Skip to main content

The impact of integrated hepatitis B virus DNA on oncogenesis and antiviral therapy

Abstract

The global burden of hepatitis B virus (HBV) infection remains high, with chronic hepatitis B (CHB) patients facing a significantly increased risk of developing cirrhosis and hepatocellular carcinoma (HCC). The ultimate objective of antiviral therapy is to achieve a sterilizing cure for HBV. This necessitates the elimination of intrahepatic covalently closed circular DNA (cccDNA) and the complete eradication of integrated HBV DNA. This review aims to summarize the oncogenetic role of HBV integration and the significance of clearing HBV integration in sterilizing cure. It specifically focuses on the molecular mechanisms through which HBV integration leads to HCC, including modulation of the expression of proto-oncogenes and tumor suppressor genes, induction of chromosomal instability, and expression of truncated mutant HBV proteins. The review also highlights the impact of antiviral therapy in reducing HBV integration and preventing HBV-related HCC. Additionally, the review offers insights into future objectives for the treatment of CHB. Current strategies for HBV DNA integration inhibition and elimination include mainly antiviral therapies, RNA interference and gene editing technologies. Overall, HBV integration deserves further investigation and can potentially serve as a biomarker for CHB and HBV-related HCC.

Background

Chronic Hepatitis B (CHB) refers to a persistent liver infection caused by the hepatitis B virus (HBV) that lasts for more than 6 months [1]. Among these individuals with CHB, a staggering 820,000 cases experienced adverse outcomes such as liver failure, liver cirrhosis, HBV-related hepatocellular carcinoma (HCC), or other HBV-associated diseases. The risk of developing HCC in patients with CHB is significantly elevated, ranging from 10 to 100 times higher compared to non-infected individuals. Integrated HBV DNA refers to the incorporation of HBV DNA into the host cell’s genome, which is one of the important factors contributing to HBV-related carcinogenesis. HBV integration can induce genetic damage and chromosomal instability, leading to tumor progression via the activation of oncogenes or inactivation of tumor suppressor genes [2,3,4]. It may persist even after successful hepatitis B surface antigen (HBsAg) seroconversion, which can contribute to HBsAg re-positivity and increase the risk of developing HCC [5]. Remarkably, integrated HBV DNA is identified in around 85-90% of HCC cases associated with hepatitis B virus infection [6]. Targeting the HBV DNA integration process and eliminating integrated HBV DNA from the host genome becomes crucial in preventing the progression of chronic hepatitis B. The treatment of CHB now includes options such as nucleos(t)ide analogues (NAs) and interferon-alpha (IFN-α), which could effectively inhibit HBV replication and new integrations. While IFN is administered for a finite duration, NAs are typically prescribed for extended periods, often lifelong [1, 7, 8]. The primary objective of CHB treatment is to suppress HBV replication to the maximum extent possible. Hepatocyte inflammation and necrosis, liver fibrosis and hyperplasia can be attenuated by inhibiting HBV replication, thereby delaying and reducing the occurrence of severe complications such as liver failure, liver cirrhosis, and HCC [9, 10]. The types of CHB cure are categorized as functional cure (also known as clinical cure or immunological cure) and sterilizing cure (also known as virological cure). A “functional” cure was defined as sustained HBsAg loss and HBV DNA less than the lower limit of quantitation (LLOQ) 24 weeks off-treatment [11]. On the other hand, a “sterilizing” cure was defined as all traces of HBV infection would be eliminated, including cccDNA and integrated HBV DNA [11, 12]. For eligible patients, the pursuit of functional cure should be considered [7, 13, 14]. The sterilizing cure of HBV is to achieve sterilizing cure, which necessitates the elimination of intrahepatic cccDNA and the complete eradication of integrated HBV DNA. However, this remains a challenging feat due to the persistence of viral reservoirs, weak immune response, long-term medication requirements, variable treatment responses, and the presence of advanced liver disease [1, 15]. In animal models, the frequency of integration events is estimated to be approximately one in 103-104 hepatocytes [16, 17]. However, the precise mechanisms of integration remain unclear, necessitating further attention and investigation. This review aims to provide a comprehensive overview of HBV DNA integration, including its molecular mechanisms, detection methods, research models, oncogenetic roles in HCC, and potential treatment strategies for eliminating HBV DNA integration.

HBV integration occurs during HBV replication

The structure and life cycle of HBV DNA

The HBV is a DNA virus that belongs to the Hepadnaviridae family [1]. Dane particles are infectious virions characterized by a lipid membrane that encapsulates a nucleocapsid. The lipid membrane contains three HBsAg, which include large (L-HBs), middle (M-HBs), and small (S-HBs) forms. The nucleocapsid within the Dane particle is composed of hepatitis B core protein (HBc), viral polymerase (Pol), and the viral genome DNA. The HBc protein provides structural integrity to the nucleocapsid, while the viral polymerase is responsible for replicating the viral genome during the viral life cycle (Fig. 1A).

The viral genome of the HBV is a partially double-stranded DNA (dsDNA) structure. It consists of one complete coding minus(-) strand and one incomplete non-coding plus(+) strand. The viral genome contains four overlapping open reading frames (ORFs) that are responsible for encoding various viral proteins including: (1) HBV DNA polymerase (pol), which is involved in viral replication and synthesis of the viral genome. (2) HBsAg, which exists in three forms: Large, Medium, and Small. These antigens are crucial for viral attachment and entry into host cells. (3) HBV core antigen (HBcAg), providing structural integrity to the nucleocapsid of the virus. (4) HBV e antigen (HBeAg), whose exact function is not fully understood but is associated with immune tolerance and viral replication. (5) HBV X protein (HBx), a multifunctional protein involved in regulating viral replication, cell proliferation, and immune response modulation. Each of these viral proteins contributes to different aspects of the HBV life cycle, including viral replication, virion assembly, and immune evasion (Fig. 1B) [18].

After the HBV virion entering hepatocytes, the nucleocapsid is released into the cytoplasm and the relaxed circular DNA (rcDNA) enters the nucleus where it will convert into cccDNA [19,20,21,22]. The cccDNA then serves as a template for the synthesis of five transcripts (3.5Kb pregenomic RNA, 3.5Kb precore mRNA, 2.4Kb preS1 mRNA, 2.1Kb preS2/S mRNA, and 0.7Kb HBx mRNA), which are transcribed by host RNA polymerase II. Among these transcripts, the 3.5Kb pregenomic RNA (pgRNA) plays a crucial role in viral reverse transcription and replication processes. Following the utilization of pgRNA as a template, the minus(-) strand is synthesized, and subsequently, the synthesis of the plus(+) strand proceeds using the minus(-) strand as a template. This process gives rise to two products during plus(+) strand synthesis: partially circular rcDNA and double-stranded linear DNA (dslDNA). Rather than being encapsulated and secreted as virions, dslDNA has the potential to re-enter the nucleus and integrate into the human genome [5] (Fig. 1C).

Fig. 1
figure 1

The structure of HBV DNA genome and HBV life cycle. (A)The schematic diagram of Dane particle. (B)The circular schematic diagram of genotype C HBV genome. (C) After HBV Dane particles entering hepatocytes, uncoating takes place and genome is released. RcDNA is repaired to form cccDNA which is transcribed to pgRNA and 3.5Kb/3.5Kb/2.4Kb/2.1Kb/0.7Kb transcripts, HBV RNA transcripts are translated into proteins such as HBeAg, Core protein and HBx protein. Polymerase binds to the pgRNA with the recruitment of Core protein to assemble nucleocapsid and package pgRNA, pgRNA serves as the template to reverse transcription synthesize the HBV minus(-)-strand DNA. Polymerase translocates accurately to synthesize the HBV plus(+) strand DNA. Polymerase translocates mistakenly results the synthesis of dslDNA and the integration of dslDNA into the host genome

The procedure of HBV integration

During reverse transcription, the HBV DNA polymerase utilizes pgRNA as a template to transcribe the minus(-) strand DNA. This process involves the generation of DNA oligonucleotides (TGAA or GAA), which serve as primers for synthesizing the minus(-) strand. These primers are produced within a ε stem-loop structure located at the 3’ terminal of the pgRNA. During the process of extending the minus(-) strand DNA, most of the pgRNA undergoes degradation mediated by RNase H, except for the capped 5’ end of pgRNA. Remarkably, these undegraded RNA oligonucleotides play a vital role as primers for the synthesis of the plus(+) strand DNA. Among these fragments, one containing the direct repeat 1 (DR1) sequence acts as a primer for the synthesis of the plus(+) strand DNA [23,24,25]. Typically, this primer binds to the direct repeat 2 (DR2) region of the newly synthesized minus(-) strand DNA, which is complementary to the DR1 segment of the residual pgRNA. This binding guides the synthesis of plus(+) strand DNA, resulting in the formation of partially circular rcDNA (90–95%). However, if the initiation of the plus(+) strand synthesis occurs in situ without binding to the DR2 region (in-situ priming), the synthesis of double-stranded dslDNA takes place (5–10%) [26]. This small probability of primer translocation failure might be due to the mutation of DR2 region which causing reduced complementarity between DR2 region and RNA primer [27]. Another study suggests that cis-acting elements mutants in HBV genome are related to the proportion of dslDNA generation [28, 29].

Since the initial discovery of HBV integration in the early 1980s [30, 31], several hypotheses have emerged to shed light on the mechanisms underlying this integration process [32, 33]. The dslDNA provides HBV DNA fragments that can integrate into the host genome. Upon entering the nucleus, dslDNA is inserted randomly into hepatocyte chromosomes through DNA repair pathways [23]. The oxidative damage caused by hepatitis can induce DNA breakages in the host genome, creating breakpoints for the integration process [34,35,36,37,38]. Considering the limited homologous sequences between viral DNA and the human genome, the most likely mechanisms for HBV integration are non-homologous end joining (NHEJ) and microhomology-mediated end joining (MMEJ) DNA repair pathways [38, 39]. These pathways facilitate the joining of DNA ends during the repair process, allowing the integration of HBV DNA fragments into the host genome. In the NHEJ pathway, DNA breaks lacking significant homology undergo modification and subsequent ligation, leading to the generation of deletions or insertions [40]. On the other hand, MMEJ pathway is a distinct mechanism for end joining that operates separately from NHEJ [41, 42]. MMEJ relies on the presence of microhomology and utilizes longer stretches of microhomology (5–25 bp) compared to NHEJ [41]. Furthermore, the dslDNA can undergo circularization through the NHEJ DNA repair pathway. However, this process can lead to the formation of non-functional molecules due to the error-prone nature of NHEJ (Fig. 2) [43].

