15/10/02说明:此前论坛服务器频繁出错,现已更换服务器。今后论坛继续数据库备份,不备份上传附件。

肝胆相照论坛

 

 

肝胆相照论坛 论坛 学术讨论& HBV English 基于寡核苷酸的慢性 HBV 感染疗法:生物化学、机制和抗 ...
查看: 163|回复: 2
go

基于寡核苷酸的慢性 HBV 感染疗法:生物化学、机制和抗病毒 [复制链接]

Rank: 8Rank: 8

现金
62111 元 
精华
26 
帖子
30441 
注册时间
2009-10-5 
最后登录
2022-12-28 

才高八斗

1
发表于 2022-11-17 02:13 |只看该作者 |倒序浏览 |打印
基于寡核苷酸的慢性 HBV 感染疗法:生物化学、机制和抗病毒作用入门
安德鲁·威兰特
[兽人]
Replicor Inc., 6100 Royalmount Avenue, Montreal, QC H4P 2R2, 加拿大
学术编辑:Mark W. Douglas 和 Thomas Tu
病毒 2022, 14(9), 2052; https://doi.org/10.3390/v14092052
收到日期:2022 年 8 月 11 日/修订日期:2022 年 9 月 8 日/接受日期:2022 年 9 月 8 日/发布日期:2022 年 9 月 16 日
(本文属于特刊 乙型肝炎病毒:战胜古老疾病的新突破)
下载 PDF 浏览图表
引文导出
抽象的
三种基于寡核苷酸的药物正在临床开发中,用于治疗慢性 HBV 感染。反义寡核苷酸 (ASO) 和合成干扰 RNA (siRNA) 旨在降解 HBV mRNA,核酸聚合物 (NAP) 可阻止 HBV 亚病毒颗粒的组装和分泌。 ASO 和 siRNA 用于多种肝病的广泛临床开发已经建立了对其药效学、在肝脏不同组织类型中的积累、药理作用、脱靶效应以及化学修饰和递送方法如何影响这些参数的深入了解。迄今为止,这些效应对于人类研究中使用的所有 ASO 和 siRNA 都是高度保守的。近年来,由于使用的递送方法不同,对几种 ASO 和 siRNA 化合物在慢性 HBV 感染中的临床评估变得复杂。此外,这些评估没有考虑 ASO/siRNA 在其他肝脏疾病中的功能的大型临床数据库,以及其他病毒感染中已知的脱靶效应。本综述的目的是总结目前对 ASO/siRNA/NAP 药理学的理解,并将这些概念整合到这些化合物治疗慢性 HBV 感染的当前临床结果中。
关键词:午睡;麻生组织;干扰核糖核酸;乙肝表面抗原;功能性治愈;免疫刺激

Rank: 8Rank: 8

现金
62111 元 
精华
26 
帖子
30441 
注册时间
2009-10-5 
最后登录
2022-12-28 

才高八斗

2
发表于 2022-11-17 02:14 |只看该作者
Oligonucleotide-Based Therapies for Chronic HBV Infection: A Primer on Biochemistry, Mechanisms and Antiviral Effects
by Andrew Vaillant
[ORCID]
Replicor Inc., 6100 Royalmount Avenue, Montreal, QC H4P 2R2, Canada
Academic Editors: Mark W. Douglas and Thomas Tu
Viruses 2022, 14(9), 2052; https://doi.org/10.3390/v14092052
Received: 11 August 2022 / Revised: 8 September 2022 / Accepted: 8 September 2022 / Published: 16 September 2022
(This article belongs to the Special Issue Hepatitis B Virus: New Breakthroughs to Conquer an Ancient Disease)
Download PDF Browse Figures
Citation Export
Abstract
Three types of oligonucleotide-based medicines are under clinical development for the treatment of chronic HBV infection. Antisense oligonucleotides (ASOs) and synthetic interfering RNA (siRNA) are designed to degrade HBV mRNA, and nucleic acid polymers (NAPs) stop the assembly and secretion of HBV subviral particles. Extensive clinical development of ASOs and siRNA for a variety of liver diseases has established a solid understanding of their pharmacodynamics, accumulation in different tissue types in the liver, pharmacological effects, off-target effects and how chemical modifications and delivery approaches affect these parameters. These effects are highly conserved for all ASO and siRNA used in human studies to date. The clinical assessment of several ASO and siRNA compounds in chronic HBV infection in recent years is complicated by the different delivery approaches used. Moreover, these assessments have not considered the large clinical database of ASO/siRNA function in other liver diseases and known off target effects in other viral infections. The goal of this review is to summarize the current understanding of ASO/siRNA/NAP pharmacology and integrate these concepts into current clinical results for these compounds in the treatment of chronic HBV infection.
Keywords: NAP; ASO; siRNA; HBsAg; functional cure; immunostimulation

