hepcludex

Hepcludex
  • 文章类型: Journal Article
    目标:丁维肽(BLV),一流的进入抑制剂,在欧洲被批准用于治疗慢性丁型肝炎(CHD)。BLV单药治疗优于延迟治疗(W)48周,主要疗效终点,在MYR301研究(NCT03852719)中。这里,我们评估了在W96之前继续BLV治疗是否会改善病毒学和生化应答率,特别是在W24时未达到病毒学应答的患者中.
    方法:在此过程中,开放标签,随机3期研究,CHD患者(N=150)随机(1:1:1)接受BLV2(n=49)或10mg/天(n=50)治疗,每个144周,或延迟治疗48周,然后BLV10mg/天,持续96周(n=51)。联合反应被定义为检测不到丁型肝炎病毒(HDV)RNA或HDVRNA减少≥2log10IU/mL从基线和丙氨酸转氨酶(ALT)正常化。其他终点包括病毒学应答,ALT正常化,和HDVRNA的变化。
    结果:在150名患者中,143(95%)完成了96周的研讨。W48和W96之间的疗效反应得到维持和/或改善,相似的组合,病毒学,生化反应率在BLV2和10mg之间。在W24时具有次优的早期病毒学应答的患者中,43%的无应答者和82%的部分应答者在W96时实现病毒学应答。生化改善通常与病毒学反应无关。不良事件(AE)大多为轻度,无与BLV相关的严重不良事件。
    结论:长期BLV治疗可维持和/或增加病毒学和生化反应,包括那些早期病毒学应答欠佳的患者。通过W96,BLV单药治疗冠心病是安全且耐受性良好的。
    根据长达48周治疗的临床研究结果,2023年7月,丁维肽被完全批准用于欧洲慢性丁型肝炎(CHD)的治疗。从长远来看,了解bulevirtide的疗效和安全性对医疗保健提供者很重要。在这个分析中,我们证明,在CHD患者中,bulevirtide单药治疗96周与联合治疗的持续改善相关,病毒学,和生化反应以及肝脏僵硬度从第48周在2-mg和10-mg的剂量。在第24周时对bulevirtide的病毒学反应欠佳的患者也受益于持续治疗,大多数人在第96周达到病毒学应答或生化改善。
    结果:
    NCT03852719。
    OBJECTIVE: Bulevirtide (BLV), a first-in-class entry inhibitor, is approved in Europe for the treatment of chronic hepatitis delta (CHD). BLV monotherapy was superior to delayed treatment at week (W) 48, the primary efficacy endpoint, in the MYR301 study (NCT03852719). Here, we assessed if continued BLV therapy until W96 would improve virologic and biochemical response rates, particularly among patients who did not achieve virologic response at W24.
    METHODS: In this ongoing, open-label, randomized phase III study, patients with CHD (N = 150) were randomized (1:1:1) to treatment with BLV 2 mg/day (n = 49) or 10 mg/day (n = 50), each for 144 weeks, or to delayed treatment for 48 weeks followed by BLV 10 mg/day for 96 weeks (n = 51). Combined response was defined as undetectable hepatitis delta virus (HDV) RNA or a decrease in HDV RNA by ≥2 log10 IU/ml from baseline and alanine aminotransferase (ALT) normalization. Other endpoints included virologic response, ALT normalization, and change in HDV RNA.
    RESULTS: Of 150 patients, 143 (95%) completed 96 weeks of the study. Efficacy responses were maintained and/or improved between W48 and W96, with similar combined, virologic, and biochemical response rates between BLV 2 and 10 mg. Of the patients with a suboptimal early virologic response at W24, 43% of non-responders and 82% of partial responders achieved virologic response at W96. Biochemical improvement often occurred independently of virologic response. Adverse events were mostly mild, with no serious adverse events related to BLV.
    CONCLUSIONS: Virologic and biochemical responses were maintained and/or increased with longer term BLV therapy, including in those with suboptimal early virologic response. BLV monotherapy for CHD was safe and well tolerated through W96.
    UNASSIGNED: In July 2023, bulevirtide was fully approved for the treatment of chronic hepatitis delta (CHD) in Europe based on clinical study results from up to 48 weeks of treatment. Understanding the efficacy and safety of bulevirtide over the longer term is important for healthcare providers. In this analysis, we demonstrate that bulevirtide monotherapy for 96 weeks in patients with CHD was associated with continued improvements in combined, virologic, and biochemical responses as well as liver stiffness from week 48 at both the 2 mg and 10 mg doses. Patients with suboptimal virologic responses to bulevirtide at week 24 also benefited from continued therapy, with the majority achieving virologic response or biochemical improvement by week 96.
