lentiviral vector

慢病毒载体
  • 文章类型: Journal Article
    近年来,基因治疗取得了长足的进步。4000多个蛋白质编码基因与6000多种遗传疾病有关;下一代测序极大地彻底改变了遗传疾病的诊断。大多数遗传疾病被认为是非常罕见的或超罕见的,这里定义为少于1:100,000的案例,但在12种已批准的基因疗法(不包括RNA疗法)中,只有一种针对的是超微病变.本文探讨了三种适用于多种罕见遗传病的基因补充治疗方法:慢病毒载体修饰的自体CD34+造血干细胞移植,将腺相关病毒(AAV)载体全身递送至肝脏,和局部AAV递送到脑脊液和大脑。连同RNA疗法,我们为这些基因疗法提出了一个潜在的商业模式。
    Gene therapy has made considerable strides in recent years. More than 4000 protein-coding genes have been implicated in more than 6000 genetic diseases; next-generation sequencing has dramatically revolutionized the diagnosis of genetic diseases. Most genetic diseases are considered very rare or ultrarare, defined here as having fewer than 1:100,000 cases, but only one of the 12 approved gene therapies (excluding RNA therapies) targets an ultrarare disease. This article explores three gene supplementation therapy approaches suitable for various rare genetic diseases: lentiviral vector-modified autologous CD34+ hematopoietic stem cell transplantation, systemic delivery of adeno-associated virus (AAV) vectors to the liver, and local AAV delivery to the cerebrospinal fluid and brain. Together with RNA therapies, we propose a potential business model for these gene therapies.
    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
    细胞表面和可溶性胞外糖胺聚糖均已显示干扰非病毒基因递送的外源核酸递送效率,包括脂质复合物和多聚复合物介导的转染。商业上和临床试验中使用的大多数基因治疗病毒载体目前是使用基于瞬时转染的生物过程制造的。对病毒载体产品日益增长的需求,加上全球生产能力短缺,需要改进的转染技术和工艺,以最大限度地提高工艺效率和生产率。发现可溶性细胞外糖胺聚糖在悬浮适应的HEK293T细胞培养物的条件细胞培养基中积累,损害转染性能和慢病毒载体生产。特定的酶降解,硫酸软骨素,发现具有软骨素酶ABC的糖胺聚糖显着增强转染性能。此外,我们报道,与对照慢病毒载体生物过程中使用的细胞密度相比,当以更高的细胞密度培养细胞时,功能性慢病毒载体滴度显著改善;当转染前培养物补充软骨素酶ABC时,这种改善进一步增强.与现有方法相比,当转染前将细胞密度加倍时,计算出功能性慢病毒载体滴度增加71.2%,并且用0.1U/mL软骨素酶ABC处理高密度细胞培养物导致滴度进一步增加18.6%。提出了一种能有效提高转染性能的方法。
    Both cell surface and soluble extracellular glycosaminoglycans have been shown to interfere with the exogenous nucleic acid delivery efficiency of non-viral gene delivery, including lipoplex and polyplex-mediated transfection. Most gene therapy viral vectors used commercially and in clinical trials are currently manufactured using transient transfection-based bioprocesses. The growing demand for viral vector products, coupled with a global shortage in production capability, requires improved transfection technologies and processes to maximise process efficiency and productivity. Soluble extracellular glycosaminoglycans were found to accumulate in the conditioned cell culture medium of suspension adapted HEK293T cell cultures, compromising transfection performance and lentiviral vector production. The enzymatic degradation of specific, chondroitin sulphate-based, glycosaminoglycans with chondroitinase ABC was found to significantly enhance transfection performance. Additionally, we report significant improvements in functional lentiviral vector titre when cultivating cells at higher cell densities than those utilised in a control lentiviral vector bioprocess; an improvement that was further enhanced when cultures were supplemented with chondroitinase ABC prior to transfection. A 71.2% increase in functional lentiviral vector titre was calculated when doubling the cell density prior to transfection compared to the existing process and treatment of the high-density cell cultures with 0.1 U/mL chondroitinase ABC resulted in a further 18.6% increase in titre, presenting a method that can effectively enhance transfection performance.
