Broad neutralization

广泛的中和
  • 文章类型: Journal Article
    背景:猪繁殖与呼吸综合征病毒(PRRSV)是一种流行的猪病原体,近30年来,这对全球养猪业造成了不利影响。然而,由于病毒引起的免疫抑制和PRRSV的遗传多样性,目前还没有成功开发针对病毒的广泛中和策略。抗病毒肽和纳米抗体以其易于生产和在实践中的功效引起了广泛的关注。在这项研究中,通过将PRRSV特异性非中和纳米抗体与靶向PRRSV蛋白的受体结合域(RBD)的CD163衍生肽组合,开发了四种新的融合蛋白,称为纳米抗体肽缀合物(NPC)。
    结果:使用两个纳米抗体分别针对PRRSVN和nsp9成功构建了四个NPC,分别与来自猪CD163的两种抗病毒肽4H7或8H2重组。所有四种NPC均表现出与PRRSV结合的特异性能力以及以剂量依赖性方式对PRRSV的各种谱系的广泛抑制作用。NPC在Nb组分的辅助下通过CD163表位肽干扰PRRSV蛋白的RBD与PRRSV预附着阶段中的CD163的结合。NPC还在附着后阶段抑制病毒复制,抑制作用取决于NPCs中Nb部分的抗病毒功能,包括在长病毒RNA合成中的干扰,NF-κB和IFN-β激活。此外,预测了NPC-N/nsp9-4H7的中和结构域4H7的aaK31和T32位点与PRRSVGP2a的基序171NLRLTG176之间的相互作用。NPC-N/nsp9-8H2的中和结构域8H2的基序28SSS30也可以形成氢与PRRSVGP3的基序152NAFLP156结合。该研究为PRRSV蛋白的RBD的结构特征和潜在功能意义提供了有价值的见解。最后,如小鼠模型所示,体内鼻内接种12-24小时的NPC维持针对PRRSV的显著中和活性。这些发现激发了NPC作为预防措施的潜力,可以降低宿主人群针对PRRSV等呼吸道感染因子的传播风险。
    结论:本研究的目的是开发基于肽的生物活性化合物以中和各种PRRSV毒株。新的抗病毒NPC(纳米抗体肽缀合物)由靶向病毒蛋白的特异性纳米抗体和用于病毒阻断的中和CD163表位肽组成,并提供显著的抗病毒活性。该研究将极大地促进针对PRRSV的抗病毒药物的研发,并启发针对其他病毒性疾病的新策略。
    BACKGROUND: Porcine reproductive and respiratory syndrome virus (PRRSV) is a prevalent swine pathogen, which has caused adverse impact on the global swine industry for almost 30 years. However, due to the immune suppression caused by the virus and the genetic diversity in PRRSV, no virus-targeting broad neutralizing strategy has been successfully developed yet. Antiviral peptide and nanobody have attracted extensive attention with the ease in production and the efficacy in practice. In this study, four new fusion proteins named nanobody peptide conjugates (NPCs) were developed by combining PRRSV specific non-neutralizing nanobodies with CD163-derived peptides targeting the receptor binding domain (RBD) of PRRSV proteins.
    RESULTS: Four NPCs were successfully constructed using two nanobodies against PRRSV N and nsp9 individually, recombining with two antiviral peptides 4H7 or 8H2 from porcine CD163 respectively. All four NPCs demonstrated specific capability of binding to PRRSV and broad inhibitory effect against various lineages of PRRSV in a dose-dependent manner. NPCs interfere with the binding of the RBD of PRRSV proteins to CD163 in the PRRSV pre-attachment stage by CD163 epitope peptides in the assistance of Nb components. NPCs also suppress viral replication during the stage of post-attachment, and the inhibitory effects depend on the antiviral functions of Nb parts in NPCs, including the interference in long viral RNA synthesis, NF-κB and IFN-β activation. Moreover, an interaction was predicted between aa K31 and T32 sites of neutralizing domain 4H7 of NPC-N/nsp9-4H7 and the motif 171NLRLTG176 of PRRSV GP2a. The motif 28SSS30 of neutralizing domain 8H2 of NPC-N/nsp9-8H2 could also form hydrogens to bind with the motif 152NAFLP156 of PRRSV GP3. The study provides valuable insights into the structural characteristics and potential functional implications of the RBD of PRRSV proteins. Finally, as indicated in a mouse model, NPC intranasally inoculated in vivo for 12-24 h sustains the significant neutralizing activity against PRRSV. These findings inspire the potential of NPC as a preventive measure to reduce the transmission risk in the host population against respiratory infectious agents like PRRSV.