    RESULTS:
    UNASSIGNED: NCT03852719.
    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:Bulevirtide(BLV)是一流的进入抑制剂,也是欧洲唯一被批准用于慢性感染HDV患者的治疗方法。我们旨在研究BLV治疗在基线和治疗24或48周后获得的配对肝活检的疗效。
    方法:我们对来自3项临床试验的126个配对肝活检进行了联合分析。在2期临床试验MYR202中,慢性丁型肝炎患者随机接受2毫克的24周BLV,5mg或10mg/天。MYR203(2期)和MYR301(3期)患者接受48周的2mg或10mg/天BLV治疗。富马酸替诺福韦酯单药治疗或延迟治疗作为比较。通过qPCR和免疫组织化学评估病毒学参数和感染相关的宿主基因。
    结果:在第24周,中位肝内HDVRNA从基线下降为0.9Log10,2mg(n=7),1.1Log10与5mg(n=5)和1.4Log10与10mg(n=7)。在第48周,平均减少量为2.2Log10(2mg)(n=27)和2.7Log10(10mg)(n=37),而HDVRNA水平在比较臂中没有变化。值得注意的是,在所有BLV治疗组中,丁型肝炎抗原阳性肝细胞数量显著下降,同时炎性趋化因子和干扰素刺激基因的转录水平也随之下降.尽管HBsAg阳性肝细胞的丰度,辅助病毒HBV的复制和共价闭合环状DNA(cccDNA)水平较低,并且不受BLV治疗的影响。
    结论:阻断病毒进入减少肝脏炎症的迹象,并促进肝脏内HDV感染的强烈减少,因此提示部分患者可通过长期治疗达到HDV治愈。
    丁型肝炎病毒的慢性感染是最严重的病毒性肝炎,影响全世界约1200万人。进入抑制剂bulevirtide(BLV)是最近批准的唯一抗HDV药物,在临床试验和真实数据中已证明是有效和安全的。这里,我们调查了配对肝活检在基线和24或48周的治疗后,从三个临床试验,以了解药物对肝脏中的病毒和宿主参数的影响,病毒复制的位点。我们发现BLV治疗大大减少了HDV感染细胞的数量和肝脏炎症的迹象。该数据暗示阻断病毒进入改善肝脏炎症,并且延长治疗方案可能导致一些患者的HDV治愈。这一概念将为CHD患者的治疗策略和联合治疗的进一步发展提供信息。
    OBJECTIVE: Bulevirtide (BLV) is a first-in-class entry inhibitor and the only approved treatment for patients chronically infected with HDV in Europe. We aimed to investigate the efficacy of BLV treatment in paired liver biopsies obtained at baseline and after 24 or 48 weeks of treatment.
    METHODS: We performed a combined analysis of 126 paired liver biopsies derived from three clinical trials. In the phase II clinical trial MYR202, patients with chronic hepatitis D were randomised to receive 24 weeks of BLV at 2 mg, 5 mg or 10 mg/day. Patients in MYR203 (phase II) and MYR301 (phase III) received 48 weeks of BLV at 2 mg or 10 mg/day. Tenofovir disoproxil fumarate monotherapy or delayed treatment served as comparators. Virological parameters and infection-related host genes were assessed by qPCR and immunohistochemistry.
    RESULTS: At week 24, median intrahepatic HDV RNA decline from baseline was 0.9Log10 with 2 mg (n = 7), 1.1Log10 with 5 mg (n = 5) and 1.4 Log10 with 10 mg (n = 7) of BLV. At week 48, median reductions were 2.2Log10 with 2 mg (n = 27) and 2.7Log10 with 10 mg (n = 37) of BLV, while HDV RNA levels did not change in the comparator arms. Notably, a drastic decline in the number of hepatitis delta antigen-positive hepatocytes and a concomitant decrease in transcriptional levels of inflammatory chemokines and interferon-stimulated genes was determined in all BLV-treatment arms. Despite the abundance of HBsAg-positive hepatocytes, replication and covalently closed circular DNA levels of the helper virus HBV were low and remained unaffected by BLV treatment.
