lentivirus

慢病毒
  • 文章类型: Journal Article
    囊性纤维化是由囊性纤维化跨膜传导调节因子(CFTR)基因突变引起的遗传性疾病。虽然囊性纤维化是一种多器官疾病,发病和死亡的主要原因与进行性肺病有关。目前对CFTR突变的广谱对CFTR功能的影响的理解已经允许开发CFTR调节剂疗法。尽管这些疗法产生了显著的影响,仍然有很大一部分囊性纤维化患者(估计占全球囊性纤维化人口的10-15%)在遗传上不符合,或者不能容忍,目前CFTR靶向治疗,其治疗需求仍未得到满足。吸入性基因疗法提供了解决囊性纤维化患者未满足的肺部治疗需求的前景。有几种方法,包括基因添加疗法(本综述的重点),基于RNA的疗法,反义寡核苷酸和基因编辑,正在探索。已经研究了各种非病毒和病毒载体用于囊性纤维化基因添加疗法,用于肺中CFTR功能的突变不可知恢复。慢病毒载体提供了高效和持久的基因表达的前景,以及安全的潜力,与其他常用的病毒载体相比,有效地重新给药。用仙台病毒F和HN包膜蛋白(rSIV。F/HN)已被开发用于治疗囊性纤维化。有希望的临床前结果支持这种携带全长CFTR转基因(BI3720931)的载体进展为预计于2024年开始的首次人体临床试验。
    Cystic fibrosis is a genetic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. While cystic fibrosis is a multi-organ disease, the leading causes of morbidity and mortality are related to progressive lung disease. Current understanding of the effects of the broad spectrum of CFTR mutations on CFTR function has allowed for the development of CFTR modulator therapies. Despite the remarkable impact that these therapies have had, there remains a significant proportion of people with cystic fibrosis (estimated at 10-15% of the global cystic fibrosis population) who are genetically ineligible for, or intolerant to, current CFTR-targeting therapies and whose therapeutic needs remain unmet. Inhaled genetic therapies offer the prospect of addressing the unmet pulmonary treatment need in people with cystic fibrosis, with several approaches, including gene addition therapy (the focus of this review), RNA-based therapies, antisense oligonucleotides and gene editing, being explored. Various non-viral and viral vectors have been investigated for cystic fibrosis gene addition therapy for mutation-agnostic restoration of CFTR function in the lungs. Lentiviral vectors offer the prospect of highly efficient and long-lasting gene expression, and the potential to be safely and, in contrast to other commonly used viral vectors, effectively re-dosed. A third-generation lentiviral vector pseudotyped with Sendai virus F and HN envelope proteins (rSIV.F/HN) has been developed for the treatment of cystic fibrosis. Promising preclinical results support the progression of this vector carrying a full-length CFTR transgene (BI 3720931) into a first-in-human clinical trial expected to begin in 2024.
    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
    蝙蝠以其特殊的特性在哺乳动物物种中脱颖而出,包括飞行和回声定位的导航能力,通过紧张/冬眠来节约能源,藏有大量的病毒,表现出对疾病的抵抗力,在恶劣的环境条件下生存,与其他类似大小的哺乳动物相比,表现出非凡的寿命。由于几个原因,蝙蝠的体内研究具有挑战性,例如在自然环境中难以定位和捕获它们,有限的可访问性,样本量低,环境变化,长寿,繁殖率缓慢,人畜共患疾病的风险,物种保护,和伦理问题。因此,建立替代实验室模型对于研究蝙蝠中观察到的各种生理适应至关重要。从组织中获得优质细胞是成功的原代细胞衍生的关键的第一步。然而,由于分离和扩增细胞所需的资源,收集新鲜组织并立即处理样品用于细胞培养通常是不切实际的。因此,冷冻组织通常是蝙蝠原代细胞衍生的起始资源,但是冷冻组织中的细胞通常会受损,并且完整性和生存力较低。因此,从冷冻组织中分离原代细胞提出了重大挑战。在这里,我们提出了一种成功开发的方案,用于从冷冻蝙蝠翼活检中分离原代真皮成纤维细胞。这个协议标志着一个重要的里程碑,因为这是第一个专门针对从蝙蝠冷冻组织中分离成纤维细胞的方案。我们还描述了原代细胞表征的方法,通过慢病毒转导对原代细胞进行遗传操作,和稳定细胞系的发展。©2024Wiley期刊有限责任公司。基本方案1:蝙蝠翼活检采集和保存支持方案1:蝙蝠静脉穿刺采血基本方案2:从成年蝙蝠冷冻翼活检中分离原代成纤维细胞支持方案2:原代成纤维细胞培养和继代培养支持方案3:生长曲线的测定和倍增时间支持方案4:原代成纤维细胞的细胞建库和解冻基本方案3:蝙蝠原代成纤维细胞的慢病毒转导基本方案4:蝙蝠稳定的成纤维细胞线发育支持方案5:通过成纤维细胞
    Bats stand out among mammalian species for their exceptional traits, including the capacity to navigate through flight and echolocation, conserve energy through torpor/hibernation, harbor a multitude of viruses, exhibit resistance to disease, survive harsh environmental conditions, and demonstrate exceptional longevity compared to other mammals of similar size. In vivo studies of bats are challenging for several reasons, such as difficulty in locating and capturing them in their natural environments, limited accessibility, low sample size, environmental variation, long lifespans, slow reproductive rates, zoonotic disease risks, species protection, and ethical concerns. Thus, establishing alternative laboratory models is crucial for investigating the diverse physiological adaptations observed in bats. Obtaining quality cells from tissues is a critical first step for successful primary cell derivation. However, it is often impractical to collect fresh tissue and process the samples immediately for cell culture due to the resources required for isolating and expanding cells. As a result, frozen