    CONCLUSIONS: The aim of the current study was to develop a peptide based bioactive compound to neutralize various PRRSV strains. The new antiviral NPC (nanobody peptide conjugate) consists of a specific nanobody targeting the viral protein and a neutralizing CD163 epitope peptide for virus blocking and provides significant antiviral activity. The study will greatly promote the antiviral drug R&D against PRRSV and enlighten a new strategy against other viral 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
    设计新的CAR靶向乙型肝炎病毒(HBV),我们从已解决感染的患者的单个B细胞中分离出识别HBV包膜蛋白的人单克隆抗体.HBV特异性记忆B细胞通过孵育外周血单核细胞与生物素化乙型肝炎表面抗原(HBsAg)分离,然后是基于单细胞流式细胞术的活细胞分选,CD19+IgG+HBsAg+细胞。来自单个记忆B细胞的免疫球蛋白基因的扩增和测序鉴定了可变重链和轻链序列。将相应的免疫球蛋白链克隆到IgG1表达载体中并在哺乳动物细胞中表达。发现两种命名为4D06和4D08的抗体对HBsAg具有高度特异性,识别构象和线性表位,分别,并显示对所有主要HBV基因型的广泛反应性和中和能力。将4D06和4D08可变链片段克隆到具有CD28和CD3G胞内信号传导结构域的第二代CAR格式中。当在原代人T细胞上表达时,新的CAR构建体显示高功能亲合力。CAR移植的T细胞被证明是多功能的细胞因子分泌和杀死HBV阳性靶细胞。有趣的是,识别线性而非构象表位的4D08-CAR的背景激活始终很低.在慢性HBV感染的临床前模型,用4D06和4D08CAR移植的鼠T细胞显示出血清转氨酶的瞬时增加表明的靶活性,和较低数量的HBV阳性肝细胞在小鼠治疗。这项研究证明了一种有效且快速的方法,可以从少量供体细胞中鉴定病原体特异性单克隆人抗体,用于后续生成新CAR。
    To design new CARs targeting hepatitis B virus (HBV), we isolated human monoclonal antibodies recognizing the HBV envelope proteins from single B cells of a patient with a resolved infection. HBV-specific memory B cells were isolated by incubating peripheral blood mononuclear cells with biotinylated hepatitis B surface antigen (HBsAg), followed by single-cell flow cytometry-based sorting of live, CD19+ IgG+ HBsAg+ cells. Amplification and sequencing of immunoglobulin genes from single memory B cells identified variable heavy and light chain sequences. Corresponding immunoglobulin chains were cloned into IgG1 expression vectors and expressed in mammalian cells. Two antibodies named 4D06 and 4D08 were found to be highly specific for HBsAg, recognized a conformational and a linear epitope, respectively, and showed broad reactivity and neutralization capacity against all major HBV genotypes. 4D06 and 4D08 variable chain fragments were cloned into a 2nd generation CAR format with CD28 and CD3zeta intracellular signaling domains. The new CAR constructs displayed a high functional avidity when expressed on primary human T cells. CAR-grafted T cells proved to be polyfunctional regarding cytokine secretion and killed HBV-positive target cells. Interestingly, background activation of the 4D08-CAR recognizing a linear instead of a conformational epitope was consistently low. In a preclinical model of chronic HBV infection, murine T cells grafted with the 4D06 and the 4D08 CAR showed on target activity indicated by a transient increase in serum transaminases, and a lower number of HBV-positive hepatocytes in the mice treated. This study demonstrates an efficient and fast approach to identifying pathogen-specific monoclonal human antibodies from small donor cell numbers for the subsequent generation of new CARs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    尽管全球疫苗接种计划在减缓COVID-19的传播方面取得了成功,但由于能够逃避先前免疫力的新SARS-CoV-2菌株的出现,这些努力受到了阻碍。SARS-CoV-2的突变和进化产生了对疫苗开发的持续努力的需求。SARS-CoV-2刺突蛋白一直是COVID-19疫苗开发的主要目标,但它也是与宿主易感性和免疫逃避直接相关的突变的热点。我们预测新出现的突变体和选择保守表位的能力对于开发广泛中和疗法或通用疫苗至关重要。在这篇文章中,我们回顾了对COVID-19疫苗的免疫反应的一般范例,突出显示刺突蛋白的免疫表位,这些表位可能与疫苗接种引起的保护性免疫有关。具体来说,我们通过toll样受体(TLRs)分析了与免疫激活和功能相关的SARS-CoV-2Spike蛋白的结构和进化特征,B细胞,和T细胞。我们旨在提供对Spike蛋白免疫表位的全面分析,从而有助于开发广泛中和或通用疫苗接种的新策略。
