intranasal vaccination

鼻内疫苗接种
  • 文章类型: Journal Article
    这项研究的重点是使用佐剂纳米颗粒递送猪甲型流感病毒(SwIAV)的鼻内疫苗平台的开发和表征。该疫苗采用全灭活的H1N1N2SwIAV作为抗原,STING激动剂ADU-S100作为佐剂,两个表面都吸附或包封在甘露糖-壳聚糖纳米颗粒(mChit-NP)中。mChit-NP的优化包括评估尺寸,zeta电位,和细胞毒性,抗原与NP的质量比为1:9,证明了适用于鼻内接种的高负载功效和非细胞毒性特性。在一项异源H1N1猪攻击试验中,mChit-NP鼻内疫苗在呼吸道诱导交叉反应的sIgA抗体,超过那些商业SwIAV疫苗。封装的mChit-NP疫苗诱导高病毒特异性中和抗体和强大的细胞免疫反应,而吸附疫苗引发特异性高IgG和血凝素抑制抗体。重要的是,与商业猪流感疫苗相比,两种mChit-NP疫苗降低了鼻腔中攻击异源病毒的复制。总之,一种新型鼻内mChit-NP疫苗平台激活了免疫系统的两个分支,是猪流感疫苗设计的一项重大进展,证明其对猪免疫的潜在有效性。
    This study focuses on the development and characterization of an intranasal vaccine platform using adjuvanted nanoparticulate delivery of swine influenza A virus (SwIAV). The vaccine employed whole inactivated H1N2 SwIAV as an antigen and STING-agonist ADU-S100 as an adjuvant, with both surface adsorbed or encapsulated in mannose-chitosan nanoparticles (mChit-NPs). Optimization of mChit-NPs included evaluating size, zeta potential, and cytotoxicity, with a 1:9 mass ratio of antigen to NP demonstrating high loading efficacy and non-cytotoxic properties suitable for intranasal vaccination. In a heterologous H1N1 pig challenge trial, the mChit-NP intranasal vaccine induced cross-reactive sIgA antibodies in the respiratory tract, surpassing those of a commercial SwIAV vaccine. The encapsulated mChit-NP vaccine induced high virus-specific neutralizing antibody and robust cellular immune responses, while the adsorbed vaccine elicited specific high IgG and hemagglutinin inhibition antibodies. Importantly, both the mChit-NP vaccines reduced challenge heterologous viral replication in the nasal cavity higher than commercial swine influenza vaccine. In summary, a novel intranasal mChit-NP vaccine platform activated both the arms of the immune system and is a significant advancement in swine influenza vaccine design, demonstrating its potential effectiveness for pig immunization.
    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)的全球紧急情况刺激了全球广泛的努力,以开发预防严重急性呼吸道综合症冠状病毒2(SARS-CoV-2)的疫苗。我们对这一全球努力的贡献包括开发多样化的纳米载体库,作为脂质纳米颗粒(LNP)的替代品,包括纳米乳液(NE)和纳米胶囊(NC),目的是保护和递送信使核糖核酸(mRNA)用于鼻疫苗接种目的。通过一系列体外和体内实验,对各种原型进行了严格的筛选,包括细胞转染的评估,细胞毒性,和肌内施用用于蛋白质翻译的模型mRNA。因此,两个有希望的候选人被确定为鼻腔给药。其中之一是结合了可电离脂质(C12-200)和阳离子脂质(DOTAP)的NE,两者都旨在浓缩mRNA,和DOPE一起,已知这有助于内体逃逸。该NE表现出120nm的尺寸和高的正表面电荷(+50mV)。另一个候选物是包含相同组分并赋予硫酸葡聚糖壳的NC制剂。该制剂显示130nm的尺寸和中等的负表面电荷(-16mV)。在鼻内施用与所述NE和NC的优化版本相关的编码卵清蛋白(mOVA)的mRNA后,观察到稳健的抗原特异性CD8+T细胞应答.这些发现强调了NE和聚合物NC在推进mRNA疫苗开发以对抗传染病方面的潜力。
    The global emergency of coronavirus disease 2019 (COVID-19) has spurred extensive worldwide efforts to develop vaccines for protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Our contribution to this global endeavor involved the development of a diverse library of nanocarriers, as alternatives to lipid nanoparticles (LNPs), including nanoemulsions (NEs) and nanocapsules (NCs), with the aim of protecting and delivering messenger ribonucleic acid (mRNA) for nasal vaccination purposes. A wide range of prototypes underwent rigorous screening through a series of in vitro and in vivo experiments, encompassing assessments of cellular