intranasal vaccine

鼻内疫苗
  • 文章类型: Journal Article
    严重急性呼吸道综合征冠状病毒2(SARS-CoV-2),冠状病毒病(COVID-19)的病原体,2019年在全球范围内迅速传播。随着Omicron变体的出现,COVID-19进入流行阶段。鉴于秋季和冬季的病例预计会增加,实施COVID-19季节性加强疫苗的策略对于保护公众健康变得越来越有价值.这种做法已经存在用于季节性流感疫苗以对抗年度流感季节。我们的目标是研究一种易于修改的疫苗平台,用于季节性对抗SARS-CoV-2。在这项研究中,我们评估了转基因流感病毒ΔNA(RBD)作为COVID-19的鼻内疫苗候选物。对这种修饰的病毒进行工程改造,以将神经氨酸酶(NA)蛋白的编码序列替换为SARS-CoV-2的受体结合域(RBD)蛋白的膜锚定形式。我们设计了实验,使用致死(Delta)和非致死(Omicron)攻击模型评估K18-hACE2小鼠中ΔNA(RBD)的保护作用。COVID-19mRNA疫苗的对照和我们实验室先前描述的鼻内病毒样颗粒疫苗被用作比较。用表达祖先RBD的ΔNA(RBD)免疫在小鼠血清中引起高抗RBDIgG水平,肺组织中的高抗RBDIgA,并提高了Delta变异攻击后的生存率。修饰ΔNA(RBD)以表达Omicron变体RBD改变了Omicron变体攻击后小鼠肺中的变体特异性抗体应答和有限的病毒负荷。总的来说,该数据表明ΔNA(RBD)可能是产生针对SARS-CoV-2的粘膜和全身免疫的有效鼻内疫苗平台。
    Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease (COVID-19), rapidly spread across the globe in 2019. With the emergence of the Omicron variant, COVID-19 shifted into an endemic phase. Given the anticipated rise in cases during the fall and winter seasons, the strategy of implementing seasonal booster vaccines for COVID-19 is becoming increasingly valuable to protect public health. This practice already exists for seasonal influenza vaccines to combat annual influenza seasons. Our goal was to investigate an easily modifiable vaccine platform for seasonal use against SARS-CoV-2. In this study, we evaluated the genetically modified influenza virus ΔNA(RBD) as an intranasal vaccine candidate for COVID-19. This modified virus was engineered to replace the coding sequence for the neuraminidase (NA) protein with a membrane-anchored form of the receptor binding domain (RBD) protein of SARS-CoV-2. We designed experiments to assess the protection of ΔNA(RBD) in K18-hACE2 mice using lethal (Delta) and non-lethal (Omicron) challenge models. Controls of COVID-19 mRNA vaccine and our lab\'s previously described intranasal virus like particle vaccine were used as comparisons. Immunization with ΔNA(RBD) expressing ancestral RBD elicited high anti-RBD IgG levels in the serum of mice, high anti-RBD IgA in lung tissue, and improved survival after Delta variant challenge. Modifying ΔNA(RBD) to express Omicron variant RBD shifted variant-specific antibody responses and limited viral burden in the lungs of mice after Omicron variant challenge. Overall, this data suggests that ΔNA(RBD) could be an effective intranasal vaccine platform that generates mucosal and systemic immunity towards SARS-CoV-2.
