nanobodies

纳米抗体
  • 文章类型: Journal Article
    在过去的十年中,随着下一代治疗性抗体的出现,癌症治疗领域取得了显着发展。化疗等常规治疗带来了巨大挑战,包括不良副作用。单克隆抗体为更有针对性和有效的干预措施铺平了道路。从嵌合抗体到人源化和完全人抗体的进化已经导致免疫原性的降低和增强的体内耐受性。下一代抗体的出现,包括双特异性抗体,纳米抗体,抗体-药物缀合物,糖工程抗体,和抗体片段,代表了癌症治疗的飞跃.这些创新提供了更高的效力,适应性,并降低耐药性。目标验证等挑战,免疫原性,和高生产成本存在。然而,抗体工程技术的技术进步为解决这些问题提供了乐观。未来预示着范式的转变,正在进行的研究将这些强大的抗体推向最前沿,彻底改变抗击癌症的斗争,创造新的预防和治疗方法。这篇综述概述了三种下一代基于抗体的分子,即双特异性抗体,抗体-药物缀合物,和纳米抗体在癌症治疗中显示出有希望的结果。它讨论了抗体从常规形式到下一代分子的进化,以及它们在癌症治疗中的应用,生产方法,和相关的挑战。该综述旨在为研究人员提供有关下一代基于抗体的癌症疗法的不断发展的前景及其彻底改变治疗策略的潜力的见解。
    The field of cancer treatment has evolved significantly over the last decade with the emergence of next-generation therapeutic antibodies. Conventional treatments like chemotherapy pose significant challenges, including adverse side effects. Monoclonal antibodies have paved the way for more targeted and effective interventions. The evolution from chimeric to humanized and fully human antibodies has led to a reduction in immunogenicity and enhanced tolerance in vivo. The advent of next-generation antibodies, including bispecific antibodies, nanobodies, antibody-drug conjugates, glyco-engineered antibodies, and antibody fragments, represents a leap forward in cancer therapy. These innovations offer increased potency, adaptability, and reduced drug resistance. Challenges such as target validation, immunogenicity, and high production costs exist. However, technological advancements in antibody engineering techniques provide optimism for addressing these issues. The future promises a paradigm shift, where ongoing research will propel these powerful antibodies to the forefront, revolutionizing the fight against cancer and creating new preventive and curative treatments. This review provides an overview of three next-generation antibody-based molecules, namely bispecific antibodies, antibody-drug conjugates, and nanobodies that have shown promising results in cancer treatment. It discusses the evolution of antibodies from conventional forms to next-generation molecules, along with their applications in cancer treatment, production methods, and associated challenges. The review aims to offer researchers insights into the evolving landscape of next-generation antibody-based cancer therapeutics and their potential to revolutionize treatment strategies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:虽然免疫疗法彻底改变了肿瘤学领域,治疗反应性的变化限制了这些疗法的广泛适用性.免疫细胞诊断成像,特别是CD8+T细胞,动力学可以允许早期患者分层,并改善治疗疗效和安全性.在这项研究中,我们报道了一种基于纳米抗体的免疫示踪剂的开发,用于人类CD8+T细胞动力学的非侵入性SPECT和PET成像。
    方法:通过美洲驼免疫和随后的生物淘选产生靶向人CD8β的纳米抗体。前导抗人CD8β纳米抗体的特征在于结合,特异性,稳定性和毒性。铅纳米体被标记为99m,镓-68和铜-64用于通过SPECT/CT或PET/CT在人CD8转基因小鼠和非人灵长类动物中的人T细胞淋巴瘤和CD8T细胞的非侵入性成像。MC38荷瘤小鼠中CD8+T细胞的重复成像允许CD8+T细胞动力学的可视化。
    结果:基于纳米抗体的免疫示踪剂显示与人CD8的高亲和力和特异性结合,而没有不需要的免疫激活。CD8+T细胞通过SPECT和PET成像在初始和荷瘤小鼠中以及在具有高灵敏度的初始非人灵长类动物中进行非侵入性可视化。与以前的人类CD8靶向免疫示踪剂相比,基于纳米抗体的免疫示踪剂对CD8+T细胞的特异性增强和/或体内药代动力学更快。使我们能够在早期时间点跟踪人类CD8+T细胞动力学。
    结论:这项研究描述了一种更特异性的人类CD8+T细胞靶向免疫示踪剂的发展,允许通过人CD8+T细胞动力学的非侵入性成像来随访免疫治疗反应。
    OBJECTIVE: While immunotherapy has revolutionized the oncology field, variations in therapy responsiveness limit the broad applicability of these therapies. Diagnostic imaging of immune cell, and specifically CD8+ T cell, dynamics could allow early patient stratification and result in improved therapy efficacy and safety. In this study, we report the development of a nanobody-based immunotracer for non-invasive SPECT and PET imaging of human CD8+ T-cell dynamics.