transfection, cytotoxicity, and intramuscular administration of a model mRNA for protein translation. As a result, two promising candidates were identified for nasal administration. One of them was a NE incorporating a combination of an ionizable lipid (C12-200) and cationic lipid (DOTAP), both intended to condense mRNA, along with DOPE, which is known to facilitate endosomal escape. This NE exhibited a size of 120 nm and a highly positive surface charge (+ 50 mV). Another candidate was an NC formulation comprising the same components and endowed with a dextran sulfate shell. This formulation showed a size of 130 nm and a moderate negative surface charge (-16 mV). Upon intranasal administration of mRNA encoding for ovalbumin (mOVA) associated with optimized versions of the said NE and NCs, a robust antigen-specific CD8 + T cell response was observed. These findings underscore the potential of NEs and polymeric NCs in advancing mRNA vaccine development for combating infectious 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
    随着2019年冠状病毒病(COVID-19)浪潮的不断涌现,科学界已经开发出一种疫苗,可以在病毒靶向器官提供广谱保护,以抑制疾病的传播和保护疾病的发展。在本研究中,一种二价的严重急性呼吸综合征冠状病毒2(SARS-CoV-2)疫苗,含有来自武汉-1的刺突的受体结合域(RBD)蛋白和装载在纳米颗粒中的omicronBA.1,二价RBDNP,已开发。使用体内模型评估该疫苗候选物的免疫原性和保护效力。结果显示,接受鼻内cGAMP-佐剂化的二价RBD-NP疫苗的小鼠引发稳健且持久的抗体应答。刺激的抗体广泛地中和了上呼吸道和下呼吸道中的祖先菌株和关注的变体(δ和omicronBA.1)。此外,免疫小鼠在其肺组织中产生T细胞反应。重要的是,用这种疫苗候选物进行鼻内免疫有效地保护小鼠免受由武汉-1和BA.1病毒引起的鼻感染。仍然易受鼻感染的免疫小鼠没有出现任何症状。这是因为鼻腔中的活化反应显著抑制了病毒的产生。另一个词是这种鼻疫苗完全保护小鼠免受疾病发展和死亡。因此,二价RBD疫苗平台有可能发展成为抗SARS-CoV-2通用疫苗。
    With the continuous emergence of coronavirus disease 2019 (COVID-19) waves, the scientific community has developed a vaccine that offers broad-spectrum protection at virus-targeted organs for inhibiting the transmission and protection of disease development. In the present study, a bivalent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine containing receptor-binding domain (RBD) protein of spike from Wuhan-1 and omicron BA.1 loaded in nanoparticles, bivalent RBD NPs, was developed. The immunogenicity and protective efficacy of this vaccine candidate were evaluated using an in vivo model. Results showed that mice that received intranasal cGAMP-adjuvanted bivalent RBD-NPs vaccine elicited robust and durable antibody responses. The stimulated antibody broadly neutralized the ancestral strain and variants of concerns (delta and omicron BA.1) in the upper and lower respiratory tracts. Furthermore, the immunized mice developed T-cell response in their lung tissue. Importantly, intranasal immunization with this vaccine candidate efficiently protected mice from nasal infection caused by both Wuhan-1 and BA.1 viruses. Immunized mice that remained susceptible to nasal infection did not develop any symptoms. This is because activated responses in the nasal cavity significantly suppressed virus production. Another word is this nasal vaccine completely protected the mice from disease development and mortality. Therefore, the bivalent RBD vaccine platform has potential to be developed into an anti-SARS-CoV-2 universal vaccine.