The breakpoints of HBV integration in the viral genome display several distinctive characteristics: (1) Integration often takes place near the DR1 or DR2 sites. (2) Integrated HBV fragments show a range of sizes, varying from 28 bp to 3215 bp. Long integration fragments(> 2000 bp) are observed more frequently than short ones. (3) It is common to observe small deletions within viral sequences at the joining site [16, 38, 44,45,46,47]. These characteristics highlight the specific patterns and variations in HBV integration events within the viral genome. Previous studies have shown that HBV has a preference for integrating into genic regions such as exons, introns, and promoters, as well as gene-rich areas [48]. Notably, certain genes such as hTERT, MLL4, and CCNE1 have been frequently identified as targets of HBV integration [44, 48,49,50,51,52]. This biased selection of integration sites has been observed in both tumor and adjacent tissues with a higher frequency of integration occurring in tumor tissues compared to non-tumor tissues [44, 53, 54]. These findings highlight the specific genomic locations where HBV integration tends to occur and suggest its potential impact on specific genes in both tumor and non-tumor tissues.

Fig. 2
figure 2

Schematic diagram about synthesis of dslDNA and HBV DNA integration. (A) ε stem-loop structure forms and P protein primes at the ε stem-loop structure to form P-ε ribonucleoprotein (RNP) complex. The terminal protein (TP) domain of the P protein binds with the first deoxyribonucleotide in ε stem-loop structure near the 5’cap of pgRNA. After the first four(TGAA) or three (GAA) nucleotides of the new minus(-) strand DNA generated in ε stem-loop structure, the DNA oligo then transferred to DR1 at the 3’end of pgRNA with TP and the synthesis of minus(-) strand starts.(B) pgRNA is degraded by RNase H domain of P protein while the minus(-) strand is synthesizing. (C) The DNA oligomer binds to the direct repeat 2 (DR2) region of the newly synthesized minus(-) strand DNA to guide the synthesis of plus(+) strand DNA, forming a partially circular rcDNA (90–95%). (D) The RNA primer directly initiated in situ without binding to the DR2 region (5–10%), dslDNA will be generated. (E) Inflammation and oxidative stress induce host genomic DNA double-stranded breaking, which provides breakpoints for integration through NHEJ or MMEJ

HBV integration occurs early in infection

The occurrence of integration during the early stages of HBV infection has been supported by multiple studies, which aligns with experimental evidence from cell infection models and animal liver infection models. For instance, in ducklings experimentally infected with the avian hepadnavirus duck hepatitis B virus (DHBV), integrated HBV DNA was detected as early as 6 days post-infection [55]. Similarly, in the woodchuck infection model, integration of woodchuck hepatitis virus (WHV) was observed within 1–3 h post-infection, indicating immediate genomic integration of WHV DNA into hepatocytes upon natural viral invasion [45, 56]. These findings highlight the early occurrence of HBV integration and provide valuable insights into the dynamics of viral integration in different infection models. Moreover, numerous investigations utilizing primary human hepatocytes (PHH), HepaRG-NTCP, HepG2-NTCP, and Huh7-NTCP cells have consistently demonstrated rapid viral integration after infection [35]. Furuta et al. conducted a study using a chimeric mouse model consisting of human hepatocytes infected with HBV, where they found that HBV integration could occur between 23 and 49 days post-infection through MMEJ, primarily within mitochondrial DNA [57]. Furthermore, the occurrence of integration in acute hepatitis B also suggests its early onset following infection [58]. Taken together, these findings strongly indicate that integration may take place within the host genome during the initial stages of hepadnaviral infection. Considering that HBV DNA integration predominantly occurs during viral replication, it becomes crucial to hinder replication at the early stage of infection in order to prevent integration.

The integration of HBV DNA and Hepatitis promotes each other

Liver damage caused by HBV infection is characterized by persistent necrotizing inflammation accompanied by immune regulation [59]. Integrations, as an early event in HBV infection, are closely associated with ongoing immune-mediated inflammatory responses. The oxidative damage to hepatocellular DNA acts as breakpoints for dslDNA integration [60]. Multiple studies consistently report a positive correlation between the extent of hepadnavirus integration and oxidative damage [61, 62]. From another perspective, HBV-specific cytotoxic T lymphocytes (CTLs) selectively target and eliminate hepatocytes replicating HBV, leading to the preferential clonal expansion of HBV DNA-integrated hepatocytes that may evade the immune response mediated by HBV-specific CTLs [47]. Moreover, the integration of HBV DNA can trigger an inflammatory response. Integrated HBV DNA is considered a potential source of HBsAg, which is derived from both a 2.1Kb transcript and mRNA transcribed from integrated HBV DNA. The presence of HBsAg plays a crucial role in the pathogenesis of hepatitis [63]. It is widely recognized that elevated production of HBsAg contributes to T cell exhaustion, resulting in restricted or impaired T cell responses and even the elimination of T cells recognizing specific epitopes [64, 65]. Additionally, CD205 has recently been identified as a pivotal receptor involved in the capture of CpG-oligodeoxynucleotides in vivo. The enhanced expression of CD205 on Kupffer cells in HBsAg-transgenic mice may be attributed to mild inflammation associated with HBsAg [66, 67]. In previous consensus, cccDNA has been acknowledged as the primary transcriptional template for HBsAg production [68, 69]. This hypothesis is further supported by evidence documented in chimpanzees with chronic HBV infection [70]. A dynamic observation using liver biopsy specimens from CHB patients revealed that individuals with HBV S gene integration experienced a slower decline in serum HBsAg levels compared to those without such integration following prolonged therapy [71]. This finding highlights the diverse origin of HBsAg. Researchers from Switzerland, based on liver biopsies obtained from HBe(-) patients, discovered that transcriptionally active integrated HBV DNA can autonomously generate HBsAg without relying on HBV replication [72]. This result may explain why serum HBsAg level is much less correlation with HBV DNA in HBe(-) patients with a very low HBV replication state.

The interaction between HBV DNA integration and hepatitis is complex, as they mutually reinforce each other through immune responses starting from the early stages of HBV infection, ultimately leading to the development of HCC.

Examination and research models for HBV integration

With the progress of high-throughput sequencing technologies, different strategies have been implemented to enable a more precise investigation into the implications of the integration process. These strategies aid in the detection of integrated viral DNA within the host genome. The unique features of each strategy are summarized in Table 1.

Examination methods based on DNA hybridization

The integration of HBV was initially detected in HCC patient tissue and the PLC/PRF/5 cell line in 1980 through Southern Blot hybridization using HBV as a probe31. It was observed that most integration events took place at the nicked cohesive end region of HBV DNA. Moreover, Northern blot analysis revealed the presence of specific transcripts of HBsAg even in the absence of HBcAg [73]. Following these discoveries, the Southern Blot hybridization technique was employed to detect viral integration within the host cell genome [2, 74]. Subsequent investigations progressively unveiled the integration of HBV DNA in liver tissues of patients with HBV-related conditions such as HCC, acute HBV hepatitis, chronic HBV infection, and HBV-related liver cirrhosis [2]. These findings highlight use of Southern Blot hybridization as a valuable tool in studying viral integration. In addition to this approach, in situ hybridization based on the same principle as Southern Blot hybridization was utilized to identify the chromosomal sites of HBV DNA integration [75]. Subsequently, Fluorescence In Situ Hybridization (FISH) emerged as a more sensitive and specific method for detecting integrated HBV DNA, replacing the previous techniques [76, 77].

Examination methods based on PCR amplification

Recombinant plasmid vectors were utilized for the direct cloning of virus-cell junctions, allowing for a comprehensive examination of integrated HBV DNA fragments [78]. However, the presence of diverse virus-cell junctions poses significant challenges in achieving accurate and sensitive detection of HBV integration.

Table 1 Detection methods of HBV DNA integration

The detection of virus-cell junctions has been made possible through the development of various PCR-based strategies, including the Arthrobacter luteus-PCR(Alu-PCR) [95]. Alu elements, which are short interspersed nuclear elements (SINEs), are widely distributed throughout primate genomes and can be found in approximately 1,000,000 copies per human genome [96]. By utilizing a combination of HBV and Alu repetitive element primers, it is possible to amplify and sequence fragments of virus-cell DNA junctions [71, 97, 98]. However, Alu-PCR has limitations in detecting HBV integrated fragments that are located far from the Alu repeat sequence or accurately quantifying the integration junctions.

In addition to Alu-PCR, another technique called inverse nested PCR (invPCR) can be employed for amplifying virus-cell DNA junctions. This method provides an alternative approach to detect and analyze these junction fragments. In 1995, Gong et al. successfully detected DR-related integrations of wild-type DHBV in LMH-D2 cells using inv PCR, which introduced a novel protocol for detecting and characterizing integrations of DHBV derived from episomal viral DNAs [99]. This strategy was primarily designed to selectively amplify virus-cell DNA junctions near the DR sequences, as these DR sequences are recognized as preferred integration sites for hepadnaviral DNA [23, 45, 55, 99, 100]. To detect integrations in or near hypothetical sites, high-molecular-weight liver DNA was cleaved by restriction endonucleases specifically targeting and cleaving HBV DNA and host DNA at unknown sites. Subsequently, the DNA was circularized using T4 DNA ligase and further cleaved by another restriction endonuclease, resulting in the generation of linear strands. Within these strands, the viral-cellular DNA junctions were located internally, with viral fragments present at both termini. These fragments were then amplified through nested PCR utilizing virus-specific primers [16, 84]. This technique has been widely employed for the detection of integrated HBV DNA due to its high sensitivity and specificity [101,102,103]. However, it is important to note that this method can only detect DNA sequences in close proximity to the junctions and is heavily reliant on restriction endonucleases [47].

Examination methods based on high-throughput sequencing technology

Whole-genome sequencing (WGS) and whole-exome sequencing (WES) are two widely used next-generation sequencing (NGS) methods that have found extensive applications in various areas of virology research. NGS offers several advantages, including the elimination of the need for prior viral DNA information and improved sensitivity in detection. WGS allows comprehensive coverage of host genomes, enabling the identification of viral sequences [104]. On the other hand, WES provides greater depth than WGS Nanopore sequencing, but it focuses solely on coding regions 91. However, deep sequencing with significant insertions or deletions remains challenging due to the intrinsic error-prone nature and limited length of the generated sequence reads [105]. In recent years, the field of third-generation sequencing technology has witnessed a remarkable advancement, offering inherent advantages in exploring complex genomic rearrangements [106]. This technology allows the generation of complete HBV genomes in a single sequencing read, facilitating the investigation of intricate and diverse distribution patterns of rapidly mutating viral genomes [107]. By combining third-generation sequencing with the analysis of biological information, a deeper understanding of HBV integration can be achieved [106].