    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),由严重急性呼吸综合征冠状病毒-2(SARS-CoV-2)引起,成为2019年的全球疫情,深刻影响人类健康和全球经济。为了克服这一挑战,开发了各种疫苗模式并将其商业化。包括灭活疫苗,mRNA疫苗,基于腺病毒载体的疫苗,和亚单位疫苗。虽然肌内疫苗诱导高IgG水平,它们通常不能刺激呼吸系统中显著的粘膜免疫。我们使用表达SARS-CoV-2Beta变体(rK148/beta-S)的刺突蛋白的新城疫病毒(NDV)载体,并评估了在K18-hACE2转基因小鼠中用rK148/β-S鼻内接种的功效。低剂量(106.0EID50)的鼻内疫苗接种在用SARS-CoV-2Beta变体攻击后可获得86%的存活率。高剂量(107.0EID50)的给药导致肺病毒载量降低,并且针对SARS-CoV-2Beta和Delta变体的存活率为100%。在SARS-CoV-2攻击后,在接种疫苗的小鼠肺中也诱导了高水平的SARS-CoV-2尖峰特异性IgA。我们的发现表明,rK148/beta-S有望作为一种鼻内疫苗候选物,可有效诱导针对SARS-CoV-2的粘膜免疫。
    Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), emerged as a global outbreak in 2019, profoundly affecting both human health and the global economy. Various vaccine modalities were developed and commercialized to overcome this challenge, including inactivated vaccines, mRNA vaccines, adenovirus vector-based vaccines, and subunit vaccines. While intramuscular vaccines induce high IgG levels, they often fail to stimulate significant mucosal immunity in the respiratory system. We employed the Newcastle disease virus (NDV) vector expressing the spike protein of the SARS-CoV-2 Beta variant (rK148/beta-S), and evaluated the efficacy of intranasal vaccination with rK148/beta-S in K18-hACE2 transgenic mice. Intranasal vaccination with a low dose (106.0 EID50) resulted in an 86% survival rate after challenge with the SARS-CoV-2 Beta variant. Administration at a high dose (107.0 EID50) led to a reduction in lung viral load and 100% survival against the SARS-CoV-2 Beta and Delta variants. A high level of the SARS-CoV-2 spike-specific IgA was also induced in vaccinated mice lungs following the SARS-CoV-2 challenge. Our findings suggest that rK148/beta-S holds promise as an intranasal vaccine candidate that effectively induces mucosal immunity against SARS-CoV-2.
    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
    这项研究评估了到达奥地利育肥农场时超声检查和体格检查的意义和预测价值。治疗频率和平均日增重(ADG)与物理和超声检查结果有关。此外,研究了鼻内接种疫苗对一半受检小牛的影响。在育肥开始和结束时记录600头小牛的临床和超声健康状况。一半的小牛接受鼻内接种(Bovalto®Respi鼻内接种)。总的来说,44.5%的人在到达农场时经胸超声检查(TUS)显示呼吸评分(RS)异常,56.0%的人显示呼吸道疾病的迹象。对于RS和TUS,对疾病和健康进行了分类。结果显示,与健康小牛(RS中位数:1.01kg/d;TUS中位数:1.01kg/d)相比,ILL小牛的ADG较低(RS中位数:0.93kg/d;TUS中位数:0.96kg/d)。与治疗和ILL小牛(RS中位数1.01kg/d;TUS:1.02kg/d)相比,未治疗和ILL小牛(RS中位数0.90kg/d;TUS:0.93kg/d)的中位ADG较低。疫苗接种不影响生长性能或ILL的发生,虽然VAC小牛的治疗频率较低(NVAC为17.0%;VAC为11.3%)。呼吸道疾病检查方案的实施可能会对生产参数产生积极影响(例如,治疗频率和ADG)。
    This study evaluated the significance and predictive value of ultrasonographic and physical examination on arrival at an Austrian fattening farm. Treatment frequency and average daily weight gain (ADG) were related to physical and ultrasonographic examination results. Additionally, the effect of an intranasal vaccination in half of the examined calves was studied. The clinical and ultrasonographic health status 600 calves was recorded at the beginning and end of fattening. Half of the calves received an intranasal vaccination (Bovalto® Respi Intranasal). Overall, 44.5% showed an abnormal respiratory scoring (RS) and 56.0% showed signs of respiratory diseases in transthoracic ultrasonography (TUS) at arrival on the farm. For both RS and TUS, a categorization between ILL and HEALTHY was conducted. Results showed lower ADG in ILL calves (RS median: 0.93 kg/d; TUS median: 0.96 kg/d) compared to HEALTHY calves (RS median: 1.01 kg/d; TUS median: 1.01 kg/d). The median ADG was lower in not treated and ILL calves (RS median 0.90 kg/d; TUS: 0.93 kg/d) compared to treated and ILL calves (RS median 1.01 kg/d; TUS: 1.02 kg/d). Vaccination did not affect growth performance or occurrence of ILL, though treatment frequency was lower in VAC calves (17.0% in NVAC; 11.3% in VAC). The implementation of examination protocols for respiratory diseases may have a positive impact on production parameters (e.g., treatment frequency and ADG).