    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
    严重急性呼吸道综合征冠状病毒2(SARS-CoV-2)对全球人类健康构成重大威胁。开发疫苗以减少病毒对公众健康的影响至关重要,经济,和社会,规范SARS-CoV-2的传播。B型流感病毒(IBV)可用作不依赖于当前循环的A型流感病毒株的载体。在这项研究中,我们通过将受体结合域(RBD)插入非结构蛋白1(NS1)截短的基因(rIBV-NS110-RBD),构建了基于IBV的载体疫苗。随后,我们评估了它的安全性,免疫原性,和对小鼠SARS-CoV-2的保护功效,并观察到它在小鼠模型中是安全的。鼻内施用重组rIBV-NS110-RBD疫苗在小鼠中诱导高水平的SARS-CoV-2特异性IgA和IgG抗体以及T细胞介导的免疫。施用两种剂量的鼻内rIBV-NS110-RBD疫苗显著降低了小鼠的病毒载量和肺损伤。这种基于IBV的新型疫苗提供了一种控制SARS-CoV-2大流行的新方法。
    Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a significant threat to human health globally. It is crucial to develop a vaccine to reduce the effect of the virus on public health, economy, and society and regulate the transmission of SARS-CoV-2. Influenza B virus (IBV) can be used as a vector that does not rely on the current circulating influenza A strains. In this study, we constructed an IBV-based vector vaccine by inserting a receptor-binding domain (RBD) into a non-structural protein 1 (NS1)-truncated gene (rIBV-NS110-RBD). Subsequently, we assessed its safety, immunogenicity, and protective efficacy against SARS-CoV-2 in mice, and observed that it was safe in a mouse model. Intranasal administration of a recombinant rIBV-NS110-RBD vaccine induced high levels of SARS-CoV-2-specific IgA and IgG antibodies and T cell-mediated immunity in mice. Administering two doses of the intranasal rIBV-NS110-RBD vaccine significantly reduced the viral load and lung damage in mice. This novel IBV-based vaccine offers a novel approach for controlling the SARS-CoV-2 pandemic.
    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
    有效癌症治疗的标志是防止肿瘤复发和转移到远端器官,这是大多数癌症死亡的原因。然而,原发性肿瘤切除术有望治愈,因为大多数实体肿瘤在实验和临床上都显示出加速转移到包括肺在内的远端器官。在这项研究中,我们评估了我们的工程化鼻纳米疫苗(CpG-NP-Tag)在减少原发肿瘤切除导致的加速肺转移中的功效.制备胞嘧啶-磷酸-鸟嘌呤寡核苷酸[CpGODN]-缀合的纳米颗粒[NP],包封肿瘤抗原[Tag](CpG-NP-Tag),并在鼻内递送后使用同基因小鼠乳腺肿瘤模型进行体内测试。我们发现我们的鼻纳米疫苗(CpG-NP-Tag),与原发性乳腺肿瘤切除术后施用的对照NP相比,接受手术的雌性BALB/c小鼠(手术小鼠)的肺转移显着减少。在手术和非手术小鼠中对疫苗功效的评估表明,原发性肿瘤切除术减少了CD11b单核细胞来源的抑制样细胞在肺中的积累,与非手术小鼠相比,允许手术小鼠肺部疫苗引发的T细胞(IFN-γCD8T细胞)的浸润增加。这些发现表明,鼻用疫苗的靶向递送与原发性肿瘤的标准手术相结合是针对建立肺转移的合理辅助治疗。
    A hallmark of effective cancer treatment is the prevention of tumor reoccurrence and metastasis to distal organs, which are responsible for most cancer deaths. However, primary tumor resection is expected to be curative as most solid tumors have been shown both experimentally and clinically to accelerate metastasis to distal organs including the lungs. In this study, we evaluated the efficacy of our engineered nasal nano-vaccine (CpG-NP-Tag) in reducing accelerated lung metastasis resulting from primary tumor resection. Cytosine-phosphate-guanine oligonucleotide [CpG ODN]-conjugated nanoparticle [NP] encapsulating tumor antigen [Tag] (CpG-NP-Tag) was manufactured and tested in vivo using a syngeneic mouse mammary tumor model following intranasal delivery. We found that our nasal nano-vaccine (CpG-NP-Tag), compared to control NPs administered after primary mammary tumor resection, significantly reduced lung metastasis in female BALB/c mice subjected to surgery (surgery mice). An evaluation of vaccine efficacy in both surgery and non-surgery mice revealed that primary tumor resection reduces CD11b+ monocyte-derived suppressor-like cell accumulation in the lungs, allowing increased infiltration of vaccine-elicited T cells (IFN-γ CD8+ T cells) in the lungs of surgery mice compared to non-surgery mice. These findings suggest that the combination of the target delivery of a nasal vaccine in conjunction with the standard surgery of primary tumors is a plausible adjunctive treatment against the establishment of lung metastasis.