    METHODS: Nanobodies targeting human CD8β were generated by llama immunizations and subsequent biopanning. The lead anti-human CD8β nanobody was characterized on binding, specificity, stability and toxicity. The lead nanobody was labeled with technetium-99m, gallium-68 and copper-64 for non-invasive imaging of human T-cell lymphomas and CD8+ T cells in human CD8 transgenic mice and non-human primates by SPECT/CT or PET/CT. Repeated imaging of CD8+ T cells in MC38 tumor-bearing mice allowed visualization of CD8+ T-cell dynamics.
    RESULTS: The nanobody-based immunotracer showed high affinity and specific binding to human CD8 without unwanted immune activation. CD8+ T cells were non-invasively visualized by SPECT and PET imaging in naïve and tumor-bearing mice and in naïve non-human primates with high sensitivity. The nanobody-based immunotracer showed enhanced specificity for CD8+ T cells and/or faster in vivo pharmacokinetics compared to previous human CD8-targeting immunotracers, allowing us to follow human CD8+ T-cell dynamics already at early timepoints.
    CONCLUSIONS: This study describes the development of a more specific human CD8+ T-cell-targeting immunotracer, allowing follow-up of immunotherapy responses by non-invasive imaging of human CD8+ T-cell dynamics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    源自骆驼科动物和鲨鱼的纳米抗体在治疗应用中提供了独特的优势,因为它们能够结合以前无法接近的表位。传统的纳米抗体开发方法面临诸如伦理问题和抗原毒性等挑战。我们的研究提出了一种合成的,使用三核苷酸定向诱变技术,允许互补决定区(CDR)中精确的氨基酸组成,专注于CDR3的多样性。该方法避免了与其他合成抗体文库构建方法相关的常见问题,例如移码突变和终止密码子插入。通过分析FDA批准的纳米抗体和蛋白质数据库序列,我们设计了具有不同CDR3长度的子文库,并引入了氨基酸取代以提高溶解度.通过针对诸如PD-1、ATXN1和STAT3的靶标的纳米抗体的成功分离来验证我们的文库展示了用于开发高特异性和亲和力纳米抗体的通用和伦理平台,并且代表了生物技术的显著进步。
    Nanobodies derived from camelids and sharks offer unique advantages in therapeutic applications due to their ability to bind to epitopes that were previously inaccessible. Traditional methods of nanobody development face challenges such as ethical concerns and antigen toxicity. Our study presents a synthetic, phagedisplayed nanobody library using trinucleotide-directed mutagenesis technology, which allows precise amino acid composition in complementarity-determining regions (CDRs), with a focus on CDR3 diversity. This approach avoids common problems such as frameshift mutations and stop codon insertions associated with other synthetic antibody library construction methods. By analyzing FDA-approved nanobodies and Protein Data Bank sequences, we designed sub-libraries with different CDR3 lengths and introduced amino acid substitutions to improve solubility. The validation of our library through the successful isolation of nanobodies against targets such as PD-1, ATXN1 and STAT3 demonstrates a versatile and ethical platform for the development of high specificity and affinity nanobodies and represents a significant advance in biotechnology.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    蛋白质的错误折叠和聚集与各种神经退行性疾病有关,如阿尔茨海默病(AD)。小分子工程抗体,如单链片段可变(scFv)抗体和纳米抗体(NBS),近年来由于其强大的构象特异性而受到关注,穿越血脑屏障(BBB)的能力,低免疫原性,并增强与聚集体内活性位点的接近度。
    我们已经综述了包括scFvs和Nbs的治疗的最新进展,这些治疗有效和特异性地靶向病理蛋白聚集体。在MEDLINE中搜索了相关出版物,GOOGLE学者,ElsevierScienceDirect和Wiley在线图书馆。
    我们回顾了scFvs和Nbs对病理蛋白聚集体的最新特异性靶向研究。这些工程化抗体可以通过识别抗原表位或通过构象特异性来抑制错误折叠蛋白质的聚集或促进其分解。此外,我们讨论了提高工程抗体在治疗AD中的有效应用的策略。这些技术策略将为临床应用小分子抗体药物开发神经系统疾病的有效治疗奠定基础。通过合理的应用策略,小分子工程抗体有望在神经系统疾病的靶向治疗中具有显著潜力.
