文章11月
Rapid Induction of Long-Lasting Systemic and Mucosal Immunity via Thermostable Microneedle-Mediated Chitosan Oligosaccharide-Encapsulated DNA Nanoparticles
通过恒温微针诱导壳聚糖寡糖包被 DNA 纳米粒子快速诱导持久的全身和黏膜免疫力Click to copy article link
点击复制文章链接Article link copied!
点击复制文章链接Article link copied!
- Minchao Li 李敏超Minchao LiSchool of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, ChinaMore by Minchao Li
- Li Yang 李阳Li YangSchool of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, ChinaMore by Li Yang
- Congcong Wang 王聪聪Congcong WangSchool of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, ChinaMore by Congcong Wang
- Mingting Cui 崔明婷Mingting CuiSchool of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, ChinaMore by Mingting Cui
- Ziyu Wen 温子玉Ziyu WenSchool of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, ChinaMore by Ziyu Wen
- Zhiheng Liao 廖志恒Zhiheng Liao 廖志恒School of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, China
中山大学公共卫生学院(深圳);中国深圳,518107More by Zhiheng Liao
廖志恒的更多作品 - Zirong Han 韩子荣Zirong HanSchool of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, ChinaMore by Zirong Han
- Yangguo ZhaoYangguo ZhaoSchool of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, ChinaMore by Yangguo Zhao
- Bing LangBing LangSchool of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, ChinaMore by Bing Lang
- Hongzhong Chen 陈红忠Hongzhong ChenSchool of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, ChinaMore by Hongzhong Chen
- Jun Qian 钱俊Jun QianSchool of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, ChinaMore by Jun Qian
- Yuelong Shu* 舒跃龙*Yuelong Shu*E-mail: shuylong@mail.sysu.edu.cnSchool of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, ChinaKey Laboratory of Pathogen Infection Prevention and Control (MOE), State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 102629, P.R. ChinaMore by Yuelong Shu
- Xiaowei Zeng* 曾小伟*Xiaowei Zeng 曾小伟*E-mail: zengxw23@mail.sysu.edu.cnSchool of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
中山大学药学院(深圳),中国深圳 518107
*E-mail:zengxw23@mail.sysu.edu.cnMore by Xiaowei Zeng
曾小伟的更多作品 - Caijun Sun* 孙才军*Caijun Sun*E-mail: suncaijun@mail.sysu.edu.cnSchool of Public Health (Shenzhen), Sun Yat-sen University; Shenzhen, 518107, ChinaKey Laboratory of Tropical Disease Control (Sun Yat-sen university), Ministry of Education, Guangzhou 514400, ChinaSchool of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University; Guangdong 518107, ChinaMore by Caijun Sun
Abstract 摘要
点击复制章节链接Section link copied!
Most existing vaccines, delivered by intramuscular injection (IM), are typically associated with stringent storage requirements under cold-chain distribution and professional administration by medical personnel and often result in the induction of weak mucosal immunity. In this context, we reported a microneedle (MN) patch to deliver chitosan oligosaccharide (COS)-encapsulated DNA vaccines (DNA@COS) encoding spike and nucleocapsid proteins of SARS-CoV-2 as a vaccination technology. Compared with IM immunization, intradermal administration via the MN-mediated DNA vaccine effectively induces a comparable level of neutralizing antibody against SARS-CoV-2 variants. Surprisingly, we found that MN-mediated intradermal immunization elicited superior systemic and mucosal T cell immunity with enhanced magnitude, polyfunctionality, and persistence. Importantly, the DNA@COS nanoparticle vaccine loaded in an MN patch can be stored at room temperature for at least 1 month without a significant decrease of its immunogenicity. Mechanically, our strategy enhanced dendritic cell maturation and antiviral immunity by activating the cGAS-STING-mediated IFN signaling pathway. In conclusion, this work provides valuable insights for the rapid development of an easy-to-administer and thermostable technology for mucosal vaccines.
现有的大多数疫苗都是通过肌肉注射(IM)的方式递送的,通常都有严格的冷链配送和医务人员专业管理的储存要求,而且往往会导致诱导出微弱的粘膜免疫。在此背景下,我们报道了一种微针(MN)贴片,用于递送壳聚糖寡糖(COS)包裹的编码SARS-CoV-2尖头蛋白和核壳蛋白的DNA疫苗(DNA@COS),作为一种疫苗接种技术。与即时免疫相比,通过 MN 介导的 DNA 疫苗皮内注射能有效诱导出与 SARS-CoV-2 变体相当水平的中和抗体。令人惊讶的是,我们发现以 MN 为介导的皮内免疫可诱导出卓越的全身和粘膜 T 细胞免疫,且免疫强度、多功能性和持久性均有所增强。重要的是,载入 MN 贴片的 DNA@COS 纳米粒子疫苗可在室温下保存至少 1 个月,其免疫原性不会明显降低。从机制上讲,我们的策略通过激活 cGAS-STING 介导的 IFN 信号通路,增强了树突状细胞成熟和抗病毒免疫力。总之,这项工作为快速开发易于给药且可恒温的粘膜疫苗技术提供了宝贵的见解。
This publication is licensed under the terms of your
institutional subscription.
Request reuse permissions.
本出版物根据您的机构订阅条款获得许可。申请重用许可。
版权所有 © 2023 美国化学学会
Introduction 导言
点击复制章节链接Section link copied!
冠状病毒病 2019(COVID-19)、痘(以前称为猴痘)和禽流感等传染病的频繁出现已成为全球公共卫生的巨大危机。为了最终控制这些传染病,在普通人群中大规模接种疫苗被认为是建立群体免疫力的最具成本效益的干预措施。迄今为止,世界卫生组织已批准了至少 15 种 COVID-19 疫苗用于临床,其中大多数通过肌肉注射(IM)给药。然而,IM 疫苗接种有几个缺点,通常需要严格的冷链配送储存条件和医务人员的专业接种,而且通常会导致黏膜免疫力低下。(1-3)此外,许多人由于对针头的恐惧而不愿意接种这种疫苗方式,这源于 IM 带来的创伤和疼痛。4,5)COVID-19 大流行凸显了对能够快速开发粘膜靶向疫苗技术的平台的迫切需求。因此,探索可提高不同人群疫苗接种效果和覆盖率的替代疫苗模式具有重要意义。
最近,使用生物相容性微针(MN)进行透皮给药已成为一种快速发展的给药方式。(6,7)微针矩阵由微米大小的针头阵列组成,提供了一种通过皮肤输送活性物质的无痛方式。(8)值得注意的是,针的长度从 50 微米到 1100 微米不等,可以有效地将抗原递送到表皮和真皮区域,这些区域含有丰富的朗格汉斯细胞和树突状细胞,从而诱发强大的免疫反应。(9,10)同时,基于 MN 的疫苗只需稍加培训即可在家中方便地自行接种,从而降低了交叉感染的风险和大流行期间大规模疫苗接种带来的事故。(11)因此,基于 MN 的给药系统正成为一种前景广阔的疫苗模式。然而,目前仍不清楚 MN 介导的皮内免疫是否能诱导粘膜组织的免疫反应。
由于疫苗的主要成分是不稳定的生物大分子,包括蛋白质亚基、DNA、mRNA 或重组病毒颗粒,因此基于 MN 的疫苗的生产过程可能需要比基于 MN 的化学药物更严格的条件。因此,利用适当的材料来有效保护负载的生物活性化合物至关重要。我们之前的研究表明,壳聚糖低聚糖(COS)是一种带正电荷的化合物,它能包裹并保护带负电荷的 DNA 疫苗或重组病毒疫苗,形成的纳米颗粒(NPs)能激发强大的体液免疫和细胞免疫。(12,13)最重要的是,颗粒的粘附性和佐剂特性使其具有诱导强烈粘膜免疫的潜在能力。(12)基于这些发现,我们报告了一种 COVID-19 疫苗模式,利用 MN 贴片递送 COS 封装的 DNA 疫苗(DNA@COS),编码尖头蛋白(S)和核壳蛋白(N)。与传统的即时免疫相比,我们的研究结果表明,MN介导的皮内给药能诱导更强的全身和粘膜免疫。此外,我们还探讨了 MN 介导的 DNA 疫苗增强免疫力的内在机制。总之,本研究为快速开发一种可成功诱导粘膜免疫的易注射、有效、无痛、可恒温的疫苗模式提供了宝贵的见解。
Results 成果
点击复制章节链接Section link copied!
Preparation and Characterization of COS-Encapsulated DNA Nanoparticles
COS 包囊 DNA 纳米粒子的制备与表征
带正电荷的 COS 和带负电荷的质粒 DNA 疫苗可以自组装形成纳米颗粒(DNA@COS NPs),因为它们之间存在离子相互作用(图 1A)。因此,如方法部分所述,我们获得了 pVAX-S@COS、pVAX-N@COS 和 pVAX-S+N@COS NPs。裸 pVAX-S 和 pVAX-N 质粒 DNA 的 zeta 电位分别为 -20.87 ± 3.50 和 -16.15 ± 0.90 mV。然而,加入带正电荷的 COS(55.54 ± 2.22 mV)后,pVAX-S@COS 和 pVAX-N@COS 的电荷发生了反转,zeta 电位分别为 31.88 ± 0.78 和 30.65 ± 0.04 mV(图 S1)。这些结果证实,COS 和 DNA 可通过正负电荷相互作用形成 NPs。透射电子显微镜(TEM)显示,pVAX-S@COS、pVAX-N@COS 和 pVAX-S+N@COS 呈球形(图 1B和图 S1),粒径分别约为 130.53 ± 0.26、137.27 ± 1.04 和 159.87 ± 1.52 nm(图 1C和图 S1)。然后,我们评估了 COS 对 DNA 质粒的保护能力。将裸质粒 DNA 或 DNA@COS NPs 与限制性内切酶孵育不同时间。结果表明,COS处理能完全保护DNA质粒不被消化,而裸DNA质粒在限制性内切酶位点被切成两段(图S2),证实了COS包被DNA疫苗能有效保护DNA质粒不被DNase I降解。我们还研究了 COS 处理对不同细胞系(HEK-293T、DC 2.4 和 Raw 264)的毒性。7)的细胞计数试剂盒-8(CCK8)检测,结果表明,COS 处理量达到 100 μg/mL 时不会表现出明显的细胞毒性(图 S3)。
随后,在将 pVAX-S@COS、pVAX-N@COS 和 pVAX-S+N@COS 与 293T 细胞培养后,使用 Western bolt 检测法确认了蛋白质的表达。结果表明,DNA@COS 适当表达了相应的尖峰蛋白和核壳蛋白,且分子量符合预期(图 1D,E)。为了进一步研究 COS 包被 DNA NPs 的转染和吸收效率,将 pVAX-GFP@COS NPs 与 HEK-293T 细胞培养 48 h 并检测转染效率。如图 1F,G 和图 S4 所示,DNA@COS NPs 的转染效率呈剂量依赖性增加。pVAX-GFP@COS NPs 的表达也在小鼠免疫细胞系中得到了验证,包括 DC 2.4(小鼠骨髓衍生树突状细胞)和 RAW264.7(小鼠单核巨噬细胞白血病细胞),尽管这些免疫细胞的体外转染效率较低(图 S5)。我们还通过检测 DNA 纳米粒子疫苗的转染效率,评估了其在室温储存 30 天的 MN 中的恒温性。如图 1G和图 S6 所示,储存在 MN 中的 DNA@COS 纳米粒子疫苗的转染效率与新鲜制备的 NPs 相当,但略低于后者。pVAX-luci@COS NPs 编码的荧光素酶可催化底物荧光素氧化产生生物发光,从而以剂量依赖性方式增加发光强度(图 1H)。 如图1I和图S7所示,pVAX-S@COS NPs与HEK-293T细胞培养4小时后,细胞对DNA质粒的摄取显著增加,摄取效率一直保持到8小时。鉴于 COS 封装提高了 DNA 质粒的细胞摄取和转染效率,我们进一步研究了 COS 封装是否能介导内/溶酶体逸出。我们用荧光染料(Cy5)标记了DNA质粒,并用LysoTracker Green染色了内/溶酶体。结果表明,用Cy5-pVAX-S@COS NPs培养细胞1小时和2小时后,观察到大量黄色染色(红绿荧光重叠)(图1J左),红绿曲线下的重叠面积也一直大于4小时和8小时时的重叠面积(图1J右),表明转染后1小时和2小时时pVAX-S@COS NPs被有效地转运到内/溶酶体。然而,在转染后 4 小时和 8 小时,pVAX-S@COS 与内/溶酶体之间的共定位显著降低(图 1J),表明 pVAX-S@COS 在转染后 4 小时可以有效地从内/溶酶体中逸出。与此相一致,1 和 2 h 时的皮尔逊相关系数值也明显高于 4 和 8 h 时(图 S8)。 因此,COS 封装的 DNA NPs 有效提高了转染效率、细胞摄取率和内/溶酶体逃逸率,有利于 DNA 介导的外来蛋白质的持久表达。
Preparation and Characterization of the MN-Mediated DNA@COS Nanoparticles
MN 介导的 DNA@COS 纳米粒子的制备与表征
在我们的研究中,采用离心灌注法将 pVAX-S+N@COS NPs 装入 MN(图 2A)。一块工程 MN 的基底半径为 600 μm,高度为 1000 μm,有 72 个针尖。为了探索 DNA@COS 在 MN 中的分布,我们用红色荧光染料(罗丹明,Rho)标记 COS,制备 DNA@COS-Rho NPs。如图 2B所示,pVAX-S+N@COS-Rho NPs 集中在 MN 的顶端,长度约为 300 μm。然后,对改良后的 MN 贴片进行了测试,以确定其是否有足够的机械强度刺穿皮肤,从而将 DNA@COS NPs 送入皮下组织和体内细胞。结果(图 2C)显示,它能有效穿透小鼠皮肤,含有 DNA@COS NPs 的 MN 顶端在 5 分钟内完全溶解,在皮肤上留下微米大小的孔。pVAX-S+N@COS NPs 的负载并不影响 MN 的机械强度,包括空白 MN、新鲜制备的 MN 和储存的 MN。此外,我们制备的所有 MN 的机械强度都超过了 1 N/针,这表明它们有能力穿透皮肤(图 2D、E)。为了进一步验证载入 MN 的 DNA 的有效递送,我们在 MN 给药前后对 DNA 含量进行了测量。给药前,20 微克由 COS 封装的 DNA 质粒被载入 MN(表 S1),结果表明载入的 DNA 得到了大量有效的递送,递送效率高达 88.6%(图 2F)。 此外,我们还通过评估给药前后 Rho 标记的 COS 含量,验证了 DNA@COS NPs 的给药效率。同样,在评估 COS-Rho 的递送效率时,我们也观察到了 87.3% 的高效率(图 2G)。这些发现凸显了 MN 作为高效 DNA 运送平台的潜力。为了证实这种改性 MN 的溶解特性,我们进行了一次体外实验,发现 MN 顶端在 20 秒内就溶解在磷酸盐缓冲盐水(PBS,pH = 7.4)中,而背衬层则保持相对完整(图 2H)。然后,我们对溶解在 PBS 中的 MN 中的 DNA@COS NPs 进行了表征。如图 S9A、B 所示,NPs 的 zeta 电位没有明显变化,溶解后的 NPs 平均尺寸略有增加,这可能是由于 MN 溶解在 PBS 中后部分 NPs 粘附所致。此外,从 MN 中溶解的 NPs 的转染效率也与新鲜制备的 NPs 相似(图 S9C,D)。
接下来,我们研究了含有 DNA@COS NPs 的修饰 MN 能否在体内适当表达目标蛋白。以 pVAX-luci@COS IM 组为阳性对照,将含有 pVAX-luci@COS 的 MN 贴片置于小鼠裸露的背部皮肤上,然后使用体内成像系统对生物发光进行量化。与传统皮内给药组的结果相比,虽然没有统计学差异,但在 MN 介导的皮内给药组,24 小时和 48 小时的荧光素酶表达水平有升高的趋势(图 2I-K)。这些结果表明,含有 DNA@COS NPs 的改性 MN 制作成功。
Induction of Effective Systemic and Mucosal Humoral Immune Responses by the Thermostable MN-Mediated DNA@COS Nanoparticle Vaccine
热稳定 MN 介导的 DNA@COS 纳米粒子疫苗诱导有效的全身和黏膜体液免疫反应
在验证了 MN 介导的 DNA@COS NPs 可通过皮内给药适当表达编码抗原后,我们进一步研究了它们在小鼠体内的免疫原性(图 3A)。为了研究其热稳定性,我们将载入 MN 的 DNA@COS NPs 在室温下保存了 30 天。然后用新鲜制备的 MN 和储存的 MN 对小鼠进行免疫。通过酶联免疫吸附试验对 SARS-CoV-2 棘突蛋白和核壳蛋白的抗原特异性 IgG 反应进行定量。结果显示,在第21天和第42天,MN介导的pVAX-S+N@COS NPs能有效诱导S特异性IgG抗体(图3B)和N特异性IgG抗体(图3C),这与pVAX-S+N@COS IM组的结果相似。加强免疫后抗体水平进一步提高(图 3B、C)。重要的是,与接种新鲜制备的 MN 疫苗的小鼠相比,接种两剂室温保存 30 天的 pVAX-S+N@COS NPs 的小鼠仍表现出相似的尖峰和核壳特异性抗体水平(图 3B、C)。接下来,我们使用基于 SARS-CoV-2 伪病毒的中和试验评估了血清抗体的中和能力。与传统的DNA@COS纳米颗粒疫苗IM免疫相似,我们基于SARS-CoV-2原型的S蛋白的MN介导的DNA@COS纳米颗粒疫苗在第21天和第42天能显著抑制SARS-CoV-2原型和β变种的感染,但不能抑制SARS-CoV-2δ和Ω变种的感染(图3D,E和图S10A,B)。 此外,用新鲜制备的 MN 和储存的 MN 免疫小鼠的血清抗体中和能力相似。因此,这些研究结果表明,我们改良的 MN 介导的 DNA@COS 纳米粒子疫苗可在室温下保持其活性和免疫原性至少 1 个月,从而避免了冷链配送下的严格储存条件,这对提高疫苗的覆盖率非常有帮助。
结果还显示,我们的策略在第21天和第42天诱导了IgG2c亚类抗体的T辅助细胞1(Th1)偏向性反应,但没有诱导IgG1亚类抗体的Th2偏向性反应(图3F),这与之前的一项研究一致,该研究显示壳聚糖材料增强了抗原特异性Th1反应。(14)重要的是,在第 21 天和第 42 天,pVAX-S+N@COS MN 组的 S 特异性 IgA 抗体水平显著高于 pVAX-S+N@COS IM 组(图 3G),这表明 MN 介导的 DNA@COS 纳米颗粒疫苗能有效激发强效粘膜免疫,而粘膜免疫作为抵御病毒入侵的第一道防线发挥着至关重要的作用。
Induction of a Robust Systemic and Mucosal T Cellular Immunity by MN-Mediated DNA@COS Nanoparticle Vaccine
MN 介导的 DNA@COS 纳米粒子疫苗诱导强大的全身和黏膜 T 细胞免疫力
先前的研究显示,在 COVID-19 康复期患者中发现了高频率的 SARS-CoV-2 特异性 T 细胞,包括 100% 的 CD4+T 细胞和 70% 的 CD8+T 细胞免疫反应(15),这意味着细胞免疫反应在控制病毒复制方面发挥着关键作用。此外,与抗体反应相比,T 细胞免疫反应通常对病毒变体更为保守,这表明通过靶向广泛交叉反应的 T 细胞表位开发通用 COVID-19 疫苗是有潜力的。(16)为了评估 SARS-CoV-2 特异性 T 细胞反应,收集了免疫小鼠的脾脏和肺淋巴细胞进行 IFN-γ ELISpot 检测。初次免疫后,与 pVAX-S+N@COS IM 组相比,pVAX-S+N@COS MN 组的 S1 特异性 IFN-γ+T 细胞应答频率增加了 4.2 倍。同样,S2 特异性 IFN-γ+T 细胞应答的频率也增加了 8.8 倍(图 4A)。与 pVAX-S+N@COS IM 组相比,pVAX-S+N@COS-MN 组的 S1 特异性 IFN-γ+T 细胞应答增加了 5.2 倍,S2 特异性 IFN-γ+T 细胞应答增加了 4.5 倍(图 4B)。同时,与 IM 组相比,MN 组的 N 特异性 IFN-γ+T 细胞应答有更高的趋势,尽管这一差异没有统计学意义。
为了评估在局部粘膜组织引起的细胞免疫,我们从免疫小鼠体内分离出肺淋巴细胞,并对其进行了 IFN-γ ELISpot 检测。值得注意的是,与 pVAX-S+N@COS IM 组相比,我们发现在第 21 天时,pVAX-S+N@COS-MN 组肺粘膜组织中的 S2 特异性 IFN-γ+T 细胞反应频率更高。虽然在S1特异性和N特异性IFN-γ+ T细胞反应中未观察到统计学意义,但总体趋势支持pVAX-S+N@COS-MN组引起的免疫反应增强(图4C)。在第 42 天第二次加强免疫后,我们在肺组织中观察到了更强的粘膜 T 细胞免疫反应。与 IM 组相比,MN 组再次出现了更强的 S2 特异性 T 细胞免疫反应,尽管 S1 特异性 T 细胞免疫反应没有显著差异,但呈上升趋势(图 4D),这与第 21 天在肺组织中观察到的数据相似(图 4C)。这一观察结果的内在机制值得在今后的研究中加以澄清。
考虑到记忆 T 细胞亚群在快速清除病毒和长期保护性免疫中的关键作用,我们进一步研究了这种改良的 MN 介导 DNA@COS 纳米粒子疫苗策略所改变的记忆 T 细胞亚群的表型(图 4E)。结果显示,pVAX-S+N@COS-MN组中S1特异性中心记忆T亚群(Tcm,CD44+CD62L+)和S2特异性效应记忆T亚群(Tem,CD44+CD62L-)的比例明显高于pVAX-S+N@COS IM组(图4F)。
MN-Mediated Intracutaneous but Not Intramuscular Immunization Induces Stronger Multifunctional T Cell Responses
MN 介导的皮内免疫而非肌内免疫可诱导更强的多功能 T 细胞反应
最近的研究表明,能同时分泌多种细胞因子(IFNγ+/IL-2+/TNFα+/CD107a+)的多功能 T 细胞通常能更好地控制病毒感染。因此,我们接下来通过多参数细胞内细胞因子染色法(ICS)检测了功能性 T 细胞的频率(图 5A)。图 5B中显示了 IFN-γ、IL-2 和 TNF-α 的产生以及 CD8+T 细胞亚群中 CD107a 的动员的 ICS 代表图。pVAX-S+N@COS MN组中S1-、S2-和N特异性CD8+IFN-γ+ T细胞的频率明显高于pVAX-S+N@COS IM组(图5C、F、I)。对 CD4+T 细胞亚群也有类似的观察结果(图 S11 和 S12)。此外,在 S1 肽池刺激下,pVAX-S+N@COS MN 组 CD8+IL2+、CD8+TNF-α+ 和 CD8+IL2+TNF-α+T 细胞的频率明显高于 pVAX-S+N@COS IM 组(图 5D、G、J)。 进一步分析还发现,与pVAX-S+N@COS IM组相比,pVAX-S+N@COS MN组的多功能CD8+T细胞同时表达IFN-γ、IL-2和TNF-α的频率更高(图5L)。接下来,我们检测了免疫后CD8+ T细胞表面的CD107a动员,因为CD107a表达的上调与CD8+ T细胞的颗粒酶杀伤活性相关,因此CD8+CD107a+ T细胞可以代表细胞毒性T淋巴细胞。不出所料,pVAX-S+N@COS MN 组 CD8+CD107a+T 细胞的频率明显高于 pVAX-S+N@COS IM 组(图 5E,H,K)。
为了进一步验证 CD8+T 细胞反应的功能性,我们根据淋巴细胞在第 42 天受到 S1、S2 和 N 肽池刺激时的集合样本进行了 t 分布随机邻接嵌入(t-SNE)分析。G1群代表这些在多功能CD8+ T细胞区域分泌IFN-γ、IL-2和TNF-α的细胞,而G2群代表这些在总CD8+ T细胞区域分泌IFN-γ、IL-2和TNF-α的细胞(图5M和图S13)。结果显示,与 pVAX-S+N@COS IM 组相比,pVAX-S+N@COS MN 组的总 CD8+T 细胞应答分泌 IFN-γ、IL-2 和 TNF-α 的功能更强。综上所述,这些研究结果表明,我们的改良MN介导的DNA@COS纳米颗粒疫苗能产生更强、更多功能性的细胞免疫。
Induction of a Long-Lasting Humoral and Cellular T Cellular Immunity by MN-Mediated DNA@COS Nanoparticle Vaccine
MN 介导的 DNA@COS 纳米粒子疫苗诱导持久的体液免疫和细胞 T 细胞免疫
考虑到针对病毒感染的长效免疫反应的重要性,我们评估了 DNA@COS 纳米粒子疫苗通过 MN 介导的皮内给药诱导的抗原特异性长效免疫反应。我们对体液免疫和细胞免疫反应进行了监测,直至第二次加强免疫后 6 周。结果显示,在第 70 天时,MN 介导的皮内给药诱导的 S 特异性抗体和 N 特异性抗体水平与 IM 组相当(图 6A,B)。值得注意的是,与 IM 组相比,MN 介导的皮内给药诱导了更强的全身和粘膜 T 细胞免疫,尤其是在 S1 和 S2 肽池刺激下(图 6C,D)。ICS 检测进一步证实了上述结果,MN 组抗原特异性 CD4+和 CD8+ IFN-γ+T 细胞的频率也更高(图 6E,F)。这些数据表明,改良的 MN 介导的疫苗接种策略能有效诱导持久的体液和细胞 T 细胞免疫。
Enhancement of T Cellular Immunity by Promoting the Maturation of Antigen-Presenting Cells via cGAS-STING-Dependent Signaling Pathway
通过 cGAS-STING 依赖性信号通路促进抗原递呈细胞成熟从而增强 T 细胞免疫力
我们进一步研究了改良MN介导的DNA@COS纳米粒子疫苗诱导T细胞免疫的幅度和多功能性增强的机制。先前的研究表明,壳聚糖能诱导线粒体源性活性氧(ROS)的产生,通过触发 I 型干扰素(IFNs)信号传导导致 DCs 成熟。(14)在本研究中,我们使用 ROS 检测试剂盒(DCF-DA)测定了 DNA@COS 纳米粒子疫苗产生的 ROS,结果显示 COS 处理促进了 ROS 的产生(图 S14)。为了解释这一现象,我们测量了线粒体膜电位(MMP)对COS刺激的反应,发现与COS孵育的DC 2.4细胞仍能保持MMP的稳定性,从而防止细胞凋亡(图S15)。
值得注意的是,我们观察到 COS 培养后 DC 2.4 细胞中 IFNs 和促炎细胞因子(包括 IFN-β(图 7A,B)、IL-6(图 7C,D)和 TNF-α(图 7E,F))的分泌量大幅增加,RT-PCR 和 ELISA 检测都证实了这一点。与这一观察结果相一致的是,COS处理也导致了DC成熟标志物表达的显著上调,包括CD40、CD86和MHC II(图7G-I和图S16)。这些数据表明,DNA@COS 纳米粒子疫苗通过激活 IFN 表达和促进抗原递呈细胞(APCs)的成熟增强了抗病毒免疫力。
接下来,我们进一步阐明了DNA@COS纳米颗粒疫苗激活IFN表达的潜在信号通路。考虑到壳聚糖可能触发cGAS-STING通路激活多种IFN刺激基因(ISGs)以调节抗病毒免疫,(14)因此我们在本研究中调查了该信号通路的下游转录产物。结果显示,COS 处理后,干扰素调节因子 3(IRF3)和信号转导和激活因子 1(STAT1)在 DC 2.4 细胞中的磷酸化显著增强(图 7J)。与此相反,用环孢素 A(CsA)预先孵育 DC 2.4 细胞,可减弱 COS 促进 IRF3 和 STAT1 磷酸化的能力(图 7J)。环孢素 A 是一种已知的抑制剂,可阻止线粒体氧化应激导致的线粒体 DNA 片段释放到细胞膜中。与上述发现一致的是,经 CsA 处理后,COS 诱导的 IFN-β、IL-6 和 TNF-α 等细胞因子的分泌显著减少(图 7K-M),CD40、CD86 和 MHC II 等 DC 细胞成熟标志物也显著减少(图 7N-P和图 S17)。
总之,这些数据表明,DNA@COS 纳米颗粒疫苗通过 cGAS-STING 依赖性信号通路激活 IFN 表达和促进 APC 成熟,从而增强了抗病毒先天免疫力,并使改良 MN 介导的 DNA@COS 纳米颗粒疫苗具有更强的诱导细胞免疫的能力(图 8)。
Discussion 讨论
点击复制章节链接Section link copied!
在本研究中,我们发现与传统的皮内免疫相比,以MN为介导的皮内注射DNA@COS纳米颗粒疫苗能激发更强的全身和粘膜T细胞免疫。同时,这种疫苗方式还能诱导有效的体液免疫,并产生针对 SARS-CoV-2 原型及其变种的中和抗体。值得注意的是,成人的皮肤表面积通常约为 1.5 至 2平方米。皮肤除了具有物理防御功能外,还是一个重要的免疫器官,容纳各种免疫细胞,包括朗格汉斯细胞、真皮树突状细胞、巨噬细胞、肥大细胞以及皮肤淋巴结中的 T 淋巴细胞和 B 淋巴细胞。(17)因此,基于 MN 的递送系统可将负载的成分引向皮肤内的局部微环境,有助于以相对较低的疫苗剂量达到较高的抗原浓度,从而提高诱导抗原特异性免疫反应的能力。(18)此外,MN 插入产生的机械应力可能会激活局部先天性免疫反应。(19)那么,本研究中使用的 COS 材料也可发挥佐剂作用,促进 APCs 成熟,上调主要组织相容性复合体(MHC)分子和协同刺激分子(包括 CD40 和 CD86)的表达,进而触发抗原特异性适应性免疫反应。 更重要的是,MN 介导的 DNA@COS NPs 在室温下储存至少 1 个月后仍能稳定地保持其免疫原性,这表明这种疫苗可以在无冷链条件下运输,从而大大提高疫苗接种覆盖率,尤其是在偏远和发展中地区。
值得注意的是,我们的策略激发了多功能 CD8+T 细胞强有力的细胞毒性 T 细胞(CTL)反应,这种细胞毒性 T 细胞通过识别由 MHC 呈现在细胞表面的病毒多肽,具有靶向和消除感染细胞的能力,这一点已得到广泛认可。(20)最近,越来越多的研究证明,T 细胞免疫可在控制病毒复制和疾病进展方面发挥重要作用。例如,在 COVID-19 的康复者中经常可以发现强大的抗原特异性 T 细胞免疫和丰富的记忆 T 细胞。(21)此外,在一些 SARS-CoV 康复者中,抗原特异性记忆 T 细胞可长期存活超过 17 年。(22)此外,与 B 细胞表位介导的抗体反应相比,T 细胞表位介导的免疫反应通常更为保守,且对病毒变体具有交叉反应性。(23)一些研究发现,SARS-CoV-2 穗状变异株的氨基酸变化并不影响 S 特异性 T 细胞免疫反应,S 抗原和非 S 抗原中的 T 细胞表位在 SARS-CoV-2 变异株中保持不变。(24-26)然而,目前可用的 COVID-19 疫苗大多针对 S 抗原产生中和抗体。(27)迄今为止,频繁出现的 SARS-CoV-2 变体可迅速逃脱 S 特异性中和抗体的免疫识别,这严重削弱了疫苗的效力,并导致突破性感染。 (28)因此,通过针对SARS-CoV-2变种的T细胞免疫来开发下一代COVID-19疫苗具有重要意义。在本研究中,我们构建了同时编码 SARS-CoV-2 穗状抗原和核头状抗原的 DNA@COS 纳米颗粒疫苗。之所以选择将 SARS-CoV-2 核头状(N)蛋白包括在内,是因为它的高度保守性,这意味着它可能是开发具有广泛保护性的 COVID-19 疫苗的一个有吸引力的免疫原成分(15,29-31)。(15,29-31)事实上,基于 N 抗原的疫苗可诱导小鼠和猕猴产生保护性 T 细胞免疫反应,并有效保护它们免受 SARS-CoV-2 感染。(32,33)此外,S 抗原和 N 抗原的组合在小鼠和猕猴体内引起了强烈的中和抗体,并扩大了 T 细胞免疫。这种综合免疫与保护作用有关,包括防止体重减轻,减少 SARS-CoV-2 及其变种感染后的呼吸道症状和肺损伤。(34,35)与这些研究结果一致,我们的 MN 给药 DNA@COS 纳米颗粒疫苗编码 S 抗原和 N 抗原的组合,既能诱导高水平的中和抗体,又能诱导强大的多功能 T 细胞免疫反应。
令人印象深刻的是,我们的数据支持这些 MN 释放的 DNA@COS NPs 有可能在局部粘膜组织引起强烈的免疫反应,肺组织中高频率的 SARS-CoV-2 特异性 IFN-γ+T 细胞和血清中高水平的 S 特异性 IgA 抗体就是证明。众所周知,许多病原体主要通过粘膜表面进入人体,包括呼吸道、皮肤上皮和生殖道,因此粘膜免疫通常被认为是抵御此类传染性病原体的第一道防线。(2)因此,有必要开发下一代粘膜疫苗,通过激发粘膜免疫来提供卓越的保护。然而,大多数已获批准的疫苗都是为诱导全身免疫而设计的,只有少数疫苗旨在诱导粘膜免疫。其中,牛痘疫苗通过皮肤划痕接种根除了致命的空气传播天花病毒,创造了抗击传染病的奇迹,这意味着皮内免疫常规是一种既能诱导保护性全身免疫,又能诱导粘膜免疫的有前途的策略。(36)研究表明,肺部和皮肤淋巴结(SLN)DC 激活的 T 细胞之间存在相似的贩运分子(如 CCR4、(37)CXCR3、(38)和 CCR5(38,39)配体)表达模式,这表明 SLN DC 激活的 T 细胞有可能进入肺部,而肺部 DC 激活的 T 细胞有可能进入皮肤。 因此,进一步的研究应侧重于揭示这种 MN 介导的皮内免疫诱导强大粘膜免疫的内在机制。
Conclusions 结论
点击复制章节链接Section link copied!
