neutralizing monoclonal antibodies

中和单克隆抗体
  • 文章类型: Journal Article
    细胞培养物中抗体中和的效力已被用作选择针对严重急性呼吸综合征冠状病毒2(SARS-CoV-2)的抗体进行临床开发的关键标准。由于其他方面也可能影响体内保护程度,我们比较了两种针对SARS-CoV-2受体结合域(RBD)不同表位的中和单克隆抗体(TRES6和4C12)在恒河猴预防性治疗中的疗效.用TRES6治疗的所有四只动物在用AlphaSARS-CoV-2变体鼻口咽攻击后2天上呼吸道的病毒载量降低。挑战后2天开始,赋予对TRES6抗性的突变在两只恒河猴中占主导地位,两种动物都无法保持减少的病毒载量。与其在攻击当天较低的血清中和滴度一致,与TRES6相比,使用4C12进行预防在第2天抑制病毒载量的效率较低.然而,挑战一周后,4C12治疗动物下呼吸道的平均病毒载量低于TRES6组,并且在鼻或咽拭子的病毒分离株中未检测到赋予4C12耐药性的突变.因此,抗性的遗传障碍似乎是抗SARS-CoV-2单克隆抗体预防效果的关键参数。此外,呼吸道分泌物中的抗体浓度与血清中的抗体浓度的比较显示,与它们在上呼吸道分泌物中的出现相比,4C12抗体在呼吸道分泌物中的分布减少,并且抗体在支气管肺泡灌洗液中的出现延迟。IMPORTANCE单克隆抗体是预防和治疗急性病毒感染的有力工具。因此,它们是首批获得许可用于治疗严重急性呼吸道综合征冠状病毒2(SARS-CoV-2)的治疗剂之一.通常,选择用于临床开发的抗体的主要标准是它们在细胞培养物中的中和效力。通过比较针对SARS-CoV-2的Spike蛋白的两种抗体,我们现在观察到,在细胞培养物中更有效地中和SARS-CoV-2的抗体在较小程度上抑制了受攻击的恒河猴的病毒载量。攻击病毒的突变体异常迅速出现,失去了对抗体的敏感性,被确定为抗体在恒河猴中功效降低的主要原因。因此,病毒对抗体抗性的遗传障碍也影响其功效。
    The potency of antibody neutralization in cell culture has been used as the key criterion for selection of antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) for clinical development. As other aspects may also influence the degree of protection in vivo, we compared the efficacy of two neutralizing monoclonal antibodies (TRES6 and 4C12) targeting different epitopes of the receptor binding domain (RBD) of SARS-CoV-2 in a prophylactic setting in rhesus monkeys. All four animals treated with TRES6 had reduced viral loads in the upper respiratory tract 2 days after naso-oropharyngeal challenge with the Alpha SARS-CoV-2 variant. Starting 2 days after challenge, mutations conferring resistance to TRES6 were dominant in two of the rhesus monkeys, with both animals failing to maintain reduced viral loads. Consistent with its lower serum neutralization titer at the day of challenge, prophylaxis with 4C12 tended to suppress viral load at day 2 less efficiently than TRES6. However, a week after challenge, mean viral loads in the lower respiratory tract in 4C12-treated animals were lower than in the TRES6 group and no mutations conferring resistance to 4C12 could be detected in viral isolates from nasal or throat swabs. Thus, genetic barrier to resistance seems to be a critical parameter for the efficacy of prophylaxis with monoclonal antibodies against SARS-CoV-2. Furthermore, comparison of antibody concentrations in respiratory secretions to those in serum shows reduced distribution of the 4C12 antibody into respiratory secretions and a delay in the appearance of antibodies in bronchoalveolar lavage fluid compared to their appearance in secretions of the upper respiratory tract.IMPORTANCEMonoclonal antibodies are a powerful tool for the prophylaxis and treatment of acute viral infections. Hence, they were one of the first therapeutic agents licensed for the treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Oftentimes, the main criterion for the selection of antibodies for clinical development is their potency of neutralization in cell culture. By comparing two antibodies targeting the Spike protein of SARS-CoV-2, we now observed that the antibody that neutralized SARS-CoV-2 more efficiently in cell culture suppressed viral load in challenged rhesus monkeys to a lesser extent. Extraordinary rapid emergence of mutants of the challenge virus, which had lost their sensitivity to the antibody, was identified as the major reason for the reduced efficacy of the antibody in rhesus monkeys. Therefore, the viral genetic barrier to resistance to antibodies also affects their efficacy.