    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
    造血干细胞基因治疗(HSCGT)是治疗神经退行性疾病的一种有前途的治疗策略,代谢紊乱。该方法涉及通过慢病毒介导的转导(TD)将缺失基因离体引入患者自身的干细胞。一旦移植回一个完全有条件的病人,这些基因修饰的HSC可以重新填充血液系统并产生功能性蛋白质,患者先前不存在或无功能,然后可以交叉校正体细胞器官和中枢神经系统中其他受影响的细胞。我们以前开发了一种用于治疗II型粘多糖贮积症(MPSII)(亨特综合征)的HSCGT方法,由艾杜糖醛酸-2-硫酸酯酶(IDS)基因突变引起的衰弱性小儿溶酶体疾病,导致乙酰肝素和硫酸皮肤素的积累,导致严重的神经变性,骨骼异常,和心肺疾病。在HSCGT概念验证研究中,使用与脑靶向肽ApoEII融合的慢病毒IDS(IDS。ApoEII),我们能够使MPSII小鼠的脑病理学和行为正常化。在这里,我们提出了一个优化和验证的良好生产实践造血干细胞TD协议为MPSII准备首次在人的研究。包含TEsLentiBOOST和硫酸鱼精蛋白可将TD效率显着提高至少3倍,而不会引起不良毒性,从而减少所需的矢量数量。
    Hematopoietic stem cell gene therapy (HSCGT) is a promising therapeutic strategy for the treatment of neurodegenerative, metabolic disorders. The approach involves the ex vivo introduction of a missing gene into patients\' own stem cells via lentiviral-mediated transduction (TD). Once transplanted back into a fully conditioned patient, these genetically modified HSCs can repopulate the blood system and produce the functional protein, previously absent or non-functional in the patient, which can then cross-correct other affected cells in somatic organs and the central nervous system. We previously developed an HSCGT approach for the treatment of Mucopolysaccharidosis type II (MPSII) (Hunter syndrome), a debilitating pediatric lysosomal disorder caused by mutations in the iduronate-2-sulphatase (IDS) gene, leading to the accumulation of heparan and dermatan sulfate, which causes severe neurodegeneration, skeletal abnormalities, and cardiorespiratory disease. In HSCGT proof-of-concept studies using lentiviral IDS fused to a brain-targeting peptide ApoEII (IDS.ApoEII), we were able to normalize brain pathology and behavior of MPSII mice. Here we present an optimized and validated good manufacturing practice hematopoietic stem cell TD protocol for MPSII in preparation for first-in-man studies. Inclusion of TEs LentiBOOST and protamine sulfate significantly improved TD efficiency by at least 3-fold without causing adverse toxicity, thereby reducing vector quantity required.
    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
    利用慢病毒载体(LV)的基因递送系统需要高转导效率以成功应用于人类基因治疗。假型可以扩大病毒嗜性,扩大LV的使用范围。虽然水泡性口炎病毒G(VSV-G)单假型LV通常使用,双重假型化的使用频率较低,因为它增加了复杂性。在这项研究中,我们研究了与VSV-G和仙台病毒血凝素-神经氨酸酶(SeV-HN)糖蛋白的表型混合的异源双重假型LV的潜力,称为V/HN-LV。我们的发现表明V/HN-LV在小鼠的各种细胞系中的转导效率显著提高,食蟹猴,和人类与单独使用VSV-G的假型LV相比。值得注意的是,V/HN-LV在人体细胞中显示出更高的转导效率,包括造血干细胞.野生型SeV-HN有效掺入V/HN-LV取决于VSV-G。SeV-HN从VSV-G中去除唾液酸,VSV-G的去唾液酸化增加了V/HN-LV的感染性。此外,V/HN-LV获得了识别唾液酸的能力,特别是宿主细胞上的N-乙酰神经氨酸,增强LV感染性。总的来说,VSV-G和SeV-HN协同提高LV转导效率,拓宽其取向,表明它们在基因传递中的潜在用途。
    A gene delivery system utilizing lentiviral vectors (LVs) requires high transduction efficiency for successful application in human gene therapy. Pseudotyping allows viral tropism to be expanded, widening the usage of LVs. While vesicular stomatitis virus G (VSV-G) single-pseudotyped LVs are commonly used, dual-pseudotyping is less frequently employed because of its increased complexity. In this study, we examined the potential of phenotypically mixed heterologous dual-pseudotyped LVs with VSV-G and Sendai virus hemagglutinin-neuraminidase (SeV-HN) glycoproteins, termed V/HN-LV. Our findings demonstrated the significantly improved transduction efficiency of V/HN-LV in various cell lines of mice, cynomolgus monkeys, and humans compared with LV pseudotyped with VSV-G alone. Notably, V/HN-LV showed higher transduction efficiency in human cells, including hematopoietic stem cells. The efficient incorporation of wild-type SeV-HN into V/HN-LV depended on VSV-G. SeV-HN removed sialic acid from VSV-G, and the desialylation of VSV-G increased V/HN-LV infectivity. Furthermore, V/HN-LV acquired the ability to recognize sialic acid, particularly N-acetylneuraminic acid on the host cell, enhancing LV infectivity. Overall, VSV-G and SeV-HN synergistically improve LV transduction efficiency and broaden its tropism, indicating their potential use in gene delivery.
