rAAV production

  • 文章类型: Journal Article
    重组腺相关病毒(rAAV)载体是基因治疗中最有前途的病毒载体之一。然而,人胚肾(HEK)细胞的制造能力有限是rAAV商业化的障碍。我们研究了内质网(ER)蛋白加工和凋亡基因对HEK293细胞中瞬时rAAV产生的影响。我们根据先前的转录组学研究选择了四个候选基因:XBP1,GADD34/PPP1R15A,HSPA6和BCL2。这些基因稳定整合到HEK293宿主细胞中。传统的三质粒瞬时转染用于评估载体生产能力以及过表达的稳定池和亲本细胞的质量。我们表明,在HEK293T细胞中,XBP1,HSPA6和GADD34的过表达使rAAV生产率增加高达100%,并使rAAV的比生产率增加高达78%。此外,当亲本细胞生产力高时,观察到与ER蛋白加工基因过表达相关的更显著的改善,但是在低产量条件下没有检测到实质性的变化。我们还证实了不同血清型(AAV2和AAV8)和不同细胞系(HEK293T和HEK293)的基因组滴度提高;然而,改善的程度可能有所不同。这项研究揭示了ER蛋白加工途径在病毒颗粒合成中的重要性,衣壳组件,和矢量生产。关键点:•内质网(ER)蛋白加工(XBP1,HSPA6和GADD34)的上调导致rAAV生产力最大100%的增加,并且在HEK293T细胞中特定rAAV生产力最大78%的提高•可以在不同的HEK293细胞系中验证生产力的提高,并可用于生产各种AAV血清型,尽管增强的程度可能略有不同•当亲本细胞生产力高时,观察到与过表达ER蛋白加工基因相关的更明显的改善。在低产量条件下注意到最小的变化。
    The recombinant adeno-associated virus (rAAV) vector is among the most promising viral vectors in gene therapy. However, the limited manufacturing capacity in human embryonic kidney (HEK) cells is a barrier to rAAV commercialization. We investigated the impact of endoplasmic reticulum (ER) protein processing and apoptotic genes on transient rAAV production in HEK293 cells. We selected four candidate genes based on prior transcriptomic studies: XBP1, GADD34 / PPP1R15A, HSPA6, and BCL2. These genes were stably integrated into HEK293 host cells. Traditional triple-plasmid transient transfection was used to assess the vector production capability and the quality of both the overexpressed stable pools and the parental cells. We show that the overexpression of XBP1, HSPA6, and GADD34 increases rAAV productivity by up to 100% and increases specific rAAV productivity by up to 78% in HEK293T cells. Additionally, more prominent improvement associated with ER protein processing gene overexpression was observed when parental cell productivity was high, but no substantial variation was detected under low-producing conditions. We also confirmed genome titer improvement across different serotypes (AAV2 and AAV8) and different cell lines (HEK293T and HEK293); however, the extent of improvement may vary. This study unveiled the importance of ER protein processing pathways in viral particle synthesis, capsid assembly, and vector production. KEY POINTS: • Upregulation of endoplasmic reticulum (ER) protein processing (XBP1, HSPA6, and GADD34) leads to a maximum 100% increase in rAAV productivity and a maximum 78% boost in specific rAAV productivity in HEK293T cells • The enhancement in productivity can be validated across different HEK293 cell lines and can be used for the production of various AAV serotypes, although the extent of the enhancement might vary slightly • The more pronounced improvements linked to overexpressing ER protein processing genes were observed when parental cell productivity was high, with minimal variation noted under low-producing conditions.
    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
    昆虫细胞-杆状病毒表达载体(IC-BEV)平台使得能够进行小规模研究和大规模商业生产重组蛋白和治疗性生物制品,包括基于重组腺相关病毒(rAAV)的基因递送载体。由于其简单性,该平台的广泛使用可与其他基于哺乳动物细胞系的平台相媲美。高产,可比较的质量属性,和强大的生物处理功能。在这一章中,我们描述了使用One-Bac平台的最新修改之一的rAAV生产方案,其由携带AAVRep2/Cap5基因的稳定转化的Sf9细胞系组成,所述AAVRep2/Cap5基因在感染后被单个重组杆状病毒表达载体诱导,所述载体含有感兴趣的转基因(rAAV基因组)。总体方案包括基本步骤,包括rBEV工作库存准备,rAAV生产,和基于离心的细胞培养裂解物的澄清。相同的协议也可以应用于使用传统的Three-Bac的rAAV载体生产,两个Bac,和Mono-Bac平台,无需进行重大更改。
    The insect cell-baculovirus expression vector (IC-BEV) platform has enabled small research-scale and large commercial-scale production of recombinant proteins and therapeutic biologics including recombinant adeno-associated virus (rAAV)-based gene delivery vectors. The wide use of this platform is comparable with other mammalian cell line-based platforms due to its simplicity, high-yield, comparable quality attributes, and robust bioprocessing features. In this chapter, we describe a rAAV production protocol employing one of the recent modifications of the One-Bac platform that consists of a stable transformed Sf9 cell line carrying AAV Rep2/Cap5 genes that are induced upon infection with a single recombinant baculovirus expression vector harboring the transgene of interest (rAAV genome). The overall protocol consists of essential steps including rBEV working stock preparation, rAAV production, and centrifugation-based clarification of cell culture lysate. The same protocol can also be applied for rAAV vector production using traditional Three-Bac, Two-Bac, and Mono-Bac platforms without requiring significant changes.