Research models for HBV DNA integration

Comprehensive investigations into HBV integration face challenges due to the limited availability of human non-tumor liver tissues at all stages of HBV infection, especially compared to HCC tissues. Additionally, the scarcity of suitable models for studying HBV infection further hampers research on HBV integrations. To overcome these limitations, several in vitro studies have utilized PHH, HepaRG-NTCP, HepG2-NTCP, and Huh7-NTCP cells to investigate the mechanisms and timing of HBV DNA integration [35, 57, 77]. These cell-based models offer valuable insights into HBV integration. Furthermore, other hepadnavirus-infected animal models have also contributed significantly. For example, studies using ducklings infected with DHBV and woodchucks infected with WHV have provided important contributions to our understanding of HBV integration [45, 55, 56]. These animal models offer insights that complement the in vitro studies and enhance our overall understanding of HBV integration. Since HBV integration in both genomic DNA and RNA transcripts was observed in various cell lines including HepG2.2.15, HepAD38, PLC/PRF/5, DE19, MHCC97H, MHCC97L, MHCCLM3 cells as well as Huh1 and Hep3B cells, [88, 92, 93] therefore, HBV-related HCC cell lines could also be utilized as the cell model for HBV integration. Animal infection models, including chimpanzees, human liver chimeric mice, Tupaia, and hNTCP-expressing macaques, have been utilized to study HBV infection. These models demonstrate susceptibility to chronic HBV infection and can generate clonally expanded hepatocytes that contain integrated viral DNA [17, 108,109,110].

HBV DNA integration induces HCC

Previously, it was suggested that integrated HBV DNA had no discernible function due to its random distribution and lack of requirement in HBV replication. However, over the past decade, numerous studies have shown the significant impact of HBV DNA integrations on both HBV infection and carcinogenesis (Fig. 3) [5, 6, 32, 36, 46, 52, 101, 99, 111,112,113,114,115,116,117,118,119,120,121]. Therefore, conducting more research to understand the relationships between integration translocations in host genes, fragments of HBV genome, and carcinogenetic mechanism of integration are of great clinical significance [112]. The integration of HBV DNA into the host genome is an early event that precedes clonal tumor expansion [122, 123] and the presence of integration events indicates their potential role as precursors to tumor development in patients with chronic hepatitis and during the acute infection stage [35, 58, 124, 125]. HBV DNA integration primarily contributes to HCC through three mechanisms: (1) modulation of the expression or function of proto-oncogenes and tumor suppressor genes, (2) induction of chromosomal instability, and (3) expression of integrated mutant HBV proteins [54].

Fig. 3
figure 3

Schematic diagram of HBV DNA integration from chronic HBV infection to hepatocellular carcinoma. (A) Initially HBV DNA randomly integrates into host genome. (B) The infected hepatocytes are eliminated by host immune response. Infected hepatocytes with favorable integrations survive and clonally expand. (C) Hepatocytes with integrations expand. When integration happens near/into HCC-related genes, HCC initiating cells may occur. (D) HCC initiating cells expand and HCC cells with carcinogenetic integrations appear. (E) HCC cells with carcinogenetic integrations expand leading to the development of HCC

HBV DNA integration modulates cancer-related genes

The integration of HBV in the human genome was observed to have a distinct distribution pattern in tumors compared to non-tumor tissues, with a tendency for enrichment around cancer driver genes [118]. The chromosomal locus 11q13.3 has a significant tendency to serve as a recurring site for HBV integration [126]. This specific genomic region contains crucial oncogenic driver genes, namely CCND1 and FGF19, which are frequently amplified in HCC [127]. Additionally, the expression levels of cancer-associated genes, such as hTERT, KMT2B, MLL4, CCNE1 and PAK3, were found to be up-regulated in tumor tissues compared to their corresponding normal counterparts [44, 128, 129]. Telomeres, which enhance telomerase activity, play a crucial role in maintaining genome stability. Additionally, the upregulation of hTERT has been extensively reported [130]. Furthermore, studies investigating hTERT integration sites have shown that HBV DNA integration at the hTERT promoter is pivotal in the overexpression of the hTERT gene [131, 132].

Integration events involving mitochondrial DNA have been identified in tissue samples obtained from both tumor and non-tumor areas of HCC patients. This new finding highlights mitochondrial DNA as a newly recognized target of HBV integration, causing mitochondrial instability and dysfunction. Consequently, this contributes to the development and progression of HCC [133]. In a recent study, the coexistence of two distinct HCC subtypes was observed in a patient with HBV infection, with no identical integration sites detected. This finding suggests that the multicentric occurrence of HCC may be attributed to diverse HBV DNA integration events [120].

HBV DNA integration induces chromosomal instability

Chromosomal instability is a fundamental characteristic of human cancer, and it is closely associated with unfavorable prognosis, metastasis, and resistance to therapeutic interventions [134]. The breakpoints of HBV integration have been found to be correlated with an increased level of copy-number variation [44]. This observation highlights the potential contribution of HBV integration to the chromosomal instability observed in the HCC genome [38, 53].

One study suggests that HBV has a preferential integration site in the human genome, particularly fragile sites and CpG islands [38]. These are regions of the genome that are prone to rearrangements and genetic alterations, which can lead to the development of cancer. HBV integration into these regions can also lead to epigenetic instability, which can further contribute to the development of HCC [135]. Additionally, HBV integration events were observed to be enriched in the proximity of telomeres, which play a crucial role in maintaining genome stability. Dysfunction of telomeres can lead to extensive DNA rearrangements, deletions, and amplification, all of which are commonly associated with the development of cancer [136].

HBV DNA integration expresses truncated HBV proteins

Truncated HBs and HBx proteins, derived from integration fragments of HBV DNA, are recognized as significant contributors to the development of HCC [137]. Truncated preS2/S sequences within hepatocytes, commonly observed in integrated HBV DNA, have been implicated in promoting HCC progression through multiple pathways [138,139,140]. The accumulation of truncated mutant HBsAg induces endoplasmic reticulum stress, leading to the generation of reactive oxygen species, oxidative stress, and DNA damage [141]. Moreover, the down-regulated expression of TGFBI induced by truncated HBsAg in the TGF-β/Smad signaling pathway also contributes to carcinogenesis [142]. Additionally, the truncated S protein impedes the G1/S phase cell cycle checkpoint by suppressing the expression of the p53-p21 axis [143]. Multiple truncated HBx proteins, particularly those with C-terminal truncation (ct-HBx), have been identified to exert diverse functions in HCC, including the induction of stem cell-like characteristics, inhibition of apoptosis, and promotion of HCC invasion and metastasis [144,145,146,147,148,149,150].

The incidence of HCC is significantly higher in males compared to females, with a ratio of approximately 4:1 but the reasons for the gender bias are unclear. Some certain integration sites of HBV can be identified as human somatic risk loci for HBV integration (VIMs). The enriched transcription factors in VIMs are involved in DNA repair and the androgen receptor (AR) signaling pathway. There are significant interactions between the AR pathway and the complement system. These interactions, along with the X-linked ZXDB regulon that includes albumin (ALB), may contribute to the male predominance observed in HCC [151]. However, additional research is required to confirm the association between HBV integration and male predominance in HCC. Furthermore, studying the underlying mechanisms of integrated HBV DNA in promoting HCC can aid in the development of more targeted therapeutic strategies for HCC and provide novel biomarkers for monitoring its occurrence.

The quest for non-invasive biomarkers in HBV DNA integration during HCC development

Until now, the quantification of integrations has predominantly been conducted in liver tissues. However, liver biopsy is an invasive procedure associated with inherent risks. Alternative biomarkers such as serum levels of HBV core-related antigen (HBcrAg) and HBV RNA may serve as indicators of transcriptional activity specific to cccDNA, as they are expected to be exclusively generated from cccDNA rather than integrated HBV DNA due to the absence of a promoter that initiates core RNA transcription [22, 152,153,154]. Therefore, further investigation is needed to identify specific serum biomarkers for HBV DNA integration. Cell-free DNA (cfDNA) is an emerging noninvasive blood biomarker that is used to assess tumor progression, evaluate prognosis, diagnose diseases, and monitor response to treatment [155]. Recent studies have reported the detection of HBV integration in circulating cfDNA from both HCC and liver cirrhosis patients’ plasma [89]. Since cfDNA primarily originates from dying tumor cells, the release of cfDNAs from non-HCC liver tissues is considerably lower compared to HCC liver tissues [156]. As a result, cfDNA is more suitable for monitoring HBV integration in HCC development. In a study on the early recurrence of HCC after surgical resection, researchers found that plasma virus-host chimera DNA (vh-DNA) could serve as a biomarker for detecting residual tumor cells and predicting recurrence [94]. Detecting sequence-unknown vh-DNA directly from cfDNA requires a sensitive NGS approach with a standardized workflow and appropriate cutoff values, along with a population study to ensure sensitivity and specificity, incorporating known tumor-related somatic mutations [157].

The importance of early intervention

HBV DNA integration has the potential to generate a portion of HBsAg and contribute to HCC development, making early intervention for HBV infection crucial. While NAs may not eradicate integrated HBV DNA, initiating treatment at an early stage can reduce the occurrence of integrations, potentially reducing oncogenic mutations. The continuous suppression of the virus through effective treatment significantly lowers the risk of oncogenic mutations. Functional cure, achieved through sustained virus suppression, greatly diminishes the likelihood of carcinogenic mutations. The elimination of cccDNA, the viral reservoir in hepatocytes, is essential in preventing HBV reactivation and relapse. The ultimate objective in managing HBV infection is to achieve a sterilizing cure, which involves the complete eradication of cccDNA and integrated HBV DNA from the host genome. Strategies aimed at accomplishing this goal include utilizing antiviral agents that specifically target and eliminate integrated HBV DNA from the host cells.

The slow decline, or no decline of serum HBsAg levels during NAs treatment may be due to the ongoing production of HBsAg from integrated HBV DNA, particularly in HBeAg-negative patients [158]. Recent studies have confirmed the presence of integrant-derived RNAs (id-RNAs) and 5’-human-HBV-3’ transcripts originating from integrated HBV DNA in serum [159]. Initiating treatment at an early stage may enhance the likelihood of achieving a functional cure by reducing HBV DNA integration. Additionally, quantifying integrations in these patients can help identify factors that contribute to the slow clearance of HBsAg.

Exploring strategies for HBV DNA integration inhibition and elimination: current progress and future directions

The integration of HBV DNA can contribute to both neoplasia and a portion of HBsAg production. The elimination of integrated HBV DNA is also regarded as a critical measure for achieving complete eradication of HBV [1]. Inhibiting HBV DNA integration at an early stage holds immense importance and has consistently garnered significant attention from researchers in this field [160, 161]. The efficacies of current strategies to eliminate HBV DNA integration are concluded in Table 2.

Table 2 Efficacies of current strategies for HBV DNA integration inhibition and elimination

The commonly used treatment, such as NAs, has been recognized for its efficacy in inhibiting the production of integrated HBV DNA [71]. NAs are effective in suppressing HBV replication, thereby reducing the generation of integrated viral DNA resulting from viral replication. After entecavir (ETV) treatment, the pattern of HBV integration appears to be more random and irregular, potentially contributing to a decreased risk of HCC [162]. In a recent study, researchers procured liver tissue specimens from individuals diagnosed with chronic hepatitis B before the initiation of NAs treatment. Subsequently, they obtained liver tissue samples from the same individuals after five and ten years of continuous NAs treatment. This longitudinal analysis revealed a gradual reduction in the frequency of HBV integration events within the liver tissue over the specified treatment durations [167]. A plausible mechanism underlying this phenomenon is that NAs effectively suppress viral replication, while concomitant normal hepatocyte regeneration results in the gradual dilution of the frequency of viral integration events. However, it is important to note that NAs do not have any effect on eliminating cccDNA or integrated DNA [79, 168].