    UNASSIGNED: The misfolding and aggregation of proteins are associated with various neurodegenerative diseases, such as Alzheimer\'s disease (AD). The small-molecule engineered antibodies, such as single-chain fragment variable (scFv) antibodies and nanobodies (Nbs), have gained attention in recent years due to their strong conformational specificity, ability to cross the blood-brain barrier (BBB), low immunogenicity, and enhanced proximity to active sites within aggregates.
    UNASSIGNED: We have reviewed recent advances in therapies involving scFvs and Nbs that efficiently and specifically target pathological protein aggregates. Relevant publications were searched for in MEDLINE, GOOGLE SCHOLAR, Elsevier ScienceDirect and Wiley Online Library.
    UNASSIGNED: We reviewed the recent and specific targeting of pathological protein aggregates by scFvs and Nbs. These engineered antibodies can inhibit the aggregation or promote the disassembly of misfolded proteins by recognizing antigenic epitopes or through conformational specificity. Additionally, we discuss strategies for improving the effective application of engineered antibodies in treating AD. These technological strategies will lay the foundation for the clinical application of small-molecule antibody drugs in developing effective treatments for neurological diseases. Through rational application strategies, small-molecule engineered antibodies are expected to have significant potential in targeted therapy for neurological disorders.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    玉米致死性坏死(MLN)是由玉米褪绿斑驳病毒(MCMV)引起的一种玉米病害,一种导致30-100%产量损失的potyvirus。本研究旨在分离针对MCMV外壳蛋白(CP)的纳米抗体,以诊断MLN。在大肠杆菌中表达的MCMVCP用于美洲驼免疫。VHH(即重链的可变重链结构域)基因片段从从免疫美洲驼抽取的血液中制备,并用于在大肠杆菌TG1细胞中产生文库。通过三轮噬菌体展示和针对MCMVCP淘选选择MCMV特异性纳米抗体。最终在大肠杆菌WK6细胞中表达并纯化所选择的纳米抗体。鉴定了11个MCMV特异性纳米抗体,并显示可在受感染的玉米植物中检测MCMV。因此,我们的结果表明,从用MCMVCP免疫的美洲驼中分离的纳米抗体可以区分感染的和健康的玉米植物,有可能开发负担得起的MCMV检测协议。
    Maize lethal necrosis (MLN) is a maize disease caused by the maize chlorotic mottle virus (MCMV), a potyvirus which causes yield losses of 30-100%. The present study aimed to isolate nanobodies against the MCMV coat protein (CP) for the diagnosis of MLN. MCMV CP expressed in Escherichia coli was used for llama immunization. VHH (i.e. variable heavy domain of heavy chain) gene fragments were prepared from blood drawn from the immunized llama and used to generate a library in E. coli TG1 cells. MCMV specific nanobodies were selected by three rounds of phage display and panning against MCMV CP. The selected nanobodies were finally expressed in E. coli WK6 cells and purified. Eleven MCMV-specific nanobodies were identified and shown to detect MCMV in infected maize plants. Thus, our results show that nanobodies isolated from llama immunized with MCMV CP can distinguish infected and healthy maize plants, potentially enabling development of affordable MCMV detection protocols.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    单结构域抗体(sdAb)或纳米抗体由于其小尺寸(〜15kDa)和在生物衍生疗法中的多种应用而受到广泛关注。随着许多现代生物技术突破应用于抗体工程和设计,纳米体的热稳定性或熔化温度(Tm)是其成功利用的关键。在这项研究中,我们提出了TEMPRO,这是一种使用计算方法估计纳米体Tm的预测建模方法。我们的方法集成了各种纳米抗体生物物理特征,包括进化尺度建模(ESM)嵌入,NetSurfP3结构预测,来自AlphaFold2的每个sdAb区域的pLDDT评分以及每个序列的物理化学特征。这种方法通过我们的组合数据集进行了验证,该数据集包含567个独特序列,这些序列具有来自手动管理的内部数据和最近发布的纳米抗体数据库的相应实验Tm值。NbThermo.我们的结果表明,蛋白质嵌入在可靠地预测sdAb的Tm方面的功效,平均绝对误差(MAE)为4.03°C,均方根误差(RMSE)为5.66°C,从而为各种生物医学和治疗应用的纳米抗体的优化提供了有价值的工具。此外,我们已经使用实验确定的来自NbThermo中未发现的纳米抗体的Tms验证了模型的性能。这种预测模型不仅增强了纳米体热稳定性预测,但也提供了使用嵌入作为促进下游蛋白质分析更广泛适用性的工具的有用观点。
