MT: Oligonucleotides: Therapies and Applications

MT : 寡核苷酸 : 治疗和应用
  • 文章类型: Journal Article
    反义寡核苷酸(ASO)是用于不治之症的治疗方式。然而,全身注射gapmer型ASO会导致类相关毒性,包括活化部分凝血活酶时间(aPTT)的延长和血小板减少。我们先前报道,与ASO相比,胆固醇缀合的DNA/RNA异源双链寡核苷酸(Chol-HDOs)表现出显著增强的基因沉默效应,甚至在中枢神经系统中,穿过血脑屏障.在本研究中,我们最初评估了HDO结构对类别相关毒性的影响.HDO结构改善了与ASO相关的类毒性,但在某种程度上仍然存在。作为进一步的解毒剂,我们开发了人工阳离子寡肽,L-2,4-二氨基丁酸低聚物(DabOs),它与HDO的A型双螺旋结构的主沟中的磷酸盐结合。DabO/Chol-HDO复合物在小鼠中显示出显著改善的aPTT延长和血小板减少症,同时维持基因沉默功效。此外,与DabOs结合有效预防脑梗死,在静脉注射高剂量Chol-HDO的小鼠中经常观察到的情况。这些方法,将HDO技术与DabO相结合,在减少毒性方面比传统策略有明显的优势。因此,DabO/HDO复合物是克服与治疗性ASO相关的类毒性的有前途的平台。
    Antisense oligonucleotides (ASOs) are a therapeutic modality for incurable diseases. However, systemic injection of gapmer-type ASOs causes class-related toxicities, including prolongation of activated partial thromboplastin time (aPTT) and thrombocytopenia. We previously reported that cholesterol-conjugated DNA/RNA heteroduplex oligonucleotides (Chol-HDOs) exhibit significantly enhanced gene-silencing effects compared to ASOs, even in the central nervous system, by crossing the blood-brain barrier. In the present study, we initially evaluated the effect of the HDO structure on class-related toxicities. The HDO structure ameliorated the class-related toxicities associated with ASOs, but they remained to some extent. As a further antidote, we have developed artificial cationic oligopeptides, L-2,4-diaminobutanoic acid oligomers (DabOs), which bind to the phosphates in the major groove of the A-type double-helical structure of HDOs. The DabO/Chol-HDO complex showed significantly improved aPTT prolongation and thrombocytopenia in mice while maintaining gene-silencing efficacy. Moreover, the conjugation with DabOs effectively prevented cerebral infarction, a condition frequently observed in mice intravenously injected with high-dose Chol-HDO. These approaches, combining HDO technology with DabOs, offer distinct advantages over conventional strategies in reducing toxicities. Consequently, the DabO/HDO complex represents a promising platform for overcoming the class-related toxicities associated with therapeutic ASOs.
    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
    C9ORF72的内含子之一内的六核苷酸(G4C2)重复扩增(HRE)是肌萎缩性侧索硬化症(ALS)和额颞叶痴呆(FTD)的主要遗传原因。C9ORF72单倍不足,RNA病灶的形成,和二肽重复(DPR)蛋白的产生已被提出作为疾病的机制。这里,我们报道了C9ORF72驱动的ALS/FTD的疾病修饰siRNA的第一个例子。使用来自C9-ALS/FTD小鼠模型的报告基因测定和原代皮质神经元的组合,我们筛选了一组超过150种靶向不同C9ORF72转录变体的完全化学稳定的siRNA.我们证明了在报告分析中siRNA功效与天然环境之间缺乏相关性;发现含有重复序列的C9ORF72mRNA变体优先定位于细胞核,因此C9ORF72mRNA可及性和细胞内定位对功能性RNAi具有主要影响。使用C9-ALS/FTD小鼠模型,我们证明了靶向C9ORF72mRNA变体的二价siRNA特异性或非选择性地降低了C9ORF72mRNA的表达并显着降低了DPR蛋白。有趣的是,沉默所有C9ORF72mRNA转录物的siRNA在去除核内mRNA聚集体方面比仅靶向含有HRE的C9ORF72mRNA转录物更有效。合并,这些数据支持基于RNAi的C9ORF72降解作为潜在的治疗范例.