    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
    在临床前研究中,多种疫苗平台已被用于开发鼻SARS-CoV-2疫苗。主要的管道是病毒载体作为蛋白质疫苗。其中,几种以病毒为载体的疫苗已进入临床开发.然而,这些临床研究报告了一些不令人满意的结果.面对这样的紧急状况,利用其他技术快速开发下一代鼻内COVID-19疫苗势在必行。纳米基鼻内疫苗已经成为对抗呼吸道传染病的方法。利用纳米技术的力量,这些疫苗提供了对病原体的非侵入性但有效的防御,包括COVID-19的威胁。基于纳米颗粒的疫苗粘膜递送技术的改进,如脂质纳米颗粒,聚合物纳米颗粒,无机纳米颗粒等.,不仅提供稳定性和控释,而且增强粘膜粘附,有效地克服了传统疫苗的局限性。因此,在这次审查中,我们概述了鼻内疫苗的评估,并强调了当前的障碍。接下来,总结了基于纳米平台的现代输送系统。最后讨论了基于纳米平台的鼻内疫苗临床应用面临的挑战。
    Multiple vaccine platforms have been employed to develop the nasal SARS-CoV-2 vaccines in preclinical studies, and the dominating pipelines are viral vectored as protein-based vaccines. Among them, several viral vectored-based vaccines have entered clinical development. Nevertheless, some unsatisfactory results were reported in these clinical studies. In the face of such urgent situations, it is imperative to rapidly develop the next-generation intranasal COVID-19 vaccine utilizing other technologies. Nanobased intranasal vaccines have emerged as an approach against respiratory infectious diseases. Harnessing the power of nanotechnology, these vaccines offer a noninvasive yet potent defense against pathogens, including the threat of COVID-19. The improvements made in vaccine mucosal delivery technologies based on nanoparticles, such as lipid nanoparticles, polymeric nanoparticles, inorganic nanoparticles etc., not only provide stability and controlled release but also enhance mucosal adhesion, effectively overcoming the limitations of conventional vaccines. Hence, in this review, we overview the evaluation of intranasal vaccine and highlight the current barriers. Next, the modern delivery systems based on nanoplatforms are summarized. The challenges in clinical application of nanoplatform based intranasal vaccine are finally discussed.
    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
    流感是一种高度传染性的呼吸道疾病,估计每年有300万至500万例严重疾病病例。虽然大多数流感疫苗是通过注射肠胃外给药的,一个缺点是它们在感染部位不会产生强烈的免疫反应,这在大流行中可能变得很重要。鼻内疫苗可以产生局部和全身保护性免疫反应,可以降低成本,加强行政管理的便捷性。先前的研究表明,肠胃外施用的携带M2e蛋白(OMV-M2e)序列的外膜囊泡(OMV)可防止小鼠和雪貂的甲型/PR8流感攻击。在目前的研究中,我们测量了OMV-M2e疫苗鼻内途径对小鼠流感A/PR8株的有效性.我们在用OMV-M2e鼻内接种的小鼠中观察到攻击后的高抗M2eIgG和IgA滴度。此外,我们在接种疫苗的小鼠中观察到Th1/Tc1偏差,Th17/Tc17反应增加,两者均与A/PR8攻击后的生存率相关,并显着降低肺病毒滴度。我们得出的结论是,0MV-M2e疫苗的鼻内途径施用是产生针对甲型流感的保护的有希望的方法,因为它导致与病毒攻击的存活相关的促炎免疫应答增加。
    Influenza is a highly contagious respiratory disease, resulting in an estimated 3 to 5 million cases of severe illness annually. While most influenza vaccines are administered parenterally via injection, one shortcoming is that they do not generate a strong immune response at the site of infection, which can become important in a pandemic. Intranasal vaccines can generate both local and systemic protective immune responses, can reduce costs, and enhance ease of administration. Previous studies showed that parenterally administered outer membrane vesicles (OMVs) that carry sequences of the M2e protein (OMV-M2e) protect against influenza A/PR8 challenge in mice and ferrets. In the current study, we measured the effectiveness of the intranasal route of the OMV-M2e vaccine against the influenza A/PR8 strain in mice. We observed high anti-M2e IgG and IgA titers post-challenge in mice vaccinated intranasally with OMV-M2e. In addition, we observed a Th1/Tc1 bias in the vaccinated mice, and an increased Th17/Tc17 response, both of which correlated with survival to A/PR8 challenge and significantly lower lung viral titers. We conclude that the intranasal-route administration of the OMV-M2e vaccine is a promising approach toward generating protection against influenza A as it leads to an increased proinflammatory immune response correlating with survival to viral challenge.