    CONCLUSIONS: Blocking viral entry diminishes signs of liver inflammation and promotes a strong reduction of HDV infection within the liver, thus suggesting that some patients may achieve HDV cure with long-term treatment.
    UNASSIGNED: Chronic infection with HDV causes the most severe form of viral hepatitis, affecting approximately 12 million people worldwide. The entry inhibitor bulevirtide (BLV) is the only recently approved anti-HDV drug, which has proven efficacious and safe in clinical trials and real-word data. Here, we investigated paired liver biopsies at baseline and after 24 or 48 weeks of treatment from three clinical trials to understand the effect of the drug on viral and host parameters in the liver, the site of viral replication. We found that BLV treatment strongly reduces the number of HDV-infected cells and signs of liver inflammation. This data implies that blocking viral entry ameliorates liver inflammation and that prolonged treatment regimens might lead to HDV cure in some patients. This concept will guide the further development of therapeutic strategies and combination treatments for patients with CHD.
    BACKGROUND: NCT03546621, NCT02888106, NCT03852719.
    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
    全球约有2.5亿人长期感染乙型肝炎病毒,使他们容易感染丁型肝炎病毒。重复感染导致病毒性肝炎的最严重形式,从而使疾病的进程急剧恶化。直到最近,唯一可用的治疗包括干扰素-α,仅适用于少数患者。2020年7月,EMA授予了整个欧盟的Hepcludex有条件营销授权。这种一流的进入抑制剂提供了防止传播以获得控制并最终参与治愈乙型肝炎和D型肝炎的承诺。Hepcludex是了解病毒生命周期如何产生新的治疗选择的一个例子。牛磺胆酸钠共转运多肽,病毒受体和Hepcludex的靶标,并对目前研究的其他丁型肝炎治疗靶点进行综述。法尼基转移酶抑制剂,如Lonafarnib,靶向HDV生命周期中的另一个重要分子,代表丁型肝炎治疗的一个有希望的目标。法尼基转移酶将法尼基(异戊二烯基)基团连接到带有C末端Ca1a2X的蛋白质上(C:半胱氨酸,a:脂肪族氨基酸,X:C端氨基酸)基序,如大型丁型肝炎病毒抗原。这种修饰使得HBV/HDV颗粒和病毒包膜蛋白能够相互作用。Lonafarnib,防止这种包裹,已经在临床试验中进行了测试。在丁型肝炎治疗中需要考虑以乙型肝炎病毒的生命周期为目标,以便从两种共存的感染中治愈患者。核酸聚合物以尚未理解的方式靶向乙型肝炎生命周期。了解丁型肝炎病毒治疗的可能目标对于改善和发展HDV患者迫切需要的足够治疗是不可避免的。
    An approximate number of 250 million people worldwide are chronically infected with hepatitis B virus, making them susceptible to a coinfection with hepatitis D virus. The superinfection causes the most severe form of a viral hepatitis and thus drastically worsens the course of the disease. Until recently, the only available therapy consisted of interferon-α, only eligible for a minority of patients. In July 2020, the EMA granted Hepcludex conditional marketing authorization throughout the European Union. This first-in-class entry inhibitor offers the promise to prevent the spread in order to gain control and eventually participate in curing hepatitis B and D. Hepcludex is an example of how understanding the viral lifecycle can give rise to new therapy options. Sodium taurocholate co-transporting polypeptide, the virus receptor and the target of Hepcludex, and other targets of hepatitis D therapy currently researched are reviewed in this work. Farnesyltransferase inhibitors such as Lonafarnib, targeting another essential molecule in the HDV life cycle, represent a promising target for hepatitis D therapy. Farnesyltransferase attaches a farnesyl (isoprenyl) group to proteins carrying a C-terminal Ca1a2X (C: cysteine, a: aliphatic amino acid, X: C-terminal amino acid) motif like the large hepatitis D virus antigen. This modification enables the interaction of the HBV/HDV particle and the virus envelope proteins. Lonafarnib, which prevents this envelopment, has been tested in clinical trials. Targeting the lifecycle of the hepatitis B virus needs to be considered in hepatitis D therapy in order to cure a patient from both coexisting infections. Nucleic acid polymers target the hepatitis B lifecycle in a manner that is not yet understood. Understanding the possible targets of the hepatitis D virus therapy is inevitable for the improvement and development of a sufficient therapy that HDV patients are desperately in need of.