    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)仍然是影响撒哈拉以南非洲地区家禽的危重疾病。在一些国家,反复爆发对当地经济和粮食安全产生重大影响。最近,我们从新城疫病毒(NDV)的埃塞俄比亚分离株中开发了编码融合蛋白的腺病毒载体疫苗。设计了腺病毒载体,在加拿大国际发展研究中心(IDRC)资助的家畜疫苗创新基金倡议的背景下开发了制造工艺。工业相关的重组疫苗技术平台正在转移到国家兽医研究所(埃塞俄比亚)用于兽医应用。这里,提出了一种使用在搅拌罐生物反应器中培养的HEK293SF悬浮细胞用于疫苗生产的制造方法。考虑到供应链的局限性,评估了无血清培养基选择的选择.流线型的下游过程,包括过滤,超滤,并开发了浓缩步骤。培养上清液中的高体积产量(感染滴度高达5×109TCID50/mL),最终制剂以1010TCID50/mL制备,液体或冻干形式。该液体制剂适合并安全用于粘膜疫苗接种,并且在37°C下稳定1周。液体和冻干制剂在4°C储存6个月后都是稳定的。我们证明,通过鼻腔滴注腺病毒载体可以保护鸡免受NDV的致命攻击。总的来说,开发了腺病毒载体疫苗的生产工艺,使用方便的递送途径确定保护剂量。建立配方和储存条件,并实施质量控制方案。
    Newcastle disease (ND) remains a critical disease affecting poultry in sub-Saharan Africa. In some countries, repeated outbreaks have a major impact on local economies and food security. Recently, we developed an adenovirus-vectored vaccine encoding the Fusion protein from an Ethiopian isolate of Newcastle disease virus (NDV). The adenoviral vector was designed, and a manufacturing process was developed in the context of the Livestock Vaccine Innovation Fund initiative funded by the International Development Research Centre (IDRC) of Canada. The industrially relevant recombinant vaccine technology platform is being transferred to the National Veterinary Institute (Ethiopia) for veterinary applications. Here, a manufacturing process using HEK293SF suspension cells cultured in stirred-tank bioreactors for the vaccine production is proposed. Taking into consideration supply chain limitations, options for serum-free media selection were evaluated. A streamlined downstream process including a filtration, an ultrafiltration, and a concentration step was developed. With high volumetric yields (infectious titers up to 5 × 109 TCID50/mL) in the culture supernatant, the final formulations were prepared at 1010 TCID50/mL, either in liquid or lyophilized forms. The liquid formulation was suitable and safe for mucosal vaccination and was stable for 1 week at 37 °C. Both the liquid and lyophilized formulations were stable after 6 months of storage at 4 °C. We demonstrate that the instillation of the adenoviral vector through the nasal cavity can confer protection to chickens against a lethal challenge with NDV. Overall, a manufacturing process for the adenovirus-vectored vaccine was developed, and protective doses were determined using a convenient route of delivery. Formulation and storage conditions were established, and quality control protocols were implemented.