In comparison to NAs treatment, therapies that target innate immunity, such as IFN-α, are more likely to possess the potency to eliminate cccDNA [192]. This therapeutic approach has shown success in inducing a functional cure among a minority of patients with CHB in clinical settings [193]. Reports indicate that patients who are functionally cured and exhibit intrahepatic HBsAg possess higher levels of integrated HBV DNA than those without intrahepatic HBsAg. Interestingly, a certain subset of these patients maintains transcriptional activity of the integrated viral DNA [170, 171]. Utilizing spatial transcriptome sequencing, it was found that transcriptionally active HBV integration is relatively low in patients who have cleared HBsAg. In addition, there’s a close correlation between the level of intrahepatic cccDNA and virus integration events [194]. IFN-α has been shown to indirectly reduce the synthesis of pgRNA, which is vital for HBV DNA integration. Nevertheless, research is currently scant on whether IFN-α can completely eliminate integration. Thus, further exploration in this field is warranted.

The utilization of CRISPR/Cas9 has shown effectiveness in eliminating both HBV cccDNA and integrated HBV DNA [15, 176]. When selecting target sequences, it is important to optimize them to maximize the elimination of viral genes while minimizing potential damage to the human genome [195]. Following this principle, sequence design could focus on targeting the full-length 3,175-bp HBV DNA sequence [174]. Additionally, studies have explored targeting specific open reading frames of HBV, such as the S and X regions [175]. Both approaches hold promising potential for achieving a radical cure. However, the clinical application of CRISPR/Cas9 technology is currently limited due to factors like off-target cleavage and the risk of inducing genome instability when cutting integrated HBV DNA. A recently devised technique, involving the concurrent administration of Cas9 mRNA and guide RNAs, demonstrates its efficacy in modifying HBV integration DNA in mouse and tree shrew models, exhibiting a notable absence of liver enzyme elevation and minimal off-target effects [177]. A separate study put forth the hypothesis that pre-existing viral integrations within clonal HBV-infected hepatocytes could be eliminated during liver damage in patients with CHB. The researchers observed a negative correlation between the types and frequencies of breakpoints and the grade score for liver inflammation activity, providing support for this hypothesis [196].

It is intriguing to note that the majority of HBV transcripts show consistent termination sites within the viral genome, creating a unique opportunity to leverage RNA silencing mechanisms [70, 197]. RNA interference agents have emerged as a novel strategy for eradicating integrated DNA, with the potential to comprehensively influence the viral life cycle by downregulating all virus-generated mRNA [158]. One such agent, ARC-520, has shown promising results in reducing viral proteins, RNA, and DNA, leading to a surprising decrease in integrated HBV DNA in both chimpanzees and patients. However, it does not directly affect cccDNA [182].

Zinc-finger nucleases (ZFNs) or transcription activator-like effector nucleases (TALENs) have shown potential in manipulating HBV cccDNA in cellular models, which may help attenuate integration events [15, 161]. As our knowledge of cccDNA formation continues to grow, future therapeutic strategies could target nuclear enzymes, histones, and other essential components that play a crucial role in cccDNA generation [22].

Conclusion

HBV integration refers to the insertion of DNA fragments derived from HBV into the human genome [119]. The integration of HBV DNA into the human genome has been extensively studied, revealing its confirmed carcinogenic potential in various experimental models. While integration events occur early during HBV infection, their exact role in the development of HCC is yet to be fully verified.

In the future, the establishment of comprehensive animal models that encapsulate the entire HBV infection process is pivotal. Such models will afford a more nuanced exploration of the ramifications of integrated HBV DNA on the hepatocytic transformation into a carcinogenic phenotype. Moreover, the development of pragmatic and cost-efficient methodologies for detecting integrations, coupled with the identification of pertinent serological markers denoting their presence, will significantly augment our capacity to appraise the potential for attaining a functional cure.

Furthermore, given that integrated HBV DNA contributes to the production of HBsAg and may impede the realization of a functional cure, prospective research should concentrate on discerning novel serological markers that more accurately signify the presence of integrations. This is particularly imperative in patients subjected to NAs treatment, where the absence of correlation between HBsAg levels and serum HBV DNA poses challenges in monitoring the efficacy of antiviral therapy.

Moreover, for the advancement of the field, the prioritization of clinical trials assessing the efficacy of diverse treatments in expediting the clearance of HBV integrations is essential. The exploration of innovative therapeutic modalities tailored specifically to target integrated HBV DNA will be instrumental in achieving comprehensive elimination. Thus, forthcoming research endeavors should be strategically oriented toward these pivotal domains to unravel the intricacies of HBV DNA integration and pave the way for more efficacious therapeutic interventions.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

HBV:

Hepatitis B virus

CHB:

Chronic hepatitis B

HCC:

Hepatocellular carcinoma

cccDNA:

Covalently closed circular DNA

NAs:

Nucleos(t)ide analogues

IFN:

Interferon

HBsAg:

Hepatitis surface antigen

HBc:

Hepatitis B core protein

Pol:

Polymerase

dsDNA:

Double-stranded DNA

ORFs:

Open reading frames

HBcAg:

HBV core antigen

HBeAg:

HBV e antigen

HBx:

HBV X protein

rcDNA:

Relaxed circular DNA

pgRNA:

Pregenomic RNA

dslDNA:

Double-stranded linear DNA

DR1:

Direct repeat 1

DR2:

Direct repeat 2

NHEJ:

Non-homologous end joining

MMEJ:

Microhomology-mediated end joining

RNP:

Ribonucleoprotein

TP:

Terminal protein

DHBV:

Duck hepatitis B virus

WHV:

Woodchuck hepatitis virus

PHH:

Primary human hepatocytes

CTLs:

Cytotoxic T lymphocytes

FISH:

Fluorescence In Situ Hybridization

Alu-PCR:

Arthrobacter luteus-PCR

SINEs:

Short interspersed nuclear elements

invPCR:

Inverse nested PCR

WGS:

Whole-genome sequencing

WES:

Whole-exome sequencing

NGS:

Next-generation sequencing

ct-HBx:

c-terminal truncation HBV X protein

AR:

Androgen receptor

ALB:

Albumin

HBcrAg:

HBV core-related antigen

cfDNA:

Cell-free DNA

id-RNAs:

Integrant-derived RNAs

ETV:

Entecavir

ZFNs:

Zinc-finger nucleases

TALENs:

Transcription activator-like effector nucleases

References

  1. Cornberg M, Lok AS, Terrault NA, Zoulim F, Faculty E-AHTEC. Mar. Guidance for design and endpoints of clinical trials in chronic hepatitis B - Report from the 2019 EASL-AASLD HBV Treatment Endpoints Conference(double dagger). J Hepatol. 2020;72(3):539–557.

  2. Shafritz DASD, Sherman HI, Hadziyannis SJ, Kew MC. Integration of hepatitis B virus DNA into the genome of liver cells in chronic liver disease and hepatocellular carcinoma. Studies in percutaneous liver biopsies and post-mortem tissue specimens. N Engl J Med. 1981;305:1067–73.

    Article  CAS  PubMed  Google Scholar 

  3. Meyer M, Wiedorn KH, Hofschneider PH, Koshy R, Caselmann WH. A chromosome 17:7 translocation is associated with a hepatitis B virus DNA integration in human hepatocellular carcinoma DNA. Hepatology. 1992;15(4):665–71.

    Article  CAS  PubMed  Google Scholar 

  4. Wang Y, Lau SH, Sham JS-T, Wu M-C, Wang T, Guan X-Y. Characterization of HBV integrants in 14 hepatocellular carcinomas: association of truncated X gene and hepatocellular carcinogenesis. Oncogene. 2004;23(1):142–8.

    Article  CAS  PubMed  Google Scholar 

  5. Levrero M, Zucman-Rossi J. Mechanisms of HBV-induced hepatocellular carcinoma. J Hepatol Apr. 2016;64(1 Suppl):S84–101.

    Article  CAS  Google Scholar 

  6. Hai H, Tamori A, Kawada N. Role of hepatitis B virus DNA integration in human hepatocarcinogenesis. World J Gastroenterol May. 2014;28(20):6236–43.

    Article  Google Scholar 

  7. Lok AS, McMahon BJ, Brown RS Jr., et al. Antiviral therapy for chronic hepatitis B viral infection in adults: a systematic review and meta-analysis. Hepatol Jan. 2016;63(1):284–306.

    Article  CAS  Google Scholar 

  8. Lee HW, Lee JS, Ahn SH. Hepatitis B Virus Cure: targets and future therapies. Int J Mol Sci Dec 28 2020;22(1).

  9. Wang M, Qian M, Fu R, et al. The Impact of Nucleos(t)ide analogs off-Therapy among Chronic Hepatitis B patients: a systematic review and Meta-analysis. Front Public Health. 2021;9:709220.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Sorrell MF, Belongia EA, Costa J, et al. National Institutes of Health consensus development conference statement: management of hepatitis B. Hepatology. 2009;49(S5):S4–12.

  11. Ghany MG, Buti M, Lampertico P, Lee HM, Faculty A-EH-HTEC. Guidance on treatment endpoints and study design for clinical trials a iming to achieve cure in chronic hepatitis B and D: Report from the 20 22 AASLD-EASL HBV-HDV Treatment Endpoints Conference. J Hepatol. 2023/11// 2023;79(5):1254–1269.

  12. Asselah T, Loureiro D, Boyer N, Mansouri A. Targets and future direct-acting antiviral approaches to achieve hepatitis B virus cure. Lancet Gastroenterol Hepatol. 2019;4(11):883–92.

    Article  PubMed  Google Scholar 

  13. Liaw YF, Kao JH, Piratvisuth T, et al. Asian-pacific consensus statement on the management of chronic hepatitis B: a 2012 update. Hepatol Int Jun. 2012;6(3):531–61.

    Article  Google Scholar 

  14. Moreno-Cubero E, Del Arco RTS, Pena-Asensio J, de Villalobos ES, Miquel J, Larrubia JR. Is it possible to stop nucleos(t)ide analogue treatment in chronic hepatitis B patients? World J Gastroenterol May. 2018;7(17):1825–38.

    Article  Google Scholar 

  15. Fanning GC, Zoulim F, Hou J, Bertoletti A. Therapeutic strategies for hepatitis B virus infection: towards a cure. Nat Rev Drug Discovery. 2019;18(11):827–44.

    Article  CAS  PubMed  Google Scholar 

  16. Yang W, Summers J. Integration of Hepadnavirus DNA in Infected Liver: Evidence for a Linear Precursor. 1999, 73, 9710–9717. J Virol. 1999;73:9710–9717.