    Despite the success of global vaccination programs in slowing the spread of COVID-19, these efforts have been hindered by the emergence of new SARS-CoV-2 strains capable of evading prior immunity. The mutation and evolution of SARS-CoV-2 have created a demand for persistent efforts in vaccine development. SARS-CoV-2 Spike protein has been the primary target for COVID-19 vaccine development, but it is also the hotspot of mutations directly involved in host susceptibility and immune evasion. Our ability to predict emerging mutants and select conserved epitopes is critical for the development of a broadly neutralizing therapy or a universal vaccine. In this article, we review the general paradigm of immune responses to COVID-19 vaccines, highlighting the immunological epitopes of Spike protein that are likely associated with eliciting protective immunity resulting from vaccination. Specifically, we analyze the structural and evolutionary characteristics of the SARS-CoV-2 Spike protein related to immune activation and function via the toll-like receptors (TLRs), B cells, and T cells. We aim to provide a comprehensive analysis of immune epitopes of Spike protein, thereby contributing to the development of new strategies for broad neutralization or universal vaccination.
    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
    SARS-CoV-2变种的不断涌现,特别是新流通的OmicronXBB亚变体,导致抗体的中和效力和宽度显著降低。在这项研究中,我们报道了SARS-CoV-2人中和抗体,1G11,它能有效和广泛地中和各种变体,包括Omicron亚变体BA.4/5和BF.7,但被最近出现的BQ.1.1和XBB所回避。与BA4/5尖峰三聚体复合的1G11的低温电子显微镜结构分析显示,1G11,3类nAb,识别与S309和LY-CoV1404相似的表位。在结构上,发现BQ.1.1和XBB亚变体中的K444T和V445P突变破坏1G11和刺突蛋白之间的界面,导致抗体逃避。1G11被进一步证明通过多种机制介导中和,包括受体结合阻断,穗间交联,Fc介导的ADCC和ADCP。总的来说,这些发现为更好地理解中和抗体逃避提供了见解,并强调了通过对现有抗体进行基于结构的修饰进行广泛中和的潜力。重要性持续的COVID-19大流行的特征是出现了新的SARS-CoV-2变体,包括高度传播的OmicronXBB亚谱系,已显示对中和抗体(nAbs)的显著抗性。这种耐药性导致疫苗效力下降,因此导致突破性感染和再感染,不断威胁公众健康。迄今为止,几乎所有可用的治疗nAbs,包括那些根据紧急使用授权nAbs授权的,以前在临床上对早期菌株有用的,最近发现对新出现的变体无效。在这项研究中,我们提供了一个全面的结构基础,包括本研究中的1G11和注意到的LY-CoV1404,被新出现的SARS-CoV-2变体所回避。
    OBJECTIVE: The ongoing COVID-19 pandemic has been characterized by the emergence of new SARS-CoV-2 variants including the highly transmissible Omicron XBB sublineages, which have shown significant resistance to neutralizing antibodies (nAbs). This resistance has led to decreased vaccine effectiveness and therefore result in breakthrough infections and reinfections, which continuously threaten public health. To date, almost all available therapeutic nAbs, including those authorized under Emergency Use Authorization nAbs that were previously clinically useful against early strains, have recently been found to be ineffective against newly emerging variants. In this study, we provide a comprehensive structural basis about how the Class 3 nAbs, including 1G11 in this study and noted LY-CoV1404, are evaded by the newly emerged SARS-CoV-2 variants.