    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:疫苗接种是预防幼儿流感感染和并发症的主要方法。我们评估了单剂量MEDI3250(鼻内,四价,流感减毒活疫苗)在2016/17流感季节期间在健康的日本儿童中使用。
    方法:在这个多中心中,随机化,双盲,3期研究(jRCT2080223345),2-18岁的参与者接受MEDI3250或安慰剂(2:1),按年龄分层(2-6岁,7-18岁)。主要和次要终点是由流行的野生型毒株或疫苗匹配的毒株引起的确诊的流感症状发作的发生率。分别。安全性结果包括不良事件(AE)和疫苗相关AE的发生率。
    结果:总体而言,910名参与者接受MEDI3250(n=608)或安慰剂(n=302)。对于主要终点(无论流感亚型),流感发病的发生率为25.5%(MEDI3250)和35.9%(安慰剂);相对风险降低,28.8%(95%置信区间,12.5%至42.0%)。对于次要终点(疫苗匹配菌株),发病率为10.9%(MEDI3250)和17.2%(安慰剂);相对风险降低,36.6%(95%置信区间,6.5%至56.8%)。在67.6%(MEDI3250)和63.6%(安慰剂)中发生了诱发的AE。大多数事件为轻度;流鼻涕最常见(59.2%[MEDI3250]和52.6%[安慰剂])。36.0%(MEDI3250)和33.1%(安慰剂)发生非请求性不良事件。最常见的不请自来的疫苗相关不良事件是腹泻(2.3%,两组)。
    结论:与安慰剂相比,MEDI3250对日本儿童流感发作具有更大的预防作用;相对于MEDI3250的先前临床和上市后研究,未观察到新的安全性信号。
    BACKGROUND: Vaccination is the primary method of preventing influenza infection and complications in young children. We evaluated the efficacy and safety of a single dose of MEDI3250 (intranasal, quadrivalent, live attenuated influenza vaccine) in healthy Japanese children during the 2016/17 influenza season.
    METHODS: In this multicenter, randomized, double-blind, phase 3 study (jRCT2080223345), participants aged 2-18 years received MEDI3250 or placebo (2:1), stratified by age (2-6 years, 7-18 years). The primary and secondary endpoints were the incidence of confirmed symptomatic onset of influenza caused by a circulating wild-type strain or by a vaccine-matched strain, respectively. Safety outcomes included the incidence of adverse events (AEs) and vaccine-related AEs.
    RESULTS: Overall, 910 participants received MEDI3250 (n = 608) or placebo (n = 302). For the primary endpoint (regardless of the influenza subtype), the incidence of influenza onset was 25.5 % (MEDI3250) and 35.9 % (placebo); relative risk reduction, 28.8 % (95 % confidence interval, 12.5 %-42.0 %). For the secondary endpoint (vaccine-matched strain), the incidence was 10.9 % (MEDI3250) and 17.2 % (placebo); relative risk reduction, 36.6 % (95 % confidence interval, 6.5 %-56.8 %). Solicited AEs occurred in 67.6 % (MEDI3250) and 63.6 % (placebo). Most events were mild; nasal discharge was most common (59.2 % [MEDI3250] and 52.6 % [placebo]). Unsolicited AEs occurred in 36.0 % (MEDI3250) and 33.1 % (placebo). The most common unsolicited vaccine-related AE was diarrhea (2.3 %, both groups).