    Single-domain antibodies (sdAbs) or nanobodies have received widespread attention due to their small size (~ 15 kDa) and diverse applications in bio-derived therapeutics. As many modern biotechnology breakthroughs are applied to antibody engineering and design, nanobody thermostability or melting temperature (Tm) is crucial for their successful utilization. In this study, we present TEMPRO which is a predictive modeling approach for estimating the Tm of nanobodies using computational methods. Our methodology integrates various nanobody biophysical features to include Evolutionary Scale Modeling (ESM) embeddings, NetSurfP3 structural predictions, pLDDT scores per sdAb region from AlphaFold2, and each sequence\'s physicochemical characteristics. This approach is validated with our combined dataset containing 567 unique sequences with corresponding experimental Tm values from a manually curated internal data and a recently published nanobody database, NbThermo. Our results indicate the efficacy of protein embeddings in reliably predicting the Tm of sdAbs with mean absolute error (MAE) of 4.03 °C and root mean squared error (RMSE) of 5.66 °C, thus offering a valuable tool for the optimization of nanobodies for various biomedical and therapeutic applications. Moreover, we have validated the models\' performance using experimentally determined Tms from nanobodies not found in NbThermo. This predictive model not only enhances nanobody thermostability prediction, but also provides a useful perspective of using embeddings as a tool for facilitating a broader applicability of downstream protein analyses.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    近年来,在发现用于治疗中枢神经系统(CNS)疾病的治疗性蛋白质和肽方面取得了快速进展。然而,它们的临床应用受到诸如低生物膜渗透性等挑战的极大阻碍,稳定性差,循环时间短,和强大的血脑屏障(BBB)。最近,在了解BBB的动力学和开发将蛋白质和肽递送到CNS的有效方法方面已经取得了实质性的进步。特别是通过使用各种纳米粒子。在这里,我们概述了在生理和病理条件下对BBB的最新认识,强调它们对大脑药物输送的影响。我们总结了先进的策略,并阐明了将蛋白质和肽输送到大脑的潜在机制。我们重点介绍了纳米载体在通过BBB杂交治疗CNS疾病中的发展和应用。我们还对当前策略的局限性和障碍提出了批评意见,并对未来的研究提出了展望。
    Recent years have witnessed rapid progress in the discovery of therapeutic proteins and peptides for the treatment of central nervous system (CNS) diseases. However, their clinical applications have been considerably hindered by challenges such as low biomembrane permeability, poor stability, short circulation time, and the formidable blood-brain barrier (BBB). Recently, substantial improvements have been made in understanding the dynamics of the BBB and developing efficient approaches for delivering proteins and peptides to the CNS, especially by using various nanoparticles. Herein, we present an overview of the up-to-date understanding of the BBB under physiological and pathological conditions, emphasizing their effects on brain drug delivery. We summarize advanced strategies and elucidate the underlying mechanisms for delivering proteins and peptides to the brain. We highlight the developments and applications of nanocarriers in treating CNS diseases via BBB crossing. We also provide critical opinions on the limitations and obstacles of the current strategies and put forward prospects for future research.