    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
    SARS-CoV-2,一种呼吸道病毒,在过去三年中,对全球卫生和经济产生了巨大影响。基于抗体的治疗最初是成功的,但后来由于病毒刺突蛋白中突变的积累而失败。因此,使抗体能够抵抗病毒逃逸的策略具有重要意义。这里,我们设计了一种分泌型免疫球蛋白A(SIgA)格式的双特异性SARS-CoV-2中和纳米抗体,命名为S2-3-IgA2m2,对SARS-CoV-1,SARS-CoV-2及其相关变体(VOCs)(包括XBB和BQ.1.1)显示出广泛而有效的中和作用。在中和XBB中,S2-3-IgA2m2比其亲本IgG对应物更有效~1800倍。当通过鼻递送施用时,S2-3-IgA2m2在小鼠肺中稳定至少三天。在被BA.5感染的仓鼠中,以1mg/kg的三个鼻内剂量的S2-3-IgA2m2显著降低了病毒RNA载量并完全消除了气管和肺中的感染性颗粒。值得注意的是,即使在1毫克/千克的单剂量,通过鼻内途径预防性施用的S2-3-IgA2m2显著降低了气道病毒RNA载量和感染性颗粒。本研究为抗击SARS-CoV-2提供了有效的武器,提出了克服病毒逃逸的新策略,并为快速反应未来“SARS-CoV-3”的潜在爆发奠定了战略储备。
    SARS-CoV-2, a type of respiratory virus, has exerted a great impact on global health and economy over the past three years. Antibody-based therapy was initially successful but later failed due to the accumulation of mutations in the spike protein of the virus. Strategies that enable antibodies to resist virus escape are therefore of great significance. Here, we engineer a bispecific SARS-CoV-2 neutralizing nanobody in secretory Immunoglobulin A (SIgA) format, named S2-3-IgA2m2, which shows broad and potent neutralization against SARS-CoV-1, SARS-CoV-2 and its variants of concern (VOCs) including XBB and BQ.1.1. S2-3-IgA2m2 is ∼1800-fold more potent than its parental IgG counterpart in neutralizing XBB. S2-3-IgA2m2 is stable in mouse lungs at least for three days when administrated by nasal delivery. In hamsters infected with BA.5, three intranasal doses of S2-3-IgA2m2 at 1 mg/kg significantly reduce viral RNA loads and completely eliminate infectious particles in the trachea and lungs. Notably, even at single dose of 1 mg/kg, S2-3-IgA2m2 prophylactically administered through the intranasal route drastically reduces airway viral RNA loads and infectious particles. This study provides an effective weapon combating SARS-CoV-2, proposes a new strategy overcoming the virus escape, and lays strategic reserves for rapid response to potential future outbreaks of \"SARS-CoV-3\".