我们以 MN 为介导的 DNA@COS 纳米粒子策略可激发全身和粘膜 T 细胞免疫,并具有更高的强度、多功能性和持久性。然而,这项研究也存在一些局限性。例如,本研究没有将皮内注射 DNA@COS NPs 作为另一个对照组。要阐明 MN 介导的皮内免疫诱导强大 T 细胞免疫的确切机制,还需要进一步的研究。此外,由于高级生物安全实验室的资源有限,针对 SARS-CoV-2 感染的挑战实验没有在动物模型中进行评估。总之,这项工作为快速开发一种易于注射、有效、无痛、可恒温的疫苗模式以诱导粘膜免疫提供了一种前景广阔的策略。
Methods 方法
点击复制章节链接Section link copied!
Materials 材料
Dulbecco's modified eagle medium (DMEM)、Roswell Park Memorial Institute (RPMI) 1640 培养基、青霉素/链霉素、胎牛血清 (FBS)、胰蛋白酶-EDTA 和二甲基亚砜 (DMSO) 购自 Gibco 公司(美国纽约大岛)。50× TAE 缓冲液、琼脂糖凝胶、PBS、脱脂奶粉、4% 多聚甲醛、含吐温-20 的三相缓冲盐水(TBST)、Triton X-100 和 4′,6-二脒基-2-苯基吲哚(DAPI)购自 Biosharp(中国北京)。壳聚糖低聚糖(COS)、蔗糖和聚乙烯醇 1795(PVA)、12-肉豆蔻酸 13-乙酸磷脂(PMA)、离子霉素、海参素 A(ConA)、d-荧光素钾盐、环孢素 A(CsA)、胶原酶 I 和 DNase I 购自 Sigma(美国密苏里州圣路易斯市)。罗丹明修饰的 COS(COS-Rho)购自西安瑞希生物科技有限公司(中国西安)。(中国西安)。双喹啉酸 (BCA) 检测试剂盒、SDS-PAGE 凝胶和 NBT/BCIP 试剂购自 Thermo Fisher Scientific (Waltham, MA, USA)。Hifair III 第一链 cDNA 试剂盒、SYBR green qPCR 超级混合物和细胞计数试剂盒-8(CCK8)购自 Yeasen(中国上海)。IFN-γ ELISpot 96 孔板、聚偏二氟乙烯(PVDF)、3,3′,5,5′-四甲基联苯胺(TMB)和终止缓冲液购自 Merck Millipore (Burlington, MA, USA)。HindIII和XbaI限制性内切酶购自新英格兰生物实验室有限公司(中国北京)。(中国北京)。GelRed 购自 Transgen 公司(中国北京)。小鼠淋巴细胞分离液和红细胞裂解缓冲液购自达科威生物技术有限公司(中国深圳)。(中国深圳)。亮光荧光素酶检测系统购自 Vazyme 公司(中国南京)。 Label IT 核酸标记试剂盒购自 Mirus 公司(中国北京)。小鼠 IFN-β 酶联免疫吸附试剂盒、小鼠 IL-6 酶联免疫吸附试剂盒和小鼠 TNF-α 酶联免疫吸附试剂盒购自 Elabscience(武汉)。ROS检测试剂盒(DCF-DA)和线粒体膜电位检测试剂盒购自百优泰(上海)生物科技有限公司。EZ-press RNA 纯化试剂盒购自 EZBioscience(Roseville, MN, USA)。EndoFree Maxi 质粒试剂盒购自天根公司(北京)。小鼠 IFN-γ ELISPOT 抗体对购自 U-CyTech(荷兰)。碱性磷酸酶结合链霉亲和素、brefeldin A、cytofix/cytoperm、FITC-抗小鼠-CD3 抗体、Precp-抗小鼠-CD4 抗体、PE-cy7-抗小鼠-CD8a 抗体、APC-抗小鼠-IFN-γ 抗体、BV605-anti-mouse-IL-2 antibody、PE-anti-mouse-TNF-α antibody、BV421-anti-mouse-CD107a antibody、BV421-anti-mouse-CD62L antibody 和 PE-anti-mouse-CD44 antibody 购自 BD Biosciences (San Jose, CA, USA)。APC-anti-mouse-CD40 抗体、BV605-anti-mouse-CD86 抗体和 PE-anti-mouse-MHCII 抗体购自 BioLegend (San Diego, CA, USA)。兔单克隆抗 IRF3 抗体、兔单克隆抗磷酸-IRF3 抗体、兔单克隆抗 STAT1 抗体、兔单克隆抗磷酸-STAT1 抗体、小鼠单克隆抗 GAPDH 抗体、HRP-conjugated anti-rabbit IgG antibody, or anti-mouse IgG antibody, HRP-conjugated anti-mouse IgA, HRP-conjugated anti-mouse IgG1, and HRP-conjugated anti-mouse IgG2c were purchased from Abcam (Cambridge, UK)。兔单克隆抗尖峰抗体和兔单克隆抗核壳抗体购自 ABclonal 公司(武汉)。 重组 SARS-COV-2 核壳蛋白和重组 SARS-COV-2 穗状病毒蛋白购自 SinoBiological(中国北京)。SARS-CoV-2 穗状病毒及其变种伪型病毒和变种由 VectorBuilder(中国广州)提供。SARS-CoV-2尖峰蛋白和核壳蛋白肽池购自 GeneScript(中国南京)。
Plasmid 质粒
SARS-CoV-2 结构蛋白序列来自美国国家生物技术信息中心(NCBI),包括 S 蛋白序列(YP_009724390.1)和 N 蛋白序列(YP_009724397.2)。蛋白质序列是根据哺乳动物细胞的首选密码子用法优化的,如前所述(40),并由三高生物技术公司(中国上海)化学合成。然后利用限制性内切酶HindIII和XbaI将经过密码子优化的 S 和 N 基因克隆到 pVAX-1 表达载体(Invitrogen 公司)中。pVAX-GFP 和 pVAX-luciferase 质粒保存在本实验室。
Cell Lines 细胞系
HEK-293T 细胞、DC 2.4 细胞和 293T-ACE2 细胞在含有 10% FBS 和 1% 青霉素/链霉素的完全 DMEM 中培养,培养温度为 37 °C,培养箱中含 5%CO2。原始 264.7 细胞在含有 10% FBS 和 1% 青霉素/链霉素的完全 RPMI 1640 中培养,培养温度为 37 °C,培养箱中含 5%CO2。
Mice 小鼠
所有动物实验均严格按照中山大学动物实验委员会的指导原则(SYSU-IACUC-2022-001967)进行。成年雌性BALB/c小鼠(6-8周龄)购自广东省医学实验动物中心。
Preparation and Characterization of the DNA@COS Nanoparticle Vaccine
DNA@COS 纳米粒子疫苗的制备与表征
DNA@COS NPs 的制备方法基于我们之前报道的方法,并做了一些修改。(12,13)简而言之,将 2 毫克 COS 溶于 2 毫克/毫升的 PBS 溶液中。在涡旋条件下,将等体积的 COS 溶液缓慢加入 DNA 溶液(500 μg/mL)中,然后涡旋 30 秒。用同样的方法将 pVAX-S 溶液换成 pVAX-N 溶液,制备 pVAX-N@COS。将 pVAX-S 溶液和 pVAX-N 溶液按 1:1 的浓度比混合后,用上述方法制备 pVAX-S+N@COS。将形成的 DNA 纳米粒子疫苗溶液浓缩并用超速离心法纯化,直到离心液中检测不到游离 DNA,确认 NP 纯化完成。透射电子显微镜(TEM)观察了 NPs 的外观。使用 zeta 电位分析仪(美国布鲁克海文仪器公司)测量 DNA 纳米粒子疫苗的流体力学尺寸和 zeta 电位。
为了确定 COS 封装 DNA 疫苗(含 1 μg DNA 质粒)对限制性内切酶的保护能力,将 1 μLHindIII和 1 μLXbaI限制性内切酶与 NPs 混合,在 37 ℃ 下孵育 0、15 分钟、30 分钟、1 小时和 2 小时,然后上载到 1% 琼脂糖凝胶上。这些凝胶在 1×TAE 缓冲液中以 120 V 电压运行 20 分钟。
Cytotoxicity Assay 细胞毒性试验
HEK-293T 细胞、DC 2.4 细胞和 Raw 264.7 细胞用于评估细胞毒性。将悬浮于含有 10% 胎牛血清的 DMEM 或 RPMI 1640 培养基中的细胞以 1 ×104 个/孔的密度播种到 96 孔微孔板中。用无血清 DMEM 培养基稀释壳聚糖,并加入不同浓度的 COS(0、0.5、1、2、4、8、10、20、50 和 100 μg/mL),用 Millex-GP 无菌注射器过滤器(0.22 μm)过滤。然后将该溶液以 100 μL 的最终体积添加到不同浓度的平板中。24 小时后,加入 10 μL CCK8 试剂,4 小时后,用微孔板阅读器(Synergy HTX,Biotek)读取 OD450 值。
In Vitro Evaluation of Uptake and Transfection Efficiency of DNA@COS Nanoparticles into Cells
体外评估细胞对 DNA@COS 纳米颗粒的吸收和转染效率
为了评估DNA@COS NPs在HEK-293T细胞中的细胞吸收情况,根据Label IT核酸标记试剂盒用荧光染料(Cy5)标记了DNA成分。简言之,将 293T 细胞与 DNA@COS NPs 共同培养 2、4 和 8 小时,使用激光扫描共聚焦显微镜(CLSM)(LSN880,德国蔡司公司)观察 NPs 的吸收情况,并用流式细胞仪(CytoFLEX S,贝克曼公司)进行量化。
用不同浓度的 pVAX-GFP 包被 COS(含 0.5、1、2 和 4 μg/mL DNA 质粒)处理 HEK-293T 细胞。48 小时后,使用倒置荧光显微镜(Axio Vert.A.1,卡尔蔡司)对处理过的细胞进行荧光信号成像。然后用流式细胞仪量化 GFP 的表达效率,并用 FlowJo 软件(X 版)进行分析。同样,用封装在 COS 中的 pVAX-luciferase 处理 HEK-293T 细胞,并使用微孔板发光检测器(GloMax 96,Promega 公司)和亮光荧光素酶检测系统对处理过的细胞进行生物发光信号成像。
为了检测 S 和 N 蛋白的表达,将 HEK-293T 细胞置于 6 孔板中培养 24 小时,然后用裸 pVAX-S、裸 pVAX-N、pVAX-S@COS、pVAX-N@COS 或 pVAX-S+N@COS(4 μg 质粒)处理细胞 48 小时。然后裂解这些细胞,在变性和还原条件下用十二烷基硫酸钠-聚丙烯酰胺凝胶电泳(SDS-PAGE)分离蛋白质。蛋白质被转移到 PVDF 膜上。在 TBST 中用 5% 脱脂奶封闭 1 小时后,用兔单克隆抗尖头抗体、兔单克隆抗核壳抗体和小鼠单克隆抗 GAPDH 抗体(1:2000 稀释)在 4 ℃ 下孵育过夜。最后用化学发光辣根过氧化物酶(HRP)底物显影。
DNA@COS Nanoparticles Escaping the Lysosome-Mediated Degradation
DNA@COS纳米颗粒摆脱溶酶体介导的降解作用
制备 DNA@COS 纳米粒子疫苗(含 2 μg/mL Cy5 标记的 DNA 质粒),并分别与 HEK-293T 细胞共培养 1、2、4 和 8 小时。室温下用 4% 多聚甲醛固定细胞 30 分钟,然后清洗。用溶酶体染色剂在 37 ℃ 下染色 HEK-293T 细胞的溶酶体 30 分钟。洗涤细胞,室温下用 DAPI 染色细胞核 10 分钟。使用 CLSM(LSN880,Zeiss,德国)观察溶酶追踪器和 NPs 的共定位。
Detection of Reactive Oxygen Species (ROS) and Mitochondrial Membrane Potential Change by DNA@COS
利用 DNA@COS 检测活性氧 (ROS) 和线粒体膜电位变化
ROS 检测试剂盒(DCF-DA)用于测定 ROS 的生成。将 DC 2.4 细胞与不同浓度的 COS(0、16 和 50 μg/mL)共孵育 2 小时后,加入 DCF-DA 探针继续共孵育 2 小时,并通过 CLSM 观察不同组中 ROS 的产生情况。
线粒体膜电位检测试剂盒采用 JC-1 检测线粒体膜电位。16 μg/mL COS 分别与 DC 2.4 细胞共培养 2、4 和 8 小时。用 JC-1 染色 DC 2.4 细胞,并用 CLSM 观察线粒体膜电位。
Dendritic Cells Activation and Antigen Presentation by DNA@COS
DNA@COS 激活树突状细胞并呈现抗原
将 DC 2.4 细胞以 2 ×105 个/孔的密度接种到 24 孔板上,培养 6 小时后,将培养基更换为含有 COS(4、8 和 16 μg/mL)的新鲜 DMEM 完全培养基。培养 24 小时后,收集、洗涤细胞并用 BV605-抗小鼠 CD86、APC-抗小鼠 CD40 和 PE-抗小鼠 MHC II 染色。最后,将细胞重悬于 PBS 中,用流式细胞仪进行分析。同样,收获细胞并使用 EZ-press RNA 纯化试剂盒提取总 RNA,然后使用 Hifair III 1st Strand cDNA 试剂盒和 SYBR green qPCR super mix 通过 RT-qPCR 检测 IFN-γ、IL-6 和 TNF-α 等细胞因子的表达。引物见表 S2。收集细胞培养上清,使用小鼠细胞因子酶联免疫吸附试剂盒检测细胞因子的分泌,包括 IFN-γ、IL-6 和 TNF-α。
为了研究细胞活化的机制,在用 COS(4、8 和 16 μg/mL)处理 DC 2.4 细胞 24 小时之前,先用 CsA(20 μM)处理 40 分钟,然后用 ELISA 测定 IFN-γ、IL-6 和 TNF-α 的浓度。同样,CD40、CD86 和 MHC II 的表达也是通过流式细胞术测定的。为了确定 cGAS-STING 依赖性途径的激活情况,在用 16 μg/mL 的 COS 处理 DC 2.4 细胞 4 小时之前,先用 CsA 处理 DC 2.4 细胞 40 分钟;然后裂解这些细胞,在变性和还原条件下用 SDS-PAGE 分离蛋白质。蛋白质被转移到 PVDF 膜上。在 TBST 中用 5%脱脂牛奶封闭 1 小时后,用兔单克隆抗 IRF3 抗体、兔单克隆抗磷酸-IRF3 抗体、兔单克隆抗 STAT1 抗体、兔单克隆抗磷酸-STAT1 抗体和小鼠单克隆抗 GAPDH 抗体以 1:2000 稀释度在 4 ℃ 孵育过夜。最后用化学发光 HRP 底物显影。
Preparation and Characteristics of the MN Patch-Based Vaccine
MN 贴片疫苗的制备和特性
MN 补丁采用离心灌注 PDMS 模具法制备。(41,42)简而言之,将 DNA@COS 纳米粒子疫苗悬浮在含 20% 蔗糖的水溶液中,然后用移液管向 PDMS 模具中加入 200 μL 溶液,离心 3 分钟(4500 rpm,4 °C)以填充空腔。用移液管移去残留的悬浮液。随后,在 25 °C 下离心 30 分钟使模具干燥。然后,向模具中加入 18% 的 PVA 水溶液,离心 10 分钟(4500 转/分,4 °C),并在干燥器中储存过夜,以获得 MN 补丁。
为了测试 MN 补丁的机械强度,我们将其切成小块,针尖朝上。然后用双面胶带将其垂直固定在纹理分析仪(Brookfield CT3,美国)的底座上。在 MNs 的上方放置一个压缩模具,以 1 mm/s 的速度施加垂直压缩力。压缩触发值设定为 0.07 N,压缩距离设定为 400 μm。在此过程中,对 MN 的力大小和变形进行了记录。
为了进一步验证载入 MNs 的 DNA 的有效输送,20 微克用罗丹明标记的包封在 COS 中的 DNA 质粒被载入 MNs。随后,测量了给药前后 MN 中 DNA 和 COS-Rho 的含量。使用 Nanodrop(赛默飞世尔科技公司,美国马萨诸塞州沃尔瑟姆)检测 OD260 值,对每个 MN 片段上的 DNA 负载量进行量化。用 ELISA 分析仪(Tecan/Infinite E plex,瑞士)通过荧光对罗丹明标记的 COS 量进行量化。
为了证实 DNA@COS NP 负载在 MN 中的表达,将含有 pVAX-荧光素酶@COS(20 μg 质粒)的 MN 贴片贴在 6-8 周大的雌性 BALB/c 小鼠裸露的背部皮肤上,然后在腹腔注射 200 μLd-luciferin 钾盐(150 μg/mL)底物的情况下,使用体内成像系统对这些小鼠在不同时间点(24 和 48 h)进行成像和量化。作为对照,在小鼠右股四头肌肌肉注射单独的 pVAX-荧光素酶@COS(20 μg 质粒)。
至于含有 DNA@COS NPs 的 MN 贴片的质量控制,我们使用 Nanodrop(Thermo Fisher Scientific, Waltham, MA, USA)检测了 MN 在 PBS 缓冲液中溶解后的 OD260 值,从而量化了每个 MN 贴片上负载的 DNA 量。在这项研究中,每个贴片上负载的 DNA 平均值为 19.372 ± 0.616 μg(表 S1)。
Animal Experiments 动物实验
将 6-8 周大的雌性 BALB/c 小鼠随机分为四组(每组 10 只)。第 1 组每只小鼠右腿股四头肌肌肉注射 20 μg pVAX-empty@COS。第 2 组每只小鼠使用 MN 贴片在裸露皮肤上注射 20 微克 pVAX-empty@COS。第 3 组小鼠右腿股四头肌肌肉注射 20 微克 pVAX-S+N@COS(每个质粒 10 微克)。同样,第 4 组每只小鼠使用 MN 贴片在裸露皮肤上注射 20 μg pVAX-S+N@COS(每个质粒 10 μg)。简言之,先对小鼠背部进行脱毛处理,然后用异氟烷麻醉小鼠。20 分钟后,待 MN 完全溶解后撕下。每组小鼠在第 0 周和第 2 周接受初次免疫,然后在第 4 周进行加强免疫。第 21 天和第 42 天,每组五只小鼠吸入异氟醚麻醉后颈椎脱位处死。从小鼠眼眶采集血样,在 4 °C 下以 8000 rpm 离心 10 分钟后收集上清液。为去除补体和其他可能影响细胞生长的活性物质,小鼠血清在 56 ℃ 下加热灭活 30 分钟。灭活的血清用于以下免疫测定。磨碎脾脏并用 75 μm 细胞滤网过滤。使用小鼠淋巴细胞分离液分离脾脏淋巴细胞。然后加入红细胞裂解缓冲液,在 4 ℃ 温育 5 分钟,并用 RPMI 1640 冲洗。 淋巴细胞用RPMI 1640悬浮,添加10% FBS、2 mMl-谷氨酰胺和1%青霉素/链霉素。将肺组织切块,用胶原酶 I(1 mg/mL)和 DNase I(20 U/mL)在 37 ℃ 下消化 1 小时,然后按照分离脾淋巴细胞的方法分离肺淋巴细胞。
ELISA Assay 酶联免疫吸附试验
SARS-CoV-2 棘突特异性抗体滴度(包括 IgG、IgA、IgG1 和 IgG2c)和核壳特异性 IgG 抗体滴度由 ELISA 法检测。(43,44)将 SARS-CoV-2 S 蛋白或 N 蛋白稀释至 1 μg/mL 并涂布到 96 孔 ELISA 板上,在 4 °C 下过夜。然后,用 PBS 冲洗 ELISA 板,并用 PBST 溶液中的 5% 脱脂牛奶在 37 ℃ 下阻断 1 小时。然后用 100 μL 2 倍序列稀释的样品(从 1:20 开始)培养 2 小时。然后,洗孔,加入 100 μL HRP 结合的抗小鼠 IgG 抗体(稀释度:1:5000)、HRP 结合的抗小鼠 IgA(稀释度:1:5000)、HRP 结合的抗小鼠 IgG1(稀释度:1:5000)和 HRP 结合的抗小鼠 IgG2c(稀释度:1:5000),37 ℃ 再孵育 1 小时。最后,洗板并用 100 μL TMB 溶液避光孵育 25 分钟。随后,用 1 MH2SO4 停止反应,并用微孔板阅读器读取 OD450 值。终点滴度定义为 OD 值比空白孔平均值高出一倍标准偏差的最低稀释度。
Neutralization Assay 中和试验
中和试验按照我们之前报道的方法进行。(43,44)简言之,在 96 孔细胞培养板中加入 50 μL 稀释度为 1:20 的小鼠血清,与 50 μL 假型 SARS-CoV-2 病毒(2 ×104PFU)在 37 °C 下孵育 1 小时,然后在每个孔中加入 2 ×104 个293T-ACE2 细胞(100 μL 加入 5% FBS 的 DMEM)。在 37 °C 和 5%CO2下培养 48 小时后,移取 100 μL 细胞培养上清液。每孔加入 100 μL 亮光荧光素酶试剂并孵育 2 分钟。孵育后,用移液管将每个孔混合 10 次,然后将 150 μL 混合物转移到白板上,使用微孔板发光检测器测量荧光素酶活性。
IFN-γ ELISpot Assay IFN-γ ELISpot 检测法
IFN-γ ELISpot 检测采用我们之前报道过的方法。(45)简单地说,用 PBS 冲洗 96 孔板,然后用纯化的大鼠抗小鼠 IFN-γ 单克隆抗体涂布,4 ℃ 过夜。将小鼠脾淋巴细胞和肺淋巴细胞按每孔 5 ×105 的比例接种到孔板中。然后每组用 4 mg/mL 的多肽池(包括 S1、S2 和 N 蛋白)孵育,DMSO 作为阴性对照,ConA(10 μg/mL)作为阳性对照。孵育 24 小时后,用生物素化的检测抗体孵育平板,并用碱性磷酸酶结合的链霉亲和素和 NBT/BCIP 试剂显色。最后,用 ELISpot 阅读器(Mabtech IRIS 53,瑞典)对斑点进行计数。
Intracellular Cytokine Staining (ICS)
细胞内细胞因子染色(ICS)
ICS 检测采用我们之前报道过的方法。(46)简而言之,将小鼠脾脏淋巴细胞按每孔2×106 个的比例接种到 96 孔板中,然后与 4 mg/mL 的肽池一起在 37 ℃ 下孵育 2 小时,DMSO 作为阴性对照,ConA(10 μg/mL)作为阳性对照。然后加入布雷非德菌素 A 并在 37 ℃ 下培养 16 小时。收获细胞并用抗小鼠 CD3-FITC、CD4-Percp 和 CD8-PE-Cy7 避光染色 30 分钟。然后加入 cytofix/cytoperm 使混合物透化,避光 30 分钟。然后用抗小鼠 IFN-γ-APC、IL-2-BV605、TNF-α-PE 和 CD107a-BV421 进行染色,4 ℃ 避光 1 小时。使用 CytExpert 软件对样本进行流式细胞术定量分析。
同样,为了测定脾脏淋巴细胞在接种DNA纳米颗粒疫苗后的记忆T细胞,将2×106个小鼠脾脏淋巴细胞接种到96孔板中,与4 mg/mL的肽池在37 °C下孵育24小时,DMSO作为阴性对照。收获细胞并用抗小鼠 CD3-FITC、CD4-Percp、CD8-PE-Cy7、CD44-PE 和 CD62L-BV421 避光染色 30 分钟。清洗样本并用流式细胞仪定量。
Statistical Analysis 统计分析
使用 GraphPad Prism 8.0 版进行统计分析和图表展示。多组(>2)间比较采用单因素方差分析,两组间比较采用双尾非配对学生t检验。数据以平均值 ± SEM 表示,P 值小于 0.05 视为具有统计学意义(*p < 0.05,**p < 0.01,***p < 0.001,****p< 0.0001;ns,无意义)。
Supporting Information 辅助信息
点击复制章节链接Section link copied!
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsnano.3c09521.
辅助信息可从https://pubs.acs.org/doi/10.1021/acsnano.3c09521 免费获取。
Supplementary figures: physiochemical characterization of COS-engineered DNA vaccine; stability of COS-encapsulated DNA vaccine; the COS showed no obvious cell toxicity to different cell lines; in vitro transfection efficacy of DNA nanoparticle vaccine; in vitro transfection efficacy of DNA nanoparticle vaccine in DC 2.4 and RAW264.7 cells; cellular uptake of DNA nanoparticle vaccine in HEK-293T cells determined by flow cytometry; co-localization observed by CLSM of the Cy5-labeled pVAX-S (red) and LysoTracker (green) in HEK-293T cells; physiochemical and biological characterization of DNA@COS-MN dissolving in PBS; neutralization efficiency of mice serum against SARS-CoV-2 variants delta and omicron with dilution at 1:20 at day 42; characterization of DNA nanoparticle vaccine in MN stored at room temperature for 30 days; SARS-CoV-2-specific cellular immune responses after MN-mediated DNA nanoparticle vaccine at day 21; SARS-CoV-2-specific cellular immune responses after MN-mediated DNA nanoparticle vaccine at day 42; T-SNE analysis of concatenated data from group S+N@COS-IM and group S+N@COS-MN for stimulation with S1, S2, and N peptide pool, showing density plots for each condition; detection of ROS by fluorescence of DCFH-DA in the DC 2.4 cells incubated with different concentration of COS; change of MMP of DC 2.4 cells at different times after incubation with COS; CD40, CD86, and MHC II expression in DC 2.4 cells incubated with 16 μg/mL COS for 24 h in the presence or absence of CsA; Supplementary table: primer sequence for qPCR to verify the DC 2.4 cells activation after COS treatment (PDF)
补图:COS 工程 DNA 疫苗的理化特性;COS 封装 DNA 疫苗的稳定性;COS 对不同细胞系无明显细胞毒性;DNA 纳米颗粒疫苗的体外转染效果;DNA 纳米颗粒疫苗在 DC 2.4 和 RAW264.7 细胞中的体外转染效果;流式细胞仪测定 DNA 纳米颗粒疫苗在 HEK-293T 细胞中的细胞摄取量;CLSM 观察到 Cy5 标记的 pVAX-S(pVAX-S)与 COS 的共定位。7细胞的体外转染效果;流式细胞仪测定DNA纳米颗粒疫苗在HEK-293T细胞中的细胞摄取量;CLSM观察到Cy5标记的pVAX-S(红色)和LysoTracker(绿色)在HEK-293T细胞中的共定位;溶于PBS的DNA@COS-MN的理化和生物学特性;小鼠血清对SARS-CoV-2变体delta和ocmicron的中和效率,稀释度为1:在第 42 天时,小鼠血清对 SARS-CoV-2 变体 delta 和 omicron 的中和效率(稀释度为 1:20);DNA 纳米粒子疫苗在 MN 中的特性(在室温下储存 30 天);在第 21 天时,MN 介导的 DNA 纳米粒子疫苗对 SARS-CoV-2 的特异性细胞免疫反应;在第 42 天时,MN 介导的 DNA 纳米粒子疫苗对 SARS-CoV-2 的特异性细胞免疫反应;S+N@COS-IM组和S+N@COS-MN组在S1、S2和N肽池刺激下的T-SNE分析,显示每种条件下的密度图;用DCFH-DA荧光检测DC 2中的ROS。4 细胞中的 ROS 的荧光检测;与 COS 培养后不同时间 DC 2.4 细胞 MMP 的变化;在 CsA 存在或不存在的情况下,与 16 μg/mL COS 培养 24 小时的 DC 2.4 细胞中 CD40、CD86 和 MHC II 的表达;补充表:用于验证 COS 处理后 DC 2.4 细胞活化的 qPCR 引物序列(PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
Figures 3A and 8 were created with BioRender.com.
References
This article references 46 other publications.
- 1Eyawo, O.; Viens, A. M.; Ugoji, U. C. Lockdowns and low- and middle-income countries: building a feasible, effective, and ethical COVID-19 response strategy. Global Health 2021, 17 (1), 13 DOI: 10.1186/s12992-021-00662-yGoogle Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3srivFaitw%253D%253D&md5=952791be0312758e4ab48595f3c5ec7dLockdowns and low- and middle-income countries: building a feasible, effective, and ethical COVID-19 response strategyEyawo Oghenowede; Viens A M; Ugoji Uchechukwu ChidiebereGlobalization and health (2021), 17 (1), 13 ISSN:.Lockdowns can be an effective pandemic response strategy that can buy much needed time to slow disease transmission and adequately scale up preventative, diagnostic, and treatment capacities. However, the broad restrictive measures typically associated with lockdowns, though effective, also comes at a cost - imposing significant social and economic burdens on individuals and societies, especially for those in low- and middle-income countries (LMICs). Like most high-income countries (HICs), many LMICs initially adopted broad lockdown strategies for COVID-19 in the first wave of the pandemic. While many HICs experiencing subsequent waves have returned to employing lockdown strategies until they can receive the first shipments of COVID-19 vaccine, many LMICs will likely have to wait much longer to get comparable access for their own citizens. In leaving LMICs vulnerable to subsequent waves for a longer period of time without vaccines, there is a risk LMICs will be tempted to re-impose lockdown measures in the meantime. In response to the urgent need for more policy development around the contextual challenges involved in employing such measures, we propose some strategies LMICs could adopt for safe and responsible lockdown entrance/exit or to avoid re-imposing coercive restrictive lockdown measures altogether.