    A hexanucleotide (G4C2) repeat expansion (HRE) within intron one of C9ORF72 is the leading genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). C9ORF72 haploinsufficiency, formation of RNA foci, and production of dipeptide repeat (DPR) proteins have been proposed as mechanisms of disease. Here, we report the first example of disease-modifying siRNAs for C9ORF72 driven ALS/FTD. Using a combination of reporter assay and primary cortical neurons derived from a C9-ALS/FTD mouse model, we screened a panel of more than 150 fully chemically stabilized siRNAs targeting different C9ORF72 transcriptional variants. We demonstrate the lack of correlation between siRNA efficacy in reporter assay versus native environment; repeat-containing C9ORF72 mRNA variants are found to preferentially localize to the nucleus, and thus C9ORF72 mRNA accessibility and intracellular localization have a dominant impact on functional RNAi. Using a C9-ALS/FTD mouse model, we demonstrate that divalent siRNAs targeting C9ORF72 mRNA variants specifically or non-selectively reduce the expression of C9ORF72 mRNA and significantly reduce DPR proteins. Interestingly, siRNA silencing all C9ORF72 mRNA transcripts was more effective in removing intranuclear mRNA aggregates than targeting only HRE-containing C9ORF72 mRNA transcripts. Combined, these data support RNAi-based degradation of C9ORF72 as a potential therapeutic paradigm.
    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
    CD4+T细胞在mRNA脂质纳米颗粒(LNP)疫苗诱导保护性CD8+T细胞中的作用是未知的。我们使用了缺乏或不缺乏CD4+T细胞的B6或Tlr9-/-小鼠和LNP疫苗,这些疫苗装载了编码异位病毒(ECTV)MHCI类H-2Kb限制性免疫显性CD8+T细胞表位TSYKFESV(TSYKFESVmRNA-LNPs)或ECTVEVM158蛋白的mRNA,其中含有TSYKFESV(EVM-158mRNA-LNP)。在用10μg两种疫苗进行初免和加强后,Kb-TSYKFESV特异性CD8+T细胞在加强(dpb)后29天(两种mRNA-LNP)或90天(EVM158mRNA-LNP)完全保护雄性和雌性小鼠免受ECTV的影响,而与CD4+T细胞无关。然而,在29dpb与1μgmRNA-LNPs,男性的Kb-TSYKFESV特异性CD8+T细胞的频率较低,并且受到保护的程度远低于女性,也独立于CD4+T细胞。在90dpb与1μgEVM158mRNA-LNP,男性和女性中Kb-TSYKFESV特异性CD8+T细胞的频率相似,两者都同样受到ECTV的部分保护,独立于CD4+T细胞。因此,在mRNA-LNP疫苗的最佳或次优剂量下,CD4+T细胞帮助不需要诱导特异性针对显性表位的保护性抗痘病毒CD8+T细胞。在次优剂量下,保护男性需要更多的时间来发展。
    The role of CD4+ T cells in the induction of protective CD8+ T cells by mRNA lipid nanoparticle (LNP) vaccines is unknown. We used B6 or Tlr9 -/- mice depleted or not of CD4+ T cells and LNP vaccines loaded with mRNAs encoding the ectromelia virus (ECTV) MHC class I H-2 Kb-restricted immunodominant CD8+ T cell epitope TSYKFESV (TSYKFESV mRNA-LNPs) or the ECTV EVM158 protein, which contains TSYKFESV (EVM-158 mRNA-LNPs). Following prime and boost with 10 μg of either vaccine, Kb-TSYKFESV-specific CD8+ T cells fully protected male and female mice from ECTV at 29 (both mRNA-LNPs) or 90 days (EVM158 mRNA-LNPs) post boost (dpb) independently of CD4+ T cells. However, at 29 dpb with 1 μg mRNA-LNPs, males had lower frequencies of Kb-TSYKFESV-specific CD8+ T cells and were much less well protected than females from ECTV, also independently of CD4+ T cells. At 90 dpb with 1 μg EVM158 mRNA-LNPs, the frequencies of Kb-TSYKFESV-specific CD8+ T cells in males and females were similar, and both were similarly partially protected from ECTV, independently of CD4+ T cells. Therefore, at optimal or suboptimal doses of mRNA-LNP vaccines, CD4+ T cell help is unnecessary to induce protective anti-poxvirus CD8+ T cells specific to a dominant epitope. At suboptimal doses, protection of males requires more time to develop.