tissue is typically the starting resource for bat primary cell derivation, but cells in frozen tissue are usually damaged and have low integrity and viability. Isolating primary cells from frozen tissues thus poses a significant challenge. Herein, we present a successfully developed protocol for isolating primary dermal fibroblasts from frozen bat wing biopsies. This protocol marks a significant milestone, as this is the first protocol specifically focused on fibroblast isolation from bat frozen tissue. We also describe methods for primary cell characterization, genetic manipulation of primary cells through lentivirus transduction, and the development of stable cell lines. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Bat wing biopsy collection and preservation Support Protocol 1: Blood collection from bat venipuncture Basic Protocol 2: Isolation of primary fibroblasts from adult bat frozen wing biopsy Support Protocol 2: Primary fibroblast culture and subculture Support Protocol 3: Determination of growth curve and doubling time Support Protocol 4: Cell banking and thawing of primary fibroblasts Basic Protocol 3: Lentiviral transduction of bat primary fibroblasts Basic Protocol 4: Bat stable fibroblast cell line development Support Protocol 5: Bat fibroblast validation by immunofluorescence staining Basic Protocol 5: Chromosome counting.
    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
    在研究将基因递送载体靶向特定细胞类型的方法时,我们研究了使用针对SARS-CoV-2刺突蛋白受体结合域的纳米抗体来指导刺突表达细胞的慢病毒感染的潜力。使用四种不同的方法,我们发现具有表面暴露的纳米抗体结构域的慢病毒选择性感染表达Spike的细胞.靶向依赖于Spike蛋白的融合功能,并且符合一个模型,在该模型中,纳米抗体与Spike蛋白结合会触发Spike融合机制。纳米抗体-刺突相互作用还能够指导细胞-细胞融合和通过刺突假型慢病毒载体选择性感染表达纳米抗体的细胞。重要的是,感染SARS-CoV-2的细胞被用嵌合纳米抗体蛋白假型的慢病毒载体有效和选择性地感染。我们的结果表明,通过使用适当的纳米抗体或病毒受体模拟物,可以将任何形成合胞体的病毒感染的细胞靶向用于基因递送。以这种方式修饰的载体可证明可用于将免疫调节剂递送至感染病灶以减轻病毒感染的影响。重要意义我们已经发现,用针对SARS-CoV-2刺突蛋白的纳米抗体修饰的慢病毒在其表面上选择性地感染表达刺突的细胞。感染依赖于纳米抗体的特异性和Spike蛋白的融合功能,符合反向融合模型,其中纳米抗体与Spike结合会触发Spike融合机制。纳米抗体-刺突相互作用还可以驱动细胞-细胞融合和携带刺突蛋白的病毒感染表达纳米抗体的细胞。重要的是,感染SARS-CoV-2的细胞被纳米抗体修饰的慢病毒选择性感染。这些结果表明,被表达活性受体结合融合蛋白的任何病毒感染的细胞可以被载体靶向,用于递送货物以减轻感染。
    While investigating methods to target gene delivery vectors to specific cell types, we examined the potential of using a nanobody against the SARS-CoV-2 Spike protein receptor-binding domain to direct lentivirus infection of Spike-expressing cells. Using four different approaches, we found that lentiviruses with surface-exposed nanobody domains selectively infect Spike-expressing cells. Targeting is dependent on the fusion function of the Spike protein, and conforms to a model in which nanobody binding to the Spike protein triggers the Spike fusion machinery. The nanobody-Spike interaction also is capable of directing cell-cell fusion and the selective infection of nanobody-expressing cells by Spike-pseudotyped lentivirus vectors. Significantly, cells infected with SARS-CoV-2 are efficiently and selectively infected by lentivirus vectors pseudotyped with a chimeric nanobody protein. Our results suggest that cells infected by any virus that forms syncytia may be targeted for gene delivery by using an appropriate nanobody or virus receptor mimic. Vectors modified in this fashion may prove useful in the delivery of immunomodulators to infected foci to mitigate the effects of viral infections.IMPORTANCEWe have discovered that lentiviruses decorated on their surfaces with a nanobody against the SARS-CoV-2 Spike protein selectively infect Spike-expressing cells. Infection is dependent on the specificity of the nanobody and the fusion function of the Spike protein and conforms to a reverse fusion model, in which nanobody binding to Spike triggers the Spike fusion machinery. The nanobody-Spike interaction also can drive cell-cell fusion and infection of nanobody-expressing cells with viruses carrying the Spike protein. Importantly, cells infected with SARS-CoV-2 are selectively infected with nanobody-decorated lentiviruses. These results suggest that cells infected by any virus that expresses an active receptor-binding fusion protein may be targeted by vectors for delivery of cargoes to mitigate infections.