- 2Feng, F.; Wen, Z.; Chen, J.; Yuan, Y.; Wang, C.; Sun, C. Strategies to Develop a Mucosa-Targeting Vaccine against Emerging Infectious Diseases. Viruses 2022, 14 (3), 520, DOI: 10.3390/v14030520Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XosVGms7Y%253D&md5=08f02a7c628671ba18325f453d4a1258Strategies to Develop a Mucosa-Targeting Vaccine against Emerging Infectious DiseasesFeng, Fengling; Wen, Ziyu; Chen, Jiaoshan; Yuan, Yue; Wang, Congcong; Sun, CaijunViruses (2022), 14 (3), 520CODEN: VIRUBR; ISSN:1999-4915. (MDPI AG)A review. Numerous pathogenic microbes, including viruses, bacteria, and fungi, usually infect the host through the mucosal surfaces of the respiratory tract, gastrointestinal tract, and reproductive tract. The mucosa is well known to provide the first line of host defense against pathogen entry by phys., chem., biol., and immunol. barriers, and therefore, mucosa-targeting vaccination is emerging as a promising strategy for conferring superior protection. However, there are still many challenges to be solved to develop an effective mucosal vaccine, such as poor adhesion to the mucosal surface, insufficient uptake to break through the mucus, and the difficulty in avoiding strong degrdn. through the gastrointestinal tract. Recently, increasing efforts to overcome these issues have been made, and we herein summarize the latest findings on these strategies to develop mucosa-targeting vaccines, including a novel needle-free mucosa-targeting route, the development of mucosa-targeting vectors, the administration of mucosal adjuvants, encapsulating vaccines into nanoparticle formulations, and antigen design to conjugate with mucosa-targeting ligands. Our work will highlight the importance of further developing mucosal vaccine technol. to combat the frequent outbreaks of infectious diseases.
- 3Seaman, C. P.; Kahn, A. L.; Kristensen, D.; Steinglass, R.; Spasenoska, D.; Scott, N.; Morgan, C. Controlled temperature chain for vaccination in low- and middle-income countries: a realist evidence synthesis. Bull. World Health Organ. 2022, 100 (8), 491– 502, DOI: 10.2471/BLT.21.287696Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2Mbntleksg%253D%253D&md5=a6628201b75aa48018813d21fcfd8468Controlled temperature chain for vaccination in low- and middle-income countries: a realist evidence synthesisSeaman Christopher P; Scott Nick; Kahn Anna-Lea; Kristensen Debra; Steinglass Robert; Spasenoska Dijana; Morgan ChristopherBulletin of the World Health Organization (2022), 100 (8), 491-502 ISSN:.Objective: To evaluate the evidence describing how the controlled temperature chain approach for vaccination could lead to improved equitable immunization coverage in low- and middle-income countries. Methods: We created a theory of change construct from the Controlled temperature chain: strategic roadmap for priority vaccines 2017-2020, containing four domains: (i) uptake and demand for the approach; (ii) compliance and safe use of the approach; (iii) programmatic efficiency gains from the approach; and (iv) improved equitable immunization coverage. To verify and improve the theory of change, we applied a realist review method to analyse published descriptions of controlled temperature chain or closely related experiences. Findings: We evaluated 34 articles, describing 22 unique controlled temperature chain or closely related experiences across four World Health Organization regions. We identified a strong demand for this approach among service delivery providers; however, generating an equal level of demand among policy-makers requires greater evidence on economic benefits and on vaccination coverage gains, and use case definitions. Consistent evidence supported safety of the approach when integrated into special vaccination programmes. Feasible training and supervision supported providers in complying with protocols. Time-savings were the main evidence for efficiency gains, while cost-saving data were minimal. Improved equitable coverage was reported where vaccine storage beyond the cold chain enabled access to hard-to-reach populations. No evidence indicated an inferior vaccine effectiveness nor increased adverse event rates for vaccines delivered under the approach. Conclusion: Synthesized evidence broadly supported the initial theory of change. Addressing evidence gaps on economic benefits and coverage gains may increase future uptake.
- 4Freeman, D.; Lambe, S.; Yu, L. M.; Freeman, J.; Chadwick, A.; Vaccari, C.; Waite, F.; Rosebrock, L.; Petit, A.; Vanderslott, S.; Lewandowsky, S.; Larkin, M.; Innocenti, S.; McShane, H.; Pollard, A. J.; Loe, B. S. Injection fears and COVID-19 vaccine hesitancy. Psychol. Med. 2023, 53 (4), 1185– 1195, DOI: 10.1017/S0033291721002609Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2c7ms1yhsg%253D%253D&md5=e6ceea4c19eb9e344e83d0e80d4e36d1Injection fears and COVID-19 vaccine hesitancyFreeman Daniel; Lambe Sinead; Freeman Jason; Waite Felicity; Rosebrock Laina; Petit Ariane; Freeman Daniel; Lambe Sinead; Waite Felicity; Rosebrock Laina; Petit Ariane; Freeman Daniel; Waite Felicity; Yu Ly-Mee; Chadwick Andrew; Vaccari Cristian; Vanderslott Samantha; Pollard Andrew J; Lewandowsky Stephan; Larkin Michael; Innocenti Stefania; McShane Helen; McShane Helen; Pollard Andrew J; Loe Bao ShengPsychological medicine (2023), 53 (4), 1185-1195 ISSN:.BACKGROUND: When vaccination depends on injection, it is plausible that the blood-injection-injury cluster of fears may contribute to hesitancy. Our primary aim was to estimate in the UK adult population the proportion of COVID-19 vaccine hesitancy explained by blood-injection-injury fears. METHODS: In total, 15 014 UK adults, quota sampled to match the population for age, gender, ethnicity, income and region, took part (19 January-5 February 2021) in a non-probability online survey. The Oxford COVID-19 Vaccine Hesitancy Scale assessed intent to be vaccinated. Two scales (Specific Phobia Scale-blood-injection-injury phobia and Medical Fear Survey-injections and blood subscale) assessed blood-injection-injury fears. Four items from these scales were used to create a factor score specifically for injection fears. RESULTS: In total, 3927 (26.2%) screened positive for blood-injection-injury phobia. Individuals screening positive (22.0%) were more likely to report COVID-19 vaccine hesitancy compared to individuals screening negative (11.5%), odds ratio = 2.18, 95% confidence interval (CI) 1.97-2.40, p < 0.001. The population attributable fraction (PAF) indicated that if blood-injection-injury phobia were absent then this may prevent 11.5% of all instances of vaccine hesitancy, AF = 0.11; 95% CI 0.09-0.14, p < 0.001. COVID-19 vaccine hesitancy was associated with higher scores on the Specific Phobia Scale, r = 0.22, p < 0.001, Medical Fear Survey, r = 0.23, p = <0.001 and injection fears, r = 0.25, p < 0.001. Injection fears were higher in youth and in Black and Asian ethnic groups, and explained a small degree of why vaccine hesitancy is higher in these groups. CONCLUSIONS: Across the adult population, blood-injection-injury fears may explain approximately 10% of cases of COVID-19 vaccine hesitancy. Addressing such fears will likely improve the effectiveness of vaccination programmes.
- 5Wang, H.; Cui, M.; Li, S.; Wu, F.; Jiang, S.; Chen, H.; Yuan, J.; Sun, C. Perception and willingness toward various immunization routes for COVID-19 vaccines: a cross-sectional survey in China. Front. Public Health 2023, 11, 1192709, DOI: 10.3389/fpubh.2023.1192709Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2sfmsVyrsA%253D%253D&md5=bd5836833032ac03af5ed4330ddc3addPerception and willingness toward various immunization routes for COVID-19 vaccines: a cross-sectional survey in ChinaWang Haohang; Cui Mingting; Li Shunran; Wu Fan; Sun Caijun; Jiang Shiqiang; Yuan Jianhui; Chen Hongbiao; Sun Caijun; Sun CaijunFrontiers in public health (2023), 11 (), 1192709 ISSN:.Background: To date, most vaccines, including the COVID-19 vaccine, are mainly administered by intramuscular injection, which might lead to vaccine hesitancy in some populations due to needle fear. Alternatively, needle-free immunization technology is extensively developed to improve the efficacy and acceptance of vaccination. However, there is no study to report the perception and willingness toward various immunization routes of the COVID-19 vaccine in the general population. Methods: A cross-sectional survey was conducted nationwide using an online questionnaire. Bivariate analyses were undertaken to assess variable associations among the participants who reported a hesitancy to receive the COVID-19 booster vaccination. Multivariable logistic regression with a backward step-wise approach was used to analyze the predicted factors associated with the willingness to receive the COVID-19 booster vaccination. Results: A total of 3,244 valid respondents were included in this survey, and 63.2% of participants thought they had a good understanding of intramuscular injection, but only 20.7, 9.2, 9.4, and 6.0% of participants had a self-perceived good understanding of inhalation vaccine, nasal spray vaccine, oral vaccine, and microneedle patch vaccine. Correspondingly, there was high acceptance for intramuscular injection (76.5%), followed by oral inhalation (64.4%) and nasal spray (43.0%). Those participants who were only willing to receive an intramuscular vaccine had less vaccine knowledge (OR = 0.78; 95% CI: 0.65-0.94) than those who were willing to receive a needle-free vaccine (OR = 1.97; 95% CI: 1.52-2.57). Some factors were found to be associated with vaccine hesitancy toward booster COVID-19 vaccination. Conclusion: Needle-free vaccination is a promising technology for the next generation of vaccines, but we found that intramuscular injection was still the most acceptable immunization route in this survey. One major reason might be that most people lack knowledge about needle-free vaccination. We should strengthen the publicity of needle-free vaccination technology, and thus improve the acceptance and coverage of vaccination in different populations.
- 6Yang, Y.; Li, Z.; Huang, P.; Lin, J.; Li, J.; Shi, K.; Lin, J.; Hu, J.; Zhao, Z.; Yu, Y.; Chen, H.; Zeng, X.; Mei, L. Rapidly separating dissolving microneedles with sustained-release colchicine and stabilized uricase for simplified long-term gout management. Acta Pharm. Sin. B 2023, 13 (8), 3454– 3470, DOI: 10.1016/j.apsb.2023.02.011Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXlsVaqtb0%253D&md5=f2880f0139f6a9b8d607bea788c7b77cRapidly separating dissolving microneedles with sustained-release colchicine and stabilized uricase for simplified long-term gout managementYang, Yao; Li, Zimu; Huang, Ping; Lin, Jiachan; Li, Jinyuan; Shi, Kexin; Lin, Jiahui; Hu, Jingwen; Zhao, Zhuoxian; Yu, Yongkang; Chen, Hongzhong; Zeng, Xiaowei; Mei, LinActa Pharmaceutica Sinica B (2023), 13 (8), 3454-3470CODEN: APSBCW; ISSN:2211-3835. (Elsevier B.V.)Despite growing prevalence and incidence, the management of gout remains suboptimal. The intermittent nature of the gout makes the long-term urate-lowering therapy (ULT) particularly important for gout management. However, patients are reluctant to take medication day after day to manage incurable occasional gout flares, and suffer from possible long-term toxicity. Therefore, a safe and easy-to-operate drug delivery system with simple prepn. for the long-term management of gout is very necessary. Here, a chitosan-contg. sustained-release microneedle system co-loaded with colchicine and uricase liposomes were fabricated to achieve this goal. This microneedle system was confirmed to successfully deliver the drug to the skin and maintain a one-week drug retention. Furthermore, its powerful therapeutic potency to manage gout was investigated in both acute gouty and chronic gouty models. Besides, the drug co-delivery system could help avoid long-term daily oral colchicine, a drug with a narrow therapeutic index. This system also avoids mass injection of uricase by improving its stability, enhancing the clin. application value of uricase. In general, this two-drug system reduces the dosage of uricase and colchicine and improves the patient's compliance, which has a strong clin. translation.
- 7Yang, L.; Yang, Y.; Chen, H.; Mei, L.; Zeng, X. Polymeric microneedle-mediated sustained release systems: Design strategies and promising applications for drug delivery. Asian J. Pharm. Sci. 2022, 17 (1), 70– 86, DOI: 10.1016/j.ajps.2021.07.002Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2MzhtlWgtw%253D%253D&md5=5e08045861ec3452358788976beebaa5Polymeric microneedle-mediated sustained release systems: Design strategies and promising applications for drug deliveryYang Li; Yang Yao; Chen Hongzhong; Mei Lin; Zeng Xiaowei; Mei LinAsian journal of pharmaceutical sciences (2022), 17 (1), 70-86 ISSN:.Parenteral sustained release drug formulations, acting as preferable platforms for long-term exposure therapy, have been wildly used in clinical practice. However, most of these delivery systems must be given by hypodermic injection. Therefore, issues including needle-phobic, needle-stick injuries and inappropriate reuse of needles would hamper the further applications of these delivery platforms. Microneedles (MNs) as a potential alternative system for hypodermic needles can benefit from minimally invasive and self-administration. Recently, polymeric microneedle-mediated sustained release systems (MN@SRS) have opened up a new way for treatment of many diseases. Here, we reviewed the recent researches in MN@SRS for transdermal delivery, and summed up its typical design strategies and applications in various diseases therapy, particularly focusing on the applications in contraception, infection, cancer, diabetes, and subcutaneous disease. An overview of the present clinical translation difficulties and future outlook of MN@SRS was also provided.
- 8Menon, I.; Bagwe, P.; Gomes, K. B.; Bajaj, L.; Gala, R.; Uddin, M. N.; D’Souza, M. J.; Zughaier, S. M. Microneedles: A New Generation Vaccine Delivery System. Micromachines 2021, 12 (4), 435, DOI: 10.3390/mi12040435Google ScholarThere is no corresponding record for this reference.
- 9Larraneta, E.; Lutton, R. E. M.; Woolfson, A. D.; Donnelly, R. F. Microneedle arrays as transdermal and intradermal drug delivery systems: Materials science, manufacture and commercial development. Mater. Sci. Eng. R-Reports 2016, 104, 1– 32, DOI: 10.1016/j.mser.2016.03.001Google ScholarThere is no corresponding record for this reference.
- 10Iwata, H.; Kakita, K.; Imafuku, K.; Takashima, S.; Haga, N.; Yamaguchi, Y.; Taguchi, K.; Oyamada, T. Safety and dose-sparing effect of Japanese encephalitis vaccine administered by microneedle patch in uninfected, healthy adults (MNA-J): a randomised, partly blinded, active-controlled, phase 1 trial. Lancet Microbe 2022, 3 (2), E96– E104, DOI: 10.1016/S2666-5247(21)00269-XGoogle Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XlvV2nsbk%253D&md5=bdb5041bfb4bd6d8f1c8a6cc4677d276Safety and dose-sparing effect of Japanese encephalitis vaccine administered by microneedle patch in uninfected, healthy adults (MNA-J): a randomised, partly blinded, active-controlled, phase 1 trialIwata, Hiroaki; Kakita, Kosuke; Imafuku, Keisuke; Takashima, Shota; Haga, Naoya; Yamaguchi, Yasuyuki; Taguchi, Kenji; Oyamada, TakayoshiLancet Microbe (2022), 3 (2), e96-e104CODEN: LMAIAR; ISSN:2666-5247. (Elsevier Ltd.)Background It is unclear whether microneedle vaccinations of Japanese encephalitis virus can induce sufficient neutralising antibodies and reduce the amt. of vaccine needed. We aimed to assess the safety and dose-sparing effect of a microneedle vaccine patch against Japanese encephalitis in healthy individuals who are naive to both the vaccine and natural infection. Methods The MNA-J study was a randomised, partly blinded, active-controlled, phase 1 clin. trial at Hokkaido University (Sapporo, Japan) that enrolled healthy adults aged 20-34 years with no history of Japanese encephalitis vaccination nor of infection as confirmed by seronegativity. We excluded individuals who had been infected with or vaccinated against Japanese encephalitis. Eligible participants were randomly assigned (1:1:1) to one of three groups to receive inactivated Japanese encephalitis vaccine administered twice, 3 wk apart, by either 2·5 μg per injection by s.c. injection, 0·63 μg per patch by high-dose microneedle array (MNA-25%), or 0·25 μg per patch by low-dose microneedle array (MNA-10%). The randomisation sequence, using stratification by cohort and blocks of six, was computer-generated by a statistician who was unaware of group assignment. After administration, the remaining amt. of unadministered vaccine was measured by ELISA and calcd. as the delivered amt. of vaccine. The primary outcome was the neutralising antibody titer at day 42 after first immunization. Successful seroconversion was defined as post-vaccination titers of 1·3 (log10) or higher in individuals whose pre-vaccination titers had been less than 1 (log10). Findings Between Aug 31 and Sept 2, 2019, 39 participants were enrolled and each was randomly assigned to a group (n = 13 per group). No serious adverse events were obsd. All participants in the microneedle array groups had a localised erythematous reaction. The amt. of vaccine delivered by microneedle array to each participant was 0·63-1·15 μg (50-92%) of the full 1·26 μg for the MNA-25% group and 0·25-0·41 μg (51-84%) of the full 0·50 μg for the MNA-10% group. All participants demonstrated seroconversion at day 42, and the mean titers (log10) were 2·55 for MNA-25%, 2·04 for MNA-10%, and 2·08 for s.c. injection. Interpretation A microneedle patch of the Japanese encephalitis vaccine is safe, well tolerated, and immunogenically effective. The dose-sparing effect suggests a significant potential to reduce the amt. of immunogens needed. However, improved delivery is needed to make it more tolerable and user friendly.
- 11Vander Straeten, A.; Sarmadi, M.; Daristotle, J. L.; Kanelli, M.; Tostanoski, L. H.; Collins, J.; Pardeshi, A.; Han, J.; Varshney, D.; Eshaghi, B.; Garcia, J.; Forster, T. A.; Li, G.; Menon, N.; Pyon, S. L.; Zhang, L.; Jacob-Dolan, C.; Powers, O. C.; Hall, K.; Alsaiari, S. K.; Wolf, M.; Tibbitt, M. W.; Farra, R.; Barouch, D. H.; Langer, R.; Jaklenec, A. A microneedle vaccine printer for thermostable COVID-19 mRNA vaccines. Nat. Biotechnol. 2023, DOI: 10.1038/s41587-023-01774-zGoogle ScholarThere is no corresponding record for this reference.
- 12Feng, F.; Hao, H.; Zhao, J.; Li, Y.; Zhang, Y.; Li, R.; Wen, Z.; Wu, C.; Li, M.; Li, P.; Chen, L.; Tang, R.; Wang, X.; Sun, C. Shell-mediated phagocytosis to reshape viral-vectored vaccine-induced immunity. Biomaterials 2021, 276, 121062 DOI: 10.1016/j.biomaterials.2021.121062Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvVaitLzM&md5=567d4ffcfc8d20b7cff865ba033d62abShell-mediated phagocytosis to reshape viral-vectored vaccine-induced immunityFeng, Fengling; Hao, Haibin; Zhao, Jin; Li, Yanjun; Zhang, Ying; Li, Ruiting; Wen, Ziyu; Wu, Chunxiu; Li, Minchao; Li, Pingchao; Chen, Ling; Tang, Ruikang; Wang, Xiaoyu; Sun, CaijunBiomaterials (2021), 276 (), 121062CODEN: BIMADU; ISSN:0142-9612. (Elsevier Ltd.)Adenovirus (Ad) has been extensively developed as a gene delivery vector, but the potential side effect caused by systematic immunization remains one major obstacle for its clin. application. Needle-free mucosal immunization with Ad-based vaccine shows advantages but still faces poor mucosal responses. We herein report that the chem. engineering of single live viral-based vaccine effectively modulated the location and pattern of the subsequently elicited immunity. Through precisely assembly of functional materials onto single live Ad particle, the modified virus entered host cell in a phagocytosis-dependent manner, which is completely distinct from the receptor-mediated entry of native Ad. RNA-Seq data further demonstrated that the modified Ad-induced innate immunity was sharply reshaped via phagocytosis-related pathway, therefore promoting the activation and mature of antigen presentation cells (APC). Moreover, the functional shell enabled the modified Ad-based vector with enhanced muco-adhesion to nasal tissues in mice, and then prolonged resident time onto mucosal surface, leading to the robust mucosal IgA prodn. and T cell immunity at local and even remote mucosal-assocd. lymphoid tissues. This study demonstrated that vaccine-induced immunity can be well modulated by chem. engineering, and this method provides the rational design for needle-free mucosa-targeting vaccine against a variety of emerging infectious diseases.
- 13Sun, C. J.; Pan, S. P.; Xie, Q. X.; Xiao, L. J. Preparation of chitosan-plasmid DNA nanoparticles encoding zona pellucida glycoprotein-3alpha and its expression in mouse. Mol. Reprod. Dev. 2004, 68 (2), 182– 8, DOI: 10.1002/mrd.20058Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjvVyjsrs%253D&md5=b0f09ad9675e0919f4c6c23ffb08f770Preparation of chitosan-plasmid DNA nanoparticles encoding zona pellucida glycoprotein-3α and its expression in mouseSun, Cai-Jun; Pan, Shan-Pei; Xie, Qi-Xuan; Xiao, Luan-JuanMolecular Reproduction and Development (2004), 68 (2), 182-188CODEN: MREDEE; ISSN:1040-452X. (Wiley-Liss, Inc.)In the present study, the porcine zona pellucida (ZP)-3α eukaryotic expression vector pVAX1-pZP3α was constructed by genetic recombinant technol., then the recombinant plasmid was encapsulated in nanoparticles with chitosan, and the imaging of chitosan/pVAX1-pZP3α nanoparticles by At. Force Microscope (AFM) was processed. Feeding mouse with those microencapsulation by gastric larvae, and after 5 days, detecting its expression in mouse intestine by RT-PCR and indirect immunofluorescence (IIF). Results show that the porcine ZP-3α eukaryotic expression vector pVAX1-pZP3α had been constructed correctly, and the chitosan-DNA expressing ZP microencapsulation was prepd. successfully. After 5 days of feeding mouse, the transcription and expression of those DNA vaccines were found in mouse alvine chorion. The prepn. of chitosan/pVAX1-pZP3α plasmid DNA nanoparticles and its expression in mice will help to investigate the feasibility of ZP DNA vaccine to induce oviduct local mucosal immunity against ZP to block the fertilization without causing ovarian dysfunction, which will provide new ideas and ways for research and exploiting more effective, more convenient oral contraceptive vaccines.
- 14Carroll, E. C.; Jin, L.; Mori, A.; Munoz-Wolf, N.; Oleszycka, E.; Moran, H. B. T.; Mansouri, S.; McEntee, C. P.; Lambe, E.; Agger, E. M.; Andersen, P.; Cunningham, C.; Hertzog, P.; Fitzgerald, K. A.; Bowie, A. G.; Lavelle, E. C. The Vaccine Adjuvant Chitosan Promotes Cellular Immunity via DNA Sensor cGAS-STING-Dependent Induction of Type I Interferons. Immunity 2016, 44 (3), 597– 608, DOI: 10.1016/j.immuni.2016.02.004Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XjsVeht7o%253D&md5=4dbaaba95c8d0222008c9a3804203cdaThe Vaccine Adjuvant Chitosan Promotes Cellular Immunity via DNA Sensor cGAS-STING-Dependent Induction of Type I InterferonsCarroll, Elizabeth. C.; Jin, Lei; Mori, Andres; Munoz-Wolf, Natalia; Oleszycka, Ewa; Moran, Hannah B. T.; Mansouri, Samira; McEntee, Craig P.; Lambe, Eimear; Agger, Else Marie; Andersen, Peter; Cunningham, Colm; Hertzog, Paul; Fitzgerald, Katherine A.; Bowie, Andrew G.; Lavelle, Ed C.Immunity (2016), 44 (3), 597-608CODEN: IUNIEH; ISSN:1074-7613. (Elsevier Inc.)The cationic polysaccharide chitosan is an attractive candidate adjuvant capable of driving potent cell-mediated immunity, but the mechanism by which it acts is not clear. We show that chitosan promotes dendritic cell maturation by inducing type I interferons (IFNs) and enhances antigen-specific T helper 1 (Th1) responses in a type I IFN receptor-dependent manner. The induction of type I IFNs, IFN-stimulated genes and dendritic cell maturation by chitosan required the cytoplasmic DNA sensor cGAS and STING, implicating this pathway in dendritic cell activation. Addnl., this process was dependent on mitochondrial reactive oxygen species and the presence of cytoplasmic DNA. Chitosan-mediated enhancement of antigen specific Th1 and IgG2c responses following vaccination was dependent on both cGAS and STING. These findings demonstrate that a cationic polymer can engage the STING-cGAS pathway to trigger innate and adaptive immune responses.
- 15Grifoni, A.; Weiskopf, D.; Ramirez, S. I.; Mateus, J.; Dan, J. M.; Moderbacher, C. R.; Rawlings, S. A.; Sutherland, A.; Premkumar, L.; Jadi, R. S.; Marrama, D.; de Silva, A. M.; Frazier, A.; Carlin, A. F.; Greenbaum, J. A.; Peters, B.; Krammer, F.; Smith, D. M.; Crotty, S.; Sette, A. Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell 2020, 181 (7), 1489– 1501 e15, DOI: 10.1016/j.cell.2020.05.015Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVOmu73N&md5=6f89bf52ae2d734af758163a4940b356Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed IndividualsGrifoni, Alba; Weiskopf, Daniela; Ramirez, Sydney I.; Mateus, Jose; Dan, Jennifer M.; Moderbacher, Carolyn Rydyznski; Rawlings, Stephen A.; Sutherland, Aaron; Premkumar, Lakshmanane; Jadi, Ramesh S.; Marrama, Daniel; de Silva, Aravinda M.; Frazier, April; Carlin, Aaron F.; Greenbaum, Jason A.; Peters, Bjoern; Krammer, Florian; Smith, Davey M.; Crotty, Shane; Sette, AlessandroCell (Cambridge, MA, United States) (2020), 181 (7), 1489-1501.e15CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Understanding adaptive immunity to SARS-CoV-2 is important for vaccine development, interpreting coronavirus disease 2019 (COVID-19) pathogenesis, and calibration of pandemic control measures. Using HLA class I and II predicted peptide "megapools," circulating SARS-CoV-2-specific CD8+ and CD4+ T cells were identified in ∼70% and 100% of COVID-19 convalescent patients, resp. CD4+ T cell responses to spike, the main target of most vaccine efforts, were robust and correlated with the magnitude of the anti-SARS-CoV-2 IgG and IgA titers. The M, spike, and N proteins each accounted for 11%-27% of the total CD4+ response, with addnl. responses commonly targeting nsp3, nsp4, ORF3a, and ORF8, among others. For CD8+ T cells, spike and M were recognized, with at least eight SARS-CoV-2 ORFs targeted. Importantly, we detected SARS-CoV-2-reactive CD4+ T cells in ∼40%-60% of unexposed individuals, suggesting cross-reactive T cell recognition between circulating "common cold" coronaviruses and SARS-CoV-2.
- 16Moss, P. The T cell immune response against SARS-CoV-2. Nat. Immunol. 2022, 23 (2), 186– 193, DOI: 10.1038/s41590-021-01122-wGoogle Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XisFOmtbg%253D&md5=a87a8b6da6c9977edfdb5950c491470eThe T cell immune response against SARS-CoV-2Moss, PaulNature Immunology (2022), 23 (2), 186-193CODEN: NIAMCZ; ISSN:1529-2908. (Nature Portfolio)The adaptive immune response is a major determinant of the clin. outcome after SARS-CoV-2 infection and underpins vaccine efficacy. T cell responses develop early and correlate with protection but are relatively impaired in severe disease and are assocd. with intense activation and lymphopenia. A subset of T cells primed against seasonal coronaviruses cross reacts with SARS-CoV-2 and may contribute to clin. protection, particularly in early life. T cell memory encompasses broad recognition of viral proteins, estd. at around 30 epitopes within each individual, and seems to be well sustained so far. This breadth of recognition can limit the impact of individual viral mutations and is likely to underpin protection against severe disease from viral variants, including Omicron. Current COVID-19 vaccines elicit robust T cell responses that likely contribute to remarkable protection against hospitalization or death, and novel or heterologous regimens offer the potential to further enhance cellular responses. T cell immunity plays a central role in the control of SARS-CoV-2 and its importance may have been relatively underestimated thus far.
- 17Sheng, T.; Luo, B.; Zhang, W.; Ge, X.; Yu, J.; Zhang, Y.; Gu, Z. Microneedle-Mediated Vaccination: Innovation and Translation. Adv. Drug Deliv. Rev. 2021, 179, 113919 DOI: 10.1016/j.addr.2021.113919Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvVaisbbO&md5=9aa9c3552eb579bfbb0471663108d957Microneedle-Mediated Vaccination: Innovation and TranslationSheng, Tao; Luo, Bowen; Zhang, Wentao; Ge, Xinyang; Yu, Jicheng; Zhang, Yuqi; Gu, ZhenAdvanced Drug Delivery Reviews (2021), 179 (), 113919CODEN: ADDREP; ISSN:0169-409X. (Elsevier B.V.)A review. Vaccine administration by s.c. or i.m. injection is the most commonly prescribed route for inoculation, however, it is often assocd. with some deficiencies such as low compliance, high professionalism, and risk of infection. Therefore, the application of microneedles for vaccine delivery has gained widespread interests in the past few years due to its high compliance, minimal invasiveness, and convenience. This review focuses on recent advances in the development and application of microneedles for vaccination based on different delivery strategies, and introduces the current status of microneedle-mediated vaccination in clin. translation. The prospects for its application including opportunities and challenges are further discussed.
- 18Korkmaz, E.; Balmert, S. C.; Carey, C. D.; Erdos, G.; Falo, L. D., Jr. Emerging skin-targeted drug delivery strategies to engineer immunity: A focus on infectious diseases. Expert Opin. Drug Deliv. 2021, 18 (2), 151– 167, DOI: 10.1080/17425247.2021.1823964Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvF2ku7%252FJ&md5=fbb20d79eb7860bdd55f522a7d84f43dEmerging skin-targeted drug delivery strategies to engineer immunity: A focus on infectious diseasesKorkmaz, Emrullah; Balmert, Stephen C.; Carey, Cara Donahue; Erdos, Geza; Falo, Louis D. JrExpert Opinion on Drug Delivery (2021), 18 (2), 151-167CODEN: EODDAW; ISSN:1742-5247. (Taylor & Francis Ltd.)A review. Introduction: Infectious pathogens are global disrupters. Progress in biomedical science and technol. has expanded the public health arsenal against infectious diseases. Specifically, vaccination has reduced the burden of infectious pathogens. Engineering systemic immunity by harnessing the cutaneous immune network has been particularly attractive since the skin is an easily accessible immune-responsive organ. Recent advances in skin-targeted drug delivery strategies have enabled safe, patient-friendly, and controlled deployment of vaccines to cutaneous microenvironments for inducing long-lived pathogen-specific immunity to mitigate infectious diseases, including COVID-19. Areas covered: This review briefly discusses the basics of cutaneous immunomodulation and provides a concise overview of emerging skin-targeted drug delivery systems that enable safe, minimally invasive, and effective intracutaneous administration of vaccines for engineering systemic immune responses to combat infectious diseases. Expert opinion: In-situ engineering of the cutaneous microenvironment using emerging skin-targeted vaccine delivery systems offers remarkable potential to develop diverse immunization strategies against pathogens. Mechanistic studies with std. correlates of vaccine efficacy will be important to compare innovative intracutaneous drug delivery strategies to each other and to existing clin. approaches. Cost-benefit analyses will be necessary for developing effective commercialization strategies. Significant involvement of industry and/or government will be imperative for successfully bringing novel skin-targeted vaccine delivery methods to market for their widespread use.