    CONCLUSIONS: MEDI3250 had a greater preventive effect against influenza onset in Japanese children than placebo; no new safety signals were observed relative to previous clinical and post-marketing studies of MEDI3250.
    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
    嵌合蛋白,由SARS-CoV-2的保守核衣壳(N)和S2蛋白的两个片段形成,在大肠杆菌中作为重组构建体获得。N片段属于C末端结构域,而S2片段在刺突蛋白的融合后构象中跨越纤维结构。产生的蛋白质,名为S2NDH,能够形成10纳米的球形颗粒,其在与CpGODN-39M混合时形成聚集体。两种制剂均被阳性COVID-19人血清识别。通过鼻内途径施用的S2NDH+ODN-39M制剂在Balb/c小鼠中产生高度免疫原性。它在血清和支气管肺泡液中诱导交叉反应性抗N体液免疫,在Th1模式下。细胞介导的免疫(CMI)也很广泛,甚至对SARS-CoV-1的N蛋白也有阳性反应。然而,未获得针对S2区的中和抗体(NAb)或CMI。作为替代,RBD蛋白作为NAb的诱导物包含在制剂中。通过鼻内途径对小鼠进行评估后,S2NDH+ODN-39M制剂相对于RBD检测到明显的佐剂作用。针对SARS-CoV-2和SARS-CoV-1诱导高水平的NAb。双价配方S2NDH+ODN-39M+RBD,通过鼻内途径给药,构成了一个有吸引力的建议作为加强疫苗的sbecovirus范围。
    A chimeric protein, formed by two fragments of the conserved nucleocapsid (N) and S2 proteins from SARS-CoV-2, was obtained as a recombinant construct in Escherichia coli. The N fragment belongs to the C-terminal domain whereas the S2 fragment spans the fibre structure in the post-fusion conformation of the spike protein. The resultant protein, named S2NDH, was able to form spherical particles of 10 nm, which forms aggregates upon mixture with the CpG ODN-39M. Both preparations were recognized by positive COVID-19 human sera. The S2NDH + ODN-39M formulation administered by the intranasal route resulted highly immunogenic in Balb/c mice. It induced cross-reactive anti-N humoral immunity in both sera and bronchoalveolar fluids, under a Th1 pattern. The cell-mediated immunity (CMI) was also broad, with positive response even against the N protein of SARS-CoV-1. However, neither neutralizing antibodies (NAb) nor CMI against the S2 region were obtained. As alternative, the RBD protein was included in the formulation as inducer of NAb. Upon evaluation in mice by the intranasal route, a clear adjuvant effect was detected for the S2NDH + ODN-39M preparation over RBD. High levels of NAb were induced against SARS-CoV-2 and SARS-CoV-1. The bivalent formulation S2NDH + ODN-39M + RBD, administered by the intranasal route, constitutes an attractive proposal as booster vaccine of sarbecovirus scope.