  17. Allweiss L, Volz T, Giersch K, et al. Proliferation of primary human hepatocytes and prevention of hepatitis B virus reinfection efficiently deplete nuclear cccDNA in vivo. Gut. 2018;67(3):542–52.

    Article  CAS  PubMed  Google Scholar 

  18. Tsukuda S, Watashi K. Hepatitis B virus biology and life cycle. Antiviral Res. 2020;(1872–9096 (Electronic)).

  19. Königer C, Wingert I, Marsmann M, Rösler C, Beck J, Nassal M. Involvement of the host DNA-repair enzyme TDP2 in formation of the covalently closed circular DNA persistence reservoir of hepatitis B viruses. Proceedings of the National Academy of Sciences. 2014;111(40).

  20. Hu J, Qi Y, Gao Z et al. DNA polymerase κ is a Key Cellular factor for the formation of covalently closed circular DNA of Hepatitis B Virus. PLoS Pathog. 2016;12(10).

  21. Siddiqui A, Kitamura K, Que L et al. Flap endonuclease 1 is involved in cccDNA formation in the hepatitis B virus. PLoS Pathog. 2018;14(6).

  22. Hu QLRY. J ea. The role of host DNA ligases in hepadnavirus covalently closed circular DNA formation. PLoS Pathogens. 2017;13(12):e1006784.

  23. Bill CA, Summers J, Genomic DNA. Double-strand breaks are targets for Hepadnaviral DNA Integration. Proc Natl Acad Sci USA. 2004;101:11135–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Freitas N, Cunha C, Menne S, Gudima SO. Envelope proteins derived from naturally integrated hepatitis B virus DNA support assembly and release of infectious hepatitis delta virus particles. J Virol May. 2014;88(10):5742–54.

    Article  Google Scholar 

  25. Blondot ML, Bruss V, Kann M. Intracellular transport and egress of hepatitis B virus. J Hepatol Apr. 2016;64(1 Suppl):S49–59.

    Article  CAS  Google Scholar 

  26. S S, DD L, D G. Mutations affecting hepadnavirus plus-strand DNA synthesis dissociate primer cleavage from translocation and reveal the origin of linear viral DNA. J Virol. 1991;65(3):1255–62.

  27. KM H DDL. The sequence of the RNA primer and the DNA template influence the initiation of plus-strand DNA synthesis in hepatitis B virus. J Mol Biol. 2007;370(3):471–80.

    Article  Google Scholar 

  28. Lee J, Shin MK, Lee HJ, Yoon G, Ryu WS. Three novel cis-acting elements required for efficient plus-strand DNA synthesis of the hepatitis B virus genome. J Virol Jul. 2004;78(14):7455–64.

    Article  CAS  Google Scholar 

  29. Lewellyn EB, Loeb DD. Base pairing between cis-acting sequences contributes to template switching during plus-strand DNA synthesis in human hepatitis B virus. J Virol Jun. 2007;81(12):6207–15.

    Article  CAS  Google Scholar 

  30. Shafritz DA, Shouval D, Sherman HI, Hadziyannis SJ, Kew MC. Integration of Hepatitis B Virus DNA into the genome of liver cells in Chronic Liver Disease and Hepatocellular Carcinoma. Studies in Percutaneous Liver biopsies and Post-mortem tissue specimens. N Engl J Med. 1981;305:1067–73.

    Article  CAS  PubMed  Google Scholar 

  31. C B, C P, B R AL. Presence of integrated hepatitis B virus DNA sequences in cellular DNA of human hepatocellular carcinoma. Nature. 1980;286:533–5.

    Article  Google Scholar 

  32. Bousali M, Karamitros T, Hepatitis B. Virus integration into transcriptionally active loci and HBV-Associated Hepatocellular Carcinoma. Microorganisms Jan 24 2022;10(2).

  33. Bousali M, Papatheodoridis G, Paraskevis D, Karamitros T, Hepatitis B, Virus DNA. Integration, chronic infections and Hepatocellular Carcinoma. Microorganisms. 2021;9(8).

  34. Tu T, Urban S. Virus entry and its inhibition to prevent and treat hepatitis B and hepatitis D virus infections. Curr Opin Virol Jun. 2018;30:68–79.

    Article  CAS  Google Scholar 

  35. Tu T, Budzinska MA, Vondran FWR, Shackel NA, Urban S, Hepatitis B, Virus DNA. Integration occurs early in the viral life cycle in an in vitro infection model via Sodium Taurocholate Cotransporting polypeptide-dependent uptake of enveloped virus particles. J Virol Jun 1 2018;92(11).

  36. Cui X, Wei W, Wang C et al. Studies on the correlation between mutation and integration of HBV in hepatocellular carcinoma. Biosci Rep Aug 28 2020;40(8).

  37. Tu T, Budzinska MA, Shackel NA, Urban S. HBV DNA integration: Molecular mechanisms and clinical implications. Viruses Apr 10 2017;9(4).

  38. Zhao LH, Liu X, Yan HX, et al. Genomic and oncogenic preference of HBV integration in hepatocellular carcinoma. Nat Commun Oct. 2016;5:7:12992.

    Article  Google Scholar 

  39. Lau C-C, Sun T, Ching Arthur KK, et al. Viral-human chimeric transcript predisposes risk to Liver Cancer Development and Progression. Cancer Cell. 2014;25(3):335–49.

    Article  CAS  PubMed  Google Scholar 

  40. Lieber MR. The mechanism of human nonhomologous DNA end joining. J Biol Chem. 2008;283(1):1–5.

    Article  CAS  PubMed  Google Scholar 

  41. Ma J-L, Kim EM, Haber JE, Lee SE. Yeast Mre11 and Rad1 proteins define a Ku-Independent mechanism to repair double-strand breaks lacking overlapping end sequences. Mol Cell Biol. 2003;23(23):8820–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Dh HVB. e. Microhomology-mediated mechanisms underlie non-recurrent disease-causing microdeletions of the FOXL2 gene or its regulatory domain. PLoS Genet. 2013;9(3):e1003358.

    Article  Google Scholar 

  43. Nassal M. HBV cccDNA: viral persistence reservoir and key obstacle for a cure of chronic hepatitis B. Gut Dec. 2015;64(12):1972–84.

    Article  CAS  Google Scholar 

  44. Sung WK, Zheng H, Li S, et al. Genome-wide survey of recurrent HBV integration in hepatocellular carcinoma. Nat Genet May. 2012;27(7):765–9.

    Article  Google Scholar 

  45. Chauhan R, Churchill ND, Mulrooney-Cousins PM, Michalak TI. Initial sites of hepadnavirus integration into host genome in human hepatocytes and in the woodchuck model of hepatitis B-associated hepatocellular carcinoma. Oncog Apr. 2017;17(4):e317.

    Google Scholar 

  46. Yang L, Ye S, Zhao X, et al. Molecular characterization of HBV DNA integration in patients with Hepatitis and Hepatocellular Carcinoma. J Cancer. 2018;9(18):3225–35.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Mason WS, Liu C, Aldrich CE, Litwin S, Yeh MM. Clonal expansion of normal-appearing human hepatocytes during chronic hepatitis B virus infection. J Virol Aug. 2010;84(16):8308–15.

    Article  CAS  Google Scholar 

  48. Li X, Zhang J, Yang Z, et al. The function of targeted host genes determines the oncogenicity of HBV integration in hepatocellular carcinoma. J Hepatol May. 2014;60(5):975–84.

    Article  Google Scholar 

  49. Horikawa I, Barrett JC. Transcriptional regulation of the telomerase hTERT gene as a target for cellular and viral oncogenic mechanisms. Carcinogenesis. 2003;24(7):1167–76.

    Article  CAS  PubMed  Google Scholar 

  50. Paterlini-Bréchot P, Saigo K, Murakami Y, et al. Hepatitis B virus-related insertional mutagenesis occurs frequently in human liver cancers and recurrently targets human telomerase gene. Oncogene. 2003;22(25):3911–6.

    Article  PubMed  Google Scholar 

  51. Saigo K, Yoshida K, Ikeda R, et al. Integration of hepatitis B virus DNA into the myeloid/lymphoid or mixed-lineage leukemia (MLL4) gene and rearrangements of MLL4 in human hepatocellular carcinoma. Hum Mutat. 2008;29(5):703–8.

    Article  CAS  PubMed  Google Scholar 

  52. Murakami Y, Saigo K, Takashima H, et al. Large scaled analysis of hepatitis B virus (HBV) DNA integration in HBV related hepatocellular carcinomas. Gut Aug. 2005;54(8):1162–8.

    Article  CAS  Google Scholar 

  53. Jiang Z, Jhunjhunwala S, Liu J, et al. The effects of hepatitis B virus integration into the genomes of hepatocellular carcinoma patients. Genome Res. 2012;22(4):593–601.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Budzinska MA, Shackel NA, Urban S, Tu T. Cellular genomic sites of Hepatitis B Virus DNA integration. Genes (Basel). Jul 20 2018;9(7).

  55. W Y. Integration of hepadnavirus DNA in infected liver: evidence for a linear precursor. J Virol. 1999;73:9710–7.

    Article  Google Scholar 

  56. Chauhan R, Shimizu Y, Watashi K, Wakita T, Fukasawa M, Michalak TI. Retrotransposon elements among initial sites of hepatitis B virus integration into human genome in the HepG2-NTCP cell infection model. Cancer Genet Jun. 2019;235–236:39–56.

    Article  Google Scholar 

  57. Mayuko Furuta HT, Yuichi Shiraishi. Characterization of HBV integration patterns and timing in liver cancer and HBV-infected livers. Oncotarget. 2018;9(38):25075–88.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Kimbi GCKA, Kew MC. Integration of hepatitis B virus DNA into chromosomal DNA during acute hepatitis B. World J Gastroenterol. 2005;11:6416–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Tsukuda S, Watashi K. Hepatitis B virus biology and life cycle. Antiviral Res. 2020;182.

  60. Hagen TM, Huang S, Curnutte J et al. Extensive oxidative DNA damage in hepatocytes of transgenic mice with chronic active hepatitis destined to develop hepatocellular carcinoma. Proc Natl Acad Sci U S A. 1994/12/20/ 91(26):12808-12.

  61. Dandri M, Burda MR, Bürkle A et al. Increase in de novo HBV DNA integrations in response to oxidative DNA damage or inhibition of poly(ADP-ribosyl)ation. Hepatology (Baltimore, Md). 2002/1// 35(1):217 – 23.

  62. Petersen J, Dandri M, Bürkle A, Zhang L, Rogler CE. Increase in the frequency of hepadnavirus DNA integrations by oxidativ e DNA damage and inhibition of DNA repair. Journal of virology. 1997/7// 1997;71(7):5455-63.

  63. Yuan T, Jiang Y, Li M, Li W. Chronic hepatitis B surface antigen seroclearance-related immune factors. Hepatol Res Jan. 2017;47(1):49–59.