    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
    在慢病毒载体(LVV)生产过程中生产细胞的转导导致70-90%的活颗粒损失。这个过程被称为反向转导,它是LVV包膜蛋白之间相互作用的结果,VSV-G,LDL受体位于生产细胞膜上,允许慢病毒载体转导。避免LVV制造中的逆向转换对于提高净产量至关重要,因此,生产过程的效率。这里,我们描述了一种定量生产细胞转导的方法和三种不同的策略,专注于VSV-G和LDLR之间的相互作用,旨在减少反向转导。
    Transduction of producer cells during lentiviral vector (LVV) production causes the loss of 70-90% of viable particles. This process is called retro-transduction and it is a consequence of the interaction between the LVV envelope protein, VSV-G, and the LDL receptor located on the producer cell membrane, allowing lentiviral vector transduction. Avoiding retro-transduction in LVV manufacturing is crucial to improve net production and, therefore, the efficiency of the production process. Here, we describe a method for quantifying the transduction of producer cells and three different strategies that, focused on the interaction between VSV-G and the LDLR, aim to reduce retro-transduction.
    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
    CRISPR介导的文库筛选的应用通过揭示病毒-宿主相互作用的复杂性从根本上改变了功能基因组学。该方案描述了使用CRISPR介导的文库筛选来鉴定调节对PEDV感染的先天免疫应答的关键功能基因。我们详细介绍了一个循序渐进的过程,从设计和构建靶向涉及先天免疫的基因的定制CRISPR敲除文库开始,到使用慢病毒载体将这些构建体有效递送到细胞中。随后,我们概述了病毒攻击后识别功能基因的过程,包括使用下一代测序(NGS),分析和鉴定对感染反应改变的敲除细胞。这种综合方法为免疫学和病毒学研究人员提供了资源和强大的框架,用于揭示宿主-病原体相互作用的遗传基础以及先天免疫系统对病毒入侵的武器库。
    The application of CRISPR-mediated library screening has fundamentally transformed functional genomics by revealing the complexity of virus-host interactions. This protocol describes the use of CRISPR-mediated library screening to identify key functional genes regulating the innate immune response to PEDV infection. We detail a step-by-step process, starting from the design and construction of a customized CRISPR knockout library targeting genes involved in innate immunity to the effective delivery of these constructs into cells using lentiviral vectors. Subsequently, we outline the process of identifying functional genes postviral attack, including the use of next-generation sequencing (NGS), to analyze and identify knockout cells that exhibit altered responses to infection. This integrated approach provides researchers in immunology and virology with a resource and a robust framework for uncovering the genetic basis of host-pathogen interactions and the arsenal of the innate immune system against viral invasions.