    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
    重组腺相关病毒(rAAV)是一种突出的基因传递载体,为新的基因治疗方法开辟了有希望的机会。然而,目前主要的病毒载体生产平台,哺乳动物细胞中的三重转染,可能无法满足日益增长的需求。因此,非常需要从宿主细胞的角度理解生产瓶颈,并改造细胞以更有利于和耐受病毒载体的生产,从而有效地增强rAAV制造。在这次审查中,我们对rAAV生产中涉及的复杂细胞过程进行了全面探索,包括不同的阶段,如质粒进入细胞质,质粒运输和核递送,rAAV结构/非结构蛋白表达,病毒衣壳组装,基因组复制,基因组包装,和rAAV释放/分泌。每个阶段都总结了支持病毒复制的宿主细胞作为制造工厂或表现出针对病毒生产的防御行为的基本生物学知识。从宿主细胞和材料的角度出发的控制策略(例如,AAV质粒)基于分子特征的表征和我们对AAV病毒生命周期的现有知识,作为我们的见解提出,在人胚肾(HEK)细胞中产生rAAV和其它病毒载体。
    Recombinant adeno-associated virus (rAAV) is one of the prominent gene delivery vehicles that has opened promising opportunities for novel gene therapeutic approaches. However, the current major viral vector production platform, triple transfection in mammalian cells, may not meet the increasing demand. Thus, it is highly required to understand production bottlenecks from the host cell perspective and engineer the cells to be more favorable and tolerant to viral vector production, thereby effectively enhancing rAAV manufacturing. In this review, we provided a comprehensive exploration of the intricate cellular process involved in rAAV production, encompassing various stages such as plasmid entry to the cytoplasm, plasmid trafficking and nuclear delivery, rAAV structural/non-structural protein expression, viral capsid assembly, genome replication, genome packaging, and rAAV release/secretion. The knowledge in the fundamental biology of host cells supporting viral replication as manufacturing factories or exhibiting defending behaviors against viral production is summarized for each stage. The control strategies from the perspectives of host cell and materials (e.g., AAV plasmids) are proposed as our insights based on the characterization of molecular features and our existing knowledge of the AAV viral life cycle, rAAV and other viral vector production in the Human embryonic kidney (HEK) cells.
    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
    基于重组腺相关病毒(rAAV)的基因治疗的发展呈指数级增长,因此,当前的rAAV制造平台需要更高效以满足不断增长的需求。病毒生产对细胞底物有很大的需求,能源,和机械;因此,病毒的生产在很大程度上依赖于宿主细胞的生理学。转录组学,作为机械驱动的工具,用于鉴定显著调节的途径并研究用于支持rAAV生产的宿主细胞的细胞特征。这项研究通过比较亲本人胚胎肾细胞(HEK293)中随时间的病毒产生培养物与非产生培养物,研究了在各自培养基中培养的两种细胞系的转录组学特征。结果表明,宿主细胞的先天免疫应答信号通路(例如,RIG-I样受体信号通路,Toll样受体信号通路,胞质DNA传感途径,JAK-STAT信号通路)显著富集和上调。这伴随着宿主细胞应激反应,包括内质网应激,自噬,和病毒生产中的细胞凋亡。相比之下,在病毒生产的后期,脂肪酸代谢和中性氨基酸转运被下调。我们的转录组学分析揭示了rAAV生产的细胞系独立特征,并作为未来针对生产力提高的进一步研究的重要参考。
    The development of gene therapies based on recombinant adeno-associated viruses (rAAVs) has grown exponentially, so the current rAAV manufacturing platform needs to be more efficient to satisfy rising demands. Viral production exerts great demand on cellular substrates, energy, and machinery; therefore, viral production relies heavily on the physiology of the host cell. Transcriptomics, as a mechanism-driven tool, was applied to identify significantly regulated pathways and to study cellular features of the host cell for supporting rAAV production. This study investigated the transcriptomic features of two cell lines cultured in their respective media by comparing viral-producing cultures with non-producing cultures over time in parental human embryonic kidney cells (HEK293). The results demonstrate that the innate immune response signaling pathways of host cells (e.g., RIG-I-like receptor signaling pathway, Toll-like receptor signaling pathway, cytosolic DNA sensing pathway, JAK-STAT signaling pathway) were significantly enriched and upregulated. This was accompanied by the host cellular stress responses, including endoplasmic reticulum stress, autophagy, and apoptosis in viral production. In contrast, fatty acid metabolism and neutral amino acid transport were downregulated in the late phase of viral production. Our transcriptomics analysis reveals the cell-line independent signatures for rAAV production and serves as a significant reference for further studies targeting the productivity improvement in the future.