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    尿液,生物医学研究和临床诊断中生物标志物的常见来源,最近引起了新的兴趣。由于其细胞外囊泡(EV)内容物的存在,它最近已成为研究的焦点。已发现这些uEV反映肾脏的生理和病理状况,尿路上皮,和前列腺组织,可以说明进一步的分子过程,导致该领域的研究迅速扩展。在这项工作中,我们介绍了基于免疫亲和力的uEVs分离方法相对于通过差异超速离心进行的金标准纯化方法的优势[在纯度和抗原存在方面。通过将特异性抗体与官能化的聚甲基丙烯酸酯聚合物结合,免疫亲和方法变得可行。流式细胞术显示显著的荧光变化,验证标记(CD9,CD63,CD81)的存在,并确认分离方法的有效性。显微镜评估已经表明,囊泡的形态保持完整并且对应于uEV的预期形状和尺寸。描述的协议很便宜,快,易于处理,具有良好的重现性,并可应用于进一步的生物样品。
    Urine, a common source of biological markers in biomedical research and clinical diagnosis, has recently generated a new wave of interest. It has recently become a focus of study due to the presence of its content of extracellular vesicles (EVs). These uEVs have been found to reflect physiological and pathological conditions in kidney, urothelial, and prostate tissue and can illustrate further molecular processes, leading to a rapid expansion of research in this field In this work, we present the advantages of an immunoaffinity-based method for uEVs\' isolation with respect to the gold standard purification approach performed by differential ultracentrifugation [in terms of purity and antigen presence. The immunoaffinity method was made feasible by combining specific antibodies with a functionalized polymethacrylate polymer. Flow cytometry indicated a significant fluorescence shift, validating the presence of the markers (CD9, CD63, CD81) and confirming the effectiveness of the isolation method. Microscopy evaluations have shown that the morphology of the vesicles remained intact and corresponded to the expected shapes and dimensions of uEVs. The described protocol is inexpensive, fast, easy to process, has good reproducibility, and can be applied to further biological samples.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    炎症性肠病(IBD)的特征是慢性肠道炎症,无法治愈,治疗选择有限,通常具有全身性副作用。在这项研究中,我们开发了一种靶标特异性系统,通过工程化益生菌大肠杆菌Nissle1917(EcN)来潜在地治疗IBD。我们的模块化系统包括三个组件:能够检测炎症生物标志物一氧化氮(NO)的基于转录因子的传感器(NorR),1型溶血素分泌系统,和由人源化抗TNFα纳米抗体文库组成的治疗货物。尽管灵敏度降低了,我们的系统对NO表现出浓度依赖性反应,成功分泌具有与常用药物阿达木单抗相当的结合亲和力的功能性纳米抗体,如通过酶联免疫吸附测定和体外测定所证实的。这个新验证的纳米抗体库扩展了EcN治疗能力。所采用的分泌系统,也是第一次在EcN中出现,可以进一步适合作为筛选和纯化目的蛋白的平台。此外,我们提供了一个数学框架来评估工程益生菌系统中的关键参数,包括相关分子的产生和扩散,细菌定植率,和粒子相互作用。这种综合方法扩展了基于EcN的疗法的合成生物学工具箱,提供新颖的零件,电路,以及炎症热点可调节反应的模型。
    Inflammatory bowel disease (IBD) is characterized by chronic intestinal inflammation with no cure and limited treatment options that often have systemic side effects. In this study, we developed a target-specific system to potentially treat IBD by engineering the probiotic bacterium Escherichia coli Nissle 1917 (EcN). Our modular system comprises three components: a transcription factor-based sensor (NorR) capable of detecting the inflammation biomarker nitric oxide (NO), a type 1 hemolysin secretion system, and a therapeutic cargo consisting of a library of humanized anti-TNFα nanobodies. Despite a reduction in sensitivity, our system demonstrated a concentration-dependent response to