COVID-19 drug treatment

COVID - 19 药物治疗
  • 文章类型: Journal Article
    人们越来越担心,由H5N1流感病毒引起的禽类严重呼吸道疾病(禽流感或“禽流感”),可能会更广泛地传播到人类并引起大流行。在这里,我们讨论了与人类感染高致病性H5N1亚型禽流感病毒有关的临床问题,并与从SARS-CoV-2感染研究中获得的最新信息进行了临床比较。首先,我们认为感染H5N1病毒的人可能会增加心血管事件.像SARS-CoV-2感染,H5N1感染可能导致内皮功能障碍和相关的促凝血和血栓形成状态,通过这种机制,感染可能会增加心血管疾病的发病率,特别是在先前存在心血管疾病的脆弱个体中。其次,我们讨论了他汀类药物使用的潜在有益作用,在预防和治疗甲型流感(H5N1)患者中,这对SARS-CoV-2感染引起的COVID-19的治疗有利。
    人们担心由高致病性禽流感A(H5N1)病毒引起的禽流感可能会更广泛地传播到人类,并导致大流行的H5N1感染可能会导致内皮功能障碍,并通过这种机制,与SARS-CoV-2感染一样,它可能会增加心血管疾病的发病率和死亡率。使用他汀类药物可降低甲型H5N1禽流感患者的心血管发病率和死亡率,正如在患有COVID-19的患者中发现的那样。
    There is growing concern that the severe respiratory disease in birds (avian influenza or \'bird flu\') caused by the H5N1 influenza virus, might potentially spread more widely to humans and cause a pandemic. Here we discuss clinical issues related to human infections by the highly pathogenic H5N1 subtype of the avian influenza A virus and make a clinical comparison with recent information obtained from studies of SARS-CoV-2 infection. Firstly, we consider the potential increase in cardiovascular events in humans infected with the H5N1 virus. Like SARS-CoV-2 infection, H5N1 infection may result in endothelial dysfunction and the associated procoagulant and prothrombotic state, and via this mechanism, the infection can potentially increase cardiovascular morbidity, especially in vulnerable individuals with pre-existing cardiovascular disease. Secondly, we discuss the potential beneficial role of statin use, both in the prophylaxis and the treatment of individuals with influenza A(H5N1), as was found favorable for the treatment of COVID-19 caused by SARS-CoV-2 infection.
    There is a concern that avian influenza caused by the highly pathogenic avian influenza A(H5N1) virus might potentially spread more widely to humans and result in a pandemicH5N1 infection may result in endothelial dysfunction and via this mechanism, it can potentially increase cardiovascular morbidity and mortality as has occurred with SARS-CoV-2 infection.There is a potential advantage of the use of statins to reduce cardiovascular morbidity and mortality in patients with avian influenza A(H5N1), as has been found in patients suffering from COVID-19.
    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
    2023年12月,法国是第一个授予Sipavibart(AZD3152,一种研究性长效单克隆抗体)作为免疫功能低下个体的COVID-19暴露前预防治疗的国家。第一批接受Sipavibart的患者有不同的特征,但他们都高度免疫功能低下,经常伴有低球蛋白血症和其他慢性疾病。未报告不良事件。
    France was the first country to grant Sipavibart (AZD3152, an investigational long-acting monoclonal antibody) as a COVID-19 pre-exposure prophylaxis treatment in immunocompromised individuals in December 2023. The first patients to receive Sipavibart had different profiles, but they were all highly immunocompromised with frequently associated hypogammaglobulinemia and other chronic conditions. No adverse event was reported.
