1, or HOPS VPS39 to Rab7-RILP, thereby facilitating minus-end transport of autophagosomes and autolysosome formation. Acidification inhibition and dysfunction of cholesterol-rich lysosomes impair autophagy flux by inhibiting autolysosome degradation, which promotes the accumulation of RSV fusion protein. RSV-F storage is nearly abolished after cholesterol depletion or knockdown of LDLR. Most importantly, the knockout of LDLR effectively inhibits RSV infection in vivo. These findings elucidate the molecular mechanism of how RSV co-regulates lysosomal cholesterol reprogramming and autophagy and reveal LDLR as a novel target for anti-RSV drug development.
    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
    人偏肺病毒融合蛋白(hMPVPre-F)的预融合构象对于引发最有效的中和抗体是关键的,并且是针对hMPV呼吸道感染的有效疫苗的优选免疫原。在这里,我们显示F蛋白中的额外切割事件允许三聚体的闭合和正确折叠。因此,我们设计了F蛋白进行双重切割,这使得能够在天然折叠的原聚体界面处筛选Pre-F稳定取代。为了识别这些替代,我们开发了一个AI卷积分类器,它成功地预测了复杂的极点相互作用,通常被基于物理的方法和视觉检测所忽略。额外处理的组合,界面区域的稳定和膜近端茎的稳定,产生Pre-F蛋白疫苗候选物,而不需要表现出高表达产量和热稳定性的异源三聚化结构域。Cryo-EM分析显示完整的胞外域结构,包括茎,以及新鉴定的切割的C末端与相邻的原聚体的特异性相互作用。重要的是,该蛋白诱导高和交叉中和抗体反应,导致棉鼠对hMPV攻击的几乎完全保护,使高度稳定,双切割的hMPVPre-F三聚体是一种有吸引力的疫苗候选物。
    The prefusion conformation of human metapneumovirus fusion protein (hMPV Pre-F) is critical for eliciting the most potent neutralizing antibodies and is the preferred immunogen for an efficacious vaccine against hMPV respiratory infections. Here we show that an additional cleavage event in the F protein allows closure and correct folding of the trimer. We therefore engineered the F protein to undergo double cleavage, which enabled screening for Pre-F stabilizing substitutions at the natively folded protomer interfaces. To identify these substitutions, we developed an AI convolutional classifier that successfully predicts complex polar interactions often overlooked by physics-based methods and visual inspection. The combination of additional processing, stabilization of interface regions and stabilization of the membrane-proximal stem, resulted in a Pre-F protein vaccine candidate without the need for a heterologous trimerization domain that exhibited high expression yields and thermostability. Cryo-EM analysis shows the complete ectodomain structure, including the stem, and a specific interaction of the newly identified cleaved C-terminus with the adjacent protomer. Importantly, the protein induces high and cross-neutralizing antibody responses resulting in near complete protection against hMPV challenge in cotton rats, making the highly stable, double-cleaved hMPV Pre-F trimer an attractive vaccine candidate.