    Article  CAS  Google Scholar 

  64. Boni C, Fisicaro P, Valdatta C, et al. Characterization of hepatitis B virus (HBV)-specific T-cell dysfunction in chronic HBV infection. J Virol Apr. 2007;81(8):4215–25.

    Article  CAS  Google Scholar 

  65. Wu S, Yi W, Gao Y, et al. Immune mechanisms underlying Hepatitis B Surface Antigen Seroclearance in Chronic Hepatitis B patients with viral coinfection. Front Immunol. 2022;13:893512.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Yong L, Li M, Gao Y et al. Identification of pro-inflammatory CD205 + macrophages in livers of hepatitis B virus transgenic mice and patients with chronic hepatitis B. Sci Rep. 2017;7(1).

  67. Hou X, Hao X, Zheng M, et al. CD205-TLR9-IL-12 axis contributes to CpG-induced oversensitive liver injury in HBsAg transgenic mice by promoting the interaction of NKT cells with Kupffer cells. Cell Mol Immunol. 2016;14(8):675–84.

    Article  PubMed  PubMed Central  Google Scholar 

  68. N M, H S. Hepatitis B virus replication. Trends Microbiol. 1993;1(6):221–8.

  69. Omata M, Yokosuka O, Imazeki F, et al. Correlation of hepatitis B virus DNA and antigens in the liver. A study in chronic liver disease. Gastroenterol Jan. 1987;92(1):192–6.

    CAS  Google Scholar 

  70. Wooddell CI, Yuen M-F, Chan HL-Y et al. RNAi-based treatment of chronically infected patients and chimpanzees reveals that integrated hepatitis B virus DNA is a source of HBsAg. Sci Transl Med. 2017;9(eaan0241.).

  71. Hu B, Wang R, Fu J, et al. Integration of hepatitis B virus S gene impacts on hepatitis B surface antigen levels in patients with antiviral therapy. J Gastroenterol Hepatol Jul. 2018;33(7):1389–96.

    Article  CAS  Google Scholar 

  72. Meier MA, Calabrese D, Suslov A, Terracciano LM, Heim MH, Wieland S. Ubiquitous expression of HBsAg from integrated HBV DNA in patients with low viral load. J Hepatol Oct. 2021;75(4):840–7.

    Article  CAS  Google Scholar 

  73. JC E, LB PGPV, WJ R. Integration of hepatitis B virus sequences and their expression in a human hepatoma cell. Nature. 1980;286:535–8.

    Article  Google Scholar 

  74. Yaginuma KKH, Kobayashi M, Morishima T, Matsuyama K, Koike K. Multiple integration site of hepatitis B virus DNA in hepatocellular carcinoma and chronic active hepatitis tissues from children. J Virol. 1987;61:1808–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Tokino T, Matsubara K. Chromosomal sites for hepatitis B virus integration in human hepatocellular carcinoma. J Virol. 1991;65:6761–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. MM AG. Fluorescence in situ hybridization: uses and limitations. Semin Hematol. 2000;37(4):0037–1963. (Print).

    Google Scholar 

  77. TH H, QJ Z, QD X, LP Z. Presence and integration of HBV DNA in mouse oocytes. World J Gastroenterol. 2005;11(Print):1007–9327.

    Google Scholar 

  78. HINO O, OHTAKE K, ROGLER CE. Features of two Hepatitis B Virus (HBV) DNA integrations suggest mechanisms of HBV Integration. J Virol. 1989;63:2638–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. B C, H M, Scotto Jea. State of hepatitis B virus DNA in hepatocytes of patients with hepatitis B surface antigen-positive and -negative liver diseases. Proc Natl Acad Sci U S A. 1981;78(6):3906–10.

    Article  Google Scholar 

  80. PJ B, SW S. Detection of integration during active replication of hepatitis B virus in the liver. J Med Virol. 1985;16(1):47–54.

    Article  Google Scholar 

  81. O Y, F I SK. State of HBV DNA in HBsAg-negative, anti-HCV-positive hepatocellular carcinoma: existence of HBV DNA possibly as nonintegrated form with analysis by Alu-HBV DNA PCR and conventional HBV PCR. Med Virol. 2001;64(4):410–8.

    Article  Google Scholar 

  82. Murakami Y, Minami M, Daimon Y, Okanoue T. Hepatitis B virus DNA in liver, serum, and peripheral blood mononuclear cells after the clearance of serum hepatitis B virus surface antigen. J Med Virol. 2004;72(2):203–14.

    Article  PubMed  Google Scholar 

  83. Tu H. Gao Hf Fau - Ma G-h, Ma Gh Fau - Liu Y, Liu Y. [Identification of hepatitis B virus integration sites in hepatocellular carcinoma tissues from patients with chronic hepatitis B]. (0376–2491 (Print)).

  84. Tu T, Jilbert AR. Detection of hepatocyte clones containing Integrated Hepatitis B Virus DNA using Inverse Nested PCR. Methods Mol Biol. 2017;1540:97–118.

    Article  CAS  PubMed  Google Scholar 

  85. Mason WS, Low HC, Xu C, et al. Detection of clonally expanded hepatocytes in chimpanzees with chronic hepatitis B virus infection. J Virol Sep. 2009;83(17):8396–408.

    Article  CAS  Google Scholar 

  86. Ruan P, Dai X, Sun J, et al. Integration of hepatitis B virus DNA into p21-activated kinase 3 (PAK3) gene in HepG2.2.15 cells. Virus Genes Apr. 2020;56(2):168–73.

    Article  CAS  Google Scholar 

  87. Choo KB, Liu MS, Chang PC, et al. Analysis of six distinct Integrated Hepatitis B virus sequences cloned from the Cellular DNA of a human Hepatocellular Carcinoma. Virology. 1986;154:405–8.

    Article  CAS  PubMed  Google Scholar 

  88. Ramirez R, van Buuren N, Gamelin L, et al. Targeted long-read sequencing reveals Comprehensive Architecture, Burden, and Transcriptional signatures from Hepatitis B Virus-Associated Integrations and translocations in Hepatocellular Carcinoma Cell lines. J Virol Sep. 2021;9(19):e0029921.

    Article  Google Scholar 

  89. Zheng B, Liu X-L, Fan R, et al. The Landscape of cell-free HBV Integrations and mutations in cirrhosis and Hepatocellular Carcinoma patients. Clin Cancer Res. 2021;27(13):3772–83.

    Article  CAS  PubMed  Google Scholar 

  90. Wang A, Wu L, Lin J et al. Whole-exome sequencing reveals the origin and evolution of hepato-cholangiocarcinoma. Nat Commun. 2018;9(1).

  91. Svicher V, Salpini R, Piermatteo L, et al. Whole exome HBV DNA integration is independent of the intrahepatic HBV reservoir in HBeAg-negative chronic hepatitis B. Gut Dec. 2021;70(12):2337–48.

    Article  CAS  Google Scholar 

  92. Hu X, Jiang J, Ni C, et al. HBV Integration-mediated cell apoptosis in HepG2.2.15. J Cancer. 2019;10(17):4142–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Ishii T, Tamura A, Shibata T et al. Analysis of HBV Genomes Integrated into the Genomes of Human Hepatoma PLC/PRF/5 cells by HBV sequence capture-based next-generation sequencing. Genes (Basel) Jun 18 2020;11(6).

  94. Li CL, Ho MC, Lin YY, et al. Cell-free virus-host chimera DNA from Hepatitis B Virus Integration sites as a circulating biomarker of Hepatocellular Cancer. Hepatol Dec. 2020;72(6):2063–76.

    Article  CAS  Google Scholar 

  95. Razavi-Shearer D, Gamkrelidze I, Nguyen MH, et al. Global prevalence, treatment, and prevention of hepatitis B virus infection in 2016: a modelling study. Lancet Gastroenterol Hepatol. 2018;3(6):383–403.

    Article  Google Scholar 

  96. Lander ESLL, Birren B, Nusbaum C, et al. Initial sequencing and analysis of the human genome. Nature. 2001;409:860–921.

    Article  CAS  PubMed  Google Scholar 

  97. Erken R, Loukachov V, van Dort K, et al. Quantified integrated hepatitis B virus is related to viral activity in patients with chronic hepatitis B. Hepatol Jul. 2022;76(1):196–206.

    Article  CAS  Google Scholar 

  98. Larsson SB, Tripodi G, Raimondo G, et al. Integration of hepatitis B virus DNA in chronically infected patients assessed by Alu-PCR. J Med Virol Oct. 2018;90(10):1568–75.

    Article  CAS  Google Scholar 

  99. Gong SS, Jensen AD, Wang H. Rogler duck hepatitis B virus integrations in LMH chicken hepatoma cells: identification and characterization of new episomally derived integrations. J Virol. 1995;69:8102–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Yan H, Yang Y, Zhang L, et al. Characterization of the genotype and integration patterns of hepatitis B virus in early- and late‐onset hepatocellular carcinoma. Hepatology. 2015;61(6):1821–31.

    Article  CAS  PubMed  Google Scholar 

  101. Budzinska MA, Shackel NA, Urban S, Tu T. Sequence analysis of integrated hepatitis B virus DNA during HBeAg-seroconversion. Emerg Microbes Infect Aug. 2018;8(1):142.

    Google Scholar 

  102. Mason WS, Gill US, Litwin S, et al. HBV DNA integration and clonal hepatocyte expansion in Chronic Hepatitis B patients considered Immune Tolerant. Gastroenterol Nov. 2016;151(5):986–e9984.

    Article  CAS  Google Scholar 

  103. Tu T, Mason WS, Clouston AD, et al. Clonal expansion of hepatocytes with a selective advantage occurs during all stages of chronic hepatitis B virus infection. J Viral Hepat Sep. 2015;22(9):737–53.

    Article  CAS  Google Scholar 

  104. Li W, Qi Y, Xu H, Wei W, Cui X. Profile of different Hepatitis B virus integration frequency in hepatocellular carcinoma patients. Biochem Biophys Res Commun May. 2021;14:553:160–4.

    Article  CAS  Google Scholar 

  105. Beerenwinkel N, Gunthard HF, Roth V, Metzner KJ. Challenges and opportunities in estimating viral genetic diversity from next-generation sequencing data. Front Microbiol. 2012;3:329.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Li W, Wei W, Hou F, Xu H, Cui X. The integration model of hepatitis B virus genome in hepatocellular carcinoma cells based on high-throughput long-read sequencing. Genomics Jan. 2022;114(1):23–30.

    Article  CAS  Google Scholar 

  107. Betz-Stablein BD, Topfer A, Littlejohn M, et al. Single-molecule sequencing reveals Complex Genome Variation of Hepatitis B Virus during 15 years of chronic infection following liver transplantation. J Virol Aug. 2016;15(16):7171–83.

    Article  Google Scholar 

  108. Yang C, Ruan P, Ou C et al. Chronic hepatitis B virus infection and occurrence of hepatocellular carcinoma in tree shrews (Tupaia belangeri chinensis). Virol J. 2015;12(1).