    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:Molnupiravir和尼马特雷韦-利托那韦口服药物治疗轻度至中度COVID-19。然而,这些药物在非常老(≥80岁)的有效性,住院患者仍不清楚,限制这些抗病毒药物在这一特定组中的风险-收益评估.这项研究调查了这些抗病毒药物在降低该组COVID-19住院患者死亡率方面的有效性。
    方法:使用香港全港公共医疗数据库,我们进行了一项目标试验模拟研究,数据来自于13642名符合资格的molnupiravir试验参与者和9553名nirmatrelvir-ritonavir试验参与者.主要结果是全因死亡率。使用克隆审查加权方法将不朽的时间和混杂的偏见降至最低。通过稳定的逆概率权重调整混杂偏差后,通过汇总逻辑回归估算死亡率比值比。
    结果:莫诺比拉韦(HR:0.895,95%CI:0.826-0.970)和尼马特雷韦-利托那韦(HR:0.804,95%CI:0.678-0.955)均显示年龄最大的住院患者的死亡率风险降低。在口服抗病毒治疗和疫苗接种状态之间没有观察到显著的相互作用。对于两种molnupiravir,发起者的28天死亡风险均低于非发起者(风险差异:-1.09%,95%CI:-2.29,0.11)和尼马特雷韦-利托那韦(风险差异:-1.71%,95%CI:-3.30,-0.16)试验。无论患者先前的疫苗接种状态如何,都观察到这些药物的有效性。
    结论:Molnupiravir和nirmatrelvir-ritonavir在降低住院年龄最大的COVID-19患者的死亡风险方面是中等有效的,无论其疫苗接种状态如何。
    BACKGROUND: Molnupiravir and nirmatrelvir-ritonavir are orally administered pharmacotherapies for mild to moderate COVID-19. However, the effectiveness of these drugs among very old (≥80 years), hospitalised patients remains unclear, limiting the risk-benefit assessment of these antivirals in this specific group. This study investigates the effectiveness of these antivirals in reducing mortality among this group of hospitalised patients with COVID-19.
    METHODS: Using a territory-wide public healthcare database in Hong Kong, a target trial emulation study was conducted with data from 13 642 eligible participants for the molnupiravir trial and 9553 for the nirmatrelvir-ritonavir trial. The primary outcome was all-cause mortality. Immortal time and confounding bias was minimised using cloning-censoring-weighting approach. Mortality odds ratios were estimated by pooled logistic regression after adjusting confounding biases by stabilised inverse probability weights.
    RESULTS: Both molnupiravir (HR: 0.895, 95% CI: 0.826-0.970) and nirmatrelvir-ritonavir (HR: 0.804, 95% CI: 0.678-0.955) demonstrated moderate mortality risk reduction among oldest-old hospitalised patients. No significant interaction was observed between oral antiviral treatment and vaccination status. The 28-day risk of mortality was lower in initiators than non-initiators for both molnupiravir (risk difference: -1.09%, 95% CI: -2.29, 0.11) and nirmatrelvir-ritonavir (risk difference: -1.71%, 95% CI: -3.30, -0.16) trials. The effectiveness of these medications was observed regardless of the patients\' prior vaccination status.
    CONCLUSIONS: Molnupiravir and nirmatrelvir-ritonavir are moderately effective in reducing mortality risk among hospitalised oldest-old patients with COVID-19, regardless of their vaccination status.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    尽管中和抗体在严重急性呼吸综合征冠状病毒-2(SARS-CoV-2)的治疗中具有潜力,其在中国患者中的疗效的临床研究仍然有限。这项研究旨在研究抗病毒治疗与中和单克隆抗体联合治疗对淋巴瘤合并B细胞耗竭的患者复发性持续性SARS-CoV-2肺炎的治疗效果。对接受抗病毒尼马特雷韦/利托那韦治疗和中和抗体替沙格维单抗-西加维单抗(tix-cil)治疗的中国患者进行了前瞻性研究。主要结果是SARS-CoV-2感染的复发率。5例淋巴瘤患者反复出现SARS-CoV-2肺炎,并接受了tix-cil治疗。所有患者在SARS-CoV-2感染前一年内都有CD20单克隆抗体使用史,两名患者也有布鲁顿酪氨酸激酶(BTK)抑制剂使用史。这些患者的淋巴细胞计数明显较低,并且几乎耗尽了B细胞。所有五名患者的血清SARS-CoV-2IgG和IgM抗体检测均为阴性。在6个月的随访期内,抗病毒和tix-cil治疗后,没有患者出现SARS-CoV-2肺炎的再感染。总之,抗病毒和SARS-CoV-2中和抗体的给药显示出令人鼓舞的治疗SARS-CoV-2肺炎的淋巴瘤并发B细胞耗竭,以及中和抗体的潜在预防作用长达6个月。