    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
    肌萎缩侧索硬化(ALS),进行性神经退行性疾病,在诊断和治疗方面提出了相当大的挑战。它分为散发性和家族性肌萎缩性侧索硬化症(fALS);后者约占病例的10%,主要以常染色体显性遗传方式遗传。本文综述了fALS的分子遗传学,突出导致其发病机制的关键突变,如SOD1,FUS,C9orf72本文的核心是探索针对这些遗传畸变的反义寡核苷酸(ASO),提供有希望的治疗策略。这篇综述详细概述了fALS的潜在分子机制和ASO的潜在治疗价值。提供治疗神经退行性疾病的新见解。
    Amyotrophic lateral sclerosis (ALS), a progressive neurodegenerative disease, presents considerable challenges in both diagnosis and treatment. It is categorized into sporadic and familial amyotrophic lateral sclerosis (fALS); the latter accounts for approximately 10% of cases and is primarily inherited in an autosomal dominant manner. This review summarizes the molecular genetics of fALS, highlighting key mutations that contribute to its pathogenesis, such as mutations in SOD1, FUS, and C9orf72. Central to this discourse is exploring antisense oligonucleotides (ASOs) that target these genetic aberrations, providing a promising therapeutic strategy. This review provides a detailed overview of the molecular mechanisms underlying fALS and the potential therapeutic value of ASOs, offering new insights into treating neurodegenerative diseases.
    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
    急性髓系白血病(AML)细胞抵抗分化刺激,尽管先天免疫受体的高表达,例如Toll样受体9(TLR9)。我们先前证明了使用TLR9靶向的诱饵寡脱氧核苷酸(CpG-STAT3d)靶向信号转导和转录激活因子3(STAT3)会增加人和小鼠AML细胞的免疫原性。这里,我们阐明了体内由STAT3抑制/TLR9激活驱动的inv(16)AML重编程的分子机制。在转录水平,从静脉内给予CpG-STAT3d或白血病靶向的Stat3沉默和TLR9共刺激后的小鼠分离的AML细胞,显示类似的髓样细胞分化上调(Irf8,Cebpa,Itgam)和抗原呈递(Ciita,Il12a,B2m)相关基因伴随白血病促进Runx1的减少。单细胞转录组学显示CpG-STAT3d诱导AML细胞多向分化为单核细胞/巨噬细胞,红细胞和B细胞亚群。如体内诱导型Irf8沉默所示,IRF8上调对于白血病细胞的单核巨噬细胞分化至关重要。TLR9驱动的AML细胞重编程可能是通过下调STAT3控制的甲基化调节剂而实现的。如DNMT1和DNMT3。事实上,使用阿扎胞苷和CpG寡核苷酸的DNA甲基转移酶(DNMT)抑制的组合模拟CpG-STAT3d效应,导致AML细胞分化,T细胞活化,和系统性白血病消退。这些发现强调了双功能寡核苷酸通过同时抑制STAT3和/或DNMT释放TLR9驱动的白血病细胞分化的免疫治疗潜力。
    Acute myeloid leukemia (AML) cells resist differentiation stimuli despite high expression of innate immune receptors, such as Toll-like receptor 9 (TLR9). We previously demonstrated that targeting Signal Transducer and Activator of Transcription 3 (STAT3) using TLR9-targeted decoy oligodeoxynucleotide (CpG-STAT3d) increases immunogenicity of human and mouse AML cells. Here, we elucidated molecular mechanisms of inv(16) AML reprogramming driven by STAT3-inhibition/TLR9-activation in vivo. At the transcriptional levels, AML cells isolated from mice after intravenous administration of CpG-STAT3d or leukemia-targeted Stat3 silencing and TLR9 co-stimulation, displayed similar upregulation of myeloid cell differentiation (Irf8, Cebpa, Itgam) and antigen-presentation (Ciita, Il12a, B2m)-related genes with concomitant reduction of leukemia-promoting Runx1. Single-cell transcriptomics revealed that CpG-STAT3d induced multilineage differentiation of AML cells into monocytes/macrophages, erythroblastic and B cell subsets. As shown by an inducible Irf8 silencing in vivo, IRF8 upregulation was critical for monocyte-macrophage differentiation of leukemic cells. TLR9-driven AML cell reprogramming was likely enabled by downregulation of STAT3-controlled methylation regulators, such as DNMT1 and DNMT3. In fact, the combination of DNA methyl transferase (DNMT) inhibition using azacitidine with CpG oligonucleotides alone mimicked CpG-STAT3d effects, resulting in AML cell differentiation, T cell activation, and systemic leukemia regression. These findings highlight immunotherapeutic potential of bi-functional oligonucleotides to unleash TLR9-driven differentiation of leukemic cells by concurrent STAT3 and/or DNMT inhibition.