    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
    心脏移植与主要障碍有关,包括可供移植的器官数量有限,由于遗传差异而导致排斥的风险,和免疫抑制的负担。在这项研究中,我们证明了在离体灌注期间对心脏进行永久性基因工程的可行性。在常温EVHP的两个小时内,将编码靶向β2-微球蛋白(shβ2m)和II类反式激活因子(shCIITA)的短发夹RNA的慢病毒载体输送到移植物。在内皮细胞和心肌细胞中稳定表达的报告基因表明了高效的基因工程。值得注意的是,猪白细胞抗原(SLA)Ⅰ类和SLAⅡ类的表达水平分别下降了66%和76%,分别,在血管内皮。乳酸的评价,肌钙蛋白T,灌注液中的LDH水平和组织学分析显示,没有由慢病毒载体引起的其他细胞损伤或组织损伤。此外,未转导和慢病毒载体转导的心脏的细胞因子分泌谱(IL-6,IL-8和TNF-α)相当。这项研究证明了在不损害组织完整性的情况下离体产生基因工程心脏。SLA表达的下调可能有助于降低心脏的免疫原性并支持同种异体或异种移植后的移植物存活。
    Heart transplantation is associated with major hurdles, including the limited number of available organs for transplantation, the risk of rejection due to genetic discrepancies, and the burden of immunosuppression. In this study, we demonstrated the feasibility of permanent genetic engineering of the heart during ex vivo perfusion. Lentiviral vectors encoding for short hairpin RNAs targeting beta2-microglobulin (shβ2m) and class II transactivator (shCIITA) were delivered to the graft during two hours of normothermic EVHP. Highly efficient genetic engineering was indicated by stable reporter gene expression in endothelial cells and cardiomyocytes. Remarkably, swine leucocyte antigen (SLA) class I and SLA class II expression levels were decreased by 66% and 76%, respectively, in the vascular endothelium. Evaluation of lactate, troponin T, and LDH levels in the perfusate and histological analysis showed no additional cell injury or tissue damage caused by lentiviral vectors. Moreover, cytokine secretion profiles (IL-6, IL-8, and TNF-α) of non-transduced and lentiviral vector-transduced hearts were comparable. This study demonstrated the ex vivo generation of genetically engineered hearts without compromising tissue integrity. Downregulation of SLA expression may contribute to reduce the immunogenicity of the heart and support graft survival after allogeneic or xenogeneic transplantation.
    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
    Diamond-Blackfan贫血(DBA)是一种罕见的遗传性疾病,影响骨髓产生红细胞的能力,导致严重贫血和各种身体异常。大约75%的DBA病例涉及核糖体蛋白(RP)基因的杂合突变,将其归类为核糖体病,RPS19是最常见的突变基因。非RP突变,例如在GATA1中,也已确定。目前的治疗包括糖皮质激素,输血,和造血干细胞移植(HSCT),HSCT是唯一的治疗选择,尽管存在供体可用性和免疫并发症等挑战。基因治疗,特别是使用慢病毒载体和CRISPR/Cas9技术,作为一个有希望的替代方案出现。这篇综述探讨了基因治疗的潜力,重点关注慢病毒载体和CRISPR/Cas9技术与非整合慢病毒载体的结合,作为DBA的治愈解决方案。它突出了DBA治疗领域的变革性进步,为受这种情况影响的个人提供希望。
    Diamond-Blackfan anemia (DBA) is a rare genetic disorder affecting the bone marrow\'s ability to produce red blood cells, leading to severe anemia and various physical abnormalities. Approximately 75% of DBA cases involve heterozygous mutations in ribosomal protein (RP) genes, classifying it as a ribosomopathy, with RPS19 being the most frequently mutated gene. Non-RP mutations, such as in GATA1, have also been identified. Current treatments include glucocorticosteroids, blood transfusions, and hematopoietic stem cell transplantation (HSCT), with HSCT being the only curative option, albeit with challenges like donor availability and immunological complications. Gene therapy, particularly using lentiviral vectors and CRISPR/Cas9 technology, emerges as a promising alternative. This review explores the potential of gene therapy, focusing on lentiviral vectors and CRISPR/Cas9 technology in combination with non-integrating lentiviral vectors, as a curative solution for DBA. It highlights the transformative advancements in the treatment landscape of DBA, offering hope for individuals affected by this condition.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    在8天的过程中,使用连续稳定的生产细胞系,采用准灌注培养来加强慢病毒载体(LV)的制造。初步研究旨在确定可扩展的播种密度,具有3、4和5×104个细胞cm-2,可提供相似的感染性LV比生产率。选择3×104个细胞cm-2的种子,并且调节准灌注以使抑制代谢物积累和在37°C下的载体暴露最小化。在每天1、2和3个血管体积(VVD)时,感染性LV和物理LV的特定生产率相似。选择1个VVD以最小化下游处理量。优化后的工艺放大了50倍,达到1,264cm2烧瓶,达到相似的LV滴度。然而,扩大到6320cm2多层烧瓶,降低了滴度,可能来自次优的气体交换。在25cm2至6,320cm2烧瓶中的三个独立过程中,重现性高,感染和物理LV滴度的变异系数为7.7%±2.9%和11.9%±3.0%,分别。优化的烧瓶工艺成功转移到iCELlisNano(Cytiva)固定床生物反应器中,在1VVD下准灌注产生1.62×108TU。