    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
    了解SARS-CoV-2(导致COVID-19的病毒)的抗体反应对于理解疾病进展以及疫苗和治疗开发的重要性至关重要。高度传染性变异的出现对体液免疫构成了重大挑战,强调掌握特异性抗体的复杂性的必要性。这篇综述强调了抗体在塑造免疫反应中的关键作用及其对诊断的意义。预防,和治疗SARS-CoV-2感染。它深入研究了SARS-CoV-2抗体反应的动力学和特征,并探讨了当前基于抗体的诊断方法,讨论他们的长处,临床效用,和限制。此外,我们强调了SARS-CoV-2特异性抗体的治疗潜力,讨论各种基于抗体的疗法,如单克隆抗体,多克隆抗体,抗细胞因子,恢复期血浆,和基于高免疫球蛋白的疗法。此外,我们提供了对SARS-CoV-2疫苗的抗体反应的见解,强调中和抗体的重要性,以赋予对SARS-CoV-2的免疫力,以及新兴的关注变体(VOC)和循环Omicron亚变体。我们还强调了该领域的挑战,例如SARS-CoV-2抗体的抗体依赖性增强(ADE)的风险,并阐明了与原始抗原性蛋白酶(OAS)效应和长期COVID相关的挑战。总的来说,这篇评论旨在提供有价值的见解,这对推进敏感的诊断工具至关重要,确定有效的基于抗体的疗法,并开发有效的疫苗,以应对全球范围内不断发展的SARS-CoV-2变种的威胁。
    Understanding the antibody response to SARS-CoV-2, the virus responsible for COVID-19, is crucial to comprehending disease progression and the significance of vaccine and therapeutic development. The emergence of highly contagious variants poses a significant challenge to humoral immunity, underscoring the necessity of grasping the intricacies of specific antibodies. This review emphasizes the pivotal role of antibodies in shaping immune responses and their implications for diagnosing, preventing, and treating SARS-CoV-2 infection. It delves into the kinetics and characteristics of the antibody response to SARS-CoV-2 and explores current antibody-based diagnostics, discussing their strengths, clinical utility, and limitations. Furthermore, we underscore the therapeutic potential of SARS-CoV-2-specific antibodies, discussing various antibody-based therapies such as monoclonal antibodies, polyclonal antibodies, anti-cytokines, convalescent plasma, and hyperimmunoglobulin-based therapies. Moreover, we offer insights into antibody responses to SARS-CoV-2 vaccines, emphasizing the significance of neutralizing antibodies in order to confer immunity to SARS-CoV-2, along with emerging variants of concern (VOCs) and circulating Omicron subvariants. We also highlight challenges in the field, such as the risks of antibody-dependent enhancement (ADE) for SARS-CoV-2 antibodies, and shed light on the challenges associated with the original antigenic sin (OAS) effect and long COVID. Overall, this review intends to provide valuable insights, which are crucial to advancing sensitive diagnostic tools, identifying efficient antibody-based therapeutics, and developing effective vaccines to combat the evolving threat of SARS-CoV-2 variants on a global scale.
    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
    血清转化为严重急性呼吸道综合征冠状病毒2(SARS-CoV-2)感染的病毒学决定因素是在对2019年冠状病毒疾病高危人群(COVID-19)进行前瞻性研究的疫苗和未感染个体的事后分析中定义的。
    这项使用casirivimab和imdevimab的COVID-19预防试验(NCT04452318)于2020年7月至2021年2月进行,在广泛的疫苗供应之前。每周对未感染(SARS-CoV-2定量逆转录聚合酶链反应[RT-qPCR]阴性)和血清阴性的安慰剂治疗参与者进行28天(疗效评估期[EAP])的COVID-19症状和SARS-CoV-2感染通过RT-qPCR对鼻咽拭子样本进行RT-qPCR和抗核衣壳免疫球蛋白(Ig)G的血清状态评估。基于回归的建模,包括因果调解分析,估计病毒载量对血清转化的影响。
    在未感染和血清阴性的157/1069(14.7%)中(对于抗标IgG,抗刺药IgA,和抗核衣壳IgG)在EAP期间感染的参与者,105(65%)血清转化。血清转化者的平均(SD)最大病毒载量为7.23(1.68)log10拷贝/mL与4.8(2.2)log10拷贝/mL在那些保持血清阴性的人中;~6.0log10拷贝/mL的病毒载量更好地预测血清转化。在有症状的参与者中,最大病毒载量的平均值为7.11log10拷贝/mL,在无症状的参与者中为5.58log10拷贝/mL。血清转化和血清阴性参与者的平均可检测病毒载量持续时间更长:3.24vs1.63周。
    最大SARS-CoV-2病毒载量是血清转换和有症状的COVID-19的主要驱动因素,高病毒载量(约6.0log10拷贝/mL)可以更好地预测血清转换。血清学低估了感染率,发病率,和SARS-CoV-2感染的患病率。
    UNASSIGNED: Virologic determinants of seroconversion to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection were defined in a post hoc analysis of prospectively studied vaccine- and infection-naïve individuals at high risk for coronavirus disease 2019 (COVID-19).