    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)和人偏肺病毒(hMPV)感染造成了巨大的健康负担。使用融合前构象融合(F)蛋白,我们从人类供体中分离出一组抗F抗体。一种抗体(RSV-199)通过识别抗原位点III,可有效交叉中和8种RSV和hMPV毒株,在RSV和hMPVF中部分保守。接下来,我们确定了与RSVF三聚体结合的RSV-199的低温电子显微镜(cryo-EM)结构,hMPVF单体,和hMPVF的意外二聚体形式。这些结构揭示了RSV-199如何通过抗体重链可变区的保守相互作用与RSV和hMPVF蛋白结合,以及重链互补决定区3(HCDR3)内的变异性如何在位点III定向抗体的F蛋白界面处容纳。此外,RSV-199对棉鼠的RSVA和B株以及hMPV具有增强的保护作用。这些发现突出了RSV-199的广泛中和和治疗潜力的机制。
    Respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) infections pose a significant health burden. Using pre-fusion conformation fusion (F) proteins, we isolated a panel of anti-F antibodies from a human donor. One antibody (RSV-199) potently cross-neutralized 8 RSV and hMPV strains by recognizing antigenic site III, which is partially conserved in RSV and hMPV F. Next, we determined the cryoelectron microscopy (cryo-EM) structures of RSV-199 bound to RSV F trimers, hMPV F monomers, and an unexpected dimeric form of hMPV F. These structures revealed how RSV-199 engages both RSV and hMPV F proteins through conserved interactions of the antibody heavy-chain variable region and how variability within heavy-chain complementarity-determining region 3 (HCDR3) can be accommodated at the F protein interface in site-III-directed antibodies. Furthermore, RSV-199 offered enhanced protection against RSV A and B strains and hMPV in cotton rats. These findings highlight the mechanisms of broad neutralization and therapeutic potential of RSV-199.
    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
    2019年冠状病毒病(COVID-19)大流行仍在继续。严重急性呼吸系统综合症冠状病毒2(SARS-CoV-2)变种(VOCs)在全球范围内流行,使其对现有的疫苗和抗病毒药物具有抗性。因此,评估基于变异的广谱疫苗以优化免疫反应并提供广泛的保护性非常重要。在这项研究中,我们在使用CHO细胞的GMP级车间中基于Beta变体表达了刺突三聚体蛋白(S-TM)。用S-TM蛋白联合氢氧化铝(Al)和CpG寡核苷酸(CpG)佐剂免疫小鼠两次以评价其安全性和功效。用S-TM+Al+CpG免疫的BALB/c诱导针对武汉-Hu-1株的高中和抗体滴度(野生型,WT),Beta和Delta变体,甚至是Omicron变体。此外,与S-TM+Al组相比,S-TM+Al+CpG组有效诱导小鼠更强的Th1偏倚细胞免疫应答。此外,第二次免疫后,H11-K18hACE2小鼠受到很好的保护,免受SARS-CoV-2Beta毒株的攻击,100%的存活率。肺部病毒载量和病理病变显著降低,在小鼠脑组织中未检测到病毒。我们的候选疫苗对目前的SARS-CoV-2挥发性有机化合物是实用和有效的,这将支持其潜在的序贯免疫和初次免疫的进一步临床开发。重要性严重急性呼吸综合征冠状病毒2(SARS-CoV-2)的适应性突变的不断出现继续挑战现有疫苗和药物的使用和开发。目前正在评估能够诱导针对SARS-CoV-2变体的更高和更广泛的保护性免疫应答的基于变体的疫苗的价值。本文表明,基于Beta变体的重组预融合刺突蛋白具有高度免疫原性,可以在小鼠中诱导更强的Th1偏向性细胞免疫应答,并且可以有效地抵抗SARS-CoV-2Beta变体的攻击。重要的是,这种基于Beta的SARS-CoV-2疫苗还可以提供强大的体液免疫反应,具有针对野生型和不同关注变体(VOCs)的有效广泛的中和能力:Delta,和OmicronBA.1变体。迄今为止,这里描述的疫苗已经以中试规模生产(200L),和发展,填充过程,毒理学安全性评估也已经完成,这提供了对新出现的SARS-CoV-2变体和疫苗开发的及时反应。
    The coronavirus disease 2019 (COVID-19) pandemic is still ongoing. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs) are circulating worldwide, making it resistant to existing vaccines and antiviral drugs. Therefore, the evaluation of variant-based expanded spectrum vaccines to optimize the immune response and provide broad protectiveness is very important. In this study, we expressed spike trimer protein (S-TM) based on the Beta variant in a GMP-grade workshop using CHO cells. Mice were immunized twice with S-TM protein combined with aluminum hydroxide (Al) and CpG Oligonucleotides (CpG) adjuvant to evaluate its safety and efficacy. BALB/c immunized with S-TM + Al + CpG induced high neutralizing antibody titers against the Wuhan-Hu-1 strain (wild-type, WT), the Beta and Delta variants, and even the Omicron variant. In addition, compared with the S-TM + Al group, the S-TM + Al + CpG group effectively induced a stronger Th1-biased cell immune response in mice. Furthermore, after the second immunization, H11-K18 hACE2 mice were well protected from challenge with the SARS-CoV-2 Beta strain, with a 