    Despite the potential of neutralizing antibodies in the management of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), clinical research on its efficacy in Chinese patients remains limited. This study is aimed at investigating the therapeutic effect of combination of antiviral therapy with neutralizing monoclonal antibodies for recurrent persistent SARS-CoV-2 pneumonia in patients with lymphoma complicated by B cell depletion. A prospective study was conducted on Chinese patients who were treated with antiviral nirmatrelvir/ritonavir therapy and the neutralizing antibody tixagevimab-cilgavimab (tix-cil). The primary outcome was the rate of recurrent SARS-CoV-2 infection. Five patients with lymphoma experienced recurrent SARS-CoV-2 pneumonia and received tix-cil treatment. All patients had a history of CD20 monoclonal antibody use within the year preceding SARS-CoV-2 infection, and two patients also had a history of Bruton\'s tyrosine kinase (BTK) inhibitor use. These patients had notably low lymphocyte counts and exhibited near depletion of B cells. All five patients tested negative for serum SARS-CoV-2 IgG and IgM antibodies. None of the patients developed reinfection with SARS-CoV-2 pneumonia after antiviral and tix-cil treatment during the 6-month follow-up period. In conclusion, the administration of antiviral and SARS-CoV-2-neutralizing antibodies showed encouraging therapeutic efficacy against SARS-CoV-2 pneumonia in patients with lymphoma complicated by B cell depletion, along with the potential preventive effect of neutralizing antibodies for up to 6 months.
    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:三调蛋白(人多价免疫球蛋白[Ig]M〜23%,IgA~21%,IgG〜56%制剂)先前已与有创机械通气(IMV)的严重社区获得性肺炎患者亚群的较低死亡率相关,并且有明显的炎症迹象。ESsCOVID试验的假设是,三调节蛋白可以预防炎症驱动的严重冠状病毒疾病2019(COVID-19)进展为危重疾病甚至死亡。
    方法:将患有严重COVID-19的成年人随机分组,除标准治疗外,连续5天接受静脉输注三调节素或安慰剂。主要疗效终点是临床恶化(第6-29天)和28天全因死亡率(第1-29天)的复合。
    结果:一百六十六名患者接受了三调节蛋白(n=84)或安慰剂(n=82)。33名患者死亡,9在治疗阶段。总的来说,84.9%和76.5%的患者完成治疗和随访,分别。主要疗效终点报告33.3%的患者接受三调蛋白治疗,34.1%的患者接受安慰剂治疗(P=0.912)。在有创机械通气第29天康复的患者比例没有观察到差异。或无重症监护病房的日子。因治疗引起的不良事件发生率相当。对早期全身性炎症患者进行事后分析,排除基线时CRP高(>150mg/L)和/或D-二聚体(≥3mg/L)和/或血小板计数低(<130×109/L)的患者。三调节蛋白组中的47名患者和安慰剂组中的49名患者符合这些标准。观察到临床恶化和死亡率的差异为15.5个百分点,有利于三调蛋白(95%置信区间:-4.46,34.78;P=0.096)。
    结论:尽管总体人群的主要结局没有差异,对早期全身性炎症患者亚组的观察表明,三调节蛋白在这种情况下可能具有潜力,值得进一步研究.Esscovid是在诊所进行登记的。2020年10月6日政府。:NCT04576728。
    BACKGROUND: Trimodulin (human polyvalent immunoglobulin [Ig] M ~ 23%, IgA ~ 21%, IgG ~ 56% preparation) has previously been associated with a lower mortality rate in a subpopulation of patients with severe community-acquired pneumonia on invasive mechanical ventilation (IMV) and with clear signs of inflammation. The hypothesis for the ESsCOVID trial was that trimodulin may prevent inflammation-driven progression of severe coronavirus disease 2019 (COVID-19) to critical disease or even death.