    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
    在黑色素瘤中实施靶向分子疗法和免疫疗法极大地改善了手术干预疗效有限的患者的治疗效果。然而,大部分黑色素瘤患者仍然难以治疗或对这些新的治疗形式产生耐药性,说明需要改进。这里,我们报道,丝裂原活化蛋白(MAP)激酶途径抑制剂dabrafenib和trametinib的临床相关组合可与RIG-I激动剂诱导的免疫疗法协同作用,杀死BRAF突变的人和小鼠黑色素瘤细胞.激酶抑制不损害宿主免疫细胞中激动剂诱导的RIG-I途径的先天性免疫应答。在黑色素瘤移植小鼠模型中,三联疗法优于个别疗法.我们的研究表明,激动剂诱导的RIG-I及其合成配体3pRNA的激活可以极大地改善接受MAP激酶抑制剂的黑色素瘤患者的大部分肿瘤控制。
    The implementation of targeted molecular therapies and immunotherapy in melanoma vastly improved the therapeutic outcome in patients with limited efficacy of surgical intervention. Nevertheless, a large fraction of patients with melanoma still remain refractory or acquire resistance to these new forms of treatment, illustrating a need for improvement. Here, we report that the clinically relevant combination of mitogen-activated protein (MAP) kinase pathway inhibitors dabrafenib and trametinib synergize with RIG-I agonist-induced immunotherapy to kill BRAF-mutated human and mouse melanoma cells. Kinase inhibition did not compromise the agonist-induced innate immune response of the RIG-I pathway in host immune cells. In a melanoma transplantation mouse model, the triple therapy outperformed individual therapies. Our study suggests that agonist-induced activation of RIG-I with its synthetic ligand 3pRNA could vastly improve tumor control in a substantial fraction of patients with melanoma receiving MAP kinase inhibitors.