    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
    甲型流感病毒(IAV)是一种普遍存在且不断发展的呼吸道病原体。模拟自然感染的鼻内疫苗接种是控制IAV的有吸引力的策略。准确靶向多个保守结构域的多表位疫苗具有拓宽当前季节性流感疫苗的保护范围和降低产生逃逸突变体的风险的潜力。这里,来自基质蛋白2胞外域(M2e)和血凝素茎结构域(HA2)的多个线性表位与幽门螺杆菌铁蛋白融合,自组装纳米载体和粘膜佐剂,开发多表位纳米疫苗。通过鼻内递送,原核表达的多表位纳米疫苗引起持久的粘膜免疫,广泛的体液免疫,和强大的细胞免疫,没有任何佐剂,并赋予小鼠对IAV的H3N2和H1N1亚型的完全保护作用。重要的是,这种鼻内多表位纳米疫苗触发记忆B细胞反应,导致免疫后分泌性免疫球蛋白A(sIgA)和血清免疫球蛋白G(IgG)水平持续超过五个月。因此,这种基于铁蛋白的鼻内多表位纳米疫苗代表了一种有希望的对抗呼吸道病原体的方法。
    The influenza A virus (IAV) is a ubiquitous and continuously evolving respiratory pathogen. The intranasal vaccination mimicking natural infections is an attractive strategy for controlling IAVs. Multiepitope vaccines accurately targeting multiple conserved domains have the potential to broaden the protective scope of current seasonal influenza vaccines and reduce the risk of generating escape mutants. Here, multiple linear epitopes from the matrix protein 2 ectodomain (M2e) and the hemagglutinin stem domain (HA2) are fused with the Helicobacter pylori ferritin, a self-assembled nanocarrier and mucosal adjuvant, to develop a multiepitope nanovaccine. Through intranasal delivery, the prokaryotically expressed multiepitope nanovaccine elicits long-lasting mucosal immunity, broad humoral immunity, and robust cellular immunity without any adjuvants, and confers complete protection against H3N2 and H1N1 subtypes of IAV in mice. Importantly, this intranasal multiepitope nanovaccine triggers memory B-cell responses, resulting in secretory immunoglobulin A (sIgA) and serum immunoglobulin G (IgG) levels persisting for more than five months post-immunization. Therefore, this intranasal ferritin-based multiepitope nanovaccine represents a promising approach to combating respiratory pathogens.
    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
    呼吸道病原体的粘膜疫苗接种提供了有效的保护,因为它们刺激感染部位的局部细胞和体液免疫。目前,鼻内疫苗接种的主要限制是使用能够承受粘膜施加的恶劣环境的有效佐剂。在这里,我们描述了使用独特的生物聚合物的功效,N-二氢半乳糖壳聚糖(GC),作为抗呼吸道感染的鼻粘膜疫苗佐剂。具体来说,我们将GC与重组SARS-CoV-2三聚体刺突(S)和核衣壳(NC)蛋白混合,以鼻内接种K18-hACE2转基因小鼠,与Addavax(AV)相比,相当于MF-59。与AV相比,鼻内应用GC诱导了一个强大的,全身抗原特异性抗体反应,并增加颈部淋巴结中T细胞的数量。此外,接种GC+S+NC的动物对致命的SARS-CoV-2攻击有很大的抵抗力,并且发病率和死亡率大大降低。动物体重和行为在感染后22天恢复正常。相比之下,动物鼻内接种AV+S+NC经历了严重的体重减轻,死亡率,呼吸窘迫,感染后6天以上无存活。我们的发现表明,GC可以作为抗SARS-CoV-2和潜在的其他呼吸道病毒的有效粘膜疫苗佐剂。重要声明::我们证明了一种独特的生物聚合物,N-二氢半乳糖壳聚糖(GC),是一种有效的抗呼吸道感染的鼻粘膜疫苗佐剂。具体来说,我们将GC与重组SARS-CoV-2三聚体刺突(S)和核衣壳(NC)蛋白混合,以鼻内接种K18-hACE2转基因小鼠,与Addavax(AV)相比。与AV相比,GC诱导了一个强大的,全身抗原特异性抗体反应,并增加颈部淋巴结中T细胞的数量。大约90%的接种GCSNC的动物在致命的SARS-CoV-2攻击中幸存下来,并在感染后22天保持健康。而接种AV+S+NC的动物经历了严重的体重减轻和呼吸窘迫,全部在感染后6天内死亡。我们的发现表明,GC是针对SARS-CoV-2和潜在的其他呼吸道病毒的有效粘膜疫苗佐剂。
    Mucosal vaccinations for respiratory pathogens provide effective protection as they stimulate localized cellular and humoral immunities at the site of infection. Currently, the major limitation of intranasal vaccination is using effective adjuvants capable of withstanding the harsh environment imposed by the mucosa. Herein, we describe the efficacy of using a unique biopolymer, N-dihydrogalactochitosan (GC), as a nasal mucosal vaccine adjuvant against respiratory infections. Specifically, we mixed GC