    METHODS: Adults with severe COVID-19 were randomised to receive intravenous infusions of trimodulin or placebo for 5 consecutive days in addition to standard of care. The primary efficacy endpoint was a composite of clinical deterioration (Days 6-29) and 28-day all-cause mortality (Days 1-29).
    RESULTS: One-hundred-and-sixty-six patients received trimodulin (n = 84) or placebo (n = 82). Thirty-three patients died, nine during the treatment phase. Overall, 84.9% and 76.5% of patients completed treatment and follow-up, respectively. The primary efficacy endpoint was reported in 33.3% of patients on trimodulin and 34.1% of patients on placebo (P = 0.912). No differences were observed in the proportion of patients recovered on Day 29, days of invasive mechanical ventilation, or intensive care unit-free days. Rates of treatment-emergent adverse events were comparable. A post hoc analysis was conducted in patients with early systemic inflammation by excluding those with high CRP (> 150 mg/L) and/or D-dimer (≥ 3 mg/L) and/or low platelet counts (< 130 × 109/L) at baseline. Forty-seven patients in the trimodulin group and 49 in the placebo group met these criteria. A difference of 15.5 percentage points in clinical deterioration and mortality was observed in favour of trimodulin (95% confidence interval: -4.46, 34.78; P = 0.096).
    CONCLUSIONS: Although there was no difference in the primary outcome in the overall population, observations in a subgroup of patients with early systemic inflammation suggest that trimodulin may have potential in this setting that warrants further investigation. ESSCOVID WAS REGISTERED PROSPECTIVELY AT CLINICALTRIALS.GOV ON OCTOBER 6, 2020.: NCT04576728.
    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有能力进化出逃避疫苗和感染获得性免疫和抗病毒药物的突变。因此,在不对病毒施加选择性压力的情况下保护免受严重疾病的变体不可知治疗剂将是有价值的生物医学工具,尽管新变体的持续出现,其仍将保持其功效。这里,我们用SARS-CoV-2Delta挑战雄性恒河猴,这是高度易感动物模型中最具致病性的变体。在挑战的时候,我们还使用雾化RBD-62治疗猕猴,RBD-62是一种通过多轮SARS-CoV-2RBD体外进化而获得1000倍增强的ACE2结合亲和力的蛋白质。RBD-62治疗同样抑制病毒在上呼吸道和下呼吸道的复制,以前在临床批准的疫苗中未观察到的现象。重要的是,RBD-62不阻断病毒特异性T细胞和B细胞应答的发展,并且不引发抗药物免疫。这些数据提供了RBD-62可以从高毒力变体预防严重疾病的概念证明。
    SARS-CoV-2 has the capacity to evolve mutations that escape vaccine- and infection-acquired immunity and antiviral drugs. A variant-agnostic therapeutic agent that protects against severe disease without putting selective pressure on the virus would thus be a valuable biomedical tool that would maintain its efficacy despite the ongoing emergence of new variants. Here, we challenge male rhesus macaques with SARS-CoV-2 Delta-the most pathogenic variant in a highly susceptible animal model. At the time of challenge, we also treat the macaques with aerosolized RBD-62, a protein developed through multiple rounds of in vitro evolution of SARS-CoV-2 RBD to acquire 1000-fold enhanced ACE2 binding affinity. RBD-62 treatment equivalently suppresses virus replication in both upper and lower airways, a phenomenon not previously observed with clinically approved vaccines. Importantly, RBD-62 does not block the development of virus-specific T- and B-cell responses and does not elicit anti-drug immunity. These data provide proof-of-concept that RBD-62 can prevent severe disease from a highly virulent variant.