    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
    过敏性接触性皮炎是一种常见的职业病,治疗选择有限。趋化因子CCL22,趋化因子受体CCR4的配体,指导免疫细胞的迁移。这里,研究表明,CCL22的遗传缺陷有效改善了接触性超敏反应(CHS)的过敏反应,一种常用的过敏性接触性皮炎小鼠模型。对于CCL22的药理学抑制,通过指数富集(SELEX)通过配体的系统进化产生对鼠CCL22特异性的DNA适体。最初选择9个CCL22结合适体并在体外进行功能测试。29-ntDNA适体AJ102.29m极大地抑制了CCL22依赖性T细胞迁移,并且不会引起不期望的Toll样受体依赖性免疫激活。全身应用后,AJ102.29m有效改善了体内CHS。此外,在皮肤上局部施用适体后,CHS相关的过敏症状也减少。用AJ102.29m离体处理的皮肤的显微镜分析表明,适体可以渗透到表皮和真皮中。发现适体AJ102.29m在乳膏中的表皮应用在抑制过敏反应方面与腹膜内注射一样有效,为超出目前全身给药途径的适体治疗用途铺平了道路。
    Allergic contact dermatitis is a prevalent occupational disease with limited therapeutic options. The chemokine CCL22, a ligand of the chemokine receptor CCR4, directs the migration of immune cells. Here, it is shown that genetic deficiency of CCL22 effectively ameliorated allergic reactions in contact hypersensitivity (CHS), a commonly used mouse model of allergic contact dermatitis. For the pharmacological inhibition of CCL22, DNA aptamers specific for murine CCL22 were generated by the systematic evolution of ligands by exponential enrichment (SELEX). Nine CCL22-binding aptamers were initially selected and functionally tested in vitro. The 29-nt DNA aptamer AJ102.29m profoundly inhibited CCL22-dependent T cell migration and did not elicit undesired Toll-like receptor-dependent immune activation. AJ102.29m efficiently ameliorated CHS in vivo after systemic application. Moreover, CHS-associated allergic symptoms were also reduced following topical application of the aptamer on the skin. Microscopic analysis of skin treated with AJ102.29m ex vivo demonstrated that the aptamer could penetrate into the epidermis and dermis. The finding that epicutaneous application of the aptamer AJ102.29m in a cream was as effective in suppressing the allergic reaction as intraperitoneal injection paves the way for therapeutic use of aptamers beyond the current routes of systemic administration.
    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
    青光眼等眼部神经退行性疾病导致进行性视网膜神经节细胞(RGC)丢失,导致不可逆的视力损害。需要神经保护来在衰弱的条件下保护RGC。神经生长因子(NGF)蛋白治疗显示疗效,但与有限的生物利用度和短的半衰期作斗争。在这里,我们探索了一种通过利用基于环状RNA(circularRNA)的治疗来解决这种缺陷的新方法。我们表明,circRNAs表现出延长蛋白质表达的特殊能力,circRNA表达的NGF保护细胞免受葡萄糖剥夺。在小鼠视神经挤压模型中,脂质纳米颗粒(LNP)配制的circNGF玻璃体内给药可保护RGC和轴突免受损伤诱导的变性。它也显著优于NGF蛋白治疗而没有可检测的视网膜毒性。此外,单细胞转录组学显示LNP-circNGF多方面的治疗效果,增强与视觉感知相关的基因,同时减少与创伤相关的变化。这项研究标志着基于circRNA的疗法有望治疗眼部神经退行性疾病,并为其他眼部疾病提供了创新的干预平台。
    Ocular neurodegenerative diseases like glaucoma lead to progressive retinal ganglion cell (RGC) loss, causing irreversible vision impairment. Neuroprotection is needed to preserve RGCs across debilitating conditions. Nerve growth factor (NGF) protein therapy shows efficacy, but struggles with limited bioavailability and a short half-life. Here we explore a novel approach to address this deficiency by utilizing circular RNA (circRNA)-based therapy. We show that circRNAs exhibit an exceptional capacity for prolonged protein expression and circRNA-expressed NGF protects cells from glucose deprivation. In a mouse optic nerve crush model, lipid nanoparticle (LNP)-formulated circNGF administered intravitreally protects RGCs and axons from injury-induced degeneration. It also significantly outperforms NGF protein therapy without detectable retinal toxicity. Furthermore, single-cell transcriptomics revealed LNP-circNGF\'s multifaceted therapeutic effects, enhancing genes related to visual perception while reducing trauma-associated changes. This study signifies the promise of circRNA-based therapies for treating ocular neurodegenerative diseases and provides an innovative intervention platform for other ocular diseases.