with recombinant SARS-CoV-2 trimeric spike (S) and nucleocapsid (NC) proteins to intranasally vaccinate K18-hACE2 transgenic mice, in comparison with Addavax (AV), an MF-59 equivalent. In contrast to AV, intranasal application of GC induces a robust, systemic antigen-specific antibody response and increases the number of T cells in the cervical lymph nodes. Moreover, GC+S+NC-vaccinated animals were largely resistant to the lethal SARS-CoV-2 challenge and experienced drastically reduced morbidity and mortality, with animal weights and behavior returning to normal 22 days post-infection. In contrast, animals intranasally vaccinated with AV+S+NC experienced severe weight loss, mortality, and respiratory distress, with none surviving beyond 6 days post-infection. Our findings demonstrate that GC can serve as a potent mucosal vaccine adjuvant against SARS-CoV-2 and potentially other respiratory viruses. STATEMENT OF SIGNIFICANCE: We demonstrated that a unique biopolymer, N-dihydrogalactochitosan (GC), was an effective nasal mucosal vaccine adjuvant against respiratory infections. Specifically, we mixed GC with recombinant SARS-CoV-2 trimeric spike (S) and nucleocapsid (NC) proteins to intranasally vaccinate K18-hACE2 transgenic mice, in comparison with Addavax (AV). In contrast to AV, GC induces a robust, systemic antigen-specific antibody response and increases the number of T cells in the cervical lymph nodes. About 90 % of the GC+S+NC-vaccinated animals survived the lethal SARS-CoV-2 challenge and remained healthy 22 days post-infection, while the AV+S+NC-vaccinated animals experienced severe weight loss and respiratory distress, and all died within 6 days post-infection. Our findings demonstrate that GC is a potent mucosal vaccine adjuvant against SARS-CoV-2 and potentially other respiratory viruses.
    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
    背景:由于鼻粘膜是COVID-19的初始感染部位,因此鼻内疫苗比常规疫苗更有利。在最近的临床研究中,鼻内免疫已显示产生更高的中和抗体;然而,缺乏上气道灭菌免疫的证据。以前,我们开发了编码SARS-CoV-2(rMeV-S)刺突蛋白的重组麻疹病毒,引发针对SARS-CoV-2的体液和细胞免疫应答。
    目的:在本研究中,我们的目标是提供一项针对麻疹病毒平台和注射途径的鼻疫苗实验。
    方法:制备表达rMeV-S的重组麻疹病毒,并通过肌内或鼻内注射对叙利亚金仓鼠施用5×105PFU的rMeV-S。随后,仓鼠用1×105PFU的SARS-CoV-2接种并在感染后4天安乐死。测定血清中的中和抗体和RBD特异性IgG以及支气管肺泡灌洗液(BALF)中的RBD特异性IgA,通过定量逆转录PCR(qRT-PCR)分析和上呼吸道和肺中的病毒滴度测量来确定SARS-CoV-2的清除能力。对来自实验仓鼠的肺样品进行免疫组织化学和组织病理学检查。
    结果:rMeV-S的鼻内免疫引发保护性免疫反应,减轻病毒诱导的病理生理学,例如仓鼠的体重减少和肺重量增加。此外,肺免疫组织化学表明,鼻内rMeV-S免疫诱导有效的SARS-CoV-2清除,与病毒RNA含量相关,通过qRT-PCR确定,在肺部和鼻腔冲洗样本中,肺部SARS-CoV-2病毒滴度,洗鼻器,BALF样本,血清RBD特异性IgG浓度,和BALF中RBD特异性IgA浓度。
    结论:由于典型的病毒感染途径,基于麻疹病毒平台的鼻内疫苗是一种有前途的策略,疫苗管理的便利性,以及它引发的强烈免疫反应。
    As the nasal mucosa is the initial site of infection for COVID-19, intranasal vaccines are more favorable than conventional vaccines. In recent clinical studies, intranasal immunization has been shown to generate higher neutralizing antibodies; however, there is a lack of evidence on sterilizing immunity in the upper airway. Previously, we developed a recombinant measles virus encoding the spike protein of SARS-CoV-2 (rMeV-S), eliciting humoral and cellular immune responses against SARS-CoV-2.