    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
    Yes相关蛋白1(YAP1)和PDZ结合基序(TAZ)的转录共激活因子的激活与再生和肿瘤发生有关,因此代表了组织稳态的双刃剑。然而,YAP1/TAZ的活性如何被调节或在这些过程中导致其失调的原因仍然未知.探讨上游刺激对YAP1/TAZ细胞活性的调节,我们开发了一种高度敏感的YAP1/TAZ/TEAD响应性DNA元件(YRE),并将其整合到基于慢病毒的报告细胞系统中,以实现对体外荧光素酶活性和体内金星荧光方面的YAP1/TAZ内源性活性的敏感和特异性监测。此外,通过用TCF和NF-κB结合DNA元件代替YRE,我们证明了该报告系统对其他途径的适用性,例如Wnt/β-catenin/TCF-和IL-1β/NF-κB介导的信号传导,分别。通过对由364种已知抑制剂组成的化学化合物库进行基于细胞的报告子筛选来评估该系统的实用性,使用能够定量YAP1/TAZ和β-catenin介导的转录活性的报告子导入细胞,这导致了多种抑制剂的鉴定,包括先前已知的以及这些信号通路的新型调节剂。我们进一步证实了新的YAP1/TAZ调节剂,如钾离子载体,Janus激酶抑制剂,血小板衍生生长因子受体抑制剂,和遗传毒性应激诱导剂,改变内源性YAP1/TAZ的蛋白质水平或磷酸化及其靶基因的表达。因此,这种报告系统提供了一个强大的工具来监测感兴趣的内源性信号活动(甚至在活细胞中),并在各种细胞环境中搜索调节剂。
    The activation of yes-associated protein 1 (YAP1) and transcriptional co-activator with PDZ-binding motif (TAZ) has been implicated in both regeneration and tumorigenesis, thus representing a double-edged sword in tissue homeostasis. However, how the activity of YAP1/TAZ is regulated or what leads to its dysregulation in these processes remains unknown. To explore the upstream stimuli modulating the cellular activity of YAP1/TAZ, we developed a highly sensitive YAP1/TAZ/TEAD-responsive DNA element (YRE) and incorporated it into a lentivirus-based reporter cell system to allow for sensitive and specific monitoring of the endogenous activity of YAP1/TAZ in terms of luciferase activity in vitro and Venus fluorescence in vivo. Furthermore, by replacing YRE with TCF- and NF-κB-binding DNA elements, we demonstrated the applicability of this reporter system to other pathways such as Wnt/β-catenin/TCF- and IL-1β/NF-κB-mediated signaling, respectively. The practicality of this system was evaluated by performing cell-based reporter screening of a chemical compound library consisting of 364 known inhibitors, using reporter-introduced cells capable of quantifying YAP1/TAZ- and β-catenin-mediated transcription activities, which led to the identification of multiple inhibitors, including previously known as well as novel modulators of these signaling pathways. We further confirmed that novel YAP1/TAZ modulators, such as potassium ionophores, Janus kinase inhibitors, platelet-derived growth factor receptor inhibitors, and genotoxic stress inducers, alter the protein level or phosphorylation of endogenous YAP1/TAZ and the expression of their target genes. Thus, this reporter system provides a powerful tool to monitor endogenous signaling activities of interest (even in living cells) and search for modulators in various cellular contexts.
    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
    下一代CRISPR/Cas基因编辑工具的发明,像基地和主要编辑,用于纠正导致疾病的基因变异,为患者体内使用创造了希望,从而导致更广泛的临床翻译。为了实现这种潜力,能够将基因编辑工具套件安全有效地运送到特定细胞群体或组织中的运载工具非常需要。这里,我们描述了包膜逆转录病毒衍生颗粒作为由Cas9衍生的编辑蛋白和单向导RNA组成的“现成工作”核糖核蛋白复合物的载体的发展。我们提出了使病毒适应细胞靶向蛋白质递送的论点,并描述了十年开发期后的状态。已经在原代细胞中显示出有效的编辑,包括T细胞和造血干细胞,在体内靶向的组织中,包括老鼠的视网膜,肝脏,和大脑。新的证据表明,工程病毒衍生的纳米颗粒可以容纳基础和主要编辑,并且似乎使发芽希望,这种颗粒可以进一步开发并大规模生产以用于治疗应用。
    The invention of next-generation CRISPR/Cas gene editing tools, like base and prime editing, for correction of gene variants causing disease, has created hope for in vivo use in patients leading to wider clinical translation. To realize this potential, delivery vehicles that can ferry gene editing tool kits safely and effectively into specific cell populations or tissues are in great demand. In this review, we describe the development of enveloped retrovirus-derived particles as carriers of \"ready-to-work\" ribonucleoprotein complexes consisting of Cas9-derived editor proteins and single guide RNAs. We present arguments for adapting viruses for cell-targeted protein delivery and describe the status after a decade-long development period, which has already shown effective editing in primary cells, including T cells and hematopoietic stem cells, and in tissues targeted in vivo, including mouse retina, liver, and brain. Emerging evidence has demonstrated that engineered virus-derived nanoparticles can accommodate both base and prime editors and seems to fertilize a sprouting hope that such particles can be further developed and produced in large scale for therapeutic applications.