    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
    Chronic HDV infections cause the most severe form of viral hepatitis. HDV requires HBV envelope proteins for hepatocyte entry, particle assembly and release. Eight HDV and 8 HBV genotypes have been identified. However, there are limited data on the replication competence of different genotypes and the effect that different HBV envelopes have on virion assembly and infectivity.
    We subcloned complementary DNAs (cDNAs) of all HDV and HBV genotypes and systematically studied HDV replication, assembly and infectivity using northern blot, western blot, reverse-transcription quantitative PCR, and in-cell ELISA.
    The 8 HDV cDNA clones initiated HDV replication with noticeable differences regarding replication efficacy. The 8 HBV-HBsAg-encoding constructs all supported secretion of subviral particles, however variations in envelope protein stoichiometry and secretion efficacy were observed. Co-transfection of all HDV/HBV combinations supported particle assembly, however, the respective pseudo-typed HDVs differed with respect to assembly kinetics. The most productive combinations did not correlate with the natural geographic distribution, arguing against an evolutionary adaptation of HDV ribonucleoprotein complexes to HBV envelopes. All HDVs elicited robust and comparable innate immune responses. HBV envelope-dependent differences in the activity of the EMA-approved entry inhibitor bulevirtide were observed, however efficient inhibition could be achieved at therapeutically applied doses. Lonafarnib also showed pan-genotypic activity.
    HDVs from different genotypes replicate with variable efficacies. Variations in HDV genomes and HBV envelope proteins are both major determinants of HDV egress and entry efficacy, and consequently assembly inhibition by lonafarnib or entry inhibition by bulevirtide. These differences possibly influence HDV pathogenicity, immune responses and the efficacy of novel drug regimens.
    HDV requires the envelope protein of HBV for assembly and to infect human cells. We investigated the ability of different HDV genotypes to infect cells and replicate. We also assessed the effect that envelope proteins from different HBV genotypes had on HDV infectivity and replication. Herein, we confirmed that genotypic differences in HDV and HBV envelope proteins are major determinants of HDV assembly, de novo cell entry and consequently the efficacy of novel antivirals.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    慢性HBV感染不能通过目前的治疗方法治愈,因为它们在感染个体的肝脏中减少共价闭合环状(ccc)DNA水平的能力有限。因此,更多的cccDNA的形成和持久性的分子决定因素的理解是必要的。一个关键问题是从头核衣壳介导的补充(再进口)对感染肝细胞中cccDNA水平的贡献程度。
    我们设计了一种感染性HBV突变体,其基因组在HBV核心开放阅读框(ΔHBcHBV)的T67位置编码终止密码子。重要的是,ΔHBcHBV病毒粒子在感染时不能启动核衣壳合成。长期在体外HBV感染标志物随访9周HepG2-NTCP细胞(A3克隆)和HBVDNA定量使用新开发的,高精度PCR测定(cccDNA倒置定量PCR)。
    ΔHBc和野生型(WT)HBV导致HBV表面抗原(HBsAg)的可比表达,可以用进入抑制剂MyrcludexB阻断,通过受体牛磺胆酸钠协同转运多肽(NTCP)确认真正的感染。在原代人肝细胞中,Huh7-NTCP,HepG2-NTCP,和HepaRG-NTCP细胞,形成相当的cccDNA拷贝数。cccDNA水平,病毒RNA的转录,和HBsAg分泌在WT和ΔHBcHBV感染细胞中保持相对稳定至少9周。
    我们的结果表明,从头合成的HBc在cccDNA的转录调控中起着次要作用。重要的是,我们表明,最初形成的cccDNA在肝细胞中是稳定的,而不需要在体外感染系统中持续补充,并且不需要从头含DNA的核衣壳的贡献。因此,衣壳再导入的短期治疗靶向可能是消除慢性感染肝细胞中cccDNA的低效策略。
    乙型肝炎病毒可以在患者的一生中维持在肝脏中,导致肝损伤和癌症。我们已经确切地阐明了它是如何在被感染的细胞中维持自身的。这意味着我们对如何靶向病毒和治疗慢性感染有了更好的想法。
    UNASSIGNED: Chronic HBV infection cannot be cured by current therapeutics owing to their limited ability to reduce covalently closed circular (ccc)DNA levels in the livers of infected individuals. Therefore, greater understanding of the molecular determinants of cccDNA formation and persistence is required. One key issue is the extent to which de novo nucleocapsid-mediated replenishment (reimport) contributes to cccDNA levels in an infected hepatocyte.