- 19Depelsenaire, A. C. I.; Meliga, S. C.; McNeilly, C. L.; Pearson, F. E.; Coffey, J. W.; Haigh, O. L.; Flaim, C. J.; Frazer, I. H.; Kendall, M. A. F. Colocalization of cell death with antigen deposition in skin enhances vaccine immunogenicity. J. Invest. Dermatol. 2014, 134 (9), 2361– 2370, DOI: 10.1038/jid.2014.174Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXns1Wru70%253D&md5=49a9dd772c48f77b964d7ff389a99352Colocalization of Cell Death with Antigen Deposition in Skin Enhances Vaccine ImmunogenicityDepelsenaire, Alexandra C. I.; Meliga, Stefano C.; McNeilly, Celia L.; Pearson, Frances E.; Coffey, Jacob W.; Haigh, Oscar L.; Flaim, Christopher J.; Frazer, Ian H.; Kendall, Mark A. F.Journal of Investigative Dermatology (2014), 134 (9), 2361-2370CODEN: JIDEAE; ISSN:0022-202X. (Nature Publishing Group)Vaccines delivered to the skin by microneedles-with and without adjuvants-have increased immunogenicity with lower doses than std. vaccine delivery techniques such as i.m. or intradermal injection. However, the mechanisms underlying this skin-mediated "adjuvant" effect are not clear. Here, we show that the dynamic application of a microprojection array (the Nanopatch) to skin generates localized transient stresses invoking cell death around each projection. Nanopatch application caused significantly higher levels (∼65-fold) of cell death in murine ear skin than i.d. injection using a hypodermic needle. Measured skin cell death is assocd. with modeled stresses ∼1-10 MPa. Nanopatch-immunized groups also yielded consistently higher anti-IgG endpoint titers (up to 50-fold higher) than i.d. groups after delivery of a split virion influenza vaccine. Importantly, colocalization of cell death with nearby live skin cells and delivered antigen was necessary for immunogenicity enhancement. These results suggest a correlation between cell death caused by the Nanopatch with increased immunogenicity. We propose that the localized cell death serves as a "phys. immune enhancer" for the adjacent viable skin cells, which also receive antigen from the projections. This natural immune enhancer effect has the potential to mitigate or replace chem.-based adjuvants in vaccines.
- 20Rock, K. L.; York, I. A.; Goldberg, A. L. Post-proteasomal antigen processing for major histocompatibility complex class I presentation. Nat. Immunol. 2004, 5 (7), 670– 7, DOI: 10.1038/ni1089Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXlt1Wmu70%253D&md5=57db65f9622669eba3ef6a7393288364Post-proteasomal antigen processing for major histocompatibility complex class I presentationRock, Kenneth L.; York, Ian A.; Goldberg, Alfred L.Nature Immunology (2004), 5 (7), 670-677CODEN: NIAMCZ; ISSN:1529-2908. (Nature Publishing Group)A review. Peptides presented by major histocompatibility complex class I mols. are derived mainly from cytosolic oligopeptides generated by proteasomes during the degrdn. of intracellular proteins. Proteasomal cleavages generate the final C terminus of these epitopes. Although proteasomes may produce mature epitopes that are eight to ten residues in length, they more often generate N-extended precursors that are too long to bind to major histocompatibility complex class I mols. Such precursors are trimmed in the cytosol or in the endoplasmic reticulum by aminopeptidases that generate the N terminus of the presented epitope. Peptidases can also destroy epitopes by trimming peptides to below the size needed for presentation. In the cytosol, endopeptidases, esp. thimet oligopeptidase, and aminopeptidases degrade many proteasomal products, thereby limiting the supply of many antigenic peptides. Thus, the extent of antigen presentation depends on the balance between several proteolytic processes that may generate or destroy epitopes.
- 21Mateus, J.; Grifoni, A.; Tarke, A.; Sidney, J.; Ramirez, S. I.; Dan, J. M.; Burger, Z. C.; Rawlings, S. A.; Smith, D. M.; Phillips, E.; Mallal, S.; Lammers, M.; Rubiro, P.; Quiambao, L.; Sutherland, A.; Yu, E. D.; da Silva Antunes, R.; Greenbaum, J.; Frazier, A.; Markmann, A. J.; Premkumar, L.; de Silva, A.; Peters, B.; Crotty, S.; Sette, A.; Weiskopf, D. Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans. Science 2020, 370 (6512), 89– 94, DOI: 10.1126/science.abd3871Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvF2itLnP&md5=0ed58b534636c4e887fe4072a7d53a53Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humansMateus, Jose; Grifoni, Alba; Tarke, Alison; Sidney, John; Ramirez, Sydney I.; Dan, Jennifer M.; Burger, Zoe C.; Rawlings, Stephen A.; Smith, Davey M.; Phillips, Elizabeth; Mallal, Simon; Lammers, Marshall; Rubiro, Paul; Quiambao, Lorenzo; Sutherland, Aaron; Yu, Esther Dawen; da Silva Antunes, Ricardo; Greenbaum, Jason; Frazier, April; Markmann, Alena J.; Premkumar, Lakshmanane; de Silva, Aravinda; Peters, Bjoern; Crotty, Shane; Sette, Alessandro; Weiskopf, DanielaScience (Washington, DC, United States) (2020), 370 (6512), 89-94CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)Many unknowns exist about human immune responses to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. SARS-CoV-2 reactive CD4+ T cells have been reported in unexposed individuals, suggesting preexisting cross-reactive T cell memory in 20 to 50% of people. However, the source of those T cells has been speculative. Using human blood samples derived before the SARS-CoV-2 virus was discovered in 2019, we mapped 142 T cell epitopes across the SARS-CoV-2 genome to facilitate precise interrogation of the SARS-CoV-2 specific CD4+ T cell repertoire. We demonstrate a range of preexisting memory CD4+ T cells that are cross-reactive with comparable affinity to SARS-CoV-2 and the common cold coronaviruses human coronavirus (HCoV)-OC43, HCoV-229E, HCoV-NL63, and HCoV-HKU1. Thus, variegated T cell memory to coronaviruses that cause the common cold may underlie at least some of the extensive heterogeneity obsd. in coronavirus disease 2019 (COVID-19) disease.
- 22Le Bert, N.; Tan, A. T.; Kunasegaran, K.; Tham, C. Y. L.; Hafezi, M.; Chia, A.; Chng, M. H. Y.; Lin, M.; Tan, N.; Linster, M.; Chia, W. N.; Chen, M. I.; Wang, L. F.; Ooi, E. E.; Kalimuddin, S.; Tambyah, P. A.; Low, J. G.; Tan, Y. J.; Bertoletti, A. SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature 2020, 584 (7821), 457– 462, DOI: 10.1038/s41586-020-2550-zGoogle Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsF2ntL3J&md5=148bb07cafbec1af0570f97c662d5b0bSARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controlsLe Bert, Nina; Tan, Anthony T.; Kunasegaran, Kamini; Tham, Christine Y. L.; Hafezi, Morteza; Chia, Adeline; Chng, Melissa Hui Yen; Lin, Meiyin; Tan, Nicole; Linster, Martin; Chia, Wan Ni; Chen, Mark I-Cheng; Wang, Lin-Fa; Ooi, Eng Eong; Kalimuddin, Shirin; Tambyah, Paul Anantharajah; Low, Jenny Guek-Hong; Tan, Yee-Joo; Bertoletti, AntonioNature (London, United Kingdom) (2020), 584 (7821), 457-462CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Memory T cells induced by previous pathogens can shape susceptibility to, and the clin. severity of, subsequent infections. Little is known about the presence in humans of pre-existing memory T cells that have the potential to recognize severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We studied T cell responses against the structural (nucleocapsid (N) protein) and non-structural (NSP7 and NSP13 of ORF1) regions of SARS-CoV-2 in individuals convalescing from COVID-19 (n = 36). In all of these individuals, we found CD4 and CD8 T cells that recognized multiple regions of the N protein. Next, we showed that patients (n = 23) who recovered from SARS (the disease assocd. with SARS-CoV infection) possess long-lasting memory T cells that are reactive to the N protein of SARS-CoV 17 yr after the outbreak of SARS in 2003; these T cells displayed robust cross-reactivity to the N protein of SARS-CoV-2. We also detected SARS-CoV-2-specific T cells in individuals with no history of SARS, COVID-19, or contact with individuals who had SARS and(or) COVID-19 (n = 37). SARS-CoV-2-specific T cells in uninfected donors exhibited a different pattern of immunodominance, and frequently targeted NSP7 and NSP13 as well as the N protein. Epitope characterization of NSP7-specific T cells showed the recognition of protein fragments that are conserved among animal betacoronaviruses but have low homol. to common cold human-assocd. coronaviruses. Thus, infection with betacoronaviruses induces multi-specific and long-lasting T cell immunity against the structural N protein. Understanding how pre-existing N- and ORF1-specific T cells that are present in the general population affect the susceptibility to and pathogenesis of SARS-CoV-2 infection is important for the management of the current COVID-19 pandemic.
- 23Selin, L. K.; Brehm, M. A.; Naumov, Y. N.; Cornberg, M.; Kim, S. K.; Clute, S. C.; Welsh, R. M. Memory of mice and men: CD8+ T-cell cross-reactivity and heterologous immunity. Immunol. Rev. 2006, 211 (1), 164– 81, DOI: 10.1111/j.0105-2896.2006.00394.xGoogle Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xot1GrsLY%253D&md5=3bba66cd073e74e993f710ae2716e279Memory of mice and men: CD8+ T-cell cross-reactivity and heterologous immunitySelin, Liisa K.; Brehm, Michael A.; Naumov, Yuri N.; Cornberg, Markus; Kim, Sung-Kwon; Clute, Shalyn C.; Welsh, Raymond M.Immunological Reviews (2006), 211 (), 164-181CODEN: IMRED2; ISSN:0105-2896. (Blackwell Publishing Ltd.)A review. The main functions of memory T cells are to provide protection upon re-exposure to a pathogen and to prevent the re-emergence of low-grade persistent pathogens. Memory T cells achieve these functions through their high frequency and elevated activation state, which lead to rapid responses upon antigenic challenge. The significance and characteristics of memory CD8+ T cells in viral infections have been studied extensively. In many of these studies of T-cell memory, exptl. viral immunologists go to great lengths to assure that their animal colonies are free of endogenous pathogens in order to design reproducible expts. These exptl. results are then thought to provide the basis for our understanding of human immune responses to viruses. Although these findings can be enlightening, humans are not immunol. naive, and they often have memory T-cell populations that can cross-react with and respond to a new infectious agent or cross-react with allo-antigens and influence the success of tissue transplantation. These cross-reactive T cells can become activated and modulate the immune response and outcome of subsequent heterologous infections, a phenomenon we have termed heterologous immunity. These large memory populations are also accommodated into a finite immune system, requiring that the host makes room for each new population of memory cell. It appears that memory cells are part of a continually evolving interactive network, where with each new infection there is an alteration in the frequencies, distributions, and activities of memory cells generated in response to previous infections and allo-antigens.
- 24Riou, C.; Keeton, R.; Moyo-Gwete, T.; Hermanus, T.; Kgagudi, P.; Baguma, R.; Valley-Omar, Z.; Smith, M.; Tegally, H.; Doolabh, D.; Iranzadeh, A.; Tyers, L.; Mutavhatsindi, H.; Tincho, M. B.; Benede, N.; Marais, G.; Chinhoyi, L. R.; Mennen, M.; Skelem, S.; du Bruyn, E.; Stek, C.; de Oliveira, T.; Williamson, C.; Moore, P. L.; Wilkinson, R. J.; Ntusi, N. A. B.; Burgers, W. A. Escape from recognition of SARS-CoV-2 variant spike epitopes but overall preservation of T cell immunity. Sci. Transl. Med. 2022, 14 (631), eabj6824 DOI: 10.1126/scitranslmed.abj6824Google ScholarThere is no corresponding record for this reference.
- 25Choi, S. J.; Kim, D. U.; Noh, J. Y.; Kim, S.; Park, S. H.; Jeong, H. W.; Shin, E. C. T cell epitopes in SARS-CoV-2 proteins are substantially conserved in the Omicron variant. Cell. Mol. Immunol. 2022, 19 (3), 447– 448, DOI: 10.1038/s41423-022-00838-5Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhtlOks7k%253D&md5=60fb3bbaa3d6bb3d59da41231c064968T cell epitopes in SARS-CoV-2 proteins are substantially conserved in the Omicron variantChoi, Seong Jin; Kim, Dong-Uk; Noh, Ji Yun; Kim, Sangwoo; Park, Su-Hyung; Jeong, Hye Won; Shin, Eui-CheolCellular & Molecular Immunology (2022), 19 (3), 447-448CODEN: CMIEAO; ISSN:1672-7681. (Nature Portfolio)Our current anal. demonstrates that T cell epitopes are considerably conserved in the Omicron variant and that substantial proportions of memory T cells elicited by COVID-19 vaccination or natural infection respond to the Omicron spike. These results indicate that memory T cells may provide protective immunity during reinfection or breakthrough infection with the Omicron variant.
- 26Naranbhai, V.; Nathan, A.; Kaseke, C.; Berrios, C.; Khatri, A.; Choi, S.; Getz, M. A.; Tano-Menka, R.; Ofoman, O.; Gayton, A.; Senjobe, F.; Zhao, Z.; St Denis, K. J.; Lam, E. C.; Carrington, M.; Garcia-Beltran, W. F.; Balazs, A. B.; Walker, B. D.; Iafrate, A. J.; Gaiha, G. D. T cell reactivity to the SARS-CoV-2 Omicron variant is preserved in most but not all individuals. Cell 2022, 185 (6), 1041– 1051 e6, DOI: 10.1016/j.cell.2022.01.029Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XkslKhsLw%253D&md5=c98ae864ccadb9b24ccbfe692c7d9b94T cell reactivity to the SARS-CoV-2 Omicron variant is preserved in most but not all individualsNaranbhai, Vivek; Nathan, Anusha; Kaseke, Clarety; Berrios, Cristhian; Khatri, Ashok; Choi, Shawn; Getz, Matthew A.; Tano-Menka, Rhoda; Ofoman, Onosereme; Gayton, Alton; Senjobe, Fernando; Zhao, Zezhou; St Denis, Kerri J.; Lam, Evan C.; Carrington, Mary; Garcia-Beltran, Wilfredo F.; Balazs, Alejandro B.; Walker, Bruce D.; Iafrate, A. John; Gaiha, Gaurav D.Cell (Cambridge, MA, United States) (2022), 185 (6), 1041-1051.e6CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The SARS-CoV-2 Omicron variant (B.1.1.529) contains mutations that mediate escape from antibody responses, although the extent to which these substitutions in spike and non-spike proteins affect T cell recognition is unknown. T cell responses in individuals with prior infection, vaccination, both prior infection and vaccination, and boosted vaccination are largely preserved to Omicron spike and non-spike proteins. However, we also identify a subset of individuals (∼21%) with a >50% redn. in T cell reactivity to the Omicron spike. Evaluation of functional CD4+ and CD8+ memory T cell responses confirmed these findings and revealed that reduced recognition to Omicron spike is primarily obsd. within the CD8+ T cell compartment potentially due to escape from HLA binding. Booster vaccination enhanced T cell responses to Omicron spike. In contrast to neutralizing immunity, these findings suggest preservation of T cell responses to the Omicron variant, although with reduced reactivity in some individuals.
- 27Fan, F.; Zhang, X.; Zhang, Z.; Ding, Y.; Wang, L.; Xu, X.; Pan, Y.; Gong, F. Y.; Jiang, L.; Kang, L.; Ha, Z.; Lu, H.; Hou, J.; Kou, Z.; Zhao, G.; Wang, B.; Gao, X. M. Potent immunogenicity and broad-spectrum protection potential of microneedle array patch-based COVID-19 DNA vaccine candidates encoding dimeric RBD chimera of SARS-CoV and SARS-CoV-2 variants. Emerg. Microbes Infect. 2023, 12 (1), 2202269 DOI: 10.1080/22221751.2023.2202269Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXpt1antbk%253D&md5=46b85f5c131f557284fac7175d7cc4abPotent immunogenicity and broad-spectrum protection potential of microneedle array patch-based COVID-19 DNA vaccine candidates encoding dimeric RBD chimera of SARS-CoV and SARS-CoV-2 variantsFan, Feng; Zhang, Xin; Zhang, Zhiyu; Ding, Yuan; Wang, Limei; Xu, Xin; Pan, Yaying; Gong, Fang-Yuan; Jiang, Lin; Kang, Lingyu; Ha, Zhuo; Lu, Huijun; Hou, Jiawang; Kou, Zhihua; Zhao, Gan; Wang, Bin; Gao, Xiao-MingEmerging Microbes & Infections (2023), 12 (1), 2202269/1-2202269/16CODEN: EMIMC4; ISSN:2222-1751. (Taylor & Francis Ltd.)Breakthrough infections by SARS-CoV-2 variants pose a global challenge to COVID-19 pandemic control, and the development of more effective vaccines of broad-spectrum protection is needed. In this study, we constructed pVAX1-based plasmids encoding receptor-binding domain (RBD) chimera of SARS-CoV-1 and SARS-CoV-2 variants, including pAD1002 (encoding RBDSARS/BA1), pAD1003 (encoding RBDSARS/Beta) and pAD131 (encoding RBDBA1/Beta). Plasmids pAD1002 and pAD131 were far more immunogenic than pAD1003 in terms of eliciting RBD-specific IgG when i.m. administered without electroporation. Furthermore, dissolvable microneedle array patches (MAP) greatly enhanced the immunogenicity of these DNA constructs in mice and rabbits. MAP laden with pAD1002 (MAP-1002) significantly outperformed inactivated SARS-CoV-2 virus vaccine in inducing RBD-specific IFN-γ+ effector and memory T cells, and generated T lymphocytes of different homing patterns compared to that induced by electroporated DNA in mice. In consistence with the high titer neutralization results of MAP-1002 antisera against SARS-CoV-2 pseudoviruses, MAP-1002 protected human ACE2-transgenic mice from Omicron BA.1 challenge. Collectively, MAP-based DNA constructs encoding chimeric RBDs of SARS-CoV-1 and SARS-CoV-2 variants, as represented by MAP-1002, are potential COVID-19 vaccine candidates worthy further translational study.
- 28Castro Dopico, X.; Ols, S.; Lore, K.; Karlsson Hedestam, G. B. Immunity to SARS-CoV-2 induced by infection or vaccination. J. Intern. Med. 2022, 291 (1), 32– 50, DOI: 10.1111/joim.13372Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhslKgur7K&md5=733c3e5af1496df634afa755928c41d3Immunity to SARS-CoV-2 induced by infection or vaccinationCastro Dopico, Xaquin; Ols, Sebastian; Lore, Karin; Karlsson Hedestam, Gunilla B.Journal of Internal Medicine (2022), 291 (1), 32-50CODEN: JINMEO; ISSN:0954-6820. (Wiley-Blackwell)A review. Adaptive immune responses play crit. roles in viral clearance and protection against re-infection, and SARS-CoV-2 is no exception. What is exceptional, is the rapid characterization of the immune response to the virus performed by researchers during the first 20 mo of the pandemic. This has given us a more detailed understanding about SARS-CoV-2 than we have about many viruses that have been with us for a long time. Furthermore, effective COVID-19 vaccines were developed in record time, and their rollout worldwide is already making a significant difference, although major challenges remain in terms of equal access. The pandemic has engaged scientists and the public alike, and terms such as seroprevalence, neutralizing antibodies, antibody escape and vaccine certificates have become familiar to a broad community. Here, we review key findings concerning B cell and antibody (Ab) responses to SARS-CoV-2, focusing on non-severe cases and anti-spike (S) Ab responses in particular, the latter being central to protective immunity induced by infection or vaccination. The emergence of viral variants that have acquired mutations in S acutely highlights the need for continued characterization of both emerging variants and Ab responses against these during the evolving pathogen-immune system arms race.
- 29Dutta, N. K.; Mazumdar, K.; Gordy, J. T. The Nucleocapsid Protein of SARS-CoV-2: a Target for Vaccine Development. J. Virol. 2020, 94 (13), 647-20, DOI: 10.1128/JVI.00647-20Google ScholarThere is no corresponding record for this reference.
- 30Ni, L.; Ye, F.; Cheng, M. L.; Feng, Y.; Deng, Y. Q.; Zhao, H.; Wei, P.; Ge, J.; Gou, M.; Li, X.; Sun, L.; Cao, T.; Wang, P.; Zhou, C.; Zhang, R.; Liang, P.; Guo, H.; Wang, X.; Qin, C. F.; Chen, F.; Dong, C. Detection of SARS-CoV-2-Specific Humoral and Cellular Immunity in COVID-19 Convalescent Individuals. Immunity 2020, 52 (6), 971– 977 e3, DOI: 10.1016/j.immuni.2020.04.023Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXptlCjtb4%253D&md5=ad2de5c1a3bfd874429e20474c6dce86Detection of SARS-CoV-2-Specific Humoral and Cellular Immunity in COVID-19 Convalescent IndividualsNi, Ling; Ye, Fang; Cheng, Meng-Li; Feng, Yu; Deng, Yong-Qiang; Zhao, Hui; Wei, Peng; Ge, Jiwan; Gou, Mengting; Li, Xiaoli; Sun, Lin; Cao, Tianshu; Wang, Pengzhi; Zhou, Chao; Zhang, Rongrong; Liang, Peng; Guo, Han; Wang, Xinquan; Qin, Cheng-Feng; Chen, Fang; Dong, ChenImmunity (2020), 52 (6), 971-977.e3CODEN: IUNIEH; ISSN:1074-7613. (Elsevier Inc.)The World Health Organization has declared SARS-CoV-2 virus outbreak a worldwide pandemic. However, there is very limited understanding on the immune responses, esp. adaptive immune responses to SARS-CoV-2 infection. Here, we collected blood from COVID-19 patients who have recently become virus-free, and therefore were discharged, and detected SARS-CoV-2-specific humoral and cellular immunity in eight newly discharged patients. Follow-up anal. on another cohort of six patients 2 wk post discharge also revealed high titers of IgG antibodies. In all 14 patients tested, 13 displayed serum-neutralizing activities in a pseudotype entry assay. Notably, there was a strong correlation between neutralization antibody titers and the nos. of virus-specific T cells. Our work provides a basis for further anal. of protective immunity to SARS-CoV-2, and understanding the pathogenesis of COVID-19, esp. in the severe cases. It also has implications in developing an effective vaccine to SARS-CoV-2 infection.
- 31Peng, Y.; Mentzer, A. J.; Liu, G.; Yao, X.; Yin, Z.; Dong, D.; Dejnirattisai, W.; Rostron, T.; Supasa, P.; Liu, C.; Lopez-Camacho, C.; Slon-Campos, J.; Zhao, Y.; Stuart, D. I.; Paesen, G. C.; Grimes, J. M.; Antson, A. A.; Bayfield, O. W.; Hawkins, D.; Ker, D. S.; Wang, B.; Turtle, L.; Subramaniam, K.; Thomson, P.; Zhang, P.; Dold, C.; Ratcliff, J.; Simmonds, P.; de Silva, T.; Sopp, P.; Wellington, D.; Rajapaksa, U.; Chen, Y. L.; Salio, M.; Napolitani, G.; Paes, W.; Borrow, P.; Kessler, B. M.; Fry, J. W.; Schwabe, N. F.; Semple, M. G.; Baillie, J. K.; Moore, S. C.; Openshaw, P. J. M.; Ansari, M. A.; Dunachie, S.; Barnes, E.; Frater, J.; Kerr, G.; Goulder, P.; Lockett, T.; Levin, R.; Zhang, Y.; Jing, R.; Ho, L. P.; Oxford Immunology Network Covid-19 Response, T. c. C.; Investigators, I. C.; Cornall, R. J.; Conlon, C. P.; Klenerman, P.; Screaton, G. R.; Mongkolsapaya, J.; McMichael, A.; Knight, J. C.; Ogg, G.; Dong, T. Broad and strong memory CD4(+) and CD8(+) T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19. Nat. Immunol. 2020, 21 (11), 1336– 1345, DOI: 10.1038/s41590-020-0782-6Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhslCksbrF&md5=89639b3bc6b4cb0abd7bd1302973da74Broad and strong memory CD4+ and CD8+ T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19Peng, Yanchun; Mentzer, Alexander J.; Liu, Guihai; Yao, Xuan; Yin, Zixi; Dong, Danning; Dejnirattisai, Wanwisa; Rostron, Timothy; Supasa, Piyada; Liu, Chang; Lopez-Camacho, Cesar; Slon-Campos, Jose; Zhao, Yuguang; Stuart, David I.; Paesen, Guido C.; Grimes, Jonathan M.; Antson, Alfred A.; Bayfield, Oliver W.; Hawkins, Dorothy E. D. P.; Ker, De-Sheng; Wang, Beibei; Turtle, Lance; Subramaniam, Krishanthi; Thomson, Paul; Zhang, Ping; Dold, Christina; Ratcliff, Jeremy; Simmonds, Peter; de Silva, Thushan; Sopp, Paul; Wellington, Dannielle; Rajapaksa, Ushani; Chen, Yi-Ling; Salio, Mariolina; Napolitani, Giorgio; Paes, Wayne; Borrow, Persephone; Kessler, Benedikt M.; Fry, Jeremy W.; Schwabe, Nikolai F.; Semple, Malcolm G.; Baillie, J. Kenneth; Moore, Shona C.; Openshaw, Peter J. M.; Ansari, M. Azim; Dunachie, Susanna; Barnes, Eleanor; Frater, John; Kerr, Georgina; Goulder, Philip; Lockett, Teresa; Levin, Robert; Zhang, Yonghong; Jing, Ronghua; Ho, Ling-Pei; Cornall, Richard J.; Conlon, Christopher P.; Klenerman, Paul; Screaton, Gavin R.; Mongkolsapaya, Juthathip; McMichael, Andrew; Knight, Julian C.; Ogg, Graham; Dong, TaoNature Immunology (2020), 21 (11), 1336-1345CODEN: NIAMCZ; ISSN:1529-2908. (Nature Research)Abstr.: The development of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines and therapeutics will depend on understanding viral immunity. We studied T cell memory in 42 patients following recovery from COVID-19 (28 with mild disease and 14 with severe disease) and 16 unexposed donors, using interferon-γ-based assays with peptides spanning SARS-CoV-2 except ORF1. The breadth and magnitude of T cell responses were significantly higher in severe as compared with mild cases. Total and spike-specific T cell responses correlated with spike-specific antibody responses. We identified 41 peptides contg. CD4+ and/or CD8+ epitopes, including six immunodominant regions. Six optimized CD8+ epitopes were defined, with peptide-MHC pentamer-pos. cells displaying the central and effector memory phenotype. In mild cases, higher proportions of SARS-CoV-2-specific CD8+ T cells were obsd. The identification of T cell responses assocd. with milder disease will support an understanding of protective immunity and highlights the potential of including non-spike proteins within future COVID-19 vaccine design.
- 32Harris, P. E.; Brasel, T.; Massey, C.; Herst, C. V.; Burkholz, S.; Lloyd, P.; Blankenberg, T.; Bey, T. M.; Carback, R.; Hodge, T.; Ciotlos, S.; Wang, L.; Comer, J. E.; Rubsamen, R. M. A Synthetic Peptide CTL Vaccine Targeting Nucleocapsid Confers Protection from SARS-CoV-2 Challenge in Rhesus Macaques. Vaccines (Basel) 2021, 9 (5), 520, DOI: 10.3390/vaccines9050520Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitV2rs7rF&md5=2ae3877233432bba92c6ca98d60bef09A synthetic peptide CTL vaccine targeting nucleocapsid confers protection from SARS-CoV-2 challenge in rhesus macaquesHarris, Paul E.; Brasel, Trevor; Massey, Christopher; Herst, C. V.; Burkholz, Scott; Lloyd, Peter; Blankenberg, Tikoes; Bey, Thomas M.; Carback, Richard; Hodge, Thomas; Ciotlos, Serban; Wang, Lu; Comer, Jason E.; Rubsamen, Reid M.Vaccines (Basel, Switzerland) (2021), 9 (5), 520CODEN: VBSABP; ISSN:2076-393X. (MDPI AG)Background: Persistent transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has given rise to a COVID-19 pandemic. Several vaccines, conceived in 2020, that evoke protective spike antibody responses are being deployed in mass public health vaccination programs. Recent data suggests, however, that as sequence variation in the spike genome accumulates, some vaccines may lose efficacy. Methods: Using a macaque model of SARS-CoV-2 infection, we tested the efficacy of a peptide-based vaccine targeting MHC class I epitopes on the SARS-CoV-2 nucleocapsid protein. We administered biodegradable microspheres with synthetic peptides and adjuvants to rhesus macaques. Unvaccinated control and vaccinated macaques were challenged with 1 x 108 TCID50 units of SARS-CoV-2, followed by assessment of clin. symptoms and viral load, chest radiographs, and sampling of peripheral blood and bronchoalveolar lavage (BAL) fluid for downstream anal. Results: Vaccinated animals were free of pneumonia-like infiltrates characteristic of SARS-CoV-2 infection and presented with lower viral loads relative to controls. Gene expression in cells collected from BAL samples of vaccinated macaques revealed a unique signature assocd. with enhanced development of adaptive immune responses relative to control macaques. Conclusions: We demonstrate that a room temp. stable peptide vaccine based on known immunogenic HLA class I bound CTL epitopes from the nucleocapsid protein can provide protection against SARS-CoV-2 infection in nonhuman primates.
- 33Matchett, W. E.; Joag, V.; Stolley, J. M.; Shepherd, F. K.; Quarnstrom, C. F.; Mickelson, C. K.; Wijeyesinghe, S.; Soerens, A. G.; Becker, S.; Thiede, J. M.; Weyu, E.; O’Flanagan, S. D.; Walter, J. A.; Vu, M. N.; Menachery, V. D.; Bold, T. D.; Vezys, V.; Jenkins, M. K.; Langlois, R. A.; Masopust, D. Cutting Edge: Nucleocapsid Vaccine Elicits Spike-Independent SARS-CoV-2 Protective Immunity. J. Immunol. 2021, 207 (2), 376– 379, DOI: 10.4049/jimmunol.2100421Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFOiur%252FL&md5=b3347d5ee3b8bc3d824c7bfb0ddf1114Cutting edge: nucleocapsid vaccine elicits spike-independent SARS-CoV-2 protective immunityMatchett, William E.; Joag, Vineet; Stolley, J. Michael; Shepherd, Frances K.; Quarnstrom, Clare F.; Mickelson, Clayton K.; Wijeyesinghe, Sathi; Soerens, Andrew G.; Becker, Samuel; Thiede, Joshua M.; Weyu, Eyob; O'Flanagan, Stephen D.; Walter, Jennifer A.; Vu, Michelle N.; Menachery, Vineet D.; Bold, Tyler D.; Vezys, Vaiva; Jenkins, Marc K.; Langlois, Ryan A.; Masopust, DavidJournal of Immunology (2021), 207 (2), 376-379CODEN: JOIMA3; ISSN:0022-1767. (American Association of Immunologists)Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the COVID-19 pandemic. Neutralizing Abs target the receptor binding domain of the spike (S) protein, a focus of successful vaccine efforts. Concerns have arisen that S-specific vaccine immunity may fail to neutralize emerging variants. We show that vaccination with a human adenovirus type 5 vector expressing the SARS-CoV-2 nucleocapsid (N) protein can establish protective immunity, defined by reduced wt. loss and viral load, in both Syrian hamsters and K18-hACE2 mice. Challenge of vaccinated mice was assocd. with rapid N-specific T cell recall responses in the respiratory mucosa. This study supports the rationale for including addnl. viral Ags in SARS-CoV-2 vaccines, even if they are not a target of neutralizing Abs, to broaden epitope coverage and immune effector mechanisms.