    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
    尽管癌症免疫学的最新进展已导致许多免疫疗法的批准,在解决难以治疗的癌症方面进展甚微.在这种情况下,治疗性寡核苷酸,包括干扰RNA,反义寡核苷酸,适体,和DNA酶,与常规化学疗法相比,由于其调节基因表达和蛋白质功能的能力而具有降低的毒性,因此在癌症治疗方法中发挥了核心作用。然而,全身施用裸寡核苷酸面临许多可以通过使用有效递送系统克服的细胞外和细胞内挑战。因此,病毒和非病毒载体可以提高寡核苷酸的稳定性和细胞内摄取,增强肿瘤积累,并增加内体逃逸的可能性,同时将其他不利影响降至最低。因此,更深入地了解各种寡核苷酸的基本作用机制以及裸寡核苷酸给药带来的挑战,本文对寡核苷酸递送系统的最新进展进行了全面综述,并概述了可以影响未来肿瘤治疗的已完成和正在进行的癌症临床试验。
    Although recent advancements in cancer immunology have resulted in the approval of numerous immunotherapies, minimal progress has been observed in addressing hard-to-treat cancers. In this context, therapeutic oligonucleotides, including interfering RNAs, antisense oligonucleotides, aptamers, and DNAzymes, have gained a central role in cancer therapeutic approaches due to their capacity to regulate gene expression and protein function with reduced toxicity compared with conventional chemotherapeutics. Nevertheless, systemic administration of naked oligonucleotides faces many extra- and intracellular challenges that can be overcome by using effective delivery systems. Thus, viral and non-viral carriers can improve oligonucleotide stability and intracellular uptake, enhance tumor accumulation, and increase the probability of endosomal escape while minimizing other adverse effects. Therefore, gaining more insight into fundamental mechanisms of actions of various oligonucleotides and the challenges posed by naked oligonucleotide administration, this article provides a comprehensive review of the recent progress on oligonucleotide delivery systems and an overview of completed and ongoing cancer clinical trials that can shape future oncological treatments.
    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
    亨廷顿氏病(HD)是一种常染色体显性疾病,由HTT基因的一个拷贝中的胞嘧啶-腺嘌呤-鸟嘌呤(CAG)重复序列的扩增引起(突变的HTT,mHTT)。未受影响的HTT基因编码野生型HTT(wtHTT)蛋白,它支持对中枢神经系统的健康和功能至关重要的过程。选择性降低mHTT治疗HD可能比非选择性降低HTT的方法提供益处。因为它旨在保护WTHTT的有益活动。靶向杂合单核苷酸多态性(SNP)(其中靶向变体在mHTT基因上)是实现等位基因选择性活性的一种策略。在这里,我们研究了含硫代磷酸酯(PS)和磷酰基胍(PN)的寡核苷酸是否可以通过靶向rs362273(SNP3)来指导等位基因选择性mHTT降低.我们证明了我们的SNP3靶向分子是有效的,耐用,在小鼠模型中对mHTT的体外和体内选择性。通过与非选择性研究化合物tominersen的替代品进行比较,我们还证明,等位基因选择性分子对mHTT显示出等效的效力,同时保留wtHTT,耐久性得到改善。我们的临床前发现支持WVE-003的进步,WVE-003是一种目前正在临床试验中的研究等位基因选择性化合物(NCT05032196),用于治疗HD患者。
    Huntington\'s disease (HD) is an autosomal dominant disease caused by the expansion of cytosine-adenine-guanine (CAG) repeats in one copy of the HTT gene (mutant HTT, mHTT). The unaffected HTT gene encodes wild-type HTT (wtHTT) protein, which supports processes important for the health and function of the central nervous system. Selective lowering of mHTT for the treatment of HD may provide a benefit over nonselective HTT-lowering approaches, as it aims to preserve the beneficial activities of wtHTT. Targeting a heterozygous single-nucleotide polymorphism (SNP) where the targeted variant is on the mHTT gene is one strategy for achieving allele-selective activity. Herein, we investigated whether stereopure phosphorothioate (PS)- and phosphoryl guanidine (PN)-containing oligonucleotides can direct allele-selective mHTT lowering by targeting rs362273 (SNP3). We demonstrate that our SNP3-targeting molecules are potent, durable, and selective for mHTT in vitro and in vivo in mouse models. Through comparisons with a surrogate for the nonselective investigational compound tominersen, we also demonstrate that allele-selective molecules display equivalent potency toward mHTT with improved durability while sparing wtHTT. Our preclinical findings support the advancement of WVE-003, an investigational allele-selective compound currently in clinical testing (NCT05032196) for the treatment of patients with HD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号