    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
    重组腺相关病毒(rAAV)是将基因组革命直接转化为药物治疗的最有效的病毒载体技术。然而,rAAV病毒载体的制造在大规模生产中具有低rAAV产量和高成本的上游加工中仍然具有挑战性,限制了基于rAAV的治疗的推广。这种情况可以通过实时监测影响关键质量属性(CQA)的关键过程参数(CPP)来改善。为了实现这一目标,软测量与预测建模相结合是一种重要的策略,可用于通过实时监控关键过程变量来优化rAAV生产的上游过程。然而,开发用于rAAV生产的软传感器作为一种快速和低成本的监测方法并不是一件容易的事。这篇评论文章描述了四个挑战,并批判性地讨论了可以将软传感器应用于rAAV生产监控的可能解决方案。从数据科学家的角度来看,挑战是(i)没有AAV病毒滴度的预测变量(软传感器输入)设置,(二)多步预测,(iii)多个工艺阶段,和(iv)由机械模型组成的软测量开发。
    Recombinant adeno-associated virus (rAAV) is the most effective viral vector technology for directly translating the genomic revolution into medicinal therapies. However, the manufacturing of rAAV viral vectors remains challenging in the upstream processing with low rAAV yield in large-scale production and high cost, limiting the generalization of rAAV-based treatments. This situation can be improved by real-time monitoring of critical process parameters (CPP) that affect critical quality attributes (CQA). To achieve this aim, soft sensing combined with predictive modeling is an important strategy that can be used for optimizing the upstream process of rAAV production by monitoring critical process variables in real time. However, the development of soft sensors for rAAV production as a fast and low-cost monitoring approach is not an easy task. This review article describes four challenges and critically discusses the possible solutions that can enable the application of soft sensors for rAAV production monitoring. The challenges from a data scientist\'s perspective are (i) a predictor variable (soft-sensor inputs) set without AAV viral titer, (ii) multi-step forecasting, (iii) multiple process phases, and (iv) soft-sensor development composed of the mechanistic model.
    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
    Recombinant adeno-associated viruses (rAAVs) are excellent vectors for gene delivery. However, current Sf9/Cap-Rep packaging cell line-dependent OneBac systems still lack versatility and flexibility for large-scale production of rAAVs. In this study, we developed an improved OneBac system that includes a novel dual-function baculovirus expression vector (BEV) termed BEV/Cap-(ITR-GOI) that carries both the AAV Cap gene and rAAV genome inverted terminal repeat (ITR) sequences flanking the gene of interest (GOI), a versatile Sf9-GFP/Rep packaging cell line that harbors silent copies of the AAV2 Rep gene that can be expressed after BEV infection, and constitutively expressed green fluorescent protein (GFP) reporter genes to facilitate cell line screening. The BEV/Cap-(ITR-GOI) construct allows flexibility to switch among different Cap gene serotypes using simple BEV reconstruction, and is stable for at least five serial passages. Furthermore, the Sf9-GFP/Rep stable cell line is versatile for production of different rAAV serotypes. The yield levels for rAAV2, rAAV8, and rAAV9 exceeded 105 vector genomes (VG) per cell, which is similar to other currently available large-scale rAAV production systems. The new Bac system-derived rAAVs have biophysical properties similar to HEK293 cell-derived rAAVs, as well as high quality and activity. In summary, the novel Sf9-GFP/Rep packaging cell line-dependent OneBac system can facilitate large-scale rAAV production and rAAV-based gene therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Current large-scale recombinant adeno-associated virus (rAAV) production systems based on the baculovirus expression vector (BEV) remain complicated and cost-intensive, and they lack versatility and flexibility. Here we present a novel recombinant baculovirus integrated with all packaging elements for the production of rAAV. To optimize BEV construction, ribosome leaky-scanning mechanism was used to express AAV Rep and Cap proteins downstream of the PH and P10 promoters in the pFast.Bac.Dual vector, respectively, and the rAAV genome was inserted between the two promoters. The yields of rAAV2, rAAV8, and rAAV9 derived from the BEV-infected Sf9 cells exceeded 105 vector genomes (VG) per cell. The BEV was shown to be stable and showed no apparent decrease of rAAV yield after at least four serial passages. The rAAVs derived from the new Bac system displayed high-quality and high-transduction activity. Additionally, rAAV2 could be efficiently generated from BEV-infected beet armyworm larvae at a per-larvae yield of 2.75 ± 1.66 × 1010 VG. The rAAV2 derived from larvae showed a structure similar to the rAAV2 derived from HEK293 cells, and it also displayed high-transduction activity. In summary, the novel BEV is ideally suitable for large-scale rAAV production. Further, this study exploits a potential cost-efficient platform for rAAV production in insect larvae.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号