    Quasi-perfusion culture was employed to intensify lentiviral vector (LV) manufacturing using a continuous stable producer cell line in an 8-day process. Initial studies aimed to identify a scalable seeding density, with 3, 4, and 5 × 104 cells cm-2 providing similar specific productivities of infectious LV. Seeding at 3 × 104 cells cm-2 was selected, and the quasi-perfusion was modulated to minimize inhibitory metabolite accumulation and vector exposure at 37°C. Similar specific productivities of infectious LV and physical LV were achieved at 1, 2, and 3 vessel volumes per day (VVD), with 1 VVD selected to minimize downstream processing volumes. The optimized process was scaled 50-fold to 1,264 cm2 flasks, achieving similar LV titers. However, scaling up beyond this to a 6,320 cm2 multilayer flask reduced titers, possibly from suboptimal gas exchange. Across three independent processes in 25 cm2 to 6,320 cm2 flasks, reproducibility was high with a coefficient of variation of 7.7% ± 2.9% and 11.9% ± 3.0% for infectious and physical LV titers, respectively. The optimized flask process was successfully transferred to the iCELLis Nano (Cytiva) fixed-bed bioreactor, with quasi-perfusion at 1 VVD yielding 1.62 × 108 TU.
    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
    大规模瞬时转染在过去20年中取得了显著进展,能够有效生产各种生物制药产品,包括病毒载体。然而,与转染试剂稳定性和转染复合物制备时间相关的许多挑战仍然存在。需要新的发展和改进的转染技术来确保基于瞬时基因表达的生物过程能够满足对病毒载体日益增长的需求。在本文中,我们证明了阳离子脂质脂质体的生长,在许多基于阳离子脂质的转染过程中,可以通过采用低pH(pH6.40至pH6.75)和低盐浓度(0.2×PBS)配方来控制,促进对纳米颗粒生长动力学的改进控制和增强颗粒稳定性。与标准制备方法相比,此类复合物在延长的时间内保持促进有效转染的能力。由于两个主要原因,这些发现对于大规模制造慢病毒载体具有重要的工业应用。首先,替代制备策略可以使用更长的脂质体孵育时间,在良好的制造实践环境中促进有效的控制。第二,颗粒稳定性的改善有助于设定更宽的工艺操作范围,这将显着提高过程的鲁棒性,并最大限度地提高批次之间的控制和产品的一致性。
    Large-scale transient transfection has advanced significantly over the last 20 years, enabling the effective production of a diverse range of biopharmaceutical products, including viral vectors. However, a number of challenges specifically related to transfection reagent stability and transfection complex preparation times remain. New developments and improved transfection technologies are required to ensure that transient gene expression-based bioprocesses can meet the growing demand for viral vectors. In this paper, we demonstrate that the growth of cationic lipid-based liposomes, an essential step in many cationic lipid-based transfection processes, can be controlled through adoption of low pH (pH 6.40 to pH 6.75) and in low salt concentration (0.2× PBS) formulations, facilitating improved control over the nanoparticle growth kinetics and enhancing particle stability. Such complexes retain the ability to facilitate efficient transfection for prolonged periods compared with standard preparation methodologies. These findings have significant industrial applications for the large-scale manufacture of lentiviral vectors for two principal reasons. First, the alternative preparation strategy enables longer liposome incubation times to be used, facilitating effective control in a good manufacturing practices setting. Second, the improvement in particle stability facilitates the setting of wider process operating ranges, which will significantly improve process robustness and maximise batch-to-batch control and product consistency.