    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)的急性表现表现出败血症相关并发症的标志,反映多器官衰竭。炎性细胞因子风暴伴随着对严重急性呼吸综合征冠状病毒2(SARS-CoV-2)感染的促炎和抗炎宿主反应的失衡导致严重和严重的感染性休克。严重感染COVID-19患者的脓毒症特征包括细胞重编程和器官功能障碍,导致高死亡率,强调改善与COVID-19相关的败血症的临床护理和先进治疗干预的重要性。功能性食品和营养重要性的植物化学物质对医疗保健系统产生了令人难以置信的影响,包括预防和/或治疗慢性病。因此,在本次审查中,我们的目的是探索与COVID-19相关的脓毒症的发病机制,它破坏了机体的生理稳态,导致严重的器官损伤。此外,我们总结了一些有效的植物化学物质的多种药理特性,可用作功能性食品以及针对SARS-CoV-2感染的败血症相关并发症的营养品。本文探索的植物化学物质包括槲皮素,姜黄素,木犀草素,芹菜素,白藜芦醇,还有柚皮素,这是我们日常食物摄入的主要植物成分。我们汇总了各种研究的发现,包括人体临床试验,进一步探讨每种植物化学物质对脓毒症和COVID-19的治疗潜力,这突显了它们在脓毒症相关COVID-19发病机制中的可能重要性。我们得出的结论是,我们的评论将为探索植物化学衍生的治疗剂以预防或治疗与COVID-19相关的危及生命的败血症并发症开辟一条新的研究途径。
    The acute manifestations of coronavirus disease 2019 (COVID-19) exhibit the hallmarks of sepsis-associated complications that reflect multiple organ failure. The inflammatory cytokine storm accompanied by an imbalance in the pro-inflammatory and anti-inflammatory host response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection leads to severe and critical septic shock. The sepsis signature in severely afflicted COVID-19 patients includes cellular reprogramming and organ dysfunction that leads to high mortality rates, emphasizing the importance of improved clinical care and advanced therapeutic interventions for sepsis associated with COVID-19. Phytochemicals of functional foods and nutraceutical importance have an incredible impact on the healthcare system, which includes the prevention and/or treatment of chronic diseases. Hence, in the present review, we aim to explore the pathogenesis of sepsis associated with COVID-19 that disrupts the physiological homeostasis of the body, resulting in severe organ damage. Furthermore, we have summarized the diverse pharmacological properties of some potent phytochemicals, which can be used as functional foods as well as nutraceuticals against sepsis-associated complications of SARS-CoV-2 infection. The phytochemicals explored in this article include quercetin, curcumin, luteolin, apigenin, resveratrol, and naringenin, which are the major phytoconstituents of our daily food intake. We have compiled the findings from various studies, including clinical trials in humans, to explore more into the therapeutic potential of each phytochemical against sepsis and COVID-19, which highlights their possible importance in sepsis-associated COVID-19 pathogenesis. We conclude that our review will open a new research avenue for exploring phytochemical-derived therapeutic agents for preventing or treating the life-threatening complications of sepsis associated with 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
    全球有近600万人死于由严重急性呼吸道综合症冠状病毒2(SARS-CoV-2)感染引起的冠状病毒病(COVID-19)爆发。尽管COVID-19疫苗在很大程度上成功地降低了疾病的严重程度和死亡人数,疫苗诱导的免疫力随着时间的推移而下降,以及新的病毒变异体或突变的持续出现,突显了开发针对SARS-CoV-2的广谱宿主介导疗法的替代策略的必要性.严重COVID-19的一个关键特征是先天免疫信号失调,最终导致大量促炎细胞因子和趋化因子的高表达和缺乏抗病毒干扰素(IFN),特别是I型(α和β)和III型(λ)。作为自然宿主防御,髓样分化初级反应蛋白,MyD88通过Toll样受体(TLRs)的信号转导途径在先天和获得性免疫反应中起关键作用,一种病原体识别受体(PRR)。然而,最近的研究表明,病毒感染会上调MyD88的表达,并通过负调节I型IFN来损害宿主的抗病毒反应。半乳糖凝集素-3(Gal3),病毒感染的另一个关键角色,已显示通过调节病毒进入和激活TLRs来调节宿主免疫应答,NLRP3炎性体,和NF-κB,导致促炎细胞因子的释放,并有助于整体炎症反应,所谓的“细胞因子风暴”。这些研究表明,对MyD88和Gal3的特异性抑制可能是一种有希望的COVID-19治疗方法。这篇综述提出了MyD88和Gal3靶向抗病毒药物发现的未来方向,强调在SARS-CoV-2感染中恢复宿主免疫力的潜力。