Rank: 8Rank: 8

现金
62111 元 
精华
26 
帖子
30441 
注册时间
2009-10-5 
最后登录
2022-12-28 

才高八斗

3
发表于 2022-11-17 02:19 |只看该作者
8. ASO/siRNA Effects In Vitro, In Vivo and in Humans in HBV Infection
HepG2.2.15 cells harbor multiple copies of the HBV genome, thus providing a genetically invariable source of HBV RNA during viral replication. Early experiments with synthetic hairpin RNA (shRNA), siRNA and ASOs demonstrated antiviral activity in this and related in vitro systems [108,109,110] but without observable reductions in established cccDNA [111]. In other in vitro systems, the development of escape mutants with exogenously introduced HBV genomes were observed [112].
In vivo assessment of ASOs and siRNA almost entirely occurred in mouse models where strong antiviral responses were observed, including rapid multi-log reductions in HBsAg [113,114,115,116,117,118]. These multilog reductions in HBsAg are uniquely distinct from the normal pattern of protein response (saturation at 75–90% reduction from baseline) to ASO and siRNA observed with other liver mRNA targets in vivo [119,120,121,122,123]. However, siRNA analysis in woodchucks (which model the genetic plasticity present in human HBV infection) showed either no antiviral response or rapid rebound in viremia with continued treatment of siRNA triggers in HBsAg or HBx. This rebound was accompanied by the appearance of siRNA escape mutant mRNAs bearing point mutations in the siRNA recognition region [124], consistent with the genetic plasticity of HBV. Importantly, one of the first siRNA studies performed in the HDI mouse model of HBV infection observed cytokine induction consistent with TLR3 activation which occurred concomitantly with antiviral responses [125]. Poly I:C is a dsRNA widely used as a TLR3 agonist and in the HDI mouse model leads to a delayed and prolonged multilog HBsAg decline which is interferon dependent [126]. Moreover, polyI:C also has antiviral effects against HBV in ducks [127] and off target stimulation of innate immunity has been shown to contribute to the antiviral effects of siRNA in the woodchuck model [128].
The first clinical evaluation of siRNA in human HBV infection was performed using LNP-formulation of three siRNA triggers (2 in HBsAg and 1 in HBx, TKM-HBV/ARB-1467). This trigger combination was based on in vivo data showing futility with single triggers and designed to efficiently target mRNA produced from cccDNA or integrated HBV DNA within hepatocytes. In a phase II study conducted in ETV/TDF suppressed chronic HBV infection using ARB-1467 at doses above those saturating for the siRNA effect, mild initial responses in viremia (HBV RNA and HBcrAg) rebounded to baseline during dosing [129,130], consistent with the effects observed in woodchucks. HBsAg response was inconsistent with an siRNA effect: a highly variable and very small HBsAg reduction averaging ~0.25 log10 IU/mL from after 4 weeks followed by similarly small additional reductions following each siRNA dose. A second LNP-formulated siRNA (ARC-520) used two triggers, both in HBsAg. Phase I studies demonstrated transient increases in MCP-1 and IL-8 following dosing [131], consistent with TLR3 stimulation. In phase II studies, HBsAg response was again hypervariable and saturated with an average 0.1 log10 IU/mL reduction from baseline at 4 weeks at the lowest dose (1 mg/kg) still several times higher than required to saturate the siRNA effect with LNP formulation (0.15 mg/kg) [132]. Small but still hypervariable increases in HBsAg decline (average ~ 0.25 log10 IU/mL from baseline) required doses 4 times higher. Interestingly, the HBsAg response to ARC-520 was stronger in HBeAg-positive than HBeAg-negative patients, consistent with immunostimulatory effects as opposed to siRNA-mediated degradation (which should be the same regardless of HBeAg status). In HBeAg-positive patients, HBsAg declines following the first dose of ARC-520 approached 1.5 log10 IU/mL from baseline [132], but this was at dosing (4 mg/kg) substantially higher than required to saturate the siRNA effect. Multiple dosing of ARC-520 showed the same hypervariable and mild HBsAg response, averaging ~0.15 log10 IU/mL after 4 doses of 1 mg/kg and 0.4 log10 IU/mL after 4 doses of 2 mg/kg [133].