    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
    镰状细胞病(SCD)基因治疗的一个主要限制是获得和获得潜在治愈的一次性治疗。由于治疗费用高。我们已经开发了一个高滴度的双功能慢病毒载体(LVV)在一个载体骨架,具有减少的大小,高载体产量,和高效的基因转移到人CD34+造血干细胞和祖细胞(HSPC)。该LVV包含表达抗镰状化βAS3-珠蛋白基因的基因座控制区核心和两个同时靶向BCL11A和ZNF410转录本的微RNA适应的短发夹RNA,以最大程度地诱导胎儿血红蛋白(HbF)表达。这种LVV诱导高水平的抗镰状血红蛋白(HbAAS3+HbF),同时减少镰状血红蛋白(HbS)。在转导的SCD患者CD34+细胞分化成红细胞时,HbS的降低和抗镰状化血红蛋白的增加阻碍了脱氧HbS聚合和红细胞在每个细胞低载体拷贝下的镰状化。红细胞血红蛋白的双重改变在体内改善了SCDBerkeley小鼠模型中的SCD表型。在低感染复数下具有高滴度和增强的HSPC转导,这种LVV将增加来自生产批次的载体的患者剂量,以降低成本,并有助于改善SCD基因治疗的可及性.
    A major limitation of gene therapy for sickle cell disease (SCD) is the availability and access to a potentially curative one-time treatment, due to high treatment costs. We have developed a high-titer bifunctional lentiviral vector (LVV) in a vector backbone that has reduced size, high vector yields, and efficient gene transfer to human CD34+ hematopoietic stem and progenitor cells (HSPCs). This LVV contains locus control region cores expressing an anti-sickling βAS3-globin gene and two microRNA-adapted short hairpin RNA simultaneously targeting BCL11A and ZNF410 transcripts to maximally induce fetal hemoglobin (HbF) expression. This LVV induces high levels of anti-sickling hemoglobins (HbAAS3 + HbF), while concurrently decreasing sickle hemoglobin (HbS). The decrease in HbS and increased anti-sickling hemoglobin impedes deoxygenated HbS polymerization and red blood cell sickling at low vector copy per cell in transduced SCD patient CD34+ cells differentiated into erythrocytes. The dual alterations in red cell hemoglobins ameliorated the SCD phenotype in the SCD Berkeley mouse model in vivo. With high titer and enhanced transduction of HSPC at a low multiplicity of infection, this LVV will increase the number of patient doses of vector from production lots to decrease costs and help improve accessibility to gene therapy for SCD.
    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
    没有基因编辑的基因沉默对于开发安全的治疗应用具有巨大的潜力。这里,我们描述了一种使用与Krüppel-AssociatedBox抑制域(ZF-Rs)融合的锌指蛋白同时抑制多个基因的新策略。这是通过优化为在造血细胞中递送ZF-R而定制的慢病毒系统来实现的。我们表明,慢病毒骨架的最佳设计对于多达三个ZF-R或两个ZF-R和一个嵌合抗原受体的多路复用至关重要。ZF-R表达对转导细胞的完整性和功能性没有影响。此外,在整个监测期间(长达10周),在体外和体内表达ZF-R的T细胞中的基因抑制是高效的,并伴有表观遗传重塑事件。最后,我们描述了一种提高ZF-R特异性的方法,以说明产生具有安全临床特征的ZF-R的途径.总之,我们成功开发了一种基于表观遗传学的细胞工程方法,用于同时调节多基因表达,绕过与DNA编辑相关的风险.
    Gene silencing without gene editing holds great potential for the development of safe therapeutic applications. Here, we describe a novel strategy to concomitantly repress multiple genes using zinc finger proteins fused to Krüppel-Associated Box repression domains (ZF-Rs). This was achieved via the optimization of a lentiviral system tailored for the delivery of ZF-Rs in hematopoietic cells. We showed that an optimal design of the lentiviral backbone is crucial to multiplex up to three ZF-Rs or two ZF-Rs and a chimeric antigen receptor. ZF-R expression had no impact on the integrity and functionality of transduced cells. Furthermore, gene repression in ZF-R-expressing T cells was highly efficient in vitro and in vivo during the entire monitoring period (up to 10 weeks), and it was accompanied by epigenetic remodeling events. Finally, we described an approach to improve ZF-R specificity to illustrate the path toward the generation of ZF-Rs with a safe clinical profile. In conclusion, we successfully developed an epigenetic-based cell engineering approach for concomitant modulation of multiple gene expressions that bypass the risks associated with DNA editing.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号