  109. Walter E, Keist R, Niederöst B, Pult I, Blum HE. Hepatitis B virus infection of tupaia hepatocytesin vitroandin vivo. Hepatology. 1996;24(1):1–5.

    CAS  PubMed  Google Scholar 

  110. Burwitz BJ, Wettengel JM, Mück-Häusl MA et al. Hepatocytic expression of human sodium-taurocholate cotransporting polypeptide enables hepatitis B virus infection of macaques. Nat Commun. 2017;8(1).

  111. Tan AT, Yang N, Lee Krishnamoorthy T, et al. Use of expression profiles of HBV-DNA Integrated into Genomes of Hepatocellular Carcinoma Cells to select T cells for Immunotherapy. Gastroenterol May. 2019;156(6):1862–e18769.

    Article  CAS  Google Scholar 

  112. Wang SH, Yeh SH, Chen PJ. Unique Features of Hepatitis B Virus-Related Hepatocellular Carcinoma in Pathogenesis and clinical significance. Cancers (Basel) May 18 2021;13(10).

  113. Kuang XJ, Jia RR, Huo RR, et al. Systematic review of risk factors of hepatocellular carcinoma after hepatitis B surface antigen seroclearance. J Viral Hepat Sep. 2018;25(9):1026–37.

    Article  CAS  Google Scholar 

  114. Zhao XL, Yang JR, Lin SZ, et al. Serum viral duplex-linear DNA proportion increases with the progression of liver disease in patients infected with HBV. Gut Mar. 2016;65(3):502–11.

    Article  CAS  Google Scholar 

  115. Jang JW, Kim JS, Kim HS, et al. Persistence of intrahepatic hepatitis B virus DNA integration in patients developing hepatocellular carcinoma after hepatitis B surface antigen seroclearance. Clin Mol Hepatol Jan. 2021;27(1):207–18.

    Article  Google Scholar 

  116. Lau KC, Joshi SS, Gao S, et al. Oncogenic HBV variants and integration are present in hepatic and lymphoid cells derived from chronic HBV patients. Cancer Lett Jun. 2020;28:480:39–47.

    Article  Google Scholar 

  117. Amaddeo G, Cao Q, Ladeiro Y, et al. Integration of tumour and viral genomic characterizations in HBV-related hepatocellular carcinomas. Gut May. 2015;64(5):820–9.

    Article  CAS  Google Scholar 

  118. Peneau C, Imbeaud S, La Bella T, et al. Hepatitis B virus integrations promote local and distant oncogenic driver alterations in hepatocellular carcinoma. Gut Mar. 2022;71(3):616–26.

    Article  CAS  Google Scholar 

  119. Yeh SH, Li CL, Lin YY, et al. Hepatitis B Virus DNA Integration drives carcinogenesis and provides a New Biomarker for HBV-related HCC. Cell Mol Gastroenterol Hepatol Jan. 2023;20(4):921–9.

    Article  Google Scholar 

  120. Lu HZYW. L ea. HBV-integrated local genomic alterations reveal multicentric independent occurrences of multifocal HCC. Clin Transl Med 2023;13(6):e1313.

  121. Alvarez EG, Demeulemeester J, Otero P, et al. Aberrant integration of Hepatitis B virus DNA promotes major restructuring of human hepatocellular carcinoma genome architecture. Nat Commun Nov. 2021;25(1):6910.

    Article  Google Scholar 

  122. Brunner SF, Roberts ND, Wylie LA, et al. Somatic mutations and clonal dynamics in healthy and cirrhotic human liver. Nature. 2019;574(7779):538–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Saitta C, Tripodi G, Barbera A, et al. Hepatitis B virus (HBV) DNA integration in patients with occult HBV infection and hepatocellular carcinoma. Liver Int Oct. 2015;35(10):2311–7.

    Article  CAS  Google Scholar 

  124. Kremsdorf D, Soussan P, Paterlini-Brechot P, Brechot C. Hepatitis B virus-related hepatocellular carcinoma: paradigms for viral-related human carcinogenesis. Oncogene Jun. 2006;26(27):3823–33.

    Article  Google Scholar 

  125. Ruan P, Dai X, Sun J, et al. Different types of viral–host junction found in HBV integration breakpoints in HBV–infected patients. Mol Med Rep Feb. 2019;19(2):1410–6.

    CAS  Google Scholar 

  126. Kojima R, Nakamoto S, Kogure T et al. Re-analysis of hepatitis B virus integration sites reveals potential new loci associated with oncogenesis in hepatocellular carcinoma. World J Virol. 2023/06/25/ 2023;12(3):209–220.

  127. Kang Hyo J, Haq F, Sung Chang O, et al. Characterization of Hepatocellular Carcinoma patients with FGF19 amplification assessed by fluorescence in situ hybridization: a large cohort study. Liver Cancer. 2019;8(1):12–23.

    Article  PubMed  Google Scholar 

  128. Cui X, Li Y, Xu H, Sun Y, Jiang S, Li W. Characteristics of Hepatitis B virus integration and mechanism of inducing chromosome translocation. NPJ Genom Med. 2023/06/02/ 2023;8(1):11.

  129. Ruan P, Zhou R, He C et al. Two fragments of HBV DNA integrated into chrX: 11009033 and its genetic regulation in HepG2.2.15. Molecular Medicine Reports. 2023/05// 2023;27(5):98.

  130. Deng Y, Chang S. Role of telomeres and telomerase in genomic instability, senescence and cancer. Lab Invest. 2007;87(11):1071–6.

    Article  CAS  PubMed  Google Scholar 

  131. Maciejowski J, de Lange T. Telomeres in cancer: tumour suppression and genome instability. Nat Rev Mol Cell Biol Mar. 2017;18(3):175–86.

    Article  CAS  Google Scholar 

  132. Sze KMF, Ho DWH, Chiu YT, et al. Hepatitis B virus–telomerase reverse transcriptase promoter integration harnesses host ELF4, resulting in Telomerase Reverse transcriptase gene transcription in Hepatocellular Carcinoma. Hepatology. 2020;73(1):23–40.

    Article  PubMed  Google Scholar 

  133. Giosa D, Lombardo D, Musolino C et al. Mitochondrial DNA is a target of HBV integration. Commun Biol. 2023/07/03/ 2023;6(1):684.

  134. Bakhoum SF, Cantley LC. The multifaceted role of chromosomal instability in Cancer and its Microenvironment. Cell. 2018;174(6):1347–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Katoh H, Shibata T, Kokubu A, et al. Epigenetic instability and chromosomal instability in Hepatocellular Carcinoma. Am J Pathol. 2006;168(4):1375–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Bailey SM. Telomeres, chromosome instability and cancer. Nucleic Acids Res. 2006;34(8):2408–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Rosato RNDB. HBV infection and host interactions: the role in viral persistence and oncogenesis. Int J Mol Sci. 2023;24(8):7651.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Tavis JE, Na B, Huang Z et al. Transgenic expression of entire Hepatitis B Virus in mice induces Hepatocarcinogenesis Independent of Chronic Liver Injury. PLoS ONE. 2011;6(10).

  139. Barone M, Spano D, D’Apolito M, et al. Gene Expression Analysis in HBV Transgenic Mouse Liver: a model to study early events related to Hepatocarcinogenesis. Mol Med. 2006;12(4–6):115–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Wang ML, Tang H. Nucleos(t)ide analogues causes HBV S gene mutations and carcinogenesis. (1499–3872 (Print)).

  141. Choi Y-M, Lee S-Y, Kim B-J. Naturally occurring Hepatitis B virus mutations leading to endoplasmic reticulum stress and their contribution to the Progression of Hepatocellular Carcinoma. Int J Mol Sci. 2019;20(3).

  142. ML W, DB W, YC T, et al. The truncated mutant HBsAg expression increases the tumorigenesis of hepatitis B. Virol J. 2018;15(1):1743–61.

    Google Scholar 

  143. SA L, K K. Nucleotide change of codon 182 in the surface gene of hepatitis B virus genotype. J Hepatol. 2012;56(1):63–9. (1600 – 0641 (Electronic)).

    Article  Google Scholar 

  144. Ma N-F, Lau SH, Hu L, et al. COOH-Terminal truncated HBV X protein plays key role in Hepatocarcinogenesis. Clin Cancer Res. 2008;14(16):5061–8.

    Article  CAS  PubMed  Google Scholar 

  145. Wu X, Ni Z, Song T et al. C-Terminal truncated HBx facilitates oncogenesis by modulating cell cycle and glucose metabolism in FXR-Deficient Hepatocellular Carcinoma. Int J Mol Sci. 2023;24(6).

  146. Sze KMF, Chu GKY, Lee JMF, Ng IOL. C-terminal truncated hepatitis B virus x protein is associated with metastasis and enhances invasiveness by c-jun/matrix metalloproteinase protein 10 activation in hepatocellular carcinoma. Hepatology. 2013;57(1):131–9.

    Article  CAS  PubMed  Google Scholar 

  147. Zhang Y, Yan Q, Gong L, et al. C-terminal truncated HBx initiates hepatocarcinogenesis by downregulating TXNIP and reprogramming glucose metabolism. Oncogene. 2020;40(6):1147–61.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Quetier I, Brezillon N, Revaud J, et al. C-terminal-truncated hepatitis B virus X protein enhances the development of diethylnitrosamine-induced hepatocellular carcinogenesis. J Gen Virol. 2015;96(3):614–25.

    Article  CAS  PubMed  Google Scholar 

  149. Zhang C, Xiao C, Ren G et al. C-terminal-truncated hepatitis B virus X protein promotes hepatocarcinogenesis by activating the MAPK pathway. Microb Pathog. 2021;159.

  150. KY N. C-terminal truncated hepatitis B virus X protein promotes hepatocellular. Oncotarget. 2016;26(7):1949–2553. (Electronic)).

    Google Scholar 

  151. Guo M, Zhao L, Jiang C et al. Multiomics analyses reveal pathological mechanisms of HBV infection and integration in liver cancer. 2023;(1096–9071 (Electronic)).

  152. Pollicino T, Caminiti G. HBV-Integration studies in the clinic: role in the natural history of infection. Viruses Feb 26 2021;13(3).

  153. Luo M, Zhou B, Hou J, Jiang D. Biomarkers for predicting nucleos(t)ide analogs discontinuation and hepatitis B virus recurrence after drug withdrawal in chronic hepatitis B patients. Hepatol Res Apr. 2022;52(4):337–51.

    Article  CAS  Google Scholar 

  154. Xie Y, Li M, Ou X, et al. HBeAg-positive patients with HBsAg < 100 IU/mL and negative HBV RNA have lower risk of virological relapse after nucleos(t)ide analogues cessation. J Gastroenterol Sep. 2021;56(9):856–67.

    Article  CAS  Google Scholar 

  155. DM M. Diagnostic and prognostic value of circulating tumor-related DNA in cancer patients. Expert Rev Mol Diagn. 2013;13(Electronic):1744–8352.