All siRNA drugs which followed ARC-520 transitioned away from LNP formulation to GalNAc conjugation (JNJ-3839, AB-729, VIR-2218 and RG6346), which is associated with accumulation of these siRNAs in KCs and LSECs. For all these GalANc-siRNA, single or multiple dose HBsAg responses were very similar (Figure 5) [134,135,136,137]. In the majority of patients, HBsAg response was either absent until 4 weeks following the first dose (similar to poly I:C response in mice) or was very mild and hypervariable with negligible HBsAg response observed at 15 days. After 4 weeks, a universal decline in HBsAg was observed in all patients saturating at 1.5–2.5 log10 IU/mL reduction from baseline. Despite delayed HBsAg declines in most patients, declines in markers of cccDNA activity (HBV DNA, HBV RNA and HBcrAg) were rapid [134], consistent with cccDNA inactivation. Extension of therapy to 48 weeks in NUC suppressed subjects with the addition a capsid assembly modulator did not significantly improve this response, and HBsAg rebound following cessation of treatment has been slow but continuous [134]. The addition of pegIFN only improves HBsAg decline seen with siRNA by an additional 0.5 log10 IU/mL [138]. To date, HBsAg loss has not occurred with any siRNA agent, except for 2 cases with ARC-520 with very low baseline HBsAg (<10 IU/mL) and with several years of exposure in the presence of ETV or TDF [139]. Importantly, HBsAg isoform response to siRNA treatment has been performed with AB-729 using the identical assay platform used for NAPs: no selective decline in the small isoform of HBsAg was observed, indicating that SVP are not targeted and that an siRNA effect is absent. Moreover, selective declines in the virus-specific large HBsAg isoform are observed, suggesting that cccDNA inactivation or other off-target effects are occurring. This is supported by additional recent observations that HBsAg response to siRNA (AB-729) is correlated with increased immunostimulatory activity [140,141].
HBsAg response to GalNAc-ASOs (RG6004/RO7062931 and GSK3389404) has also been inconsistent with an ASO effect [142,143]. No HBsAg response to RG6004 was observed with 0.5 mg/kg dosing, a dose level which yields easily observable protein reductions with Gal-NAc ASOs for other target mRNAs within 2 weeks after the first dose. Elevated dosing with RG6004 to 3 mg/kg produced a highly variable HBsAg decline between subjects which averaged only 0.4 log10 IU/mL after 8 weeks. Responses with GSK3398404 were almost identical to RG6004 in dose response, variability and average HBsAg decline. Bepirovirsen is the unconjugated variant of GSK3389404 carrying the identically modified oligonucleotide. This ASO is a 2′MOE gapmer designed to target mRNA cleavage in HBsAg but it also contains a class II CpG motif within its middle segment of DNA which is not shielded by 2′ ribose modification [79,144]. In contrast to administration of GSK3389404, where this oligonucleotide is delivered primarily to hepatocytes, administration of bepirovirsen results in the delivery of this oligonucleotide mainly to LSECs and KCs. With bepirovirsen, no HBV DNA or HBsAg response was observed in 4/6 subjects at the 150 mg dose [145], a dose which results in easily observed rapid protein reductions with unconjugated ASOs against other liver targets [146,147]. HBsAg responses were limited to the 300 mg dose, but in contrast to GSK 3389404, strong and rapid HBsAg declines (accompanied by host mediated transaminase flares) were restricted to subjects with baseline HBsAg < 1000 IU/mL [145]. In subjects with baseline HBsAg > 1000 IU/mL, HBsAg declines were indistinguishable from those observed with GSK3389404. Recently, HBsAg declines with bepirovirsen have also been correlated with its immunostimulatory activity [148]. These immunostimulatory activities have been attributed to TLR8 based on studies in mice but mice do not accurately model TLR reactivity in humans (Table 1) and this finding is at odds with the well understood biochemical properties of oligonucleotides: TLR8 reacts to unmodified ssRNA and TLR reactivity to TLR7/8 is well shielded by 2′MOE modification in bepirovirsen. Moreover, the oral TLR8 antagonist selgantolimod (GS-9688) did not elicit any HBsAg responses in human infection [149] despite clear immunostimulatory effects [149,150].
9. Conclusions and Perspective
HBsAg loss is an important milestone in the establishment of a functional cure for chronic hepatitis B infection. The molecular biology of HBV, and clinical experience with pegIFN and NAPs (the only agents currently capable of achieving HBsAg loss on therapy) indicate the antiviral effects on HBsAg required to achieve functional cure are 1) SVP particle production must be controlled and 2) HBsAg reduction > 4 log10 IU/mL from baseline must occur during therapy.