- 34Chiuppesi, F.; Nguyen, V. H.; Park, Y.; Contreras, H.; Karpinski, V.; Faircloth, K.; Nguyen, J.; Kha, M.; Johnson, D.; Martinez, J.; Iniguez, A.; Zhou, Q.; Kaltcheva, T.; Frankel, P.; Kar, S.; Sharma, A.; Andersen, H.; Lewis, M. G.; Shostak, Y.; Wussow, F.; Diamond, D. J. Synthetic multiantigen MVA vaccine COH04S1 protects against SARS-CoV-2 in Syrian hamsters and non-human primates. NPJ Vaccines 2022, 7 (1), 7, DOI: 10.1038/s41541-022-00436-6Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhvFGjt70%253D&md5=50f41db00099e194241d3b0fef03a451Synthetic multiantigen MVA vaccine COH04S1 protects against SARS-CoV-2 in Syrian hamsters and non-human primatesChiuppesi, Flavia; Nguyen, Vu H.; Park, Yoonsuh; Contreras, Heidi; Karpinski, Veronica; Faircloth, Katelyn; Nguyen, Jenny; Kha, Mindy; Johnson, Daisy; Martinez, Joy; Iniguez, Angelina; Zhou, Qiao; Kaltcheva, Teodora; Frankel, Paul; Kar, Swagata; Sharma, Ankur; Andersen, Hanne; Lewis, Mark G.; Shostak, Yuriy; Wussow, Felix; Diamond, Don J.npj Vaccines (2022), 7 (1), 7CODEN: VACCBC; ISSN:2059-0105. (Nature Portfolio)Second-generation COVID-19 vaccines could contribute to establish protective immunity against SARS-CoV-2 and its emerging variants. We developed COH04S1, a synthetic multiantigen modified vaccinia Ankara-based SARS-CoV-2 vaccine that co-expresses spike and nucleocapsid antigens. Here, we report COH04S1 vaccine efficacy in animal models. We demonstrate that i.m. or intranasal vaccination of Syrian hamsters with COH04S1 induces robust Th1-biased antigen-specific humoral immunity and cross-neutralizing antibodies (NAb) and protects against wt. loss, lower respiratory tract infection, and lung injury following intranasal SARS-CoV-2 challenge. Moreover, we demonstrate that single-dose or two-dose vaccination of non-human primates with COH04S1 induces robust antigen-specific binding antibodies, NAb, and Th1-biased T cells, protects against both upper and lower respiratory tract infection following intranasal/intratracheal SARS-CoV-2 challenge, and triggers potent post-challenge anamnestic antiviral responses. These results demonstrate COH04S1-mediated vaccine protection in animal models through different vaccination routes and dose regimens, complementing ongoing investigation of this multiantigen SARS-CoV-2 vaccine in clin. trials.
- 35Afkhami, S.; D’Agostino, M. R.; Zhang, A.; Stacey, H. D.; Marzok, A.; Kang, A.; Singh, R.; Bavananthasivam, J.; Ye, G.; Luo, X.; Wang, F.; Ang, J. C.; Zganiacz, A.; Sankar, U.; Kazhdan, N.; Koenig, J. F. E.; Phelps, A.; Gameiro, S. F.; Tang, S.; Jordana, M.; Wan, Y.; Mossman, K. L.; Jeyanathan, M.; Gillgrass, A.; Medina, M. F. C.; Smaill, F.; Lichty, B. D.; Miller, M. S.; Xing, Z. Respiratory mucosal delivery of next-generation COVID-19 vaccine provides robust protection against both ancestral and variant strains of SARS-CoV-2. Cell 2022, 185 (5), 896– 915 e19, DOI: 10.1016/j.cell.2022.02.005Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XktVGrsLg%253D&md5=0484708611d00713d5cea5f903809600Respiratory mucosal delivery of next-generation COVID-19 vaccine provides robust protection against both ancestral and variant strains of SARS-CoV-2Afkhami, Sam; D'Agostino, Michael R.; Zhang, Ali; Stacey, Hannah D.; Marzok, Art; Kang, Alisha; Singh, Ramandeep; Bavananthasivam, Jegarubee; Ye, Gluke; Luo, Xiangqian; Wang, Fuan; Ang, Jann C.; Zganiacz, Anna; Sankar, Uma; Kazhdan, Natallia; Koenig, Joshua F. E.; Phelps, Allyssa; Gameiro, Steven F.; Tang, Shangguo; Jordana, Manel; Wan, Yonghong; Mossman, Karen L.; Jeyanathan, Mangalakumari; Gillgrass, Amy; Medina, Maria Fe C.; Smaill, Fiona; Lichty, Brian D.; Miller, Matthew S.; Xing, ZhouCell (Cambridge, MA, United States) (2022), 185 (5), 896-915.e19CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The emerging SARS-CoV-2 variants of concern (VOCs) threaten the effectiveness of current COVID-19 vaccines administered i.m. and designed to only target the spike protein. There is a pressing need to develop next-generation vaccine strategies for broader and long-lasting protection. Using adenoviral vectors (Ad) of human and chimpanzee origin, we evaluated Ad-vectored trivalent COVID-19 vaccines expressing spike-1, nucleocapsid, and RdRp antigens in murine models. Single-dose intranasal immunization, particularly with chimpanzee Ad-vectored vaccine, is superior to i.m. immunization in induction of the tripartite protective immunity consisting of local and systemic antibody responses, mucosal tissue-resident memory T cells and mucosal trained innate immunity. We further show that intranasal immunization provides protection against both the ancestral SARS-CoV-2 and two VOC, B.1.1.7 and B.1.351. Our findings indicate that respiratory mucosal delivery of Ad-vectored multivalent vaccine represents an effective next-generation COVID-19 vaccine strategy to induce all-around mucosal immunity against current and future VOC.
- 36Liu, L.; Zhong, Q.; Tian, T.; Dubin, K.; Athale, S. K.; Kupper, T. S. Epidermal injury and infection during poxvirus immunization is crucial for the generation of highly protective T cell-mediated immunity. Nat. Med. 2010, 16 (2), 224– 7, DOI: 10.1038/nm.2078Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXntVGjsA%253D%253D&md5=59664e863ab882eb1803281b6d0503c4Epidermal injury and infection during poxvirus immunization is crucial for the generation of highly protective T cell-mediated immunityLiu, Luzheng; Zhong, Qiong; Tian, Tian; Dubin, Krista; Athale, Shruti K.; Kupper, Thomas S.Nature Medicine (New York, NY, United States) (2010), 16 (2), 224-227CODEN: NAMEFI; ISSN:1078-8956. (Nature Publishing Group)Variola major (smallpox) infection claimed hundreds of millions lives before it was eradicated by a simple vaccination strategy: epicutaneous application of the related orthopoxvirus vaccinia virus (VACV) to superficially injured skin (skin scarification, s.s.). However, the remarkable success of this strategy was attributed to the immunogenicity of VACV rather than to the unique mode of vaccine delivery. The authors now show that VACV immunization via s.s., but not conventional injection routes, is essential for the generation of superior T cell-mediated immune responses that provide complete protection against subsequent challenges, independent of neutralizing antibodies. Skin-resident effector memory T cells (TEM cells) provide complete protection against cutaneous challenge, whereas protection against lethal respiratory challenge requires both respiratory mucosal TEM cells and central memory T cells (TCM cells). Vaccination with recombinant VACV (rVACV) expressing a tumor antigen was protective against tumor challenge only if delivered via the s.s. route; it was ineffective if delivered by hypodermic injection. The clin. safer nonreplicative modified vaccinia Ankara virus (MVA) also generated far superior protective immunity when delivered via the s.s. route compared to i.m. injection as used in MVA clin. trials. Thus, delivery of rVACV-based vaccines, including MVA vaccines, through phys. disrupted epidermis has clear-cut advantages over conventional vaccination via hypodermic injection.
- 37Mikhak, Z.; Strassner, J. P.; Luster, A. D. Lung dendritic cells imprint T cell lung homing and promote lung immunity through the chemokine receptor CCR4. J. Exp. Med. 2013, 210 (9), 1855– 69, DOI: 10.1084/jem.20130091Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVSgsbjF&md5=59b555d9feb77183237226eac2caba00Lung dendritic cells imprint T cell lung homing and promote lung immunity through the chemokine receptor CCR4Mikhak, Zamaneh; Strassner, James P.; Luster, Andrew D.Journal of Experimental Medicine (2013), 210 (9), 1855-1869CODEN: JEMEAV; ISSN:0022-1007. (Rockefeller University Press)T cell trafficking into the lung is crit. for lung immunity, but the mechanisms that mediate T cell lung homing are not well understood. Here, we show that lung dendritic cells (DCs) imprint T cell lung homing, as lung DC-activated T cells traffic more efficiently into the lung in response to inhaled antigen and at homeostasis compared with T cells activated by DCs from other tissues. Consequently, lung DC-imprinted T cells protect against influenza more effectively than do gut and skin DC-imprinted T cells. Lung DCs imprint the expression of CCR4 on T cells, and CCR4 contributes to T cell lung imprinting. Lung DC-activated, CCR4-deficient T cells fail to traffic into the lung as efficiently and to protect against influenza as effectively as lung DC-activated, CCR4-sufficient T cells. Thus, lung DCs imprint T cell lung homing and promote lung immunity in part through CCR4.
- 38Dijkman, K.; Aguilo, N.; Boot, C.; Hofman, S. O.; Sombroek, C. C.; Vervenne, R. A. W.; Kocken, C. H. M.; Marinova, D.; Thole, J.; Rodriguez, E.; Vierboom, M. P. M.; Haanstra, K. G.; Puentes, E.; Martin, C.; Verreck, F. A. W. Pulmonary MTBVAC vaccination induces immune signatures previously correlated with prevention of tuberculosis infection. Cell Rep. Med. 2021, 2 (1), 100187 DOI: 10.1016/j.xcrm.2020.100187Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhvFWlu7fE&md5=fd1734096801c6284856455ba6c42a86Pulmonary MTBVAC vaccination induces immune signatures previously correlated with prevention of tuberculosis infectionDijkman, Karin; Aguilo, Nacho; Boot, Charelle; Hofman, Sam O.; Sombroek, Claudia C.; Vervenne, Richard A. W.; Kocken, Clemens H. M.; Marinova, Dessislava; Thole, Jelle; Rodriguez, Esteban; Vierboom, Michel P. M.; Haanstra, Krista G.; Puentes, Eugenia; Martin, Carlos; Verreck, Frank A. W.Cell Reports Medicine (2021), 2 (1), 100187CODEN: CRMEDE; ISSN:2666-3791. (Elsevier Inc.)To fight tuberculosis, better vaccination strategies are needed. Live attenuated Mycobacterium tuberculosis-derived vaccine, MTBVAC, is a promising candidate in the pipeline, proven to be safe and immunogenic in humans so far. Independent studies have shown that pulmonary mucosal delivery of Bacillus Calmette-Guerin (BCG), the only tuberculosis (TB) vaccine available today, confers superior protection over std. intradermal immunization. Here we demonstrate that mucosal MTBVAC is well tolerated, eliciting polyfunctional T helper type 17 cells, interleukin-10, and Igs in the airway and yielding a broader antigenic profile than BCG in rhesus macaques. Beyond our previous work, we show that local Igs, induced by MTBVAC and BCG, bind to M. tuberculosis and enhance pathogen uptake. Furthermore, after pulmonary vaccination, but not M. tuberculosis infection, local T cells expressed high levels of mucosal homing and tissue residency markers. Our data show that pulmonary MTBVAC administration has the potential to enhance its efficacy and justifies further exploration of mucosal vaccination strategies in preclin. efficacy studies.
- 39Pan, Y.; Liu, L.; Tian, T.; Zhao, J.; Park, C. O.; Lofftus, S. Y.; Stingley, C. A.; Yan, Y.; Mei, S.; Liu, X.; Kupper, T. S. Epicutaneous immunization with modified vaccinia Ankara viral vectors generates superior T cell immunity against a respiratory viral challenge. NPJ Vaccines 2021, 6 (1), 1, DOI: 10.1038/s41541-020-00265-5Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXht1ejsbw%253D&md5=0b0b65986a334c7270199712cd484e48Epicutaneous immunization with modified vaccinia Ankara viral vectors generates superior T cell immunity against a respiratory viral challengePan, Youdong; Liu, Luzheng; Tian, Tian; Zhao, Jingxia; Park, Chang Ook; Lofftus, Serena Y.; Stingley, Claire A.; Yan, Yu; Mei, Shenglin; Liu, Xing; Kupper, Thomas S.npj Vaccines (2021), 6 (1), 1CODEN: VACCBC; ISSN:2059-0105. (Nature Research)Abstr.: Modified Vaccinia Ankara (MVA) was recently approved as a smallpox vaccine. Variola is transmitted by respiratory droplets and MVA immunization by skin scarification (s.s.) protected mice far more effectively against lethal respiratory challenge with vaccinia virus (VACV) than any other route of delivery, and at lower doses. Comparisons of s.s. with intradermal, s.c., or i.m. routes showed that MVAOVA s.s.-generated T cells were both more abundant and transcriptionally unique. MVAOVA s.s. produced greater nos. of lung Ova-specific CD8+ TRM and was superior in protecting mice against lethal VACVOVA respiratory challenge. Nearly as many lung TRM were generated with MVAOVA s.s. immunization compared to intra-tracheal immunization with MVAOVA and both routes vaccination protected mice against lethal pulmonary challenge with VACVOVA. Strikingly, MVAOVA s.s.-generated effector T cells exhibited overlapping gene transcriptional profiles to those generated via intra-tracheal immunization. Overall, our data suggest that heterologous MVA vectors immunized via s.s. are uniquely well-suited as vaccine vectors for respiratory pathogens, which may be relevant to COVID-19. In addn., MVA delivered via s.s. could represent a more ED-sparing smallpox vaccine.
- 40Sun, C.; Zhang, L.; Zhang, M.; Liu, Y.; Zhong, M.; Ma, X.; Chen, L. Induction of balance and breadth in the immune response is beneficial for the control of SIVmac239 replication in rhesus monkeys. J. Infect. 2010, 60 (5), 371– 381, DOI: 10.1016/j.jinf.2010.03.005Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3czgs12ktA%253D%253D&md5=19532898ec9bc6d864a673ac02836494Induction of balance and breadth in the immune response is beneficial for the control of SIVmac239 replication in rhesus monkeysSun Caijun; Zhang Lei; Zhang Maochao; Liu Yichu; Zhong Miao; Ma Xin; Chen LingThe Journal of infection (2010), 60 (5), 371-81 ISSN:.OBJECTIVES: The aim of this study was to induce cellular and humoral responses with enhanced breadth and more balanced magnitude as a possible approach for an effective HIV vaccine. METHODS: All nine of the SIVmac239 genes (gag, pol, env, nef, vif, vpx, vpr, rev and tat) were optimized for mammalian expression, synthesized and cloned into recombinant adenovirus type 5 (Ad5). These vectors were used as a vaccine regimen, and the immunogenicity and immune protection of this regimen was assessed in murine and macaques. RESULTS: A vaccine regimen including all nine genes of the SIVmac239 virus was developed, and it was demonstrated that in contrast to single antigen vaccination, the total SIV antigen regimen more effectively elicited the balanced and broad immune responses in murine and macaques. Moreover, the responses afforded effective immune control against infection and replication of the highly pathogenic SIVmac239. CONCLUSIONS: Induction of balance and breadth in the immune response is beneficial in controlling SIVmac239 replication in rhesus monkeys. This study provides insight for the future development of an effective HIV vaccine.
- 41Sun, Y.; Chen, M. L.; Yang, D.; Qin, W. B.; Quan, G. L.; Wu, C. B.; Pan, X. Self-assembly nanomicelle-microneedle patches with enhanced tumor penetration for superior chemo-photothermal therapy. Nano Res. 2022, 15 (3), 2335– 2346, DOI: 10.1007/s12274-021-3817-xGoogle Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXit1WlsLjO&md5=f91936fc6da51d2ec0020eaa0b7762f6Self-assembly nanomicelle-microneedle patches with enhanced tumor penetration for superior chemo-photothermal therapySun, Ying; Chen, Minglong; Yang, Dan; Qin, Wanbing; Quan, Guilan; Wu, Chuanbin; Pan, XinNano Research (2022), 15 (3), 2335-2346CODEN: NRAEB5; ISSN:1998-0000. (Springer GmbH)Nanomedicine with high specificity has been a promising tool for cancer diagnosis and therapy. However, the successful application of nanoparticle-based superficial cancer therapy is severely hindered by restricted deep tumor tissue accumulation and penetration. Herein, a self-assembly nanomicelle dissolving microneedle (DMN) patch according to the ''nano in micro'' strategy was conducted to co-deliver a first-line chemotherapeutic agent paclitaxel (PTX), and a photosensitizer IR780 (PTX/IR780-NMs @DMNs) for chemo-photothermal synergetic melanoma therapy. Upon direct insertion into the tumor site, DMNs created a regular and multipoint three-dimensional drug depot to maximize the tumor accumulation. Accompanied by the DMN dissoln., the compn. of the needle matrixes self-assembled into nanomicelles, which could efficiently penetrate deep tumor tissue. Upon laser irradn., the nanomicelles could not only ablate tumor cells directly by photothermal conversion but also trigger PTX release to induce tumor cell apoptosis. In vivo results showed that compared with i.v. injection, IR780 delivered by PTX/IR780-NMs @DMNs was almost completely accumulated at the tumor site. The antitumor results revealed that the PTX/IR780-NMs @DMNs could effectively eliminate tumors with an 88% curable rate without any damage to normal tissues. This work provides a versatile and generalizable framework for designing self-assembly DMN-mediated combination therapy to fight against superficial cancer.
- 42Lin, S. Q.; Quan, G. L.; Hou, A. L.; Yang, P. P.; Peng, T. T.; Gu, Y. K.; Qin, W. B.; Liu, R. B.; Ma, X. Y.; Pan, X.; Liu, H.; Wang, L. L.; Wu, C. B. Strategy for hypertrophic scar therapy: Improved delivery of triamcinolone acetonide using mechanically robust tip-concentrated dissolving microneedle array. J. Controlled Release 2019, 306, 69– 82, DOI: 10.1016/j.jconrel.2019.05.038Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtV2lt7nF&md5=18b2ceb48fb16cc5fb44706e28e2cfafStrategy for hypertrophic scar therapy: Improved delivery of triamcinolone acetonide using mechanically robust tip-concentrated dissolving microneedle arrayLin, Shiqi; Quan, Guilan; Hou, Ailin; Yang, Peipei; Peng, Tingting; Gu, Yukun; Qin, Wanbing; Liu, Rongben; Ma, Xiangyu; Pan, Xin; Liu, Hu; Wang, Lili; Wu, ChuanbinJournal of Controlled Release (2019), 306 (), 69-82CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)The purpose of this study was to provide an alternative treatment for HS by establishing a novel intradermal delivery system with a dissolving microneedle array (DMNA). To produce needles of higher mech. strength for successful insertion into the compact and hard HS tissue, hydroxypropyl-B-cyclodextrin (HP-B-CD) was added into sodium hyaluronic acid (HA), the needle material. The hydrogen interaction between HP-B-CD and HA restricted the mobility of the mol. chains, and subsequently increased the elastic modulus of the complex materials. The HP-B-CD also contributed to improved loading of the hydrophobic drug mols. into the DMNA needle tips. To assess the delivery of TA to the HS site via DMNA, an HS model was established in the ventral skin of New Zealand rabbits' ears. It was found that the value of the scar elevation index was decreased to normal, together with the down regulation of mRNA expressions of Collagen I and transforming growth factor-B1 (TGF-B1) following the administration of DMNA contg. TA (TA-DMNA). Western blotting results also revealed decreased protein expressions of both Collagen I and TGF-B1. Hence, TA-DMNA appears to be a promising alternative to multi-injection of TA injection, providing a convenient and low-pain therapeutic strategy for HS treatment.
- 43Li, M.; Chen, J.; Liu, Y.; Zhao, J.; Li, Y.; Hu, Y.; Chen, Y. Q.; Sun, L.; Shu, Y.; Feng, F.; Sun, C. Rational design of AAVrh10-vectored ACE2 functional domain to broadly block the cell entry of SARS-CoV-2 variants. Antiviral Res. 2022, 205, 105383 DOI: 10.1016/j.antiviral.2022.105383Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XitVKns7jM&md5=3bd24c716edaf1bcf064d8f764122282Rational design of AAVrh10-vectored ACE2 functional domain to broadly block the cell entry of SARS-CoV-2 variantsLi, Minchao; Chen, Jiaoshan; Liu, Yajie; Zhao, Jin; Li, Yanjun; Hu, Yunqi; Chen, Yao-qing; Sun, Litao; Shu, Yuelong; Feng, Fengling; Sun, CaijunAntiviral Research (2022), 205 (), 105383CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)The frequently emerging SARS-CoV-2 variants have weakened the effectiveness of existing COVID-19 vaccines and neutralizing antibody therapy. Nevertheless, the infections of SARS-CoV-2 variants still depend on angiotensin-converting enzyme 2 (ACE2) receptor-mediated cell entry, and thus the sol. human ACE2 (shACE2) is a potential decoy for broadly blocking SARS-CoV-2 variants. In this study, we firstly generated the recombinant AAVrh10-vectored shACE2 constructs, a kind of adeno-assocd. virus (AAV) serotype with pulmonary tissue tropism, and then validated its inhibition capacity against SARS-CoV-2 infection. To further optimize the minimized ACE2 functional domain candidates, a comprehensive anal. was performed to clarify the interactions between the ACE2 orthologs from various species and the receptor binding domain (RBD) of SARS-CoV-2 spike (S) protein. Based on the key interface amino acids, we designed a series of truncated ACE2 orthologs, and then assessed their potential affinity to bind to SARS-CoV-2 variants RBD in silico. Of note, we found that the 24-83aa fragment of dog ACE2 (dACE224-83) had a higher affinity to the RBD of SARS-CoV-2 variants than that of human ACE2. Importantly, AAVrh10-vectored shACE2 or dACE224-83 constructs exhibited a broadly blockage breadth against SARS-CoV-2 prototype and variants in vitro and ex vivo. Collectively, these data highlighted a promising therapeutic strategy against SARS-CoV-2 variants.
- 44Luo, H.; Jia, T.; Chen, J.; Zeng, S.; Qiu, Z.; Wu, S.; Li, X.; Lei, Y.; Wang, X.; Wu, W.; Zhang, R.; Zou, X.; Feng, T.; Ding, R.; Zhang, Y.; Chen, Y. Q.; Sun, C.; Wang, T.; Fang, S.; Shu, Y. The Characterization of Disease Severity Associated IgG Subclasses Response in COVID-19 Patients. Front. Immunol. 2021, 12, 632814 DOI: 10.3389/fimmu.2021.632814Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXpvF2iur4%253D&md5=1237dd3ed72ee775819c45cb22464425The characterization of disease severity associated IgG subclasses response in COVID-19 patientsLuo, Huanle; Jia, Tingting; Chen, Jiamin; Zeng, Shike; Qiu, Zengzhao; Wu, Shu; Li, Xu; Lei, Yuxuan; Wang, Xin; Wu, Weihua; Zhang, Renli; Zou, Xuan; Feng, Tiejian; Ding, Ruxia; Zhang, Yue; Chen, Yao-Qing; Sun, Caijun; Wang, Tian; Fang, Shisong; Shu, YuelongFrontiers in Immunology (2021), 12 (), 632814CODEN: FIRMCW; ISSN:1664-3224. (Frontiers Media S.A.)Increasing evidence suggests that dysregulated immune responses are assocd. with the clin. outcome of coronavirus disease 2019 (COVID-19). Nucleocapsid protein (NP)-, spike (S)-, receptor binding domain (RBD)- specific Ig isotypes, IgG subclasses and neutralizing antibody (NAb) were analyzed in 123 serum from 63 hospitalized patients with severe, moderate, mild or asymptomatic COVID-19. Mild to modest correlations were found between disease severity and antigen specific IgG subclasses in serum, of which IgG1 and IgG3 were neg. assocd. with viral load in nasopharyngeal swab. Multiple cytokines were significantly related with antigen-specific Ig isotypes and IgG subclasses, and IL-1β was pos. correlated with most antibodies. Furthermore, the old patients (≤ 60 years old) had higher levels of chemokines, increased NAb activities and SARS-CoV-2 specific IgG1, and IgG3 responses and compromised T cell responses compared to the young patients (≤ 18 years old), which are related with more severe cases. Higher IgG1 and IgG3 were found in COVID-19 patients with comorbidities while biol. sex had no effect on IgG subclasses. Overall, the authors have identified diseases severity was related to higher antibodies, of which IgG subclasses had weakly neg. correlation with viral load, and cytokines were significantly assocd. with antibody response. Further, advancing age and comorbidities had obvious effect on IgG1 and IgG3.
- 45Li, P.; Wang, Q.; He, Y.; Yang, C.; Zhang, Z.; Liu, Z.; Liu, B.; Yin, L.; Cui, Y.; Hu, P.; Liu, Y.; Zheng, P.; Wang, W.; Qu, L.; Sun, C.; Guan, S.; Feng, L.; Chen, L. Booster vaccination is required to elicit and maintain COVID-19 vaccine-induced immunity in SIV-infected macaques. Emerg. Microbes Infect. 2023, 12 (1), e2136538 DOI: 10.1080/22221751.2022.2136538Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXkt1yhtro%253D&md5=53b4acd282d993f26aab9b3221932c90Booster vaccination is required to elicit and maintain COVID-19 vaccine-induced immunity in SIV-infected macaquesLi, Pingchao; Wang, Qian; He, Yizi; Yang, Chenchen; Zhang, Zhengyuan; Liu, Zijian; Liu, Bo; Yin, Li; Cui, Yilan; Hu, Peiyu; Liu, Yichu; Zheng, Pingqian; Wang, Wei; Qu, Linbing; Sun, Caijun; Guan, Suhua; Feng, Liqiang; Chen, LingEmerging Microbes & Infections (2023), 12 (1), e2136538/1-e2136538/13CODEN: EMIMC4; ISSN:2222-1751. (Taylor & Francis Ltd.)Prolonged infection and possible evolution of SARS-CoV-2 in patients living with uncontrolled HIV-1 infection highlight the importance of an effective vaccination regimen, yet the immunogenicity of COVID-19 vaccines and predictive immune biomarkers have not been well investigated. Herein, we report that the magnitude and persistence of antibody and cell-mediated immunity (CMI) elicited by an Ad5-vectored COVID-19 vaccine are impaired in SIV-infected macaques with high viral loads (> 105 genome copies per mL plasma, SIVhi) but not in macaques with low viral loads (< 105, SIVlow). After a second vaccination, the immune responses are robustly enhanced in all uninfected and SIVlow macaques. These responses also show a moderate increase in 70% SIVhi macaques but decline sharply soon after. Further anal. reveals that decreased antibody and CMI responses are assocd. with reduced circulating follicular helper T cell (TFH) counts and aberrant CD4/CD8 ratios, resp., indicating that dysregulation of CD4+ T cells by SIV infection impairs the COVID-19 vaccine-induced immunity. Ad5-vectored COVID-19 vaccine shows no impact on SIV loads or SIV-specific CMI responses. Our study underscores the necessity of frequent booster vaccinations in HIV-infected patients and provides indicative biomarkers for predicting vaccination effectiveness in these patients.
- 46Wen, Z.; Fang, C.; Liu, X.; Liu, Y.; Li, M.; Yuan, Y.; Han, Z.; Wang, C.; Zhang, T.; Sun, C. A recombinant Mycobacterium smegmatis-based surface display system for developing the T cell-based COVID-19 vaccine. Hum. Vaccines Immunother. 2023, 19 (1), 2171233 DOI: 10.1080/21645515.2023.2171233Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXjsVGku7Y%253D&md5=10974bd628fd6d5c6ea2a6876037bc54A recombinant Mycobacterium smegmatis-based surface display system for developing the T cell-based COVID-19 vaccineWen, Ziyu; Fang, Cuiting; Liu, Xinglai; Liu, Yan; Li, Minchao; Yuan, Yue; Han, Zirong; Wang, Congcong; Zhang, Tianyu; Sun, CaijunHuman Vaccines & Immunotherapeutics (2023), 19 (1), 2171233/1-2171233/12CODEN: HVIUAK; ISSN:2164-554X. (Taylor & Francis Ltd.)The immune escape mutations of SARS-CoV-2 variants emerged frequently, posing a new challenge to weaken the protective efficacy of current vaccines. Thus, the development of novel SARS-CoV-2 vaccines is of great significance for future epidemic prevention and control. We herein reported constructing the attenuated Mycobacterium smegmatis (M. smegmatis) as a bacterial surface display system to carry the spike (S) and nucleocapsid (N) of SARS-CoV-2. To mimic the native localization on the surface of viral particles, the S or N antigen was fused with truncated PE_PGRS33 protein, which is a transportation component onto the cell wall of Mycobacterium tuberculosis (M.tb). The sub-cellular fraction anal. demonstrated that S or N protein was exactly expressed onto the surface (cell wall) of the recombinant M. smegmatis. After the immunization of the M. smegmatis-based COVID-19 vaccine candidate in mice, S or N antigen-specific T cell immune responses were effectively elicited, and the subsets of central memory CD4+ T cells and CD8+ T cells were significantly induced. Further anal. showed that there were some potential cross-reactive CTL epitopes between SARS-CoV-2 and M.smegmatis. Overall, our data provided insights that M. smegmatis-based bacterial surface display system could be a suitable vector for developing T cell-based vaccines against SARS-CoV-2 and other infectious diseases.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
- SupportingSupporting0
- MentioningMentioning1
- ContrastingContrasting0
This article is cited by 1 publications.
- Hong Liu, Haolin Chen, Zeyu Yang, Zhenfu Wen, Zhan Gao, Zhijia Liu, Lixin Liu, Yongming Chen. Precision Nanovaccines for Potent Vaccination. JACS Au 2024, 4
(8)
, 2792-2810. https://doi.org/10.1021/jacsau.4c00568
了解这些指标
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles 推荐文章
Reactive Oxygen Species-Responsive Gel-Based Microneedle Patches for Prolonged and Intelligent Psoriasis Management
活性氧反应凝胶微针贴片,用于长期、智能地治疗牛皮癣Separable Microneedle Patch to Protect and Deliver DNA Nanovaccines Against COVID-19
可分离微针贴片用于保护和输送抗 COVID-19 的 DNA 纳米疫苗Microneedle Patch Delivery of PROTACs for Anti-Cancer Therapy
Multifunctional Microneedle Patches via Direct Ink Drawing of Nanocomposite Inks for Personalized Transdermal Drug Delivery
Injectable Hydrogel Mucosal Vaccine Elicits Protective Immunity against Respiratory Viruses
References
This article references 46 other publications.
- 1Eyawo, O.; Viens, A. M.; Ugoji, U. C. Lockdowns and low- and middle-income countries: building a feasible, effective, and ethical COVID-19 response strategy. Global Health 2021, 17 (1), 13 DOI: 10.1186/s12992-021-00662-y1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3srivFaitw%253D%253D&md5=952791be0312758e4ab48595f3c5ec7dLockdowns and low- and middle-income countries: building a feasible, effective, and ethical COVID-19 response strategyEyawo Oghenowede; Viens A M; Ugoji Uchechukwu ChidiebereGlobalization and health (2021), 17 (1), 13 ISSN:.Lockdowns can be an effective pandemic response strategy that can buy much needed time to slow disease transmission and adequately scale up preventative, diagnostic, and treatment capacities. However, the broad restrictive measures typically associated with lockdowns, though effective, also comes at a cost - imposing significant social and economic burdens on individuals and societies, especially for those in low- and middle-income countries (LMICs). Like most high-income countries (HICs), many LMICs initially adopted broad lockdown strategies for COVID-19 in the first wave of the pandemic. While many HICs experiencing subsequent waves have returned to employing lockdown strategies until they can receive the first shipments of COVID-19 vaccine, many LMICs will likely have to wait much longer to get comparable access for their own citizens. In leaving LMICs vulnerable to subsequent waves for a longer period of time without vaccines, there is a risk LMICs will be tempted to re-impose lockdown measures in the meantime. In response to the urgent need for more policy development around the contextual challenges involved in employing such measures, we propose some strategies LMICs could adopt for safe and responsible lockdown entrance/exit or to avoid re-imposing coercive restrictive lockdown measures altogether.