    UNASSIGNED: This phase 3 COVID-19 prevention trial (NCT04452318) with casirivimab and imdevimab was conducted in July 2020-February 2021, before widespread vaccine availability. Placebo-treated participants who were uninfected (SARS-CoV-2 quantitative reverse transcription polymerase chain reaction [RT-qPCR] negative) and seronegative were assessed weekly for 28 days (efficacy assessment period [EAP]) for COVID-19 symptoms and SARS-CoV-2 infection by RT-qPCR of nasopharyngeal swab samples and for serostatus by antinucleocapsid immunoglobulin (Ig) G. Regression-based modeling, including causal mediation analysis, estimated the effects of viral load on seroconversion.
    UNASSIGNED: Of 157/1069 (14.7%) uninfected and seronegative (for antispike IgG, antispike IgA, and antinucleocapsid IgG) participants who became infected during the EAP, 105 (65%) seroconverted. The mean (SD) maximum viral load of seroconverters was 7.23 (1.68) log10 copies/mL vs 4.8 (2.2) log10 copies/mL in those who remained seronegative; viral loads of ∼6.0 log10 copies/mL better predicted seroconversion. The mean of the maximum viral load was 7.11 log10 copies/mL in symptomatic participants vs 5.58 log10 copies/mL in asymptomatic participants. The mean duration of detectable viral load was longer in seroconverted vs seronegative participants: 3.24 vs 1.63 weeks.
    UNASSIGNED: Maximum SARS-CoV-2 viral load is a major driver of seroconversion and symptomatic COVID-19, with high viral loads (∼6.0 log10 copies/mL) better predicting seroconversion. Serology underestimates infection rates, incidence, and prevalence of SARS-CoV-2 infection.
    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
    由严重急性呼吸道综合症冠状病毒2及其关注变种(VOCs)引起的2019年冠状病毒病(COVID-19)的流行已经持续了3年多。包含恢复期血浆的抗体疗法,高免疫球蛋白,和中和单克隆抗体(mAb)应用于被动免疫治疗产生了积极的结果,并在早期COVID-19治疗中发挥了关键作用。在这次审查中,发展道路,作用机制,临床研究结果,挑战,以及与使用COVID-19恢复期血浆相关的安全性,高免疫球蛋白,和单克隆抗体进行了总结。此外,评估了应用抗VOCs抗体治疗的前景,提供应对新传染病暴发的应对策略的见解。
    The epidemic of corona virus disease 2019 (COVID-19) caused by severe acute respiratory syndrome Coronavirus 2 and its variants of concern (VOCs) has been ongoing for over 3 years. Antibody therapies encompassing convalescent plasma, hyperimmunoglobulin, and neutralizing monoclonal antibodies (mAbs) applied in passive immunotherapy have yielded positive outcomes and played a crucial role in the early COVID-19 treatment. In this review, the development path, action mechanism, clinical research results, challenges, and safety profile associated with the use of COVID-19 convalescent plasma, hyperimmunoglobulin, and mAbs were summarized. In addition, the prospects of applying antibody therapy against VOCs was assessed, offering insights into the coping strategies for facing new infectious disease outbreaks.