    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:最近批准的基于AS01E佐剂的呼吸道合胞病毒(RSV)融合前F蛋白的老年人疫苗(RSVPreF3OA)在≥60岁的人群中表现出对RSV相关疾病的高疗效。
    方法:这项在≥60岁人群中进行的3期研究评估了RSVPreF3OA疫苗接种后3年的免疫持久性。这里,我们描述了体液和细胞介导的免疫原性的中期结果,反应原性,和安全性,直到1年剂量后1。
    结果:总计,1653名参与者接种了疫苗。剂量1后一个月,中和滴度相对于剂量1前增加10.5倍(RSV-A)和7.8倍(RSV-B)。然后,滴度在第6个月下降到比剂量1前高4.4倍(RSV-A)和3.5倍(RSV-B)的水平,并保持3.1倍(RSV-A)和2.3倍(RSV-B)高于1年后的剂量1水平。RSVPreF3结合免疫球蛋白G水平和CD4+T细胞频率显示相似的动力学。62.2%和49.5%的参与者报告了可疑的给药部位和全身不良事件(主要是轻度至中度和短暂)。3.9%的参与者在剂量1后6个月内报告了严重不良事件;1例被认为与疫苗相关。
    结论:一种RSVPreF3OA剂量引起细胞介导的和RSV-A-和RSV-B-特异性体液免疫反应,随着时间的推移而下降,但保持在剂量1前水平以上至少1年。该疫苗具有良好的耐受性,具有可接受的安全性。临床试验注册。NCT04732871(ClinicalTrials.gov)。
    呼吸道合胞病毒(RSV)是老年人患病和住院的主要原因。GSK开发的老年人RSV疫苗最近获得批准。该疫苗具有良好的耐受性,并在至少1个RSV季节期间在≥60岁的成年人中提供针对RSV疾病的保护。在这项正在进行的研究中,我们正在评估免疫反应的强度和持久性,以及疫苗的安全性,5个国家/地区≥60岁的成年人接种疫苗后3年。这里,我们报告了1剂疫苗接种后1年的中期分析结果.总的来说,1653名参与者接种了疫苗。我们发现疫苗诱导了强烈的免疫反应,这在接种疫苗1个月后很明显,之后下降,但持续了至少1年。研究参与者最常报告注射部位疼痛,肌肉疼痛,疲倦,和头痛作为不良反应,大多为轻度至中度,持续时间短。一个严重的不良反应被认为与疫苗有关。在该研究中观察到的长期免疫应答与在至少1个RSV季节期间提供保护的疫苗一致。
    BACKGROUND: The recently approved AS01E-adjuvanted respiratory syncytial virus (RSV) prefusion F protein-based vaccine for older adults (RSVPreF3 OA) demonstrated high efficacy against RSV-related disease in ≥60-year-olds.
    METHODS: This ongoing phase 3 study in ≥60-year-olds evaluates immune persistence until 3 years after RSVPreF3 OA vaccination. Here, we describe interim results on humoral and cell-mediated immunogenicity, reactogenicity, and safety until 1 year post-dose 1.
    RESULTS: In total, 1653 participants were vaccinated. One month post-dose 1, neutralization titers increased 10.5-fold (RSV-A) and 7.8-fold (RSV-B) vs pre-dose 1. Titers then declined to levels 4.4-fold (RSV-A) and 3.5-fold (RSV-B) above pre-dose 1 at month 6 and remained 3.1-fold (RSV-A) and 2.3-fold (RSV-B) above pre-dose 1 levels after 1 year. RSVPreF3-binding immunoglobulin G levels and CD4+ T-cell frequencies showed similar kinetics. Solicited administration-site and systemic adverse events (mostly mild to moderate and transient) were reported by 62.2% and 49.5% of participants. Serious adverse events were reported by 3.9% of participants within 6 months post-dose 1; 1 case was considered vaccine related.
    CONCLUSIONS: One RSVPreF3 OA dose elicited cell-mediated and RSV-A- and RSV-B-specific humoral immune responses that declined over time but remained above pre-dose 1 levels for at least 1 year. The vaccine was well tolerated with an acceptable safety profile. Clinical Trials Registration. NCT04732871 (ClinicalTrials.gov).
    Respiratory syncytial virus (RSV) is a major cause of illness and hospitalization in older adults. An RSV vaccine for older adults developed by GSK was recently approved. The vaccine was well tolerated and provided protection against RSV disease in adults aged ≥60 years during at least 1 RSV season. In this ongoing study, we are evaluating the magnitude and durability of the immune response, as well as vaccine safety, until 3 years after vaccination of adults aged ≥60 years from 5 countries. Here, we report the results of an interim analysis until 1 year after vaccination with 1 dose. In total, 1653 participants were vaccinated. We found that the vaccine induced a strong immune response that was evident 1 month after vaccination, after which it declined but persisted for at least 1 year. Study participants most often reported pain at the injection site, muscle pain, tiredness, and headache as adverse reactions, which were mostly mild to moderate and of short duration. One serious adverse reaction was considered related to the vaccine. The long-term immune response that was observed in this study is consistent with the vaccine providing protection during at least 1 RSV season.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号