- 2Feng, F.; Wen, Z.; Chen, J.; Yuan, Y.; Wang, C.; Sun, C. Strategies to Develop a Mucosa-Targeting Vaccine against Emerging Infectious Diseases. Viruses 2022, 14 (3), 520, DOI: 10.3390/v140305202https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XosVGms7Y%253D&md5=08f02a7c628671ba18325f453d4a1258Strategies to Develop a Mucosa-Targeting Vaccine against Emerging Infectious DiseasesFeng, Fengling; Wen, Ziyu; Chen, Jiaoshan; Yuan, Yue; Wang, Congcong; Sun, CaijunViruses (2022), 14 (3), 520CODEN: VIRUBR; ISSN:1999-4915. (MDPI AG)A review. Numerous pathogenic microbes, including viruses, bacteria, and fungi, usually infect the host through the mucosal surfaces of the respiratory tract, gastrointestinal tract, and reproductive tract. The mucosa is well known to provide the first line of host defense against pathogen entry by phys., chem., biol., and immunol. barriers, and therefore, mucosa-targeting vaccination is emerging as a promising strategy for conferring superior protection. However, there are still many challenges to be solved to develop an effective mucosal vaccine, such as poor adhesion to the mucosal surface, insufficient uptake to break through the mucus, and the difficulty in avoiding strong degrdn. through the gastrointestinal tract. Recently, increasing efforts to overcome these issues have been made, and we herein summarize the latest findings on these strategies to develop mucosa-targeting vaccines, including a novel needle-free mucosa-targeting route, the development of mucosa-targeting vectors, the administration of mucosal adjuvants, encapsulating vaccines into nanoparticle formulations, and antigen design to conjugate with mucosa-targeting ligands. Our work will highlight the importance of further developing mucosal vaccine technol. to combat the frequent outbreaks of infectious diseases.
- 3Seaman, C. P.; Kahn, A. L.; Kristensen, D.; Steinglass, R.; Spasenoska, D.; Scott, N.; Morgan, C. Controlled temperature chain for vaccination in low- and middle-income countries: a realist evidence synthesis. Bull. World Health Organ. 2022, 100 (8), 491– 502, DOI: 10.2471/BLT.21.2876963https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2Mbntleksg%253D%253D&md5=a6628201b75aa48018813d21fcfd8468Controlled temperature chain for vaccination in low- and middle-income countries: a realist evidence synthesisSeaman Christopher P; Scott Nick; Kahn Anna-Lea; Kristensen Debra; Steinglass Robert; Spasenoska Dijana; Morgan ChristopherBulletin of the World Health Organization (2022), 100 (8), 491-502 ISSN:.Objective: To evaluate the evidence describing how the controlled temperature chain approach for vaccination could lead to improved equitable immunization coverage in low- and middle-income countries. Methods: We created a theory of change construct from the Controlled temperature chain: strategic roadmap for priority vaccines 2017-2020, containing four domains: (i) uptake and demand for the approach; (ii) compliance and safe use of the approach; (iii) programmatic efficiency gains from the approach; and (iv) improved equitable immunization coverage. To verify and improve the theory of change, we applied a realist review method to analyse published descriptions of controlled temperature chain or closely related experiences. Findings: We evaluated 34 articles, describing 22 unique controlled temperature chain or closely related experiences across four World Health Organization regions. We identified a strong demand for this approach among service delivery providers; however, generating an equal level of demand among policy-makers requires greater evidence on economic benefits and on vaccination coverage gains, and use case definitions. Consistent evidence supported safety of the approach when integrated into special vaccination programmes. Feasible training and supervision supported providers in complying with protocols. Time-savings were the main evidence for efficiency gains, while cost-saving data were minimal. Improved equitable coverage was reported where vaccine storage beyond the cold chain enabled access to hard-to-reach populations. No evidence indicated an inferior vaccine effectiveness nor increased adverse event rates for vaccines delivered under the approach. Conclusion: Synthesized evidence broadly supported the initial theory of change. Addressing evidence gaps on economic benefits and coverage gains may increase future uptake.
- 4Freeman, D.; Lambe, S.; Yu, L. M.; Freeman, J.; Chadwick, A.; Vaccari, C.; Waite, F.; Rosebrock, L.; Petit, A.; Vanderslott, S.; Lewandowsky, S.; Larkin, M.; Innocenti, S.; McShane, H.; Pollard, A. J.; Loe, B. S. Injection fears and COVID-19 vaccine hesitancy. Psychol. Med. 2023, 53 (4), 1185– 1195, DOI: 10.1017/S00332917210026094https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2c7ms1yhsg%253D%253D&md5=e6ceea4c19eb9e344e83d0e80d4e36d1Injection fears and COVID-19 vaccine hesitancyFreeman Daniel; Lambe Sinead; Freeman Jason; Waite Felicity; Rosebrock Laina; Petit Ariane; Freeman Daniel; Lambe Sinead; Waite Felicity; Rosebrock Laina; Petit Ariane; Freeman Daniel; Waite Felicity; Yu Ly-Mee; Chadwick Andrew; Vaccari Cristian; Vanderslott Samantha; Pollard Andrew J; Lewandowsky Stephan; Larkin Michael; Innocenti Stefania; McShane Helen; McShane Helen; Pollard Andrew J; Loe Bao ShengPsychological medicine (2023), 53 (4), 1185-1195 ISSN:.BACKGROUND: When vaccination depends on injection, it is plausible that the blood-injection-injury cluster of fears may contribute to hesitancy. Our primary aim was to estimate in the UK adult population the proportion of COVID-19 vaccine hesitancy explained by blood-injection-injury fears. METHODS: In total, 15 014 UK adults, quota sampled to match the population for age, gender, ethnicity, income and region, took part (19 January-5 February 2021) in a non-probability online survey. The Oxford COVID-19 Vaccine Hesitancy Scale assessed intent to be vaccinated. Two scales (Specific Phobia Scale-blood-injection-injury phobia and Medical Fear Survey-injections and blood subscale) assessed blood-injection-injury fears. Four items from these scales were used to create a factor score specifically for injection fears. RESULTS: In total, 3927 (26.2%) screened positive for blood-injection-injury phobia. Individuals screening positive (22.0%) were more likely to report COVID-19 vaccine hesitancy compared to individuals screening negative (11.5%), odds ratio = 2.18, 95% confidence interval (CI) 1.97-2.40, p < 0.001. The population attributable fraction (PAF) indicated that if blood-injection-injury phobia were absent then this may prevent 11.5% of all instances of vaccine hesitancy, AF = 0.11; 95% CI 0.09-0.14, p < 0.001. COVID-19 vaccine hesitancy was associated with higher scores on the Specific Phobia Scale, r = 0.22, p < 0.001, Medical Fear Survey, r = 0.23, p = <0.001 and injection fears, r = 0.25, p < 0.001. Injection fears were higher in youth and in Black and Asian ethnic groups, and explained a small degree of why vaccine hesitancy is higher in these groups. CONCLUSIONS: Across the adult population, blood-injection-injury fears may explain approximately 10% of cases of COVID-19 vaccine hesitancy. Addressing such fears will likely improve the effectiveness of vaccination programmes.
- 5Wang, H.; Cui, M.; Li, S.; Wu, F.; Jiang, S.; Chen, H.; Yuan, J.; Sun, C. Perception and willingness toward various immunization routes for COVID-19 vaccines: a cross-sectional survey in China. Front. Public Health 2023, 11, 1192709, DOI: 10.3389/fpubh.2023.11927095https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2sfmsVyrsA%253D%253D&md5=bd5836833032ac03af5ed4330ddc3addPerception and willingness toward various immunization routes for COVID-19 vaccines: a cross-sectional survey in ChinaWang Haohang; Cui Mingting; Li Shunran; Wu Fan; Sun Caijun; Jiang Shiqiang; Yuan Jianhui; Chen Hongbiao; Sun Caijun; Sun CaijunFrontiers in public health (2023), 11 (), 1192709 ISSN:.Background: To date, most vaccines, including the COVID-19 vaccine, are mainly administered by intramuscular injection, which might lead to vaccine hesitancy in some populations due to needle fear. Alternatively, needle-free immunization technology is extensively developed to improve the efficacy and acceptance of vaccination. However, there is no study to report the perception and willingness toward various immunization routes of the COVID-19 vaccine in the general population. Methods: A cross-sectional survey was conducted nationwide using an online questionnaire. Bivariate analyses were undertaken to assess variable associations among the participants who reported a hesitancy to receive the COVID-19 booster vaccination. Multivariable logistic regression with a backward step-wise approach was used to analyze the predicted factors associated with the willingness to receive the COVID-19 booster vaccination. Results: A total of 3,244 valid respondents were included in this survey, and 63.2% of participants thought they had a good understanding of intramuscular injection, but only 20.7, 9.2, 9.4, and 6.0% of participants had a self-perceived good understanding of inhalation vaccine, nasal spray vaccine, oral vaccine, and microneedle patch vaccine. Correspondingly, there was high acceptance for intramuscular injection (76.5%), followed by oral inhalation (64.4%) and nasal spray (43.0%). Those participants who were only willing to receive an intramuscular vaccine had less vaccine knowledge (OR = 0.78; 95% CI: 0.65-0.94) than those who were willing to receive a needle-free vaccine (OR = 1.97; 95% CI: 1.52-2.57). Some factors were found to be associated with vaccine hesitancy toward booster COVID-19 vaccination. Conclusion: Needle-free vaccination is a promising technology for the next generation of vaccines, but we found that intramuscular injection was still the most acceptable immunization route in this survey. One major reason might be that most people lack knowledge about needle-free vaccination. We should strengthen the publicity of needle-free vaccination technology, and thus improve the acceptance and coverage of vaccination in different populations.
- 6Yang, Y.; Li, Z.; Huang, P.; Lin, J.; Li, J.; Shi, K.; Lin, J.; Hu, J.; Zhao, Z.; Yu, Y.; Chen, H.; Zeng, X.; Mei, L. Rapidly separating dissolving microneedles with sustained-release colchicine and stabilized uricase for simplified long-term gout management. Acta Pharm. Sin. B 2023, 13 (8), 3454– 3470, DOI: 10.1016/j.apsb.2023.02.0116https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXlsVaqtb0%253D&md5=f2880f0139f6a9b8d607bea788c7b77cRapidly separating dissolving microneedles with sustained-release colchicine and stabilized uricase for simplified long-term gout managementYang, Yao; Li, Zimu; Huang, Ping; Lin, Jiachan; Li, Jinyuan; Shi, Kexin; Lin, Jiahui; Hu, Jingwen; Zhao, Zhuoxian; Yu, Yongkang; Chen, Hongzhong; Zeng, Xiaowei; Mei, LinActa Pharmaceutica Sinica B (2023), 13 (8), 3454-3470CODEN: APSBCW; ISSN:2211-3835. (Elsevier B.V.)Despite growing prevalence and incidence, the management of gout remains suboptimal. The intermittent nature of the gout makes the long-term urate-lowering therapy (ULT) particularly important for gout management. However, patients are reluctant to take medication day after day to manage incurable occasional gout flares, and suffer from possible long-term toxicity. Therefore, a safe and easy-to-operate drug delivery system with simple prepn. for the long-term management of gout is very necessary. Here, a chitosan-contg. sustained-release microneedle system co-loaded with colchicine and uricase liposomes were fabricated to achieve this goal. This microneedle system was confirmed to successfully deliver the drug to the skin and maintain a one-week drug retention. Furthermore, its powerful therapeutic potency to manage gout was investigated in both acute gouty and chronic gouty models. Besides, the drug co-delivery system could help avoid long-term daily oral colchicine, a drug with a narrow therapeutic index. This system also avoids mass injection of uricase by improving its stability, enhancing the clin. application value of uricase. In general, this two-drug system reduces the dosage of uricase and colchicine and improves the patient's compliance, which has a strong clin. translation.
- 7Yang, L.; Yang, Y.; Chen, H.; Mei, L.; Zeng, X. Polymeric microneedle-mediated sustained release systems: Design strategies and promising applications for drug delivery. Asian J. Pharm. Sci. 2022, 17 (1), 70– 86, DOI: 10.1016/j.ajps.2021.07.0027https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2MzhtlWgtw%253D%253D&md5=5e08045861ec3452358788976beebaa5Polymeric microneedle-mediated sustained release systems: Design strategies and promising applications for drug deliveryYang Li; Yang Yao; Chen Hongzhong; Mei Lin; Zeng Xiaowei; Mei LinAsian journal of pharmaceutical sciences (2022), 17 (1), 70-86 ISSN:.Parenteral sustained release drug formulations, acting as preferable platforms for long-term exposure therapy, have been wildly used in clinical practice. However, most of these delivery systems must be given by hypodermic injection. Therefore, issues including needle-phobic, needle-stick injuries and inappropriate reuse of needles would hamper the further applications of these delivery platforms. Microneedles (MNs) as a potential alternative system for hypodermic needles can benefit from minimally invasive and self-administration. Recently, polymeric microneedle-mediated sustained release systems (MN@SRS) have opened up a new way for treatment of many diseases. Here, we reviewed the recent researches in MN@SRS for transdermal delivery, and summed up its typical design strategies and applications in various diseases therapy, particularly focusing on the applications in contraception, infection, cancer, diabetes, and subcutaneous disease. An overview of the present clinical translation difficulties and future outlook of MN@SRS was also provided.
- 8Menon, I.; Bagwe, P.; Gomes, K. B.; Bajaj, L.; Gala, R.; Uddin, M. N.; D’Souza, M. J.; Zughaier, S. M. Microneedles: A New Generation Vaccine Delivery System. Micromachines 2021, 12 (4), 435, DOI: 10.3390/mi12040435There is no corresponding record for this reference.
- 9Larraneta, E.; Lutton, R. E. M.; Woolfson, A. D.; Donnelly, R. F. Microneedle arrays as transdermal and intradermal drug delivery systems: Materials science, manufacture and commercial development. Mater. Sci. Eng. R-Reports 2016, 104, 1– 32, DOI: 10.1016/j.mser.2016.03.001There is no corresponding record for this reference.
- 10Iwata, H.; Kakita, K.; Imafuku, K.; Takashima, S.; Haga, N.; Yamaguchi, Y.; Taguchi, K.; Oyamada, T. Safety and dose-sparing effect of Japanese encephalitis vaccine administered by microneedle patch in uninfected, healthy adults (MNA-J): a randomised, partly blinded, active-controlled, phase 1 trial. Lancet Microbe 2022, 3 (2), E96– E104, DOI: 10.1016/S2666-5247(21)00269-X10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XlvV2nsbk%253D&md5=bdb5041bfb4bd6d8f1c8a6cc4677d276Safety and dose-sparing effect of Japanese encephalitis vaccine administered by microneedle patch in uninfected, healthy adults (MNA-J): a randomised, partly blinded, active-controlled, phase 1 trialIwata, Hiroaki; Kakita, Kosuke; Imafuku, Keisuke; Takashima, Shota; Haga, Naoya; Yamaguchi, Yasuyuki; Taguchi, Kenji; Oyamada, TakayoshiLancet Microbe (2022), 3 (2), e96-e104CODEN: LMAIAR; ISSN:2666-5247. (Elsevier Ltd.)Background It is unclear whether microneedle vaccinations of Japanese encephalitis virus can induce sufficient neutralising antibodies and reduce the amt. of vaccine needed. We aimed to assess the safety and dose-sparing effect of a microneedle vaccine patch against Japanese encephalitis in healthy individuals who are naive to both the vaccine and natural infection. Methods The MNA-J study was a randomised, partly blinded, active-controlled, phase 1 clin. trial at Hokkaido University (Sapporo, Japan) that enrolled healthy adults aged 20-34 years with no history of Japanese encephalitis vaccination nor of infection as confirmed by seronegativity. We excluded individuals who had been infected with or vaccinated against Japanese encephalitis. Eligible participants were randomly assigned (1:1:1) to one of three groups to receive inactivated Japanese encephalitis vaccine administered twice, 3 wk apart, by either 2·5 μg per injection by s.c. injection, 0·63 μg per patch by high-dose microneedle array (MNA-25%), or 0·25 μg per patch by low-dose microneedle array (MNA-10%). The randomisation sequence, using stratification by cohort and blocks of six, was computer-generated by a statistician who was unaware of group assignment. After administration, the remaining amt. of unadministered vaccine was measured by ELISA and calcd. as the delivered amt. of vaccine. The primary outcome was the neutralising antibody titer at day 42 after first immunization. Successful seroconversion was defined as post-vaccination titers of 1·3 (log10) or higher in individuals whose pre-vaccination titers had been less than 1 (log10). Findings Between Aug 31 and Sept 2, 2019, 39 participants were enrolled and each was randomly assigned to a group (n = 13 per group). No serious adverse events were obsd. All participants in the microneedle array groups had a localised erythematous reaction. The amt. of vaccine delivered by microneedle array to each participant was 0·63-1·15 μg (50-92%) of the full 1·26 μg for the MNA-25% group and 0·25-0·41 μg (51-84%) of the full 0·50 μg for the MNA-10% group. All participants demonstrated seroconversion at day 42, and the mean titers (log10) were 2·55 for MNA-25%, 2·04 for MNA-10%, and 2·08 for s.c. injection. Interpretation A microneedle patch of the Japanese encephalitis vaccine is safe, well tolerated, and immunogenically effective. The dose-sparing effect suggests a significant potential to reduce the amt. of immunogens needed. However, improved delivery is needed to make it more tolerable and user friendly.
- 11Vander Straeten, A.; Sarmadi, M.; Daristotle, J. L.; Kanelli, M.; Tostanoski, L. H.; Collins, J.; Pardeshi, A.; Han, J.; Varshney, D.; Eshaghi, B.; Garcia, J.; Forster, T. A.; Li, G.; Menon, N.; Pyon, S. L.; Zhang, L.; Jacob-Dolan, C.; Powers, O. C.; Hall, K.; Alsaiari, S. K.; Wolf, M.; Tibbitt, M. W.; Farra, R.; Barouch, D. H.; Langer, R.; Jaklenec, A. A microneedle vaccine printer for thermostable COVID-19 mRNA vaccines. Nat. Biotechnol. 2023, DOI: 10.1038/s41587-023-01774-zThere is no corresponding record for this reference.
- 12Feng, F.; Hao, H.; Zhao, J.; Li, Y.; Zhang, Y.; Li, R.; Wen, Z.; Wu, C.; Li, M.; Li, P.; Chen, L.; Tang, R.; Wang, X.; Sun, C. Shell-mediated phagocytosis to reshape viral-vectored vaccine-induced immunity. Biomaterials 2021, 276, 121062 DOI: 10.1016/j.biomaterials.2021.12106212https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvVaitLzM&md5=567d4ffcfc8d20b7cff865ba033d62abShell-mediated phagocytosis to reshape viral-vectored vaccine-induced immunityFeng, Fengling; Hao, Haibin; Zhao, Jin; Li, Yanjun; Zhang, Ying; Li, Ruiting; Wen, Ziyu; Wu, Chunxiu; Li, Minchao; Li, Pingchao; Chen, Ling; Tang, Ruikang; Wang, Xiaoyu; Sun, CaijunBiomaterials (2021), 276 (), 121062CODEN: BIMADU; ISSN:0142-9612. (Elsevier Ltd.)Adenovirus (Ad) has been extensively developed as a gene delivery vector, but the potential side effect caused by systematic immunization remains one major obstacle for its clin. application. Needle-free mucosal immunization with Ad-based vaccine shows advantages but still faces poor mucosal responses. We herein report that the chem. engineering of single live viral-based vaccine effectively modulated the location and pattern of the subsequently elicited immunity. Through precisely assembly of functional materials onto single live Ad particle, the modified virus entered host cell in a phagocytosis-dependent manner, which is completely distinct from the receptor-mediated entry of native Ad. RNA-Seq data further demonstrated that the modified Ad-induced innate immunity was sharply reshaped via phagocytosis-related pathway, therefore promoting the activation and mature of antigen presentation cells (APC). Moreover, the functional shell enabled the modified Ad-based vector with enhanced muco-adhesion to nasal tissues in mice, and then prolonged resident time onto mucosal surface, leading to the robust mucosal IgA prodn. and T cell immunity at local and even remote mucosal-assocd. lymphoid tissues. This study demonstrated that vaccine-induced immunity can be well modulated by chem. engineering, and this method provides the rational design for needle-free mucosa-targeting vaccine against a variety of emerging infectious diseases.
- 13Sun, C. J.; Pan, S. P.; Xie, Q. X.; Xiao, L. J. Preparation of chitosan-plasmid DNA nanoparticles encoding zona pellucida glycoprotein-3alpha and its expression in mouse. Mol. Reprod. Dev. 2004, 68 (2), 182– 8, DOI: 10.1002/mrd.2005813https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjvVyjsrs%253D&md5=b0f09ad9675e0919f4c6c23ffb08f770Preparation of chitosan-plasmid DNA nanoparticles encoding zona pellucida glycoprotein-3α and its expression in mouseSun, Cai-Jun; Pan, Shan-Pei; Xie, Qi-Xuan; Xiao, Luan-JuanMolecular Reproduction and Development (2004), 68 (2), 182-188CODEN: MREDEE; ISSN:1040-452X. (Wiley-Liss, Inc.)In the present study, the porcine zona pellucida (ZP)-3α eukaryotic expression vector pVAX1-pZP3α was constructed by genetic recombinant technol., then the recombinant plasmid was encapsulated in nanoparticles with chitosan, and the imaging of chitosan/pVAX1-pZP3α nanoparticles by At. Force Microscope (AFM) was processed. Feeding mouse with those microencapsulation by gastric larvae, and after 5 days, detecting its expression in mouse intestine by RT-PCR and indirect immunofluorescence (IIF). Results show that the porcine ZP-3α eukaryotic expression vector pVAX1-pZP3α had been constructed correctly, and the chitosan-DNA expressing ZP microencapsulation was prepd. successfully. After 5 days of feeding mouse, the transcription and expression of those DNA vaccines were found in mouse alvine chorion. The prepn. of chitosan/pVAX1-pZP3α plasmid DNA nanoparticles and its expression in mice will help to investigate the feasibility of ZP DNA vaccine to induce oviduct local mucosal immunity against ZP to block the fertilization without causing ovarian dysfunction, which will provide new ideas and ways for research and exploiting more effective, more convenient oral contraceptive vaccines.
- 14Carroll, E. C.; Jin, L.; Mori, A.; Munoz-Wolf, N.; Oleszycka, E.; Moran, H. B. T.; Mansouri, S.; McEntee, C. P.; Lambe, E.; Agger, E. M.; Andersen, P.; Cunningham, C.; Hertzog, P.; Fitzgerald, K. A.; Bowie, A. G.; Lavelle, E. C. The Vaccine Adjuvant Chitosan Promotes Cellular Immunity via DNA Sensor cGAS-STING-Dependent Induction of Type I Interferons. Immunity 2016, 44 (3), 597– 608, DOI: 10.1016/j.immuni.2016.02.00414https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XjsVeht7o%253D&md5=4dbaaba95c8d0222008c9a3804203cdaThe Vaccine Adjuvant Chitosan Promotes Cellular Immunity via DNA Sensor cGAS-STING-Dependent Induction of Type I InterferonsCarroll, Elizabeth. C.; Jin, Lei; Mori, Andres; Munoz-Wolf, Natalia; Oleszycka, Ewa; Moran, Hannah B. T.; Mansouri, Samira; McEntee, Craig P.; Lambe, Eimear; Agger, Else Marie; Andersen, Peter; Cunningham, Colm; Hertzog, Paul; Fitzgerald, Katherine A.; Bowie, Andrew G.; Lavelle, Ed C.Immunity (2016), 44 (3), 597-608CODEN: IUNIEH; ISSN:1074-7613. (Elsevier Inc.)The cationic polysaccharide chitosan is an attractive candidate adjuvant capable of driving potent cell-mediated immunity, but the mechanism by which it acts is not clear. We show that chitosan promotes dendritic cell maturation by inducing type I interferons (IFNs) and enhances antigen-specific T helper 1 (Th1) responses in a type I IFN receptor-dependent manner. The induction of type I IFNs, IFN-stimulated genes and dendritic cell maturation by chitosan required the cytoplasmic DNA sensor cGAS and STING, implicating this pathway in dendritic cell activation. Addnl., this process was dependent on mitochondrial reactive oxygen species and the presence of cytoplasmic DNA. Chitosan-mediated enhancement of antigen specific Th1 and IgG2c responses following vaccination was dependent on both cGAS and STING. These findings demonstrate that a cationic polymer can engage the STING-cGAS pathway to trigger innate and adaptive immune responses.
- 15Grifoni, A.; Weiskopf, D.; Ramirez, S. I.; Mateus, J.; Dan, J. M.; Moderbacher, C. R.; Rawlings, S. A.; Sutherland, A.; Premkumar, L.; Jadi, R. S.; Marrama, D.; de Silva, A. M.; Frazier, A.; Carlin, A. F.; Greenbaum, J. A.; Peters, B.; Krammer, F.; Smith, D. M.; Crotty, S.; Sette, A. Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell 2020, 181 (7), 1489– 1501 e15, DOI: 10.1016/j.cell.2020.05.01515https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVOmu73N&md5=6f89bf52ae2d734af758163a4940b356Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed IndividualsGrifoni, Alba; Weiskopf, Daniela; Ramirez, Sydney I.; Mateus, Jose; Dan, Jennifer M.; Moderbacher, Carolyn Rydyznski; Rawlings, Stephen A.; Sutherland, Aaron; Premkumar, Lakshmanane; Jadi, Ramesh S.; Marrama, Daniel; de Silva, Aravinda M.; Frazier, April; Carlin, Aaron F.; Greenbaum, Jason A.; Peters, Bjoern; Krammer, Florian; Smith, Davey M.; Crotty, Shane; Sette, AlessandroCell (Cambridge, MA, United States) (2020), 181 (7), 1489-1501.e15CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Understanding adaptive immunity to SARS-CoV-2 is important for vaccine development, interpreting coronavirus disease 2019 (COVID-19) pathogenesis, and calibration of pandemic control measures. Using HLA class I and II predicted peptide "megapools," circulating SARS-CoV-2-specific CD8+ and CD4+ T cells were identified in ∼70% and 100% of COVID-19 convalescent patients, resp. CD4+ T cell responses to spike, the main target of most vaccine efforts, were robust and correlated with the magnitude of the anti-SARS-CoV-2 IgG and IgA titers. The M, spike, and N proteins each accounted for 11%-27% of the total CD4+ response, with addnl. responses commonly targeting nsp3, nsp4, ORF3a, and ORF8, among others. For CD8+ T cells, spike and M were recognized, with at least eight SARS-CoV-2 ORFs targeted. Importantly, we detected SARS-CoV-2-reactive CD4+ T cells in ∼40%-60% of unexposed individuals, suggesting cross-reactive T cell recognition between circulating "common cold" coronaviruses and SARS-CoV-2.
- 16Moss, P. The T cell immune response against SARS-CoV-2. Nat. Immunol. 2022, 23 (2), 186– 193, DOI: 10.1038/s41590-021-01122-w16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XisFOmtbg%253D&md5=a87a8b6da6c9977edfdb5950c491470eThe T cell immune response against SARS-CoV-2Moss, PaulNature Immunology (2022), 23 (2), 186-193CODEN: NIAMCZ; ISSN:1529-2908. (Nature Portfolio)The adaptive immune response is a major determinant of the clin. outcome after SARS-CoV-2 infection and underpins vaccine efficacy. T cell responses develop early and correlate with protection but are relatively impaired in severe disease and are assocd. with intense activation and lymphopenia. A subset of T cells primed against seasonal coronaviruses cross reacts with SARS-CoV-2 and may contribute to clin. protection, particularly in early life. T cell memory encompasses broad recognition of viral proteins, estd. at around 30 epitopes within each individual, and seems to be well sustained so far. This breadth of recognition can limit the impact of individual viral mutations and is likely to underpin protection against severe disease from viral variants, including Omicron. Current COVID-19 vaccines elicit robust T cell responses that likely contribute to remarkable protection against hospitalization or death, and novel or heterologous regimens offer the potential to further enhance cellular responses. T cell immunity plays a central role in the control of SARS-CoV-2 and its importance may have been relatively underestimated thus far.
- 17Sheng, T.; Luo, B.; Zhang, W.; Ge, X.; Yu, J.; Zhang, Y.; Gu, Z. Microneedle-Mediated Vaccination: Innovation and Translation. Adv. Drug Deliv. Rev. 2021, 179, 113919 DOI: 10.1016/j.addr.2021.11391917https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvVaisbbO&md5=9aa9c3552eb579bfbb0471663108d957Microneedle-Mediated Vaccination: Innovation and TranslationSheng, Tao; Luo, Bowen; Zhang, Wentao; Ge, Xinyang; Yu, Jicheng; Zhang, Yuqi; Gu, ZhenAdvanced Drug Delivery Reviews (2021), 179 (), 113919CODEN: ADDREP; ISSN:0169-409X. (Elsevier B.V.)A review. Vaccine administration by s.c. or i.m. injection is the most commonly prescribed route for inoculation, however, it is often assocd. with some deficiencies such as low compliance, high professionalism, and risk of infection. Therefore, the application of microneedles for vaccine delivery has gained widespread interests in the past few years due to its high compliance, minimal invasiveness, and convenience. This review focuses on recent advances in the development and application of microneedles for vaccination based on different delivery strategies, and introduces the current status of microneedle-mediated vaccination in clin. translation. The prospects for its application including opportunities and challenges are further discussed.
- 18Korkmaz, E.; Balmert, S. C.; Carey, C. D.; Erdos, G.; Falo, L. D., Jr. Emerging skin-targeted drug delivery strategies to engineer immunity: A focus on infectious diseases. Expert Opin. Drug Deliv. 2021, 18 (2), 151– 167, DOI: 10.1080/17425247.2021.182396418https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvF2ku7%252FJ&md5=fbb20d79eb7860bdd55f522a7d84f43dEmerging skin-targeted drug delivery strategies to engineer immunity: A focus on infectious diseasesKorkmaz, Emrullah; Balmert, Stephen C.; Carey, Cara Donahue; Erdos, Geza; Falo, Louis D. JrExpert Opinion on Drug Delivery (2021), 18 (2), 151-167CODEN: EODDAW; ISSN:1742-5247. (Taylor & Francis Ltd.)A review. Introduction: Infectious pathogens are global disrupters. Progress in biomedical science and technol. has expanded the public health arsenal against infectious diseases. Specifically, vaccination has reduced the burden of infectious pathogens. Engineering systemic immunity by harnessing the cutaneous immune network has been particularly attractive since the skin is an easily accessible immune-responsive organ. Recent advances in skin-targeted drug delivery strategies have enabled safe, patient-friendly, and controlled deployment of vaccines to cutaneous microenvironments for inducing long-lived pathogen-specific immunity to mitigate infectious diseases, including COVID-19. Areas covered: This review briefly discusses the basics of cutaneous immunomodulation and provides a concise overview of emerging skin-targeted drug delivery systems that enable safe, minimally invasive, and effective intracutaneous administration of vaccines for engineering systemic immune responses to combat infectious diseases. Expert opinion: In-situ engineering of the cutaneous microenvironment using emerging skin-targeted vaccine delivery systems offers remarkable potential to develop diverse immunization strategies against pathogens. Mechanistic studies with std. correlates of vaccine efficacy will be important to compare innovative intracutaneous drug delivery strategies to each other and to existing clin. approaches. Cost-benefit analyses will be necessary for developing effective commercialization strategies. Significant involvement of industry and/or government will be imperative for successfully bringing novel skin-targeted vaccine delivery methods to market for their widespread use.