    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
    由金黄色葡萄球菌引起的脓毒性休克患者的死亡率超过50%,尽管有适当的抗生素治疗。我们的目标是建立金黄色葡萄球菌败血性休克的兔模型,并确定新型免疫疗法是否可以预防或阻止其自然疾病进展。
    麻醉的兔子用肺保护性小潮气量通气,先进的血液动力学监测仪器,并通过金黄色葡萄球菌静脉攻击后的超声心动图和脓毒症相关生物标志物表征急性心肌功能障碍的纵向变化。为了证明这种高动力感染性休克模型在临床前药物开发中的潜在实用性,家兔随机接受抗Hla/Luk/ClfA单克隆抗体组合预防,该组合可以中和α-溶血素(Hla),双组分成孔杀白细胞素(Luk),包括Panton-Valentine杀白细胞素,杀白细胞素ED,和γ-溶血素,和结块因子A(ClfA),或不相关的同种型匹配的对照IgG(c-IgG),然后用金黄色葡萄球菌挑战。
    受到金黄色葡萄球菌挑战的兔子,但不是那些有盐水的人,出现了以心输出量(CO)升高为特征的感染性休克的高动力学状态,每搏量(SV)增加,全身血管阻力(SVR)降低,随后是以平均动脉压(MAP)快速下降为特征的致命低动力状态,中心静脉压升高,减少CO,降低SV,高架SVR,左心室射血分数降低,从而再现人葡萄球菌感染性休克的标志性临床特征。在这个模型中,与用c-IgG预处理的兔相比,用抗-Hla/Luk/ClfAmAb组合预处理的兔死亡率降低69%(P<0.001)。USA300诱导的急性循环衰竭-定义为MAP从感染前基线降低>70%-仅发生在20%(2/10)的用抗Hla/Luk/ClfAmAb组合预处理的兔子中,与100%(9/9)的用c-IgG预处理的那些相比。抗Hla/Luk/ClfAmAb组合的预防阻止了致死性低动力休克的进展,正如对高乳酸盐血症发展的显著保护所证明的那样,低碳酸血症,高钾血症,白细胞减少症,中性粒细胞减少症,单核细胞减少症,淋巴细胞减少,以及与急性心肌损伤相关的生物标志物。
    这些结果证明了机械通气兔模型的潜在实用性,该模型再现了高动力感染性休克的标志性临床特征,以及针对金黄色葡萄球菌毒力因子的免疫疗法预防葡萄球菌感染性休克的转化潜力。
    Patients with septic shock caused by Staphylococcus aureus have mortality rates exceeding 50%, despite appropriate antibiotic therapy. Our objectives were to establish a rabbit model of S. aureus septic shock and to determine whether a novel immunotherapy can prevent or halt its natural disease progression.
    Anesthetized rabbits were ventilated with lung-protective low-tidal volume, instrumented for advanced hemodynamic monitoring, and characterized for longitudinal changes in acute myocardial dysfunction by echocardiography and sepsis-associated biomarkers after S. aureus intravenous challenge. To demonstrate the potential utility of this hyperdynamic septic shock model for preclinical drug development, rabbits were randomized for prophylaxis with anti-Hla/Luk/ClfA monoclonal antibody combination that neutralizes alpha-hemolysin (Hla), the bicomponent pore-forming leukocidins (Luk) including Panton-Valentine leukocidin, leukocidin ED, and gamma-hemolysin, and clumping factor A (ClfA), or an irrelevant isotype-matched control IgG (c-IgG), and then challenged with S. aureus.
    Rabbits challenged with S. aureus, but not those with saline, developed a hyperdynamic state of septic shock characterized by elevated cardiac output (CO), increased stroke volume (SV) and reduced systemic vascular resistance (SVR), which was followed by a lethal hypodynamic state characterized by rapid decline in mean arterial pressure (MAP), increased central venous pressure, reduced CO, reduced SV, elevated SVR, and reduced left-ventricular ejection fraction, thereby reproducing the hallmark clinical features of human staphylococcal septic shock. In this model, rabbits pretreated with anti-Hla/Luk/ClfA mAb combination had 69% reduction in mortality when compared to those pretreated with c-IgG (P<0.001). USA300-induced acute circulatory failure-defined as >70% decreased in MAP from pre-infection baseline-occurred in only 20% (2/10) of rabbits pretreated with anti-Hla/Luk/ClfA mAb combination compared to 100% (9/9) of those pretreated with c-IgG. Prophylaxis with anti-Hla/Luk/ClfA mAb combination halted progression to lethal hypodynamic shock, as evidenced by significant protection against the development of hyperlactatemia, hypocapnia, hyperkalemia, leukopenia, neutropenia, monocytopenia, lymphopenia, as well as biomarkers associated with acute myocardial injury.