    UNASSIGNED: We engineered an infectious HBV mutant with a genome encoding a stop codon at position T67 in the HBV core open reading frame (ΔHBc HBV). Importantly, ΔHBc HBV virions cannot initiate nucleocapsid synthesis upon infection. Long-term in vitro HBV infection markers were followed for up for 9 weeks in HepG2-NTCP cells (A3 clone) and HBV DNA was quantified using a newly-developed, highly-precise PCR assay (cccDNA inversion quantitative PCR).
    UNASSIGNED: ΔHBc and wild-type (WT) HBV resulted in comparable expression of HBV surface antigen (HBsAg), which could be blocked using the entry inhibitor Myrcludex B, confirming bona fide infection via the receptor sodium taurocholate cotransporting polypeptide (NTCP). In primary human hepatocytes, Huh7-NTCP, HepG2-NTCP, and HepaRG-NTCP cells, comparable copy numbers of cccDNA were formed. cccDNA levels, transcription of viral RNA, and HBsAg secretion remained comparably stable in WT and ΔHBc HBV-infected cells for at least 9 weeks.
    UNASSIGNED: Our results imply that de novo synthesised HBc plays a minor role in transcriptional regulation of cccDNA. Importantly, we show that initially-formed cccDNA is stable in hepatocytes without requiring continuous replenishment in in vitro infection systems and contribution from de novo DNA-containing nucleocapsids is not required. Thus, short-term therapeutic targeting of capsid-reimport is likely an inefficient strategy in eliminating cccDNA in chronically infected hepatocytes.
    UNASSIGNED: The hepatitis B virus can maintain itself in the liver for a patient\'s lifetime, causing liver injury and cancer. We have clarified exactly how it maintains itself in an infected cell. This now means we have a better idea at how to target the virus and cure a chronic infection.
    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
    Chronic hepatitis D (CHD) is the most severe form of viral hepatitis, with rapid progression of liver-related diseases and high rates of development of hepatocellular carcinoma. The causative agent, hepatitis D virus (HDV), contains a small (approximately 1.7 kb) highly self-pairing single-strand circular RNA genome that assembles with the HDV antigen to form a ribonucleoprotein (RNP) complex. HDV depends on hepatitis B virus (HBV) envelope proteins for envelopment and de novo hepatocyte entry; however, its intracellular RNA replication is autonomous. In addition, HDV can amplify HBV independently through cell division. Cellular innate immune responses, mainly interferon (IFN) response, are crucial for controlling invading viruses, while viruses counteract these responses to favor their propagation. In contrast to HBV, HDV activates profound IFN response through the melanoma differentiation antigen 5 (MDA5) pathway. This cellular response efficiently suppresses cell-division-mediated HDV spread and, to some extent, early stages of HDV de novo infection, but only marginally impairs RNA replication in resting hepatocytes. In this review, we summarize the current knowledge on HDV structure, replication, and persistence and subsequently focus on the interplay between HDV and IFN response, including IFN activation, sensing, antiviral effects, and viral countermeasures. Finally, we discuss crosstalk with HBV.
    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
    Hepatitis B virus (HBV) is the major cause of virus-associated liver disease. Persistent HBV infection is maintained by its episomal genome (covalently closed circular DNA, cccDNA), which acts as a template for viral transcripts. The formation of cccDNA is poorly characterised due to limited ability to quantify it accurately in the presence of replicative intermediates. Here, we describe a novel cccDNA quantification assay (cccDNA inversion quantitative PCR, cinqPCR), which uses restriction enzymes to invert a DNA sequence close to the gap region of Genotype D HBV strains, including the isolate widely used in experimental studies. Importantly, cinqPCR allows simultaneous normalisation to cellular DNA in a single reaction, provides absolute copy numbers without requiring a standard curve, and has high precision, sensitivity, and specificity for cccDNA compared to previous assays. We first established that cinqPCR gives values consistent with classical approaches in both in vitro and in vivo (humanised mice) HBV infections. We then used cinqPCR to find that cccDNA is formed within 12 h post-inoculation (hpi). cccDNA formation slowed by 28 hpi despite de novo synthesis of HBV DNA, indicating inefficient conversion of new viral genomes to cccDNA within infected cells. Finally, we show that cinqPCR can be used as a 96-well screening assay. Thus, we have developed an ideal method for testing current and future anti-cccDNA therapeutics with high precision and sensitivity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号