- 19Depelsenaire, A. C. I.; Meliga, S. C.; McNeilly, C. L.; Pearson, F. E.; Coffey, J. W.; Haigh, O. L.; Flaim, C. J.; Frazer, I. H.; Kendall, M. A. F. Colocalization of cell death with antigen deposition in skin enhances vaccine immunogenicity. J. Invest. Dermatol. 2014, 134 (9), 2361– 2370, DOI: 10.1038/jid.2014.17419https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXns1Wru70%253D&md5=49a9dd772c48f77b964d7ff389a99352Colocalization of Cell Death with Antigen Deposition in Skin Enhances Vaccine ImmunogenicityDepelsenaire, Alexandra C. I.; Meliga, Stefano C.; McNeilly, Celia L.; Pearson, Frances E.; Coffey, Jacob W.; Haigh, Oscar L.; Flaim, Christopher J.; Frazer, Ian H.; Kendall, Mark A. F.Journal of Investigative Dermatology (2014), 134 (9), 2361-2370CODEN: JIDEAE; ISSN:0022-202X. (Nature Publishing Group)Vaccines delivered to the skin by microneedles-with and without adjuvants-have increased immunogenicity with lower doses than std. vaccine delivery techniques such as i.m. or intradermal injection. However, the mechanisms underlying this skin-mediated "adjuvant" effect are not clear. Here, we show that the dynamic application of a microprojection array (the Nanopatch) to skin generates localized transient stresses invoking cell death around each projection. Nanopatch application caused significantly higher levels (∼65-fold) of cell death in murine ear skin than i.d. injection using a hypodermic needle. Measured skin cell death is assocd. with modeled stresses ∼1-10 MPa. Nanopatch-immunized groups also yielded consistently higher anti-IgG endpoint titers (up to 50-fold higher) than i.d. groups after delivery of a split virion influenza vaccine. Importantly, colocalization of cell death with nearby live skin cells and delivered antigen was necessary for immunogenicity enhancement. These results suggest a correlation between cell death caused by the Nanopatch with increased immunogenicity. We propose that the localized cell death serves as a "phys. immune enhancer" for the adjacent viable skin cells, which also receive antigen from the projections. This natural immune enhancer effect has the potential to mitigate or replace chem.-based adjuvants in vaccines.
- 20Rock, K. L.; York, I. A.; Goldberg, A. L. Post-proteasomal antigen processing for major histocompatibility complex class I presentation. Nat. Immunol. 2004, 5 (7), 670– 7, DOI: 10.1038/ni108920https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXlt1Wmu70%253D&md5=57db65f9622669eba3ef6a7393288364Post-proteasomal antigen processing for major histocompatibility complex class I presentationRock, Kenneth L.; York, Ian A.; Goldberg, Alfred L.Nature Immunology (2004), 5 (7), 670-677CODEN: NIAMCZ; ISSN:1529-2908. (Nature Publishing Group)A review. Peptides presented by major histocompatibility complex class I mols. are derived mainly from cytosolic oligopeptides generated by proteasomes during the degrdn. of intracellular proteins. Proteasomal cleavages generate the final C terminus of these epitopes. Although proteasomes may produce mature epitopes that are eight to ten residues in length, they more often generate N-extended precursors that are too long to bind to major histocompatibility complex class I mols. Such precursors are trimmed in the cytosol or in the endoplasmic reticulum by aminopeptidases that generate the N terminus of the presented epitope. Peptidases can also destroy epitopes by trimming peptides to below the size needed for presentation. In the cytosol, endopeptidases, esp. thimet oligopeptidase, and aminopeptidases degrade many proteasomal products, thereby limiting the supply of many antigenic peptides. Thus, the extent of antigen presentation depends on the balance between several proteolytic processes that may generate or destroy epitopes.
- 21Mateus, J.; Grifoni, A.; Tarke, A.; Sidney, J.; Ramirez, S. I.; Dan, J. M.; Burger, Z. C.; Rawlings, S. A.; Smith, D. M.; Phillips, E.; Mallal, S.; Lammers, M.; Rubiro, P.; Quiambao, L.; Sutherland, A.; Yu, E. D.; da Silva Antunes, R.; Greenbaum, J.; Frazier, A.; Markmann, A. J.; Premkumar, L.; de Silva, A.; Peters, B.; Crotty, S.; Sette, A.; Weiskopf, D. Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans. Science 2020, 370 (6512), 89– 94, DOI: 10.1126/science.abd387121https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvF2itLnP&md5=0ed58b534636c4e887fe4072a7d53a53Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humansMateus, Jose; Grifoni, Alba; Tarke, Alison; Sidney, John; Ramirez, Sydney I.; Dan, Jennifer M.; Burger, Zoe C.; Rawlings, Stephen A.; Smith, Davey M.; Phillips, Elizabeth; Mallal, Simon; Lammers, Marshall; Rubiro, Paul; Quiambao, Lorenzo; Sutherland, Aaron; Yu, Esther Dawen; da Silva Antunes, Ricardo; Greenbaum, Jason; Frazier, April; Markmann, Alena J.; Premkumar, Lakshmanane; de Silva, Aravinda; Peters, Bjoern; Crotty, Shane; Sette, Alessandro; Weiskopf, DanielaScience (Washington, DC, United States) (2020), 370 (6512), 89-94CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)Many unknowns exist about human immune responses to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. SARS-CoV-2 reactive CD4+ T cells have been reported in unexposed individuals, suggesting preexisting cross-reactive T cell memory in 20 to 50% of people. However, the source of those T cells has been speculative. Using human blood samples derived before the SARS-CoV-2 virus was discovered in 2019, we mapped 142 T cell epitopes across the SARS-CoV-2 genome to facilitate precise interrogation of the SARS-CoV-2 specific CD4+ T cell repertoire. We demonstrate a range of preexisting memory CD4+ T cells that are cross-reactive with comparable affinity to SARS-CoV-2 and the common cold coronaviruses human coronavirus (HCoV)-OC43, HCoV-229E, HCoV-NL63, and HCoV-HKU1. Thus, variegated T cell memory to coronaviruses that cause the common cold may underlie at least some of the extensive heterogeneity obsd. in coronavirus disease 2019 (COVID-19) disease.
- 22Le Bert, N.; Tan, A. T.; Kunasegaran, K.; Tham, C. Y. L.; Hafezi, M.; Chia, A.; Chng, M. H. Y.; Lin, M.; Tan, N.; Linster, M.; Chia, W. N.; Chen, M. I.; Wang, L. F.; Ooi, E. E.; Kalimuddin, S.; Tambyah, P. A.; Low, J. G.; Tan, Y. J.; Bertoletti, A. SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature 2020, 584 (7821), 457– 462, DOI: 10.1038/s41586-020-2550-z22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsF2ntL3J&md5=148bb07cafbec1af0570f97c662d5b0bSARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controlsLe Bert, Nina; Tan, Anthony T.; Kunasegaran, Kamini; Tham, Christine Y. L.; Hafezi, Morteza; Chia, Adeline; Chng, Melissa Hui Yen; Lin, Meiyin; Tan, Nicole; Linster, Martin; Chia, Wan Ni; Chen, Mark I-Cheng; Wang, Lin-Fa; Ooi, Eng Eong; Kalimuddin, Shirin; Tambyah, Paul Anantharajah; Low, Jenny Guek-Hong; Tan, Yee-Joo; Bertoletti, AntonioNature (London, United Kingdom) (2020), 584 (7821), 457-462CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Memory T cells induced by previous pathogens can shape susceptibility to, and the clin. severity of, subsequent infections. Little is known about the presence in humans of pre-existing memory T cells that have the potential to recognize severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We studied T cell responses against the structural (nucleocapsid (N) protein) and non-structural (NSP7 and NSP13 of ORF1) regions of SARS-CoV-2 in individuals convalescing from COVID-19 (n = 36). In all of these individuals, we found CD4 and CD8 T cells that recognized multiple regions of the N protein. Next, we showed that patients (n = 23) who recovered from SARS (the disease assocd. with SARS-CoV infection) possess long-lasting memory T cells that are reactive to the N protein of SARS-CoV 17 yr after the outbreak of SARS in 2003; these T cells displayed robust cross-reactivity to the N protein of SARS-CoV-2. We also detected SARS-CoV-2-specific T cells in individuals with no history of SARS, COVID-19, or contact with individuals who had SARS and(or) COVID-19 (n = 37). SARS-CoV-2-specific T cells in uninfected donors exhibited a different pattern of immunodominance, and frequently targeted NSP7 and NSP13 as well as the N protein. Epitope characterization of NSP7-specific T cells showed the recognition of protein fragments that are conserved among animal betacoronaviruses but have low homol. to common cold human-assocd. coronaviruses. Thus, infection with betacoronaviruses induces multi-specific and long-lasting T cell immunity against the structural N protein. Understanding how pre-existing N- and ORF1-specific T cells that are present in the general population affect the susceptibility to and pathogenesis of SARS-CoV-2 infection is important for the management of the current COVID-19 pandemic.
- 23Selin, L. K.; Brehm, M. A.; Naumov, Y. N.; Cornberg, M.; Kim, S. K.; Clute, S. C.; Welsh, R. M. Memory of mice and men: CD8+ T-cell cross-reactivity and heterologous immunity. Immunol. Rev. 2006, 211 (1), 164– 81, DOI: 10.1111/j.0105-2896.2006.00394.x23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xot1GrsLY%253D&md5=3bba66cd073e74e993f710ae2716e279Memory of mice and men: CD8+ T-cell cross-reactivity and heterologous immunitySelin, Liisa K.; Brehm, Michael A.; Naumov, Yuri N.; Cornberg, Markus; Kim, Sung-Kwon; Clute, Shalyn C.; Welsh, Raymond M.Immunological Reviews (2006), 211 (), 164-181CODEN: IMRED2; ISSN:0105-2896. (Blackwell Publishing Ltd.)A review. The main functions of memory T cells are to provide protection upon re-exposure to a pathogen and to prevent the re-emergence of low-grade persistent pathogens. Memory T cells achieve these functions through their high frequency and elevated activation state, which lead to rapid responses upon antigenic challenge. The significance and characteristics of memory CD8+ T cells in viral infections have been studied extensively. In many of these studies of T-cell memory, exptl. viral immunologists go to great lengths to assure that their animal colonies are free of endogenous pathogens in order to design reproducible expts. These exptl. results are then thought to provide the basis for our understanding of human immune responses to viruses. Although these findings can be enlightening, humans are not immunol. naive, and they often have memory T-cell populations that can cross-react with and respond to a new infectious agent or cross-react with allo-antigens and influence the success of tissue transplantation. These cross-reactive T cells can become activated and modulate the immune response and outcome of subsequent heterologous infections, a phenomenon we have termed heterologous immunity. These large memory populations are also accommodated into a finite immune system, requiring that the host makes room for each new population of memory cell. It appears that memory cells are part of a continually evolving interactive network, where with each new infection there is an alteration in the frequencies, distributions, and activities of memory cells generated in response to previous infections and allo-antigens.
- 24Riou, C.; Keeton, R.; Moyo-Gwete, T.; Hermanus, T.; Kgagudi, P.; Baguma, R.; Valley-Omar, Z.; Smith, M.; Tegally, H.; Doolabh, D.; Iranzadeh, A.; Tyers, L.; Mutavhatsindi, H.; Tincho, M. B.; Benede, N.; Marais, G.; Chinhoyi, L. R.; Mennen, M.; Skelem, S.; du Bruyn, E.; Stek, C.; de Oliveira, T.; Williamson, C.; Moore, P. L.; Wilkinson, R. J.; Ntusi, N. A. B.; Burgers, W. A. Escape from recognition of SARS-CoV-2 variant spike epitopes but overall preservation of T cell immunity. Sci. Transl. Med. 2022, 14 (631), eabj6824 DOI: 10.1126/scitranslmed.abj6824There is no corresponding record for this reference.
- 25Choi, S. J.; Kim, D. U.; Noh, J. Y.; Kim, S.; Park, S. H.; Jeong, H. W.; Shin, E. C. T cell epitopes in SARS-CoV-2 proteins are substantially conserved in the Omicron variant. Cell. Mol. Immunol. 2022, 19 (3), 447– 448, DOI: 10.1038/s41423-022-00838-525https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhtlOks7k%253D&md5=60fb3bbaa3d6bb3d59da41231c064968T cell epitopes in SARS-CoV-2 proteins are substantially conserved in the Omicron variantChoi, Seong Jin; Kim, Dong-Uk; Noh, Ji Yun; Kim, Sangwoo; Park, Su-Hyung; Jeong, Hye Won; Shin, Eui-CheolCellular & Molecular Immunology (2022), 19 (3), 447-448CODEN: CMIEAO; ISSN:1672-7681. (Nature Portfolio)Our current anal. demonstrates that T cell epitopes are considerably conserved in the Omicron variant and that substantial proportions of memory T cells elicited by COVID-19 vaccination or natural infection respond to the Omicron spike. These results indicate that memory T cells may provide protective immunity during reinfection or breakthrough infection with the Omicron variant.
- 26Naranbhai, V.; Nathan, A.; Kaseke, C.; Berrios, C.; Khatri, A.; Choi, S.; Getz, M. A.; Tano-Menka, R.; Ofoman, O.; Gayton, A.; Senjobe, F.; Zhao, Z.; St Denis, K. J.; Lam, E. C.; Carrington, M.; Garcia-Beltran, W. F.; Balazs, A. B.; Walker, B. D.; Iafrate, A. J.; Gaiha, G. D. T cell reactivity to the SARS-CoV-2 Omicron variant is preserved in most but not all individuals. Cell 2022, 185 (6), 1041– 1051 e6, DOI: 10.1016/j.cell.2022.01.02926https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XkslKhsLw%253D&md5=c98ae864ccadb9b24ccbfe692c7d9b94T cell reactivity to the SARS-CoV-2 Omicron variant is preserved in most but not all individualsNaranbhai, Vivek; Nathan, Anusha; Kaseke, Clarety; Berrios, Cristhian; Khatri, Ashok; Choi, Shawn; Getz, Matthew A.; Tano-Menka, Rhoda; Ofoman, Onosereme; Gayton, Alton; Senjobe, Fernando; Zhao, Zezhou; St Denis, Kerri J.; Lam, Evan C.; Carrington, Mary; Garcia-Beltran, Wilfredo F.; Balazs, Alejandro B.; Walker, Bruce D.; Iafrate, A. John; Gaiha, Gaurav D.Cell (Cambridge, MA, United States) (2022), 185 (6), 1041-1051.e6CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The SARS-CoV-2 Omicron variant (B.1.1.529) contains mutations that mediate escape from antibody responses, although the extent to which these substitutions in spike and non-spike proteins affect T cell recognition is unknown. T cell responses in individuals with prior infection, vaccination, both prior infection and vaccination, and boosted vaccination are largely preserved to Omicron spike and non-spike proteins. However, we also identify a subset of individuals (∼21%) with a >50% redn. in T cell reactivity to the Omicron spike. Evaluation of functional CD4+ and CD8+ memory T cell responses confirmed these findings and revealed that reduced recognition to Omicron spike is primarily obsd. within the CD8+ T cell compartment potentially due to escape from HLA binding. Booster vaccination enhanced T cell responses to Omicron spike. In contrast to neutralizing immunity, these findings suggest preservation of T cell responses to the Omicron variant, although with reduced reactivity in some individuals.
- 27Fan, F.; Zhang, X.; Zhang, Z.; Ding, Y.; Wang, L.; Xu, X.; Pan, Y.; Gong, F. Y.; Jiang, L.; Kang, L.; Ha, Z.; Lu, H.; Hou, J.; Kou, Z.; Zhao, G.; Wang, B.; Gao, X. M. Potent immunogenicity and broad-spectrum protection potential of microneedle array patch-based COVID-19 DNA vaccine candidates encoding dimeric RBD chimera of SARS-CoV and SARS-CoV-2 variants. Emerg. Microbes Infect. 2023, 12 (1), 2202269 DOI: 10.1080/22221751.2023.220226927https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXpt1antbk%253D&md5=46b85f5c131f557284fac7175d7cc4abPotent immunogenicity and broad-spectrum protection potential of microneedle array patch-based COVID-19 DNA vaccine candidates encoding dimeric RBD chimera of SARS-CoV and SARS-CoV-2 variantsFan, Feng; Zhang, Xin; Zhang, Zhiyu; Ding, Yuan; Wang, Limei; Xu, Xin; Pan, Yaying; Gong, Fang-Yuan; Jiang, Lin; Kang, Lingyu; Ha, Zhuo; Lu, Huijun; Hou, Jiawang; Kou, Zhihua; Zhao, Gan; Wang, Bin; Gao, Xiao-MingEmerging Microbes & Infections (2023), 12 (1), 2202269/1-2202269/16CODEN: EMIMC4; ISSN:2222-1751. (Taylor & Francis Ltd.)Breakthrough infections by SARS-CoV-2 variants pose a global challenge to COVID-19 pandemic control, and the development of more effective vaccines of broad-spectrum protection is needed. In this study, we constructed pVAX1-based plasmids encoding receptor-binding domain (RBD) chimera of SARS-CoV-1 and SARS-CoV-2 variants, including pAD1002 (encoding RBDSARS/BA1), pAD1003 (encoding RBDSARS/Beta) and pAD131 (encoding RBDBA1/Beta). Plasmids pAD1002 and pAD131 were far more immunogenic than pAD1003 in terms of eliciting RBD-specific IgG when i.m. administered without electroporation. Furthermore, dissolvable microneedle array patches (MAP) greatly enhanced the immunogenicity of these DNA constructs in mice and rabbits. MAP laden with pAD1002 (MAP-1002) significantly outperformed inactivated SARS-CoV-2 virus vaccine in inducing RBD-specific IFN-γ+ effector and memory T cells, and generated T lymphocytes of different homing patterns compared to that induced by electroporated DNA in mice. In consistence with the high titer neutralization results of MAP-1002 antisera against SARS-CoV-2 pseudoviruses, MAP-1002 protected human ACE2-transgenic mice from Omicron BA.1 challenge. Collectively, MAP-based DNA constructs encoding chimeric RBDs of SARS-CoV-1 and SARS-CoV-2 variants, as represented by MAP-1002, are potential COVID-19 vaccine candidates worthy further translational study.
- 28Castro Dopico, X.; Ols, S.; Lore, K.; Karlsson Hedestam, G. B. Immunity to SARS-CoV-2 induced by infection or vaccination. J. Intern. Med. 2022, 291 (1), 32– 50, DOI: 10.1111/joim.1337228https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhslKgur7K&md5=733c3e5af1496df634afa755928c41d3Immunity to SARS-CoV-2 induced by infection or vaccinationCastro Dopico, Xaquin; Ols, Sebastian; Lore, Karin; Karlsson Hedestam, Gunilla B.Journal of Internal Medicine (2022), 291 (1), 32-50CODEN: JINMEO; ISSN:0954-6820. (Wiley-Blackwell)A review. Adaptive immune responses play crit. roles in viral clearance and protection against re-infection, and SARS-CoV-2 is no exception. What is exceptional, is the rapid characterization of the immune response to the virus performed by researchers during the first 20 mo of the pandemic. This has given us a more detailed understanding about SARS-CoV-2 than we have about many viruses that have been with us for a long time. Furthermore, effective COVID-19 vaccines were developed in record time, and their rollout worldwide is already making a significant difference, although major challenges remain in terms of equal access. The pandemic has engaged scientists and the public alike, and terms such as seroprevalence, neutralizing antibodies, antibody escape and vaccine certificates have become familiar to a broad community. Here, we review key findings concerning B cell and antibody (Ab) responses to SARS-CoV-2, focusing on non-severe cases and anti-spike (S) Ab responses in particular, the latter being central to protective immunity induced by infection or vaccination. The emergence of viral variants that have acquired mutations in S acutely highlights the need for continued characterization of both emerging variants and Ab responses against these during the evolving pathogen-immune system arms race.
- 29Dutta, N. K.; Mazumdar, K.; Gordy, J. T. The Nucleocapsid Protein of SARS-CoV-2: a Target for Vaccine Development. J. Virol. 2020, 94 (13), 647-20, DOI: 10.1128/JVI.00647-20There is no corresponding record for this reference.
- 30Ni, L.; Ye, F.; Cheng, M. L.; Feng, Y.; Deng, Y. Q.; Zhao, H.; Wei, P.; Ge, J.; Gou, M.; Li, X.; Sun, L.; Cao, T.; Wang, P.; Zhou, C.; Zhang, R.; Liang, P.; Guo, H.; Wang, X.; Qin, C. F.; Chen, F.; Dong, C. Detection of SARS-CoV-2-Specific Humoral and Cellular Immunity in COVID-19 Convalescent Individuals. Immunity 2020, 52 (6), 971– 977 e3, DOI: 10.1016/j.immuni.2020.04.02330https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXptlCjtb4%253D&md5=ad2de5c1a3bfd874429e20474c6dce86Detection of SARS-CoV-2-Specific Humoral and Cellular Immunity in COVID-19 Convalescent IndividualsNi, Ling; Ye, Fang; Cheng, Meng-Li; Feng, Yu; Deng, Yong-Qiang; Zhao, Hui; Wei, Peng; Ge, Jiwan; Gou, Mengting; Li, Xiaoli; Sun, Lin; Cao, Tianshu; Wang, Pengzhi; Zhou, Chao; Zhang, Rongrong; Liang, Peng; Guo, Han; Wang, Xinquan; Qin, Cheng-Feng; Chen, Fang; Dong, ChenImmunity (2020), 52 (6), 971-977.e3CODEN: IUNIEH; ISSN:1074-7613. (Elsevier Inc.)The World Health Organization has declared SARS-CoV-2 virus outbreak a worldwide pandemic. However, there is very limited understanding on the immune responses, esp. adaptive immune responses to SARS-CoV-2 infection. Here, we collected blood from COVID-19 patients who have recently become virus-free, and therefore were discharged, and detected SARS-CoV-2-specific humoral and cellular immunity in eight newly discharged patients. Follow-up anal. on another cohort of six patients 2 wk post discharge also revealed high titers of IgG antibodies. In all 14 patients tested, 13 displayed serum-neutralizing activities in a pseudotype entry assay. Notably, there was a strong correlation between neutralization antibody titers and the nos. of virus-specific T cells. Our work provides a basis for further anal. of protective immunity to SARS-CoV-2, and understanding the pathogenesis of COVID-19, esp. in the severe cases. It also has implications in developing an effective vaccine to SARS-CoV-2 infection.
- 31Peng, Y.; Mentzer, A. J.; Liu, G.; Yao, X.; Yin, Z.; Dong, D.; Dejnirattisai, W.; Rostron, T.; Supasa, P.; Liu, C.; Lopez-Camacho, C.; Slon-Campos, J.; Zhao, Y.; Stuart, D. I.; Paesen, G. C.; Grimes, J. M.; Antson, A. A.; Bayfield, O. W.; Hawkins, D.; Ker, D. S.; Wang, B.; Turtle, L.; Subramaniam, K.; Thomson, P.; Zhang, P.; Dold, C.; Ratcliff, J.; Simmonds, P.; de Silva, T.; Sopp, P.; Wellington, D.; Rajapaksa, U.; Chen, Y. L.; Salio, M.; Napolitani, G.; Paes, W.; Borrow, P.; Kessler, B. M.; Fry, J. W.; Schwabe, N. F.; Semple, M. G.; Baillie, J. K.; Moore, S. C.; Openshaw, P. J. M.; Ansari, M. A.; Dunachie, S.; Barnes, E.; Frater, J.; Kerr, G.; Goulder, P.; Lockett, T.; Levin, R.; Zhang, Y.; Jing, R.; Ho, L. P.; Oxford Immunology Network Covid-19 Response, T. c. C.; Investigators, I. C.; Cornall, R. J.; Conlon, C. P.; Klenerman, P.; Screaton, G. R.; Mongkolsapaya, J.; McMichael, A.; Knight, J. C.; Ogg, G.; Dong, T. Broad and strong memory CD4(+) and CD8(+) T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19. Nat. Immunol. 2020, 21 (11), 1336– 1345, DOI: 10.1038/s41590-020-0782-631https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhslCksbrF&md5=89639b3bc6b4cb0abd7bd1302973da74Broad and strong memory CD4+ and CD8+ T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19Peng, Yanchun; Mentzer, Alexander J.; Liu, Guihai; Yao, Xuan; Yin, Zixi; Dong, Danning; Dejnirattisai, Wanwisa; Rostron, Timothy; Supasa, Piyada; Liu, Chang; Lopez-Camacho, Cesar; Slon-Campos, Jose; Zhao, Yuguang; Stuart, David I.; Paesen, Guido C.; Grimes, Jonathan M.; Antson, Alfred A.; Bayfield, Oliver W.; Hawkins, Dorothy E. D. P.; Ker, De-Sheng; Wang, Beibei; Turtle, Lance; Subramaniam, Krishanthi; Thomson, Paul; Zhang, Ping; Dold, Christina; Ratcliff, Jeremy; Simmonds, Peter; de Silva, Thushan; Sopp, Paul; Wellington, Dannielle; Rajapaksa, Ushani; Chen, Yi-Ling; Salio, Mariolina; Napolitani, Giorgio; Paes, Wayne; Borrow, Persephone; Kessler, Benedikt M.; Fry, Jeremy W.; Schwabe, Nikolai F.; Semple, Malcolm G.; Baillie, J. Kenneth; Moore, Shona C.; Openshaw, Peter J. M.; Ansari, M. Azim; Dunachie, Susanna; Barnes, Eleanor; Frater, John; Kerr, Georgina; Goulder, Philip; Lockett, Teresa; Levin, Robert; Zhang, Yonghong; Jing, Ronghua; Ho, Ling-Pei; Cornall, Richard J.; Conlon, Christopher P.; Klenerman, Paul; Screaton, Gavin R.; Mongkolsapaya, Juthathip; McMichael, Andrew; Knight, Julian C.; Ogg, Graham; Dong, TaoNature Immunology (2020), 21 (11), 1336-1345CODEN: NIAMCZ; ISSN:1529-2908. (Nature Research)Abstr.: The development of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines and therapeutics will depend on understanding viral immunity. We studied T cell memory in 42 patients following recovery from COVID-19 (28 with mild disease and 14 with severe disease) and 16 unexposed donors, using interferon-γ-based assays with peptides spanning SARS-CoV-2 except ORF1. The breadth and magnitude of T cell responses were significantly higher in severe as compared with mild cases. Total and spike-specific T cell responses correlated with spike-specific antibody responses. We identified 41 peptides contg. CD4+ and/or CD8+ epitopes, including six immunodominant regions. Six optimized CD8+ epitopes were defined, with peptide-MHC pentamer-pos. cells displaying the central and effector memory phenotype. In mild cases, higher proportions of SARS-CoV-2-specific CD8+ T cells were obsd. The identification of T cell responses assocd. with milder disease will support an understanding of protective immunity and highlights the potential of including non-spike proteins within future COVID-19 vaccine design.
- 32Harris, P. E.; Brasel, T.; Massey, C.; Herst, C. V.; Burkholz, S.; Lloyd, P.; Blankenberg, T.; Bey, T. M.; Carback, R.; Hodge, T.; Ciotlos, S.; Wang, L.; Comer, J. E.; Rubsamen, R. M. A Synthetic Peptide CTL Vaccine Targeting Nucleocapsid Confers Protection from SARS-CoV-2 Challenge in Rhesus Macaques. Vaccines (Basel) 2021, 9 (5), 520, DOI: 10.3390/vaccines905052032https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitV2rs7rF&md5=2ae3877233432bba92c6ca98d60bef09A synthetic peptide CTL vaccine targeting nucleocapsid confers protection from SARS-CoV-2 challenge in rhesus macaquesHarris, Paul E.; Brasel, Trevor; Massey, Christopher; Herst, C. V.; Burkholz, Scott; Lloyd, Peter; Blankenberg, Tikoes; Bey, Thomas M.; Carback, Richard; Hodge, Thomas; Ciotlos, Serban; Wang, Lu; Comer, Jason E.; Rubsamen, Reid M.Vaccines (Basel, Switzerland) (2021), 9 (5), 520CODEN: VBSABP; ISSN:2076-393X. (MDPI AG)Background: Persistent transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has given rise to a COVID-19 pandemic. Several vaccines, conceived in 2020, that evoke protective spike antibody responses are being deployed in mass public health vaccination programs. Recent data suggests, however, that as sequence variation in the spike genome accumulates, some vaccines may lose efficacy. Methods: Using a macaque model of SARS-CoV-2 infection, we tested the efficacy of a peptide-based vaccine targeting MHC class I epitopes on the SARS-CoV-2 nucleocapsid protein. We administered biodegradable microspheres with synthetic peptides and adjuvants to rhesus macaques. Unvaccinated control and vaccinated macaques were challenged with 1 x 108 TCID50 units of SARS-CoV-2, followed by assessment of clin. symptoms and viral load, chest radiographs, and sampling of peripheral blood and bronchoalveolar lavage (BAL) fluid for downstream anal. Results: Vaccinated animals were free of pneumonia-like infiltrates characteristic of SARS-CoV-2 infection and presented with lower viral loads relative to controls. Gene expression in cells collected from BAL samples of vaccinated macaques revealed a unique signature assocd. with enhanced development of adaptive immune responses relative to control macaques. Conclusions: We demonstrate that a room temp. stable peptide vaccine based on known immunogenic HLA class I bound CTL epitopes from the nucleocapsid protein can provide protection against SARS-CoV-2 infection in nonhuman primates.
- 33Matchett, W. E.; Joag, V.; Stolley, J. M.; Shepherd, F. K.; Quarnstrom, C. F.; Mickelson, C. K.; Wijeyesinghe, S.; Soerens, A. G.; Becker, S.; Thiede, J. M.; Weyu, E.; O’Flanagan, S. D.; Walter, J. A.; Vu, M. N.; Menachery, V. D.; Bold, T. D.; Vezys, V.; Jenkins, M. K.; Langlois, R. A.; Masopust, D. Cutting Edge: Nucleocapsid Vaccine Elicits Spike-Independent SARS-CoV-2 Protective Immunity. J. Immunol. 2021, 207 (2), 376– 379, DOI: 10.4049/jimmunol.210042133https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFOiur%252FL&md5=b3347d5ee3b8bc3d824c7bfb0ddf1114Cutting edge: nucleocapsid vaccine elicits spike-independent SARS-CoV-2 protective immunityMatchett, William E.; Joag, Vineet; Stolley, J. Michael; Shepherd, Frances K.; Quarnstrom, Clare F.; Mickelson, Clayton K.; Wijeyesinghe, Sathi; Soerens, Andrew G.; Becker, Samuel; Thiede, Joshua M.; Weyu, Eyob; O'Flanagan, Stephen D.; Walter, Jennifer A.; Vu, Michelle N.; Menachery, Vineet D.; Bold, Tyler D.; Vezys, Vaiva; Jenkins, Marc K.; Langlois, Ryan A.; Masopust, DavidJournal of Immunology (2021), 207 (2), 376-379CODEN: JOIMA3; ISSN:0022-1767. (American Association of Immunologists)Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the COVID-19 pandemic. Neutralizing Abs target the receptor binding domain of the spike (S) protein, a focus of successful vaccine efforts. Concerns have arisen that S-specific vaccine immunity may fail to neutralize emerging variants. We show that vaccination with a human adenovirus type 5 vector expressing the SARS-CoV-2 nucleocapsid (N) protein can establish protective immunity, defined by reduced wt. loss and viral load, in both Syrian hamsters and K18-hACE2 mice. Challenge of vaccinated mice was assocd. with rapid N-specific T cell recall responses in the respiratory mucosa. This study supports the rationale for including addnl. viral Ags in SARS-CoV-2 vaccines, even if they are not a target of neutralizing Abs, to broaden epitope coverage and immune effector mechanisms.