100% survival rate. The virus load and pathological lesions in the lungs were significantly reduced, and no virus was detected in mouse brain tissue. Our vaccine candidate is practical and effective for current SARS-CoV-2 VOCs, which will support its further clinical development for potential sequential immune and primary immunization. IMPORTANCE Continuous emergence of adaptive mutations of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to challenge the use and development of existing vaccines and drugs. The value of variant-based vaccines that are capable of inducing a higher and broader protection immune response against SARS-CoV-2 variants is currently being evaluated. This article shows that a recombinant prefusion spike protein based on a Beta variant was highly immunogenic and could induced a stronger Th1-biased cell immune response in mice and was effectively protective against challenge with the SARS-CoV-2 Beta variant. Importantly, this Beta-based SARS-CoV-2 vaccine could also offer a robust humoral immune response with effectively broad neutralization ability against the wild type and different variants of concern (VOCs): the Beta, Delta, and Omicron BA.1 variants. To date, the vaccine described here has been produced in a pilot scale (200L), and the development, filling process, and toxicological safety evaluation have also been completed, which provides a timely response to the emerging SARS-CoV-2 variants and 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
    多价疫苗结合了来自系统发育差异变体的关键突变,可能是防御现有和未来SARS-CoV-2变体的有效方法。在这项研究中,我们开发了一种四价COVID-19疫苗SCTV01E,基于SARS-CoV-2变种α的三聚体刺突蛋白,Beta,Delta,和OmicronBA.1,以及基于角鲨烯的水包油佐剂SCT-VA02B。在初始BALB/c和C57BL/6J小鼠的免疫原性研究中,SCTV01E表现出最有利的免疫原性特征,以诱导针对前Omicron变体的平衡和广谱中和效力(D614G,阿尔法,Beta,和Delta)和新出现的Omicron亚变体(BA.1,BA.1.1,BA.2,BA.3和BA.4/5)。在先前用D614G单价疫苗免疫的C57BL/6J小鼠中的加强研究表明,SCTV01E对Omicron亚变体具有优异的中和能力,与D614G加强方案相比。此外,SCTV01E疫苗接种引起对SARS-CoV-2祖先菌株和Omicron刺突肽的幼稚和中枢记忆T细胞应答。一起,我们的综合免疫原性评估结果表明,SCTV01E可能成为重要的COVID-19疫苗平台,以对抗由高度免疫规避的BA4/5变体引起的激增感染.SCTV01E目前正在对头免疫原性比较3期临床研究中使用灭活疫苗和mRNA疫苗(NCT05323461)进行研究。
    Multivalent vaccines combining crucial mutations from phylogenetically divergent variants could be an effective approach to defend against existing and future SARS-CoV-2 variants. In this study, we developed a tetravalent COVID-19 vaccine SCTV01E, based on the trimeric Spike protein of SARS-CoV-2 variants Alpha, Beta, Delta, and Omicron BA.1, with a squalene-based oil-in-water adjuvant SCT-VA02B. In the immunogenicity studies in naïve BALB/c and C57BL/6J mice, SCTV01E exhibited the most favorable immunogenic characteristics to induce balanced and broad-spectrum neutralizing potencies against pre-Omicron variants (D614G, Alpha, Beta, and Delta) and newly emerging Omicron subvariants (BA.1, BA.1.1, BA.2, BA.3, and BA.4/5). Booster studies in C57BL/6J mice previously immunized with D614G monovalent vaccine demonstrated superior neutralizing capacities of SCTV01E against Omicron subvariants, compared with the D614G booster regimen. Furthermore, SCTV01E vaccination elicited naïve and central memory T cell responses to SARS-CoV-2 ancestral strain and Omicron spike peptides. Together, our comprehensive immunogenicity evaluation results indicate that SCTV01E could become an important COVID-19 vaccine platform to combat surging infections caused by the highly immune evasive BA.4/5 variants. SCTV01E is currently being studied in a head-to-head immunogenicity comparison phase 3 clinical study with inactivated and mRNA vaccines (NCT05323461).