    These results demonstrate the potential utility of a mechanically ventilated rabbit model that reproduced hallmark clinical features of hyperdynamic septic shock and the translational potential of immunotherapy targeting S. aureus virulence factors for the prevention of staphylococcal septic shock.
    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)大流行已经到了一个转折点。预防COVID-19的非药物干预措施正在取消。一般来说,疫苗接种正在增加,但是这种策略不断受到严重急性呼吸道综合征冠状病毒2(SARS-CoV-2)快速演变的挑战。值得注意的是,Omicron亚变体在全球传播至少一年,最近开发的亚变异体显示出对先前存在的免疫的强烈逃避,要么来自以前的感染,疫苗接种或两者因此,早期和适当的抗病毒药物治疗有严重COVID-19或死亡风险的患者对于降低发病率和死亡率至关重要,恢复医疗保健能力,促进恢复新常态。目前COVID-19的抗病毒治疗包括中和单克隆抗体(mAb)和直接抗病毒剂。每种药物均已被证明可用于COIVD-19的早期非卧床治疗,但由于患者的合并症而存在不同的有效性和局限性,药物特性,或抗病毒抗性。此外,在与确诊的COVID-19患者密切接触之前或之后,一些特定的单克隆抗体被用于预防COVID-19。这篇评论文章总结了在Omicron亚变体的背景下,目前可用的抗病毒药物治疗COVID-19的证据和未满足的需求。
    The coronavirus disease 2019 (COVID-19) pandemic has reached a turning point. The non-pharmaceutical interventions for preventing COVID-19 are lifting. Vaccination uptake is increasing in general, but this strategy is continuously challenged by the rapid evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Of note, the Omicron subvariants spread globally for at least one year, and the most recently developed subvariants show strong immune evasion to preexisting immunity, either from previous infection, vaccination or both. Therefore, early and appropriate antiviral agents to treat patients at risk for severe COVID-19 or death is crucial to decrease morbidities and mortalities, to restore the healthcare capacities and to facilitate a return to the new normal. Current antiviral therapy for COVID-19 consist of neutralizing monoclonal antibodies (mAbs) and direct antiviral agents. Each agent has been proved for early ambulatory treatment of COIVD-19, but suffer from variable effectiveness and limitations due to patients\' comorbidities, drug properties, or antiviral resistance. Besides, some specific mAbs are indicated for prophylaxis of COVID-19 before or after close contact with confirmed COVID-19 patients. This review article summarizes the evidence and unmet needs of the currently available antiviral agents for management of COVID-19 in the context of the Omicron subvariants.
    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
    与SARS-CoV-2治疗相关的最紧迫的挑战之一是可能更具传染性的新变种的出现,导致更严重的疾病,或者对目前的治疗和疫苗有抵抗力。SARS-CoV-2的出现导致了全球大流行,导致全球数百万人死亡。已经采取了各种策略来对抗这种病毒,包括中和单克隆抗体(mAb),CRISPR/Cas13和反义寡核苷酸(ASO)。虽然疫苗和小分子已被证明是预防严重COVID-19和降低传播率的有效手段,新病毒变种的出现对其有效性提出了挑战。单克隆抗体已显示出治疗早期COVID-19的前景,但其在严重病例中的有效性有限,新变体的出现可能会降低其结合亲和力。CRISPR/Cas13已显示出靶向必需病毒基因的潜力,但是它的效率,特异性,和运送到感染部位是主要的限制。ASO也被证明是有效的靶向病毒RNA,但它们在递送和潜在脱靶效应方面面临与CRISPR/Cas13相似的挑战.总之,这些策略的组合可能会提供更有效的抗击SARS-CoV-2的手段,未来的研究应该集中在提高效率上,特异性,并运送到感染部位。显然,这些替代疗法的持续研究和开发对于正在进行的对抗SARS-CoV-2及其潜在的未来变体的斗争至关重要。
    One of the most pressing challenges associated with SARS-CoV-2 treatment is the emergence of new variants that may be more transmissible, cause more severe disease, or be resistant to current treatments and vaccines. The emergence of SARS-CoV-2 has led to a global pandemic, resulting in millions of deaths worldwide. Various strategies have been employed to combat the virus, including neutralizing monoclonal antibodies (mAbs), CRISPR/Cas13, and antisense oligonucleotides (ASOs). While vaccines and small molecules have proven to be an effective means of preventing severe COVID-19 and reducing transmission rates, the emergence of new virus variants poses a challenge to their effectiveness. Monoclonal antibodies have shown promise in treating early-stage COVID-19, but their effectiveness is limited in severe cases and the emergence of new variants may reduce their binding affinity. CRISPR/Cas13 has shown potential in targeting essential viral genes, but its efficiency, specificity, and delivery to the site of infection are major limitations. ASOs have also been shown to be effective in targeting viral RNA, but they face similar challenges to CRISPR/Cas13 in terms of delivery and potential off-target effects. In conclusion, a combination of these strategies may provide a more effective means of combating SARS-CoV-2, and future research should focus on improving their efficiency, specificity, and delivery to the site of infection. It is evident that the continued research and development of these alternative therapies will be essential in the ongoing fight against SARS-CoV-2 and its potential future variants.