The genetic plasticity of chronic HBV infection is at odds with the notion that sequence-dependent mRNA cleavage of HBV mRNA by ASO or siRNA approaches are by themselves capable of achieving HBsAg responses below the threshold required for achieving functional cure. The failure of siRNA in preclinical development in models of HBV infection that model this genetic plasticity and the seminal siRNA assessment with ARB-1467, where rebound of viremia in the presence of three siRNA triggers occurred are consistent with these realities.
GalNAc-ASOs and GalNAc-siRNA have an identical pharmacodynamic protein response signature, susceptibility to single point mutation and share similar, very well-established threshold dosing for effects in humans. As such, clinical evaluations with ASOs in chronic HBV infection inform on the potential for siRNA effects and vice versa in human studies. GalNAc-ASOs are accompanied by little to no HBsAg response at doses saturatingthe ASO effect for other mRNA targets in the liver and at higher doses experience weak and hypervariable HBsAg responses still inconsistent with the ASO effect. Strong HBsAg responses are only observed when an ASO containing a TLR9 stimulatory motif is targeted to immunoreactive cells in the liver (bepirovirsen) and then only in patients with low baseline HBsAg (where immune functioning against HBV is more permissive). Consistent with these ASO findings in chronic HBV infection, HBsAg responses to all GalANc-siRNA are also inconsistent with the cleavage of mRNA. Rebound of infection occurs with LNP-RNA or mild HBsAg response is restricted to immunosensitive (HBeAg+) patient populations at siRNA doses far above the threshold required to saturate the siRNA effect. All GalNAc siRNAs have a delayed HBsAg response in the majority of patients which excludes an siRNA effect but is consistent with the exposure of immunoreactive siRNA in LSECs and KCs with GalNAc conjugation and with the delayed HBsAg responses observed with TLR 3 stimulation in mouse HBV models with dsRNA (poly I:C). These off target effects have been previously observed during the development of siRNA for influenza infection [151]. Additionally, siRNA has been shown to not target the production of SVP both directly by HBsAg isoform analysis and indirectly by the ubiquitously saturated HBsAg decline of 1.5–2.5 log10 IU/mL from baseline with all siRNA. SVP constitute > 99.99% of circulating HBsAg so the relatively mild HBsAg reduction threshold with siRNA indicates SVP are not being targeted.
The residence time of NAPs differs from GalNAc-ASO and GalNAc-siRNA. NAPs are rapidly cleared from the liver [52], whereas the GalNAc conjugation acts as a depot for ASO and siRNA in the liver [75]. Caution must be taken when interpreting the off-therapy antiviral responses to GalNAc-ASO and GalNAC-siRNA as the pharmacological effects of these agents can linger for months. As such, the delayed or slow rebound of HBsAg observed following cessation of GalNAc-ASO or GalNAc siRNA may reflect persistent pharmacological effects of these agents instead of real host antiviral responses.
Based on the substantial amount of available clinical data, the persisting or rapid development of escape mutants to ASO or siRNA exposure appear to restrict the effects of siRNA and ASO in human HBV infection to inactivation of cccDNA and or autophagy of HBsAg destined to form virus or subviral filaments: both activities are known to be stimulated by TLR activation. Neither ASO or siRNA appear to directly target the degradation of mRNA and consequently, the production of SVP from integrated HBV DNA. This is an important distinction as integrated HBV DNA is the source of the bulk of HBsAg in HBeAg negative chronic HBV infection [79]. On the other hand, NAPs do not directly affect viral replication but very effectively target SVP production from cccDNA and integrated HBV DNA, leading to much stronger HBsAg declines to below the limit of detection of the most sensitive experimental HBsAg assay available.
It is important to point out that the apparent TLR-stimulatory effects of ASO and siRNA are the first TLR-agonist effects to yield significant antiviral responses in chronic HBV infection. This is likely a function of their ability to activate TLR3 or TLR9 and be targeted directly to the liver and to immunoreactive non-parenchymal cells. These activities will be important to consider in future combination therapies in the pursuit of HBV functional cure. In the design and reporting of future clinical data from ASO/siRNA based drugs, individual baseline HBsAg and HBsAg responses will also be important to disclose to solidify our understanding of how ASO/siRNA technologies may participate in functional cure of HBV.
‹ 上一主题|下一主题
你需要登录后才可以回帖 登录 | 注册

肝胆相照论坛

GMT+8, 2024-5-6 04:02 , Processed in 0.013644 second(s), 11 queries , Gzip On.

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.