    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
    自2019年12月以来,严重急性呼吸道综合症冠状病毒2(SARS-CoV-2)已成为一种新兴的人类病毒,世界人口易患2019年冠状病毒病(COVID-19)。SARS-CoV-2比以前的冠状病毒具有更高的传播能力,由核糖核酸(RNA)病毒性质相关的高突变率,导致SARS-CoV-2变体在全球传播时出现。中和抗体被鉴定为针对COVID-19的即时和直接作用治疗剂。单结构域抗体(sdAb),作为具有非复杂结构和内在稳定性的小生物分子,可以获得与常规抗体相当的抗原结合能力,作为一种有吸引力的中和溶液。SARS-CoV-2刺突蛋白附着于肺上皮细胞上的人血管紧张素转换酶2(ACE2)受体以引发病毒感染,作为潜在的治疗靶点。sdAb已经显示出广泛的中和对SARS-CoV-2的各种突变,有效阻止和预防感染,同时有效阻止突变逃逸。此外,sdAb可以发展成为针对COVID-19的多价抗体或吸入性生物治疗剂。
    With severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as an emergent human virus since December 2019, the world population is susceptible to coronavirus disease 2019 (COVID-19). SARS-CoV-2 has higher transmissibility than the previous coronaviruses, associated by the ribonucleic acid (RNA) virus nature with high mutation rate, caused SARS-CoV-2 variants to arise while circulating worldwide. Neutralizing antibodies are identified as immediate and direct-acting therapeutic against COVID-19. Single-domain antibodies (sdAbs), as small biomolecules with non-complex structure and intrinsic stability, can acquire antigen-binding capabilities comparable to conventional antibodies, which serve as an attractive neutralizing solution. SARS-CoV-2 spike protein attaches to human angiotensin-converting enzyme 2 (ACE2) receptor on lung epithelial cells to initiate viral infection, serves as potential therapeutic target. sdAbs have shown broad neutralization towards SARS-CoV-2 with various mutations, effectively stop and prevent infection while efficiently block mutational escape. In addition, sdAbs can be developed into multivalent antibodies or inhaled biotherapeutics against COVID-19.
    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
    SARS-CoV-2疫苗和治疗性抗体的有效性受到病毒变体的持续出现和抗体从循环到呼吸道病毒感染部位的限制扩散的限制。这里,我们报告了在广泛中和抗体识别的Omicron变体受体结合域上鉴定出两个高度保守的区域.此外,我们产生了双特异性单结构域抗体,该抗体能够同时和协同地结合单个Omicron变体受体结合结构域上的这两个区域,如cryo-EM结构所揭示的.我们证明了这种双特异性抗体可以通过吸入施用有效地递送到肺,并且在SARS-CoV-2感染的小鼠模型中表现出精致的中和宽度和治疗功效。重要的是,这项研究还破译了刺突三聚体界面内一个不常见且高度保守的隐匿性表位,这可能对设计具有广泛保护性的SARS-CoV-2疫苗和疗法有影响.
    The effectiveness of SARS-CoV-2 vaccines and therapeutic antibodies have been limited by the continuous emergence of viral variants and by the restricted diffusion of antibodies from circulation into the sites of respiratory virus infection. Here, we report the identification of two highly conserved regions on the Omicron variant receptor-binding domain recognized by broadly neutralizing antibodies. Furthermore, we generated a bispecific single-domain antibody that was able to simultaneously and synergistically bind these two regions on a single Omicron variant receptor-binding domain as revealed by cryo-EM structures. We demonstrated that this bispecific antibody can be effectively delivered to lung via inhalation administration and exhibits exquisite neutralization breadth and therapeutic efficacy in mouse models of SARS-CoV-2 infections. Importantly, this study also deciphered an uncommon and highly conserved cryptic epitope within the spike trimeric interface that may have implications for the design of broadly protective SARS-CoV-2 vaccines and therapeutics.
    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