    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
    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
    该协议的目的是提供一个全面的,使用活细胞mt-KEIMA方法评估线粒体自噬通量的逐步指南。拟议的协议是敏感的,可重复,定量,并且易于执行。虽然线粒体自噬已经被广泛研究,目前的方法主要集中在终端测量,忽略了这个过程的动态方面。因此,这种直接的活细胞线粒体自噬追踪方案的引入使得能够实时监测线粒体选择性自噬的动力学,从而增强了就关键监管机构和过程的可逆性得出结论的能力。该测定采用慢病毒方法在原代或永生化细胞系中诱导mt-KEIMA表达。随后,使用活细胞成像系统以特定的时间间隔观察各自的线粒体自噬报告细胞,并且进一步定量允许检测线粒体自噬通量。该协议已被证明在研究线粒体自噬通量有效,包括随着时间的推移对化学诱导剂或基因修饰细胞的反应。值得注意的是,这种方法非常适合高通量筛选可能影响细胞中线粒体自噬反应的化学物质或适当的基因编辑文库.
    The purpose of this protocol is to provide a comprehensive, stepwise guide for assessing mitophagy flux utilizing a live-cell mt-KEIMA approach. The proposed protocol is sensitive, reproducible, quantitative, and easy to perform. While mitophagy has been extensively studied, current methodologies primarily focus on terminal measurements, neglecting the dynamic aspect of this process. Hence, the introduction of this straightforward live-cell mitophagy tracing protocol enables real-time monitoring of the dynamics of mitochondrial selective autophagy, thereby enhancing the ability to draw conclusions regarding key regulators and the reversibility of the process. The assay employs a lentiviral approach to induce mt-KEIMA expression in primary or immortalized cell lines. Subsequently, the respective mitophagy reporter cells are observed using a live-cell imaging system at specific time intervals, and further quantification allows the detection of mitophagy flux. This protocol has proven efficacious in investigating mitophagy flux, including responses to chemical inducers or genetically modified cells over time. Notably, this approach is well-suited for large throughput screening of chemicals or appropriate gene-editing libraries that may influence mitophagy responses in 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
    一名因ABCD1全基因缺失而患有肾上腺脑白质营养不良的9岁男孩被诊断为活动性脑肾上腺脑白质营养不良,其特征是脑MRI脱髓鞘和a增强。他接受了造血细胞移植(HCT),使用表达ABCD1的慢病毒载体转导的自体CD34细胞(eli-cel,elivaldogeneautotemcel)作为ALD-104临床试验的一部分。HCT后50天,患者的MRI显示钆分辨率;全血载体拷贝数(VCN)为0.666拷贝/mL。HCT后六个月,MRI显示钆增强的重新出现;VCN已降至0.029拷贝/mL。移植后9个月可检测到ABCD1基因产物的多克隆抗体,显示对过氧化物酶体的反应性,暗示了一种免疫反应,然而,未显示与人CD34+细胞结合的抗体。患者在基因治疗后12个月接受了一次成功的同种异体HCT,结果钆消退,脑疾病稳定,以及抗体的消失.同时VCN损失和ABCD1基因产物抗体的出现是令人感兴趣的。我们推测这与患者的整个ABCD1基因缺失有关。我们建议密切监测正在考虑基因治疗的完全缺失患者由于免疫反应而导致的基因治疗功效丧失。
    A 9-year-old boy with adrenoleukodystrophy due to ABCD1 whole-gene deletion was diagnosed with active cerebral adrenoleukodystrophy characterized by demyelination and gadolinium enhancement on brain MRI. He underwent hematopoietic cell transplant (HCT) with autologous CD34+ cells transduced with an ABCD1-expressing lentiviral vector (eli-cel [elivaldogene autotemcel]) as part of the ALD-104 clinical trial. Fifty days after HCT, the patient\'s MRI showed gadolinium resolution; the whole-blood vector copy number (VCN) was 0.666 copies/mL. Six months following HCT, an MRI showed re-emergence of gadolinium enhancement; the VCN had decreased to 0.029 copies/mL. Polyclonal antibodies to the ABCD1 gene product were detectable 9 months after transplant, showing reactivity to peroxisomes, suggesting an immune response; however, no antibody binding to human CD34+ cells could be shown. The patient underwent a successful allogeneic HCT 12 months after gene therapy with resultant gadolinium resolution, cerebral disease stabilization, and the disappearance of antibodies. The coincident VCN loss and appearance of antibody to the ABCD1 gene product is of interest, and we postulate that it is related to the patient\'s whole ABCD1 gene deletion. We suggest close monitoring of loss of gene therapy efficacy due to immune response in patients with full deletions who are considering gene therapy.
    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