    Nearly six million people worldwide have died from the coronavirus disease (COVID-19) outbreak caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Although COVID-19 vaccines are largely successful in reducing the severity of the disease and deaths, the decline in vaccine-induced immunity over time and the continuing emergence of new viral variants or mutations underscore the need for an alternative strategy for developing broad-spectrum host-mediated therapeutics against SARS-CoV-2. A key feature of severe COVID-19 is dysregulated innate immune signaling, culminating in a high expression of numerous pro-inflammatory cytokines and chemokines and a lack of antiviral interferons (IFNs), particularly type I (alpha and beta) and type III (lambda). As a natural host defense, the myeloid differentiation primary response protein, MyD88, plays pivotal roles in innate and acquired immune responses via the signal transduction pathways of Toll-like receptors (TLRs), a type of pathogen recognition receptors (PRRs). However, recent studies have highlighted that infection with viruses upregulates MyD88 expression and impairs the host antiviral response by negatively regulating type I IFN. Galectin-3 (Gal3), another key player in viral infections, has been shown to modulate the host immune response by regulating viral entry and activating TLRs, the NLRP3 inflammasome, and NF-κB, resulting in the release of pro-inflammatory cytokines and contributing to the overall inflammatory response, the so-called \"cytokine storm\". These studies suggest that the specific inhibition of MyD88 and Gal3 could be a promising therapy for COVID-19. This review presents future directions for MyD88- and Gal3-targeted antiviral drug discovery, highlighting the potential to restore host immunity in SARS-CoV-2 infections.
    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
    呼吸道病毒感染(VRTIs)是全球发病率和死亡率的主要原因之一。每年影响所有年龄组的数百万人。这些感染是由各种病原体引起的,包括鼻病毒(RV),腺病毒(AdVs),和冠状病毒(CoV),在寒冷的季节特别普遍。虽然许多VRTI是自限的,它们的频繁复发和严重健康并发症的可能性凸显了对有效治疗策略的迫切需要.病毒蛋白酶对于病毒的成熟和复制至关重要,使它们成为有希望的治疗目标。这篇综述探讨了病毒蛋白酶在呼吸道病毒生命周期中的关键作用,以及蛋白酶抑制剂作为对这些感染的战略反应的开发。抗病毒治疗的最新进展突出了蛋白酶抑制剂在减少病毒性疾病的传播和严重程度方面的有效性。特别是在正在进行的COVID-19大流行期间。它还评估了目前旨在鉴定和开发针对主要呼吸道病毒关键蛋白酶的抑制剂的努力。包括人类房车,AdVs,和(严重急性呼吸综合征冠状病毒-2)SARS-CoV-2。尽管最近发现了SARS-CoV-2,但在过去的五年中,科学界已经投入了相当多的时间和资源来研究现有的药物和开发针对病毒主要蛋白酶的新抑制剂。然而,鉴定RV和AdV蛋白酶抑制剂的研究工作有限。因此,在这里,建议利用这些知识来开发影响呼吸道的其他病毒的蛋白酶的新抑制剂或开发双重抑制剂。最后,通过详述这些抑制剂的作用机制和治疗潜力,这篇综述旨在证明它们在改变呼吸道病毒性疾病管理方面的重要作用,并为未来的研究方向提供见解。
    Respiratory viral infections (VRTIs) rank among the leading causes of global morbidity and mortality, affecting millions of individuals each year across all age groups. These infections are caused by various pathogens, including rhinoviruses (RVs), adenoviruses (AdVs), and coronaviruses (CoVs), which are particularly prevalent during colder seasons. Although many VRTIs are self-limiting, their frequent recurrence and potential for severe health complications highlight the critical need for effective therapeutic strategies. Viral proteases are crucial for the maturation and replication of viruses, making them promising therapeutic targets. This review explores the pivotal role of viral proteases in the lifecycle of respiratory viruses and the development of protease inhibitors as a strategic response to these