    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: Systematic Review
    单克隆抗体(mAb)已获得2019年轻中度冠状病毒病(COVID-19)或预防COVID-19的紧急使用授权,包括卡西利维单抗加imdevimab(C+I),bamlanivimab加etesevimab(B+E),tixagevimab加西加维玛(T+CG),和sotrovimab(S)和bebtelovimab(BEB)。进行系统评价是为了评估其有效性和安全性。PubMed,Embase,Scopus,medRxiv,bioRxiv,本研究检索了2021年1月至2022年5月期间发表的研究和FDA情况说明书,并使用与上述mAb相关的适当检索词进行数据收集.审查包括原创性研究,包括随机临床试验和发表或预印本的观察性研究。该综述中包括的研究与安慰剂或标准护理或无治疗或单克隆抗体之间以及各种剂量进行了比较。进行了数据提取,并对有效性和安全性进行了审查。本综述共纳入20项研究。与安慰剂组的7%相比,30天内的住院率为2%。用组合mAb更观察到病毒载量的显著降低。联合疗法显示出针对Gamma变体的更快的病毒学治愈。用C+I作为暴露后预防(PEP),29.0%的无症状参与者出现有症状的COVID-19。使用T+CG的暴露前预防使感染的发生率降低了77%。输液相关反应是最常见的不良事件(AE)。中和的mAb减少了轻度至中度患者的住院率,这些患者的输液相关反应是常见的AE。血清阴性患者的反应更好。大多数这些研究是在未接种疫苗的个体中进行的,并且针对Alpha,Gamma,和Delta变体。
    Monoclonal antibodies (mAbs) had received emergency use authorization for mild-to-moderate coronavirus disease 2019 (COVID-19) or for prophylaxis against COVID-19, including casirivimab plus imdevimab (C+I), bamlanivimab plus etesevimab (B+E), tixagevimab plus cilgavimab (T+CG), and sotrovimab (S) and bebtelovimab (BEB). This systematic review was done to assess the efficacy and safety of the same. PubMed, Embase, Scopus, medRxiv, bioRxiv, and FDA fact sheets were searched for the studies published between January 2021 and May 2022, and appropriate search terms related to the mentioned mAbs were used for data collection. Review included original research including randomized clinical trials and observational studies published or preprints. Studies included in the review had compared with placebo or standard of care or no treatment or mAbs with each other and also of various doses. Data extraction was done and reviewed the same for both efficacy and safety. Total of 20 studies were included in this review. The rate of hospitalization within 30 days showed ∼2% in comparison to ∼7% with placebo. Significant reduction in viral load was more observed with combination mAbs. Combination therapy showed faster virological cure against the Gamma variant. With C + I as postexposure prophylaxis (PEP), 29.0% of asymptomatic participants developed symptomatic COVID-19. Pre-exposure prophylaxis with T+CG reduced the incidence of infection by 77%. Infusion-related reaction was the most common adverse event (AE). The neutralizing mAbs reduced hospitalization in mild-to-moderate patients with infusion-related reactions as common AE. The response was better in the seronegative patients. Most of these studies were conducted in unvaccinated individuals and against Alpha, Gamma, and Delta variants.