    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
    人类副流感病毒3型(HPIV3)是一种主要的儿科呼吸道病原体,缺乏可用的疫苗或抗病毒药物。我们通过密码子对去优化(CPD)产生了活的减毒HPIV3疫苗候选物。HPIV3开放阅读框(ORFs)编码核蛋白(N),磷蛋白(P),矩阵(M),融合(F),血凝素-神经氨酸酶(HN),和聚合酶(L)被单独或组合修饰以产生12种病毒,命名为Min-N,Min-P,Min-M,Min-FHN,Min-L,Min-NP,Min-NPM,最小不良贷款,Min-PM,Min-PFHN,Min-MFHN,和Min-PMFHN。N或L的CPD严重降低了体外生长,没有进一步评估。P或M的CPD与体外干扰素(IFN)反应增加和减少有关,分别,但对病毒复制影响不大。在Vero细胞中,F和HN延迟病毒复制的CPD,但最终滴度与野生型(wt)HPIV3相当。在人肺上皮A549细胞中,CPDF和HN诱导更强的IFN应答,病毒滴度降低了100倍,F和HN蛋白的表达显着降低,而不影响N或P或蛋白质在病毒体中的相对包装。仓鼠鼻内感染后,对于携带CPDF和HN的病毒,鼻甲和肺中的复制倾向于减少最多,最大减少约10倍。尽管体内复制减少(体外CPDF和HN的表达降低),所有病毒均诱导与wt相似的血清HPIV3中和抗体滴度,并提供针对HPIV3攻击的完全保护。总之,HPIV3的CPD产生了适合进一步开发的有希望的疫苗候选物。
    Human parainfluenza virus type 3 (HPIV3) is a major pediatric respiratory pathogen lacking available vaccines or antiviral drugs. We generated live-attenuated HPIV3 vaccine candidates by codon-pair deoptimization (CPD). HPIV3 open reading frames (ORFs) encoding the nucleoprotein (N), phosphoprotein (P), matrix (M), fusion (F), hemagglutinin-neuraminidase (HN), and polymerase (L) were modified singly or in combination to generate 12 viruses designated Min-N, Min-P, Min-M, Min-FHN, Min-L, Min-NP, Min-NPM, Min-NPL, Min-PM, Min-PFHN, Min-MFHN, and Min-PMFHN. CPD of N or L severely reduced growth in vitro and was not further evaluated. CPD of P or M was associated with increased and decreased interferon (IFN) response in vitro, respectively, but had little effect on virus replication. In Vero cells, CPD of F and HN delayed virus replication, but final titers were comparable to wild-type (wt) HPIV3. In human lung epithelial A549 cells, CPD F and HN induced a stronger IFN response, viral titers were reduced 100-fold, and the expression of F and HN proteins was significantly reduced without affecting N or P or the relative packaging of proteins into virions. Following intranasal infection in hamsters, replication in the nasal turbinates and lungs tended to be the most reduced for viruses bearing CPD F and HN, with maximum reductions of approximately 10-fold. Despite decreased in vivo replication (and lower expression of CPD F and HN in vitro), all viruses induced titers of serum HPIV3-neutralizing antibodies similar to wt and provided complete protection against HPIV3 challenge. In summary, CPD of HPIV3 yielded promising vaccine candidates suitable for further 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
    粘膜疫苗有可能在呼吸道病原体进入点引发保护性免疫反应,从而防止甚至最初的种子感染。与许可的注射疫苗不同,包含蛋白质亚基的粘膜疫苗仅在开发中。与粘膜疫苗相关的主要挑战之一是鉴定和表征可以有效引发多因素粘膜免疫的安全而有效的粘膜佐剂。在这项研究中,我们测试了NanoSTING,干扰素基因刺激因子(STING)途径的内源性激活剂的脂质体制剂,环鸟苷一磷酸腺苷(cGAMP),作为粘膜佐剂。我们基于用NanoSTING佐剂化的H1抗原(Ag85b和ESAT-6的融合蛋白)配制疫苗。NanoSTING-H1的鼻内免疫在接种疫苗的动物的肺中引起强烈的T细胞应答,其特征在于(a)已知对于控制细菌感染是必需的CXCR3+KLRG1-肺驻留T细胞,(b)分泌IFNγ的CD4+T细胞,这是细胞内杀菌活性所必需的,和(c)分泌IL17的CD4+T细胞,其可以赋予针对多种临床相关的Mtb菌株的保护性免疫。在用雾化结核分枝杆菌Erdman菌株攻击后,鼻内NanoSTING-H1提供与皮下施用活的减毒牛分枝杆菌疫苗菌株Bacille-Calmette-Guérin(BCG)相当的保护。我们的结果表明,NanoSTING佐剂化的蛋白质疫苗可以引发多因素免疫反应,保护免受结核分枝杆菌感染。