infections. Recent advances in antiviral therapy have highlighted the effectiveness of protease inhibitors in curtailing the spread and severity of viral diseases, especially during the ongoing COVID-19 pandemic. It also assesses the current efforts aimed at identifying and developing inhibitors targeting key proteases from major respiratory viruses, including human RVs, AdVs, and (severe acute respiratory syndrome coronavirus-2) SARS-CoV-2. Despite the recent identification of SARS-CoV-2, within the last five years, the scientific community has devoted considerable time and resources to investigate existing drugs and develop new inhibitors targeting the virus\'s main protease. However, research efforts in identifying inhibitors of the proteases of RVs and AdVs are limited. Therefore, herein, it is proposed to utilize this knowledge to develop new inhibitors for the proteases of other viruses affecting the respiratory tract or to develop dual inhibitors. Finally, by detailing the mechanisms of action and therapeutic potentials of these inhibitors, this review aims to demonstrate their significant role in transforming the management of respiratory viral diseases and to offer insights into future research directions.
    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
    (1)背景:血管紧张素转换酶2(ACE2)是SARS-CoV-2病毒的重要功能受体。尽管感染的规模不再处于大流行水平,仍然有致命的病例。病毒感染皮肤的可能性引发了有关新预防措施的疑问。在抗SARS-CoV-2应用的背景下,抗菌纳米材料(银,银;钻石,D;氧化石墨烯,检查GO及其复合物)以评估其影响ACE2是否与病毒结合的能力。(2)方法:进行ACE2抑制竞争试验和原代成人表皮角质形成细胞(HEKa)和原代成人真皮成纤维细胞(HDFa)的体外处理,以评估纳米材料/纳米复合物的阻断能力及其对细胞的毒性。(3)结果:与单个纳米材料相比,纳米复合物发挥协同作用。HEKa细胞比HDFa细胞对Ag处理和高浓度的GO更敏感。D未观察到细胞毒性作用。在复合物中,两种碳纳米材料对Ag都有舒缓作用。(4)结论:Ag5D10和Ag5GO10纳米复合物似乎对于皮肤应用而言是最有效和安全的,以通过阻断ACE2-S结合来对抗SARS-CoV-2感染。这些纳米复合物应通过延长体内暴露来评估。预期的低特异性能够实现更广泛的应用。
    (1) Background: Angiotensin-converting enzyme 2 (ACE2) is a crucial functional receptor of the SARS-CoV-2 virus. Although the scale of infections is no longer at pandemic levels, there are still fatal cases. The potential of the virus to infect the skin raises questions about new preventive measures. In the context of anti-SARS-CoV-2 applications, the interactions of antimicrobial nanomaterials (silver, Ag; diamond, D; graphene oxide, GO and their complexes) were examined to assess their ability to affect whether ACE2 binds with the virus. (2) Methods: ACE2 inhibition competitive tests and in vitro treatments of primary human adult epidermal keratinocytes (HEKa) and primary human adult dermal fibroblasts (HDFa) were performed to assess the blocking capacity of nanomaterials/nanocomplexes and their toxicity to cells. (3) Results: The nanocomplexes exerted a synergistic effect compared to individual nanomaterials. HEKa cells were more sensitive than HDFa cells to Ag treatments and high concentrations of GO. Cytotoxic effects were not observed with D. In the complexes, both carbonic nanomaterials had a soothing effect against Ag. (4) Conclusions: The Ag5D10 and Ag5GO10 nanocomplexes seem to be most effective and safe for skin applications to combat SARS-CoV-2 infection by blocking ACE2-S binding. These nanocomplexes should be evaluated through prolonged in vivo exposure. The expected low specificity enables wider applications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号