    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
    H9N2禽流感病毒(AIV)仍然严重威胁着全球家禽业和公众健康。血凝素(HA)蛋白是AIV的必需保护性抗原,并且是中和抗体和疫苗的主要靶标。因此,在这项研究中,我们使用水稻来源的HA蛋白作为免疫原来产生单克隆抗体(mAb),并使用免疫过氧化物酶单层测定和间接酶联免疫吸附测定对它们进行筛选.八种单克隆抗体与重组H9N2AIV和HA蛋白反应良好,其中四个表现出抗血凝的有效抑制活性,而三个显示出显著的中和能力。Western印迹证实两种mAb与HA蛋白结合。使用mAb鉴定线性表位;一种新的线性表位,确定了480HKCDDQCM487。结构分析表明,新的线性表位位于HA2的C末端,靠近二硫键连接的HA1和HA2。氨基酸序列的比对显示该表位在多种H9N2AIV菌株中是高度保守的。这项研究的结果为改进疫苗和诊断策略提供了新的见解,并扩大了我们对AIV免疫反应的理解。
    The H9N2 avian influenza virus (AIV) remains a serious threat to the global poultry industry and public health. The hemagglutinin (HA) protein is an essential protective antigen of AIVs and a major target of neutralizing antibodies and vaccines. Therefore, in this study, we used rice-derived HA protein as an immunogen to generate monoclonal antibodies (mAbs) and screened them using an immunoperoxidase monolayer assay and indirect enzyme-linked immunosorbent assay. Eight mAbs reacted well with the recombinant H9N2 AIV and HA protein, four of which exhibited potent inhibitory activity against hemagglutination, while three showed remarkable neutralization capacities. Western blotting confirmed that two mAbs bound to the HA protein. Linear epitopes were identified using the mAbs; a novel linear epitope, 480HKCDDQCM487, was identified. Structural analysis revealed that the novel linear epitope is located at the C-terminus of HA2 near the disulfide bond-linked HA1 and HA2. Alignment of the amino acid sequences showed that the epitope was highly conserved among multiple H9N2 AIV strains. The results of this study provide novel insights for refining vaccine and diagnostic strategies and expand our understanding of the immune response against AIV.
    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
    COVID-19仍然是血液恶性肿瘤(HM)患者的一个相关问题。疫苗通常在主动治疗的受试者中无效。在这项对意大利GruppoMalattieEMatologichedell\'Adulto(GIMEMA)的多中心回顾性研究中,我们收集了91名接受抗刺药中和单克隆抗体(nMoAbs)治疗的无症状HM患者的数据,以确定病毒清除时间。参考一组未经治疗的无症状患者的28天的预期值。次要终点包括住院率,重症监护病房(ICU)入院,COVID-19相关死亡和安全。86例患者(95%)获得SARS-CoV-2分子拭子阴性,中位时间为18天(IQR13-26;p<0.0001)。我们没有发现根据年龄的显著差异,诊断,治疗类型,疫苗接种状态或nMoAbs类型。COVID-19进展导致的住院率为12%(11/91),2例患者(2.2%)需要入住ICU。中位随访时间为2.33个月,总死亡率为5.5%(5/91),3人死于COVID-19。副作用罕见且具有自限性。我们的数据表明,nMoAbs可以限制免疫抑制治疗对COVID-19临床进展和病毒清除时间的不利影响。最初的审判是在www上注册的。clinicaltrials.gov作为#NCT04932967。
    COVID-19 continues to be a relevant issue among patients with haematological malignancies (HM). Vaccines are frequently not effective in subjects on active treatment. In this multicentre retrospective study of Gruppo Italiano Malattie EMatologiche dell\'Adulto (GIMEMA), we collected data from 91 paucisymptomatic HM patients treated with anti-spike neutralizing monoclonal antibodies (nMoAbs) to determine time to viral clearance, referencing it to the expected value of 28 days from an historical group of untreated paucisymptomatic patients. Secondary endpoints included rate of hospitalization, intensive care unit (ICU) admission, COVID-19 related death and safety. SARS-CoV-2 molecular swab negativity was obtained in 86 patients (95%), with a median time of 18 days (IQR 13-26; p < 0.0001). We did not find significant variations according to age, diagnosis, treatment type, vaccination status or nMoAbs type. Rate of hospitalization due to COVID-19 progression was 12% (11/91), with 2 patients (2.2%) requiring ICU admission. With a median follow-up of 2.33 months, the overall mortality was 5.5% (5/91), with 3 deaths due to COVID-19. Side effects were rare and self-limiting. Our data suggest that nMoAbs can limit the detrimental effect of immunosuppressive treatments on COVID-19 clinical progression and time to viral clearance. The original trial was registered at www.clinicaltrials.gov as #NCT04932967.
    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