NO, successfully secreting functional nanobodies with binding affinities comparable to the commonly used drug Adalimumab, as confirmed by enzyme-linked immunosorbent assay and in vitro assays. This newly validated nanobody library expands EcN therapeutic capabilities. The adopted secretion system, also characterized for the first time in EcN, can be further adapted as a platform for screening and purifying proteins of interest. Additionally, we provided a mathematical framework to assess critical parameters in engineering probiotic systems, including the production and diffusion of relevant molecules, bacterial colonization rates, and particle interactions. This integrated approach expands the synthetic biology toolbox for EcN-based therapies, providing novel parts, circuits, and a model for tunable responses at inflammatory hotspots.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    基于抗体对的免疫测定平台是病原体检测领域中必不可少且有效的工具。然而,制备繁琐且检测灵敏度有限的抗体对在建立高灵敏度检测平台时面临挑战。在这项研究中,使用COVID-19测试作为案例,我们利用容易获得的纳米抗体作为检测抗体,并进一步提出了一种精确的设计概念,采用更科学和有效的筛选策略来获得超灵敏的抗体对。我们采用能够结合核衣壳(NP)或受体结合域(RBD)抗原夹心的不同抗原表位的纳米抗体作为快速检测格式的单克隆抗体(mAb)夹心的替代品,并利用时间分辨荧光(TRF)微球作为信号探针。因此,我们开发了基于多表位纳米体夹心的荧光侧流免疫分析(FLFA)条。我们的结果表明,NP抗原的检出限为12.01pg/mL,而使用我们的FLFA试条,RBD抗原的限制为6.51pg/mL。基于双单克隆抗体三明治,本文提供的值显示灵敏度提高了4到32倍,和32至256倍的增强与市售抗原侧流测定试剂盒相比。此外,我们展示了所提出的试纸的优异特性,包括它的特异性,稳定性,准确度,和可重复性,这突显了它的预期效用。的确,这些发现表明,我们建立的筛选策略以及多表位纳米抗体夹心模式提供了病原体检测领域的优化策略。
    Antibody pairs-based immunoassay platforms served as essential and effective tools in the field of pathogen detection. However, the cumbersome preparation and limited detection sensitivity of antibody pairs challenge in establishment of a highly sensitive detection platform. In this study, using COVID-19 testing as a case, we utilized readily accessible nanobodies as detection antibodies and further proposed an accurate design concept with a more scientific and efficient screening strategy to obtain ultrasensitive antibody pairs. We employed nanobodies capable of binding different antigenic epitopes of the nucleocapsid (NP) or receptor-binding domain (RBD) antigens sandwich as substitutes for monoclonal antibodies (mAbs) sandwich in fast detection formats and utilized time-resolved fluorescence (TRF) microspheres as the signal probe. Consequently, we developed a multi-epitope nanobody sandwich-based fluorescence lateral flow immunoassay (FLFA) strip. Our results suggest that the NP antigen had a detection limit of 12.01pg/mL, while the RBD antigen had a limit of 6.51 pg/mL using our FLFA strip. Based on double mAb sandwiches, the values presented herein demonstrated 4 to 32-fold enhancements in sensitivity, and 32 to 256-fold enhancements compared to commercially available antigen lateral flow assay kits. Furthermore, we demonstrated the excellent characteristics of the proposed test strip, including its specificity, stability, accuracy, and repeatability, which underscores its the prospective utility. Indeed, these findings indicate that our established screening strategy along with the multi-epitope nanobody sandwich mode provides an optimized strategy in the field of pathogen detection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号