    Mucosal vaccines have the potential to elicit protective immune responses at the point of entry of respiratory pathogens, thus preventing even the initial seed infection. Unlike licensed injectable vaccines, mucosal vaccines comprising protein subunits are only in development. One of the primary challenges associated with mucosal vaccines has been identifying and characterizing safe yet effective mucosal adjuvants that can effectively prime multi-factorial mucosal immunity. In this study, we tested NanoSTING, a liposomal formulation of the endogenous activator of the stimulator of interferon genes (STING) pathway, cyclic guanosine adenosine monophosphate (cGAMP), as a mucosal adjuvant. We formulated a vaccine based on the H1 antigen (fusion protein of Ag85b and ESAT-6) adjuvanted with NanoSTING. Intranasal immunization of NanoSTING-H1 elicited a strong T-cell response in the lung of vaccinated animals characterized by (a) CXCR3+ KLRG1- lung resident T cells that are known to be essential for controlling bacterial infection, (b) IFNγ-secreting CD4+ T cells which is necessary for intracellular bactericidal activity, and (c) IL17-secreting CD4+ T cells that can confer protective immunity against multiple clinically relevant strains of Mtb. Upon challenge with aerosolized Mycobacterium tuberculosis Erdman strain, intranasal NanoSTING-H1 provides protection comparable to subcutaneous administration of the live attenuated Mycobacterium bovis vaccine strain Bacille-Calmette-Guérin (BCG). Our results indicate that NanoSTING adjuvanted protein vaccines can elicit a multi-factorial immune response that protects from infection by M. tuberculosis.
    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
    紧急研究创新的严重急性呼吸道冠状病毒-2(SARS-CoV-2)疫苗,可以成功地防止各种新出现的变种,特别是Omicron变体及其亚变体,是必要的。这里,我们设计了一种嵌合腺病毒载体疫苗,命名为Ad5-Beta/Delta.这种疫苗是通过整合来自Delta变体的受体结合结构域而产生的,有L452R和T478K突变,变成β变体的完整刺突蛋白。用Ad5-β/Deta疫苗进行的肌内(IM)和鼻内(IN)疫苗接种均诱导了针对包含OmicronBA.5的变体的强大的广谱中和。用Ad5-Beta/Delta疫苗进行的IN免疫表现出优异的粘膜免疫,表现为较高的分泌性IgA抗体和呼吸道中更多的组织驻留记忆T细胞(TRM)。Ad5-β/δ疫苗的IM和IN递送的组合能够协同地引发更强的全身和粘膜免疫应答。此外,在两个剂量的mRNA或亚单位重组蛋白疫苗后,Ad5-Beta/Delta疫苗接种显示出更有效的增强作用,表明其在异源疫苗接种中作为加强剂的利用能力。这些结果定量的Ad5-β/Delta疫苗作为有利的疫苗可以提供相对于关注的SARS-CoV-2pre-Omicron变体和包含BA.5的Omicron亚变体的保护性免疫。
    Urgent research into innovative severe acute respiratory coronavirus-2 (SARS-CoV-2) vaccines that may successfully prevent various emerging emerged variants, particularly the Omicron variant and its subvariants, is necessary. Here, we designed a chimeric adenovirus-vectored vaccine named Ad5-Beta/Delta. This vaccine was created by incorporating the receptor-binding domain from the Delta variant, which has the L452R and T478K mutations, into the complete spike protein of the Beta variant. Both intramuscular (IM) and intranasal (IN) vaccination with Ad5-Beta/Deta vaccine induced robust broad-spectrum neutralization against Omicron BA.5-included variants. IN immunization with Ad5-Beta/Delta vaccine exhibited superior mucosal immunity, manifested by higher secretory IgA antibodies and more tissue-resident memory T cells (TRM) in respiratory tract. The combination of IM and IN delivery of the Ad5-Beta/Delta vaccine was capable of synergically eliciting stronger systemic and mucosal immune responses. Furthermore, the Ad5-Beta/Delta vaccination demonstrated more effective boosting implications after two dosages of mRNA or subunit recombinant protein vaccine, indicating its capacity for utilization as a booster shot in the heterologous vaccination. These outcomes quantified Ad5-Beta/Delta vaccine as a favorable vaccine can provide protective immunity versus SARS-CoV-2 pre-Omicron variants of concern and BA.5-included Omicron subvariants.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号