    In this study, we aim to provide an experiment on nasal vaccines focusing on a measles virus platform as well as injection routes.
    Recombinant measles viruses expressing rMeV-S were prepared, and 5 × 105 PFUs of rMeV-S were administered to Syrian golden hamsters via intramuscular or intranasal injection. Subsequently, the hamsters were challenged with inoculations of 1 × 105 PFUs of SARS-CoV-2 and euthanized 4 days post-infection. Neutralizing antibodies and RBD-specific IgG in the serum and RBD-specific IgA in the bronchoalveolar lavage fluid (BALF) were measured, and SARS-CoV-2 clearance capacity was determined via quantitative reverse-transcription PCR (qRT-PCR) analysis and viral titer measurement in the upper respiratory tract and lungs. Immunohistochemistry and histopathological examinations of lung samples from experimental hamsters were conducted.
    The intranasal immunization of rMeV-S elicits protective immune responses and alleviates virus-induced pathophysiology, such as body weight reduction and lung weight increase in hamsters. Furthermore, lung immunohistochemistry demonstrated that intranasal rMeV-S immunization induces effective SARS-CoV-2 clearance that correlates with viral RNA content, as determined by qRT-PCR, in the lung and nasal wash samples, SARS-CoV-2 viral titers in lung, nasal wash, BALF samples, serum RBD-specific IgG concentration, and RBD-specific IgA concentration in the BALF.
    An intranasal vaccine based on the measles virus platform is a promising strategy owing to the typical route of infection of the virus, the ease of administration of the vaccine, and the strong immune response it elicits.
    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
    鼻内粘膜疫苗能更有效地诱导抗SARS-CoV-2的粘膜免疫应答。这里,我们在仓鼠中显示,鼻内亚单位粘膜疫苗加强与β变体S1可以防止体重减轻,除了减少病毒载量,这不能在没有发展为COVID样疾病的猕猴中进行研究。针对病毒载量和体重减轻的保护功效与血清抗体滴度相关。检测到性别偏见,因为女性的免疫反应和对病毒载量的保护作用大于男性。我们还发现,来自武汉菌株的S1引发较低的体液免疫反应,针对β变体,并导致对β病毒攻击的保护作用降低,表明匹配抗原的重要性。粘膜疫苗在上呼吸道的更高功效以及在疫苗保护中需要考虑性别差异,这对未来改进的COVID-19疫苗的开发很重要。
    Intranasal mucosal vaccines can more effectively induce mucosal immune responses against SARS-CoV-2. Here, we show in hamsters that an intranasal subunit mucosal vaccine boost with the beta variant S1 can prevent weight loss, in addition to reducing viral load, which cannot be studied in macaques that don\'t develop COVID-like disease. Protective efficacy against both viral load and weight loss correlated with serum antibody titers. A sex bias was detected in that immune responses and protection against viral load were greater in females than males. We also found that priming with S1 from the Wuhan strain elicited lower humoral immune responses against beta variant and led to less protection against beta viral challenge, suggesting the importance of matched antigens. The greater efficacy of mucosal vaccines in the upper respiratory tract and the need to consider sex differences in vaccine protection are important in the development of future improved COVID-19 vaccines.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号