    Google Scholar 

  156. Chen W, Zhang K, Dong P, et al. Noninvasive chimeric DNA profiling identifies tumor-originated HBV integrants contributing to viral antigen expression in liver cancer. Hepatol Int May. 2020;14(3):326–37.

    Article  Google Scholar 

  157. Li C-L, Yeh S-H, Chen P-J. Circulating virus–host chimera DNAs in the clinical monitoring of Virus-related cancers. Cancers. 2022;14(10).

  158. Lindh M, Rydell GE, Larsson SB. Impact of integrated viral DNA on the goal to clear hepatitis B surface antigen with different therapeutic strategies. Curr Opin Virol Jun. 2018;30:24–31.

    Article  CAS  Google Scholar 

  159. Zaiets I, Gunewardena S, Menne S, Weinman SA, Gudima SO. Sera of Individuals Chronically Infected with Hepatitis B Virus (HBV) Contain Diverse RNA Types Produced by HBV Replication or Derived from Integrated HBV DNA. Journal of Virology. 2023/03/30/ 2023;97(3):e01950-22.

  160. Gehring AJ, Protzer U. Targeting Innate and Adaptive Immune responses to Cure Chronic HBV infection. Gastroenterology. 2019;156(2):325–37.

    Article  CAS  PubMed  Google Scholar 

  161. Maepa M, Roelofse I, Ely A, Arbuthnot P. Progress and prospects of Anti-HBV Gene Therapy Development. Int J Mol Sci. 2015;16(8):17589–610.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Zhang M, Zhang H, Cheng X, et al. Liver biopsy of chronic hepatitis B patients indicates HBV integration profile may complicate the endpoint and effect of entecavir treatment. Antiviral Res Aug. 2022;204:105363.

    Article  CAS  Google Scholar 

  163. Chen H, Mu M, Liu Q, et al. Hepatocyte endoplasmic reticulum stress inhibits Hepatitis B Virus Secretion and Delays Intracellular Hepatitis B Virus Clearance after Entecavir Treatment. Front Med (Lausanne). 2020;7:589040.

    Article  PubMed  Google Scholar 

  164. Lai CL, Wong DK, Wong GT, Seto WK, Fung J, Yuen MF. Rebound of HBV DNA after cessation of nucleos/tide analogues in chronic hepatitis B patients with undetectable covalently closed. JHEP Rep Jun. 2020;2(3):100112.

    Google Scholar 

  165. Hsu Y-C, Suri V, Nguyen MH et al. Inhibition of Viral Replication Reduces Transcriptionally Active Distinct Hepatitis B Virus Integrations With Implications on Host Gene Dysregulation. Gastroenterology. 2022/04// 2022;162(4):1160–1170.e1.

  166. Yip TC, Wong GL, Chan HL, et al. HBsAg seroclearance further reduces hepatocellular carcinoma risk after complete viral suppression with nucleos(t)ide analogues. J Hepatol Mar. 2019;70(3):361–70.

    Article  CAS  Google Scholar 

  167. Chow N, Wong D, Lai C-L et al. Effect of Antiviral Treatment on Hepatitis B Virus Integration and Hepatocyte Clonal Expansion. Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America. 2023/02/08/ 2023;76(3):e801-e809.

  168. Baran B. Nucleos(t)ide analogs in the prevention of hepatitis B virus related hepatocellular carcinoma. World J Hepatol Jul. 2015;8(13):1742–54.

    Article  Google Scholar 

  169. Wen X, Wu X, Sun Y et al. Long-term antiviral therapy is associated with changes in the profile of transcriptionally active HBV integration in the livers of patients with CHB. J Med Virol. 2024;96(6).

  170. Gan W, Gao N, Gu L et al. Reduction in Intrahepatic cccDNA and Integration of HBV in Chronic Hepatitis B Patients with a Functional Cure. Journal of Clinical and Translational Hepatology. 2023/04/28/ 2023;11(2):314–322.

  171. Gao N, Guan G, Xu G et al. Integrated HBV DNA and cccDNA maintain transcriptional activity in intrahepatic HBsAg-positive patients with functional cure following PEG-IFN-based therapy. Alimentary Pharmacology & Therapeutics. 2023/08/29/ 2023:apt.17670.

  172. Zhang J, Zhang Y, Khanal S et al. Synthetic gRNA/Cas9 ribonucleoprotein targeting HBV DNA inhibits viral replication. Journal of Medical Virology. 2023/07// 2023;95(7):e28952.

  173. Seeger C, Sohn JA, Targeting Hepatitis B. Virus with CRISPR/Cas9. Mol Ther Nucleic Acids Dec. 2014;16(12):e216.

    Article  Google Scholar 

  174. Li H, Sheng C, Wang S, et al. Removal of Integrated Hepatitis B Virus DNA using CRISPR-Cas9. Front Cell Infect Microbiol. 2017;7:91.

    Article  PubMed  PubMed Central  Google Scholar 

  175. Karimova M, Beschorner N, Dammermann W, et al. CRISPR/Cas9 nickase-mediated disruption of hepatitis B virus open reading frame S and X. Sci Rep. Sep 2015;3:5:13734.

    Article  Google Scholar 

  176. Ramanan V, Shlomai A, Cox DB, et al. CRISPR/Cas9 cleavage of viral DNA efficiently suppresses hepatitis B virus. Sci Rep Jun. 2015;2:5:10833.

    Article  Google Scholar 

  177. Yi J, Lei X, Guo F et al. Co-delivery of Cas9 mRNA and guide RNAs edits hepatitis B virus episomal and integration DNA in mouse and tree shrew models. 2023;(1872–9096 (Electronic)).

  178. Gao L, Yang J, Feng J, et al. PreS/2-21-Guided siRNA nanoparticles target to Inhibit Hepatitis B Virus infection and replication. Front Immunol. 2022;13:856463.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Hui RW, Mak LY, Seto WK, Yuen MF. RNA interference as a novel treatment strategy for chronic hepatitis B infection. Clin Mol Hepatol Jul. 2022;28(3):408–24.

    Article  Google Scholar 

  180. Teng Y, Xu Z, Zhao K, et al. Novel function of SART1 in HNF4alpha transcriptional regulation contributes to its antiviral role during HBV infection. J Hepatol Nov. 2021;75(5):1072–82.

    Article  CAS  Google Scholar 

  181. Wang J, Chen R, Zhang R, et al. The gRNA-miRNA-gRNA Ternary Cassette combining CRISPR/Cas9 with RNAi Approach strongly inhibits Hepatitis B Virus Replication. Theranostics. 2017;7(12):3090–105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Wooddell CI, Gehring AJ, Yuen MF, Given BD. RNA interference therapy for chronic Hepatitis B predicts the importance of addressing viral integration when developing Novel cure strategies. Viruses Mar 30 2021;13(4).

  183. Yuen MF, Schiefke I, Yoon JH, et al. RNA interference therapy with ARC-520 results in prolonged Hepatitis B Surface Antigen response in patients with chronic Hepatitis B infection. Hepatol Jul. 2020;72(1):19–31.

    Article  CAS  Google Scholar 

  184. T D, S N, A E, P A, K B. Improved antiviral efficacy using TALEN-mediated homology directed recombination to introduce artificial primary miRNAs into DNA of hepatitis B virus. Biochem Biophys Res Commun. 2016;478(4):1563–8.

  185. Bloom K, Ely A, Mussolino C, Cathomen T, Arbuthnot P. Inactivation of hepatitis B virus replication in cultured cells and in vivo with engineered transcription activator-like effector nucleases. Mol Ther Oct. 2013;21(10):1889–97.

    Article  CAS  Google Scholar 

  186. Chen J, Zhang W, Lin J, et al. An efficient antiviral strategy for targeting hepatitis B virus genome using transcription activator-like effector nucleases. Mol Ther Feb. 2014;22(2):303–11.

    Article  Google Scholar 

  187. Cradick TJ, Keck K, Bradshaw S, Jamieson AC, McCaffrey AP. Zinc-finger nucleases as a novel therapeutic strategy for targeting hepatitis B virus DNAs. Mol Ther May. 2010;18(5):947–54.

    Article  CAS  Google Scholar 

  188. Kasianchuk N, Dobrowolska K, Harkava S et al. Gene-editing and RNA interference in treating Hepatitis B: a review. Viruses Dec 8 2023;15(12).

  189. Schiffer JT, Swan DA, Stone D, Jerome KR. Predictors of hepatitis B cure using gene therapy to deliver DNA cleavage enzymes: a mathematical modeling approach. PLoS Comput Biol. 2013;9(7):e1003131.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Weber ND, Stone D, Jerome KR. TALENs targeting HBV: designer endonuclease therapies for viral infections. Mol Ther Oct. 2013;21(10):1819–20.

    Article  CAS  Google Scholar 

  191. Smith T, Singh P, Chmielewski KO et al. Improved specificity and safety of Anti-hepatitis B Virus TALENs using Obligate Heterodimeric FokI nuclease domains. Viruses Jul 12 2021;13(7).

  192. F Z SL. Targeting innate immunity: a new step in the development of combination therapy for chronic hepatitis B. Durantel DGastroenterology. 2013;144(7):1342–4.

    Google Scholar 

  193. Li Q, Sun B, Zhuo Y et al. Interferon and interferon-stimulated genes in HBV treatment. Front Immunol. 2022;13.

  194. Yu X, Gong Q, Yu DA-O et al. Spatial transcriptomics reveals a low extent of transcriptionally active hepatitis B virus integration in patients with HBsAg loss. Gut. 2023;(1468–3288 (Electronic)):Published online November 15, 2023.

  195. Horodecka K, Düchler M. CRISPR/Cas9: Principle, Applications, and delivery through Extracellular vesicles. Int J Mol Sci. 2021;22(11).

  196. Hu G, Huang MX, Li WY, Gan CJ, Dong WX, Peng XM. Liver damage favors the eliminations of HBV integration and clonal hepatocytes in chronic hepatitis B. Hepatol Int Feb. 2021;15(1):60–70.

    Article  Google Scholar 

  197. Hu B, Zhong L, Weng Y et al. Therapeutic siRNA: state of the art. Signal Transduct Target Therapy. 2020;5(1).

Download references

Acknowledgements

Not applicable.

Funding

This work is supported by the National Key Research and Development Program of China (No.2022YFC2304800) and 1.3.5 project for disciplines of excellence, West China Hospital, Sichuan University (No.ZYGD23030).

Author information

Authors and Affiliations

Authors

Contributions

MZ and HC conceived the manuscript; MZ, HC and HT searched for the papers and made the outline; MZ and HC wrote the initial draft; MZ, HC and HL designed and drew the figures; MZ, HC and HL checked all references and formatting; MZ and HC contributed equally to this work. All authors revised and contributed to the final version of the manuscript.

Corresponding author

Correspondence to Hong Tang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, M., Chen, H., Liu, H. et al. The impact of integrated hepatitis B virus DNA on oncogenesis and antiviral therapy. Biomark Res 12, 84 (2024). https://doi.org/10.1186/s40364-024-00611-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40364-024-00611-y

Keywords