- 34Chiuppesi, F.; Nguyen, V. H.; Park, Y.; Contreras, H.; Karpinski, V.; Faircloth, K.; Nguyen, J.; Kha, M.; Johnson, D.; Martinez, J.; Iniguez, A.; Zhou, Q.; Kaltcheva, T.; Frankel, P.; Kar, S.; Sharma, A.; Andersen, H.; Lewis, M. G.; Shostak, Y.; Wussow, F.; Diamond, D. J. Synthetic multiantigen MVA vaccine COH04S1 protects against SARS-CoV-2 in Syrian hamsters and non-human primates. NPJ Vaccines 2022, 7 (1), 7, DOI: 10.1038/s41541-022-00436-634https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhvFGjt70%253D&md5=50f41db00099e194241d3b0fef03a451Synthetic multiantigen MVA vaccine COH04S1 protects against SARS-CoV-2 in Syrian hamsters and non-human primatesChiuppesi, Flavia; Nguyen, Vu H.; Park, Yoonsuh; Contreras, Heidi; Karpinski, Veronica; Faircloth, Katelyn; Nguyen, Jenny; Kha, Mindy; Johnson, Daisy; Martinez, Joy; Iniguez, Angelina; Zhou, Qiao; Kaltcheva, Teodora; Frankel, Paul; Kar, Swagata; Sharma, Ankur; Andersen, Hanne; Lewis, Mark G.; Shostak, Yuriy; Wussow, Felix; Diamond, Don J.npj Vaccines (2022), 7 (1), 7CODEN: VACCBC; ISSN:2059-0105. (Nature Portfolio)Second-generation COVID-19 vaccines could contribute to establish protective immunity against SARS-CoV-2 and its emerging variants. We developed COH04S1, a synthetic multiantigen modified vaccinia Ankara-based SARS-CoV-2 vaccine that co-expresses spike and nucleocapsid antigens. Here, we report COH04S1 vaccine efficacy in animal models. We demonstrate that i.m. or intranasal vaccination of Syrian hamsters with COH04S1 induces robust Th1-biased antigen-specific humoral immunity and cross-neutralizing antibodies (NAb) and protects against wt. loss, lower respiratory tract infection, and lung injury following intranasal SARS-CoV-2 challenge. Moreover, we demonstrate that single-dose or two-dose vaccination of non-human primates with COH04S1 induces robust antigen-specific binding antibodies, NAb, and Th1-biased T cells, protects against both upper and lower respiratory tract infection following intranasal/intratracheal SARS-CoV-2 challenge, and triggers potent post-challenge anamnestic antiviral responses. These results demonstrate COH04S1-mediated vaccine protection in animal models through different vaccination routes and dose regimens, complementing ongoing investigation of this multiantigen SARS-CoV-2 vaccine in clin. trials.
- 35Afkhami, S.; D’Agostino, M. R.; Zhang, A.; Stacey, H. D.; Marzok, A.; Kang, A.; Singh, R.; Bavananthasivam, J.; Ye, G.; Luo, X.; Wang, F.; Ang, J. C.; Zganiacz, A.; Sankar, U.; Kazhdan, N.; Koenig, J. F. E.; Phelps, A.; Gameiro, S. F.; Tang, S.; Jordana, M.; Wan, Y.; Mossman, K. L.; Jeyanathan, M.; Gillgrass, A.; Medina, M. F. C.; Smaill, F.; Lichty, B. D.; Miller, M. S.; Xing, Z. Respiratory mucosal delivery of next-generation COVID-19 vaccine provides robust protection against both ancestral and variant strains of SARS-CoV-2. Cell 2022, 185 (5), 896– 915 e19, DOI: 10.1016/j.cell.2022.02.00535https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XktVGrsLg%253D&md5=0484708611d00713d5cea5f903809600Respiratory mucosal delivery of next-generation COVID-19 vaccine provides robust protection against both ancestral and variant strains of SARS-CoV-2Afkhami, Sam; D'Agostino, Michael R.; Zhang, Ali; Stacey, Hannah D.; Marzok, Art; Kang, Alisha; Singh, Ramandeep; Bavananthasivam, Jegarubee; Ye, Gluke; Luo, Xiangqian; Wang, Fuan; Ang, Jann C.; Zganiacz, Anna; Sankar, Uma; Kazhdan, Natallia; Koenig, Joshua F. E.; Phelps, Allyssa; Gameiro, Steven F.; Tang, Shangguo; Jordana, Manel; Wan, Yonghong; Mossman, Karen L.; Jeyanathan, Mangalakumari; Gillgrass, Amy; Medina, Maria Fe C.; Smaill, Fiona; Lichty, Brian D.; Miller, Matthew S.; Xing, ZhouCell (Cambridge, MA, United States) (2022), 185 (5), 896-915.e19CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The emerging SARS-CoV-2 variants of concern (VOCs) threaten the effectiveness of current COVID-19 vaccines administered i.m. and designed to only target the spike protein. There is a pressing need to develop next-generation vaccine strategies for broader and long-lasting protection. Using adenoviral vectors (Ad) of human and chimpanzee origin, we evaluated Ad-vectored trivalent COVID-19 vaccines expressing spike-1, nucleocapsid, and RdRp antigens in murine models. Single-dose intranasal immunization, particularly with chimpanzee Ad-vectored vaccine, is superior to i.m. immunization in induction of the tripartite protective immunity consisting of local and systemic antibody responses, mucosal tissue-resident memory T cells and mucosal trained innate immunity. We further show that intranasal immunization provides protection against both the ancestral SARS-CoV-2 and two VOC, B.1.1.7 and B.1.351. Our findings indicate that respiratory mucosal delivery of Ad-vectored multivalent vaccine represents an effective next-generation COVID-19 vaccine strategy to induce all-around mucosal immunity against current and future VOC.
- 36Liu, L.; Zhong, Q.; Tian, T.; Dubin, K.; Athale, S. K.; Kupper, T. S. Epidermal injury and infection during poxvirus immunization is crucial for the generation of highly protective T cell-mediated immunity. Nat. Med. 2010, 16 (2), 224– 7, DOI: 10.1038/nm.207836https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXntVGjsA%253D%253D&md5=59664e863ab882eb1803281b6d0503c4Epidermal injury and infection during poxvirus immunization is crucial for the generation of highly protective T cell-mediated immunityLiu, Luzheng; Zhong, Qiong; Tian, Tian; Dubin, Krista; Athale, Shruti K.; Kupper, Thomas S.Nature Medicine (New York, NY, United States) (2010), 16 (2), 224-227CODEN: NAMEFI; ISSN:1078-8956. (Nature Publishing Group)Variola major (smallpox) infection claimed hundreds of millions lives before it was eradicated by a simple vaccination strategy: epicutaneous application of the related orthopoxvirus vaccinia virus (VACV) to superficially injured skin (skin scarification, s.s.). However, the remarkable success of this strategy was attributed to the immunogenicity of VACV rather than to the unique mode of vaccine delivery. The authors now show that VACV immunization via s.s., but not conventional injection routes, is essential for the generation of superior T cell-mediated immune responses that provide complete protection against subsequent challenges, independent of neutralizing antibodies. Skin-resident effector memory T cells (TEM cells) provide complete protection against cutaneous challenge, whereas protection against lethal respiratory challenge requires both respiratory mucosal TEM cells and central memory T cells (TCM cells). Vaccination with recombinant VACV (rVACV) expressing a tumor antigen was protective against tumor challenge only if delivered via the s.s. route; it was ineffective if delivered by hypodermic injection. The clin. safer nonreplicative modified vaccinia Ankara virus (MVA) also generated far superior protective immunity when delivered via the s.s. route compared to i.m. injection as used in MVA clin. trials. Thus, delivery of rVACV-based vaccines, including MVA vaccines, through phys. disrupted epidermis has clear-cut advantages over conventional vaccination via hypodermic injection.
- 37Mikhak, Z.; Strassner, J. P.; Luster, A. D. Lung dendritic cells imprint T cell lung homing and promote lung immunity through the chemokine receptor CCR4. J. Exp. Med. 2013, 210 (9), 1855– 69, DOI: 10.1084/jem.2013009137https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVSgsbjF&md5=59b555d9feb77183237226eac2caba00Lung dendritic cells imprint T cell lung homing and promote lung immunity through the chemokine receptor CCR4Mikhak, Zamaneh; Strassner, James P.; Luster, Andrew D.Journal of Experimental Medicine (2013), 210 (9), 1855-1869CODEN: JEMEAV; ISSN:0022-1007. (Rockefeller University Press)T cell trafficking into the lung is crit. for lung immunity, but the mechanisms that mediate T cell lung homing are not well understood. Here, we show that lung dendritic cells (DCs) imprint T cell lung homing, as lung DC-activated T cells traffic more efficiently into the lung in response to inhaled antigen and at homeostasis compared with T cells activated by DCs from other tissues. Consequently, lung DC-imprinted T cells protect against influenza more effectively than do gut and skin DC-imprinted T cells. Lung DCs imprint the expression of CCR4 on T cells, and CCR4 contributes to T cell lung imprinting. Lung DC-activated, CCR4-deficient T cells fail to traffic into the lung as efficiently and to protect against influenza as effectively as lung DC-activated, CCR4-sufficient T cells. Thus, lung DCs imprint T cell lung homing and promote lung immunity in part through CCR4.
- 38Dijkman, K.; Aguilo, N.; Boot, C.; Hofman, S. O.; Sombroek, C. C.; Vervenne, R. A. W.; Kocken, C. H. M.; Marinova, D.; Thole, J.; Rodriguez, E.; Vierboom, M. P. M.; Haanstra, K. G.; Puentes, E.; Martin, C.; Verreck, F. A. W. Pulmonary MTBVAC vaccination induces immune signatures previously correlated with prevention of tuberculosis infection. Cell Rep. Med. 2021, 2 (1), 100187 DOI: 10.1016/j.xcrm.2020.10018738https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhvFWlu7fE&md5=fd1734096801c6284856455ba6c42a86Pulmonary MTBVAC vaccination induces immune signatures previously correlated with prevention of tuberculosis infectionDijkman, Karin; Aguilo, Nacho; Boot, Charelle; Hofman, Sam O.; Sombroek, Claudia C.; Vervenne, Richard A. W.; Kocken, Clemens H. M.; Marinova, Dessislava; Thole, Jelle; Rodriguez, Esteban; Vierboom, Michel P. M.; Haanstra, Krista G.; Puentes, Eugenia; Martin, Carlos; Verreck, Frank A. W.Cell Reports Medicine (2021), 2 (1), 100187CODEN: CRMEDE; ISSN:2666-3791. (Elsevier Inc.)To fight tuberculosis, better vaccination strategies are needed. Live attenuated Mycobacterium tuberculosis-derived vaccine, MTBVAC, is a promising candidate in the pipeline, proven to be safe and immunogenic in humans so far. Independent studies have shown that pulmonary mucosal delivery of Bacillus Calmette-Guerin (BCG), the only tuberculosis (TB) vaccine available today, confers superior protection over std. intradermal immunization. Here we demonstrate that mucosal MTBVAC is well tolerated, eliciting polyfunctional T helper type 17 cells, interleukin-10, and Igs in the airway and yielding a broader antigenic profile than BCG in rhesus macaques. Beyond our previous work, we show that local Igs, induced by MTBVAC and BCG, bind to M. tuberculosis and enhance pathogen uptake. Furthermore, after pulmonary vaccination, but not M. tuberculosis infection, local T cells expressed high levels of mucosal homing and tissue residency markers. Our data show that pulmonary MTBVAC administration has the potential to enhance its efficacy and justifies further exploration of mucosal vaccination strategies in preclin. efficacy studies.
- 39Pan, Y.; Liu, L.; Tian, T.; Zhao, J.; Park, C. O.; Lofftus, S. Y.; Stingley, C. A.; Yan, Y.; Mei, S.; Liu, X.; Kupper, T. S. Epicutaneous immunization with modified vaccinia Ankara viral vectors generates superior T cell immunity against a respiratory viral challenge. NPJ Vaccines 2021, 6 (1), 1, DOI: 10.1038/s41541-020-00265-539https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXht1ejsbw%253D&md5=0b0b65986a334c7270199712cd484e48Epicutaneous immunization with modified vaccinia Ankara viral vectors generates superior T cell immunity against a respiratory viral challengePan, Youdong; Liu, Luzheng; Tian, Tian; Zhao, Jingxia; Park, Chang Ook; Lofftus, Serena Y.; Stingley, Claire A.; Yan, Yu; Mei, Shenglin; Liu, Xing; Kupper, Thomas S.npj Vaccines (2021), 6 (1), 1CODEN: VACCBC; ISSN:2059-0105. (Nature Research)Abstr.: Modified Vaccinia Ankara (MVA) was recently approved as a smallpox vaccine. Variola is transmitted by respiratory droplets and MVA immunization by skin scarification (s.s.) protected mice far more effectively against lethal respiratory challenge with vaccinia virus (VACV) than any other route of delivery, and at lower doses. Comparisons of s.s. with intradermal, s.c., or i.m. routes showed that MVAOVA s.s.-generated T cells were both more abundant and transcriptionally unique. MVAOVA s.s. produced greater nos. of lung Ova-specific CD8+ TRM and was superior in protecting mice against lethal VACVOVA respiratory challenge. Nearly as many lung TRM were generated with MVAOVA s.s. immunization compared to intra-tracheal immunization with MVAOVA and both routes vaccination protected mice against lethal pulmonary challenge with VACVOVA. Strikingly, MVAOVA s.s.-generated effector T cells exhibited overlapping gene transcriptional profiles to those generated via intra-tracheal immunization. Overall, our data suggest that heterologous MVA vectors immunized via s.s. are uniquely well-suited as vaccine vectors for respiratory pathogens, which may be relevant to COVID-19. In addn., MVA delivered via s.s. could represent a more ED-sparing smallpox vaccine.
- 40Sun, C.; Zhang, L.; Zhang, M.; Liu, Y.; Zhong, M.; Ma, X.; Chen, L. Induction of balance and breadth in the immune response is beneficial for the control of SIVmac239 replication in rhesus monkeys. J. Infect. 2010, 60 (5), 371– 381, DOI: 10.1016/j.jinf.2010.03.00540https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3czgs12ktA%253D%253D&md5=19532898ec9bc6d864a673ac02836494Induction of balance and breadth in the immune response is beneficial for the control of SIVmac239 replication in rhesus monkeysSun Caijun; Zhang Lei; Zhang Maochao; Liu Yichu; Zhong Miao; Ma Xin; Chen LingThe Journal of infection (2010), 60 (5), 371-81 ISSN:.OBJECTIVES: The aim of this study was to induce cellular and humoral responses with enhanced breadth and more balanced magnitude as a possible approach for an effective HIV vaccine. METHODS: All nine of the SIVmac239 genes (gag, pol, env, nef, vif, vpx, vpr, rev and tat) were optimized for mammalian expression, synthesized and cloned into recombinant adenovirus type 5 (Ad5). These vectors were used as a vaccine regimen, and the immunogenicity and immune protection of this regimen was assessed in murine and macaques. RESULTS: A vaccine regimen including all nine genes of the SIVmac239 virus was developed, and it was demonstrated that in contrast to single antigen vaccination, the total SIV antigen regimen more effectively elicited the balanced and broad immune responses in murine and macaques. Moreover, the responses afforded effective immune control against infection and replication of the highly pathogenic SIVmac239. CONCLUSIONS: Induction of balance and breadth in the immune response is beneficial in controlling SIVmac239 replication in rhesus monkeys. This study provides insight for the future development of an effective HIV vaccine.
- 41Sun, Y.; Chen, M. L.; Yang, D.; Qin, W. B.; Quan, G. L.; Wu, C. B.; Pan, X. Self-assembly nanomicelle-microneedle patches with enhanced tumor penetration for superior chemo-photothermal therapy. Nano Res. 2022, 15 (3), 2335– 2346, DOI: 10.1007/s12274-021-3817-x41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXit1WlsLjO&md5=f91936fc6da51d2ec0020eaa0b7762f6Self-assembly nanomicelle-microneedle patches with enhanced tumor penetration for superior chemo-photothermal therapySun, Ying; Chen, Minglong; Yang, Dan; Qin, Wanbing; Quan, Guilan; Wu, Chuanbin; Pan, XinNano Research (2022), 15 (3), 2335-2346CODEN: NRAEB5; ISSN:1998-0000. (Springer GmbH)Nanomedicine with high specificity has been a promising tool for cancer diagnosis and therapy. However, the successful application of nanoparticle-based superficial cancer therapy is severely hindered by restricted deep tumor tissue accumulation and penetration. Herein, a self-assembly nanomicelle dissolving microneedle (DMN) patch according to the ''nano in micro'' strategy was conducted to co-deliver a first-line chemotherapeutic agent paclitaxel (PTX), and a photosensitizer IR780 (PTX/IR780-NMs @DMNs) for chemo-photothermal synergetic melanoma therapy. Upon direct insertion into the tumor site, DMNs created a regular and multipoint three-dimensional drug depot to maximize the tumor accumulation. Accompanied by the DMN dissoln., the compn. of the needle matrixes self-assembled into nanomicelles, which could efficiently penetrate deep tumor tissue. Upon laser irradn., the nanomicelles could not only ablate tumor cells directly by photothermal conversion but also trigger PTX release to induce tumor cell apoptosis. In vivo results showed that compared with i.v. injection, IR780 delivered by PTX/IR780-NMs @DMNs was almost completely accumulated at the tumor site. The antitumor results revealed that the PTX/IR780-NMs @DMNs could effectively eliminate tumors with an 88% curable rate without any damage to normal tissues. This work provides a versatile and generalizable framework for designing self-assembly DMN-mediated combination therapy to fight against superficial cancer.
- 42Lin, S. Q.; Quan, G. L.; Hou, A. L.; Yang, P. P.; Peng, T. T.; Gu, Y. K.; Qin, W. B.; Liu, R. B.; Ma, X. Y.; Pan, X.; Liu, H.; Wang, L. L.; Wu, C. B. Strategy for hypertrophic scar therapy: Improved delivery of triamcinolone acetonide using mechanically robust tip-concentrated dissolving microneedle array. J. Controlled Release 2019, 306, 69– 82, DOI: 10.1016/j.jconrel.2019.05.03842https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtV2lt7nF&md5=18b2ceb48fb16cc5fb44706e28e2cfafStrategy for hypertrophic scar therapy: Improved delivery of triamcinolone acetonide using mechanically robust tip-concentrated dissolving microneedle arrayLin, Shiqi; Quan, Guilan; Hou, Ailin; Yang, Peipei; Peng, Tingting; Gu, Yukun; Qin, Wanbing; Liu, Rongben; Ma, Xiangyu; Pan, Xin; Liu, Hu; Wang, Lili; Wu, ChuanbinJournal of Controlled Release (2019), 306 (), 69-82CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)The purpose of this study was to provide an alternative treatment for HS by establishing a novel intradermal delivery system with a dissolving microneedle array (DMNA). To produce needles of higher mech. strength for successful insertion into the compact and hard HS tissue, hydroxypropyl-B-cyclodextrin (HP-B-CD) was added into sodium hyaluronic acid (HA), the needle material. The hydrogen interaction between HP-B-CD and HA restricted the mobility of the mol. chains, and subsequently increased the elastic modulus of the complex materials. The HP-B-CD also contributed to improved loading of the hydrophobic drug mols. into the DMNA needle tips. To assess the delivery of TA to the HS site via DMNA, an HS model was established in the ventral skin of New Zealand rabbits' ears. It was found that the value of the scar elevation index was decreased to normal, together with the down regulation of mRNA expressions of Collagen I and transforming growth factor-B1 (TGF-B1) following the administration of DMNA contg. TA (TA-DMNA). Western blotting results also revealed decreased protein expressions of both Collagen I and TGF-B1. Hence, TA-DMNA appears to be a promising alternative to multi-injection of TA injection, providing a convenient and low-pain therapeutic strategy for HS treatment.
- 43Li, M.; Chen, J.; Liu, Y.; Zhao, J.; Li, Y.; Hu, Y.; Chen, Y. Q.; Sun, L.; Shu, Y.; Feng, F.; Sun, C. Rational design of AAVrh10-vectored ACE2 functional domain to broadly block the cell entry of SARS-CoV-2 variants. Antiviral Res. 2022, 205, 105383 DOI: 10.1016/j.antiviral.2022.10538343https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XitVKns7jM&md5=3bd24c716edaf1bcf064d8f764122282Rational design of AAVrh10-vectored ACE2 functional domain to broadly block the cell entry of SARS-CoV-2 variantsLi, Minchao; Chen, Jiaoshan; Liu, Yajie; Zhao, Jin; Li, Yanjun; Hu, Yunqi; Chen, Yao-qing; Sun, Litao; Shu, Yuelong; Feng, Fengling; Sun, CaijunAntiviral Research (2022), 205 (), 105383CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)The frequently emerging SARS-CoV-2 variants have weakened the effectiveness of existing COVID-19 vaccines and neutralizing antibody therapy. Nevertheless, the infections of SARS-CoV-2 variants still depend on angiotensin-converting enzyme 2 (ACE2) receptor-mediated cell entry, and thus the sol. human ACE2 (shACE2) is a potential decoy for broadly blocking SARS-CoV-2 variants. In this study, we firstly generated the recombinant AAVrh10-vectored shACE2 constructs, a kind of adeno-assocd. virus (AAV) serotype with pulmonary tissue tropism, and then validated its inhibition capacity against SARS-CoV-2 infection. To further optimize the minimized ACE2 functional domain candidates, a comprehensive anal. was performed to clarify the interactions between the ACE2 orthologs from various species and the receptor binding domain (RBD) of SARS-CoV-2 spike (S) protein. Based on the key interface amino acids, we designed a series of truncated ACE2 orthologs, and then assessed their potential affinity to bind to SARS-CoV-2 variants RBD in silico. Of note, we found that the 24-83aa fragment of dog ACE2 (dACE224-83) had a higher affinity to the RBD of SARS-CoV-2 variants than that of human ACE2. Importantly, AAVrh10-vectored shACE2 or dACE224-83 constructs exhibited a broadly blockage breadth against SARS-CoV-2 prototype and variants in vitro and ex vivo. Collectively, these data highlighted a promising therapeutic strategy against SARS-CoV-2 variants.
- 44Luo, H.; Jia, T.; Chen, J.; Zeng, S.; Qiu, Z.; Wu, S.; Li, X.; Lei, Y.; Wang, X.; Wu, W.; Zhang, R.; Zou, X.; Feng, T.; Ding, R.; Zhang, Y.; Chen, Y. Q.; Sun, C.; Wang, T.; Fang, S.; Shu, Y. The Characterization of Disease Severity Associated IgG Subclasses Response in COVID-19 Patients. Front. Immunol. 2021, 12, 632814 DOI: 10.3389/fimmu.2021.63281444https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXpvF2iur4%253D&md5=1237dd3ed72ee775819c45cb22464425The characterization of disease severity associated IgG subclasses response in COVID-19 patientsLuo, Huanle; Jia, Tingting; Chen, Jiamin; Zeng, Shike; Qiu, Zengzhao; Wu, Shu; Li, Xu; Lei, Yuxuan; Wang, Xin; Wu, Weihua; Zhang, Renli; Zou, Xuan; Feng, Tiejian; Ding, Ruxia; Zhang, Yue; Chen, Yao-Qing; Sun, Caijun; Wang, Tian; Fang, Shisong; Shu, YuelongFrontiers in Immunology (2021), 12 (), 632814CODEN: FIRMCW; ISSN:1664-3224. (Frontiers Media S.A.)Increasing evidence suggests that dysregulated immune responses are assocd. with the clin. outcome of coronavirus disease 2019 (COVID-19). Nucleocapsid protein (NP)-, spike (S)-, receptor binding domain (RBD)- specific Ig isotypes, IgG subclasses and neutralizing antibody (NAb) were analyzed in 123 serum from 63 hospitalized patients with severe, moderate, mild or asymptomatic COVID-19. Mild to modest correlations were found between disease severity and antigen specific IgG subclasses in serum, of which IgG1 and IgG3 were neg. assocd. with viral load in nasopharyngeal swab. Multiple cytokines were significantly related with antigen-specific Ig isotypes and IgG subclasses, and IL-1β was pos. correlated with most antibodies. Furthermore, the old patients (≤ 60 years old) had higher levels of chemokines, increased NAb activities and SARS-CoV-2 specific IgG1, and IgG3 responses and compromised T cell responses compared to the young patients (≤ 18 years old), which are related with more severe cases. Higher IgG1 and IgG3 were found in COVID-19 patients with comorbidities while biol. sex had no effect on IgG subclasses. Overall, the authors have identified diseases severity was related to higher antibodies, of which IgG subclasses had weakly neg. correlation with viral load, and cytokines were significantly assocd. with antibody response. Further, advancing age and comorbidities had obvious effect on IgG1 and IgG3.
- 45Li, P.; Wang, Q.; He, Y.; Yang, C.; Zhang, Z.; Liu, Z.; Liu, B.; Yin, L.; Cui, Y.; Hu, P.; Liu, Y.; Zheng, P.; Wang, W.; Qu, L.; Sun, C.; Guan, S.; Feng, L.; Chen, L. Booster vaccination is required to elicit and maintain COVID-19 vaccine-induced immunity in SIV-infected macaques. Emerg. Microbes Infect. 2023, 12 (1), e2136538 DOI: 10.1080/22221751.2022.213653845https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXkt1yhtro%253D&md5=53b4acd282d993f26aab9b3221932c90Booster vaccination is required to elicit and maintain COVID-19 vaccine-induced immunity in SIV-infected macaquesLi, Pingchao; Wang, Qian; He, Yizi; Yang, Chenchen; Zhang, Zhengyuan; Liu, Zijian; Liu, Bo; Yin, Li; Cui, Yilan; Hu, Peiyu; Liu, Yichu; Zheng, Pingqian; Wang, Wei; Qu, Linbing; Sun, Caijun; Guan, Suhua; Feng, Liqiang; Chen, LingEmerging Microbes & Infections (2023), 12 (1), e2136538/1-e2136538/13CODEN: EMIMC4; ISSN:2222-1751. (Taylor & Francis Ltd.)Prolonged infection and possible evolution of SARS-CoV-2 in patients living with uncontrolled HIV-1 infection highlight the importance of an effective vaccination regimen, yet the immunogenicity of COVID-19 vaccines and predictive immune biomarkers have not been well investigated. Herein, we report that the magnitude and persistence of antibody and cell-mediated immunity (CMI) elicited by an Ad5-vectored COVID-19 vaccine are impaired in SIV-infected macaques with high viral loads (> 105 genome copies per mL plasma, SIVhi) but not in macaques with low viral loads (< 105, SIVlow). After a second vaccination, the immune responses are robustly enhanced in all uninfected and SIVlow macaques. These responses also show a moderate increase in 70% SIVhi macaques but decline sharply soon after. Further anal. reveals that decreased antibody and CMI responses are assocd. with reduced circulating follicular helper T cell (TFH) counts and aberrant CD4/CD8 ratios, resp., indicating that dysregulation of CD4+ T cells by SIV infection impairs the COVID-19 vaccine-induced immunity. Ad5-vectored COVID-19 vaccine shows no impact on SIV loads or SIV-specific CMI responses. Our study underscores the necessity of frequent booster vaccinations in HIV-infected patients and provides indicative biomarkers for predicting vaccination effectiveness in these patients.
- 46Wen, Z.; Fang, C.; Liu, X.; Liu, Y.; Li, M.; Yuan, Y.; Han, Z.; Wang, C.; Zhang, T.; Sun, C. A recombinant Mycobacterium smegmatis-based surface display system for developing the T cell-based COVID-19 vaccine. Hum. Vaccines Immunother. 2023, 19 (1), 2171233 DOI: 10.1080/21645515.2023.217123346https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXjsVGku7Y%253D&md5=10974bd628fd6d5c6ea2a6876037bc54A recombinant Mycobacterium smegmatis-based surface display system for developing the T cell-based COVID-19 vaccineWen, Ziyu; Fang, Cuiting; Liu, Xinglai; Liu, Yan; Li, Minchao; Yuan, Yue; Han, Zirong; Wang, Congcong; Zhang, Tianyu; Sun, CaijunHuman Vaccines & Immunotherapeutics (2023), 19 (1), 2171233/1-2171233/12CODEN: HVIUAK; ISSN:2164-554X. (Taylor & Francis Ltd.)The immune escape mutations of SARS-CoV-2 variants emerged frequently, posing a new challenge to weaken the protective efficacy of current vaccines. Thus, the development of novel SARS-CoV-2 vaccines is of great significance for future epidemic prevention and control. We herein reported constructing the attenuated Mycobacterium smegmatis (M. smegmatis) as a bacterial surface display system to carry the spike (S) and nucleocapsid (N) of SARS-CoV-2. To mimic the native localization on the surface of viral particles, the S or N antigen was fused with truncated PE_PGRS33 protein, which is a transportation component onto the cell wall of Mycobacterium tuberculosis (M.tb). The sub-cellular fraction anal. demonstrated that S or N protein was exactly expressed onto the surface (cell wall) of the recombinant M. smegmatis. After the immunization of the M. smegmatis-based COVID-19 vaccine candidate in mice, S or N antigen-specific T cell immune responses were effectively elicited, and the subsets of central memory CD4+ T cells and CD8+ T cells were significantly induced. Further anal. showed that there were some potential cross-reactive CTL epitopes between SARS-CoV-2 and M.smegmatis. Overall, our data provided insights that M. smegmatis-based bacterial surface display system could be a suitable vector for developing T cell-based vaccines against SARS-CoV-2 and other infectious diseases.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsnano.3c09521.
Supplementary figures: physiochemical characterization of COS-engineered DNA vaccine; stability of COS-encapsulated DNA vaccine; the COS showed no obvious cell toxicity to different cell lines; in vitro transfection efficacy of DNA nanoparticle vaccine; in vitro transfection efficacy of DNA nanoparticle vaccine in DC 2.4 and RAW264.7 cells; cellular uptake of DNA nanoparticle vaccine in HEK-293T cells determined by flow cytometry; co-localization observed by CLSM of the Cy5-labeled pVAX-S (red) and LysoTracker (green) in HEK-293T cells; physiochemical and biological characterization of DNA@COS-MN dissolving in PBS; neutralization efficiency of mice serum against SARS-CoV-2 variants delta and omicron with dilution at 1:20 at day 42; characterization of DNA nanoparticle vaccine in MN stored at room temperature for 30 days; SARS-CoV-2-specific cellular immune responses after MN-mediated DNA nanoparticle vaccine at day 21; SARS-CoV-2-specific cellular immune responses after MN-mediated DNA nanoparticle vaccine at day 42; T-SNE analysis of concatenated data from group S+N@COS-IM and group S+N@COS-MN for stimulation with S1, S2, and N peptide pool, showing density plots for each condition; detection of ROS by fluorescence of DCFH-DA in the DC 2.4 cells incubated with different concentration of COS; change of MMP of DC 2.4 cells at different times after incubation with COS; CD40, CD86, and MHC II expression in DC 2.4 cells incubated with 16 μg/mL COS for 24 h in the presence or absence of CsA; Supplementary table: primer sequence for qPCR to verify the DC 2.4 cells activation after COS treatment (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.