Open Access

HBV integrated genomic characterization revealed hepatocyte genomic alterations in HBV‑related hepatocellular carcinomas

  • Authors:
    • Ming Yang
    • Guiqi Yang
    • Fengyan Li
    • Minglin Ou
    • Chunhong Li
    • Jiejing Chen
    • Hua Lin
    • Yue Zhang
    • Wen Xue
    • Yan Wu
    • Yong Xu
    • Weiguo Sui
    • Yong Dai
  • View Affiliations

  • Published online on: October 1, 2020     https://doi.org/10.3892/mco.2020.2149
  • Article Number: 79
  • Copyright: © Yang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatocellular carcinoma (HCC) is one of the most lethal malignancies that is closely associated with the Hepatitis B virus (HBV). HBV integration into host genomes can induce instability and the aberrant expression of human genomic DNA. To directly assess HBV integration breakpoints at whole genome level, four small sequencing libraries were constructed and the HBV integration profiles of four patients with HCC were characterized. In total, the current study identified 11,800,974, 11,216,998, 11,026,546 and 11,607,842 clean reads for patients 1‑3 and 4, respectively, of which 92.82, 95.95, 97.21 and 97.29% were properly aligned to the hybrid reference genome. In addition, 220 HBV integration events were detected from the tumor tissues of four patients with HCC and an average of 55 breakpoints per sample was calculated. The results indicated that HBV integration events may be implicated in HCC physiologies and diseases. The results acquired may also provide insight into the pathogenesis of HCC, which may be valuable for future HCC therapy.

Introduction

Hepatocellular carcinoma (HCC), one of the highest prevalence of liver cancer, caused by hepatitis B virus (HBV), and more than 350 million people were affected by HBV worldwide (1,2). All over the world, majority of HBV-related deaths were closely associated with HCC for each year, which indicates the third leading cause of cancer deaths (2,3). As we known, the HBV was a major etiologic agent for HCC, and prevalent in China, Southeast Asia and sub-Saharan Africa (4). Notably, an increasing number of studies indicated that HBV was implicated in tumorigenesis, of which the main mechanism as follows (3-7): i) Expression of viral proteins, in particular, from HBV gene X (HBx), to modulate cell proliferation and viability, ii) accumulation of genetic damage due to hepatic inflammation mediated by virus-specific T cells, and iii) it is intriguing that integration of HBV DNA into the host genome to alter the function of endogenous genes or induce chromosomal instability.

There are increasing evidences that the events of the HBV integration occurred in the HCC, and affected function of HCC genome. Some researchers found that HBV DNA integration distributed different chromosomal sites in the host genome (8), and they could trigger chromosomal changes, genomic instability, or changes in the expression of human genes (9). It was reported that HBV integration events are involved in chromosome fragile sites or repetitive sequences and are usually followed by local rearrangement, all of which relate to higher genomic instability (10). HBV insertion was found to target the retinoic acid receptor-β (RARB) gene or the human cyclin A2 (CCNA2) and to generate chimeric oncogenic proteins (11). Until now, all reports involving HBV DNA integration implied that integration plays a role in the transformation (12-14). HBV integration into the host cell genome has also been reported, which resulted in gene mutations, insertions, deletions or rearrangements of the host genome (15-17). Recently, a great number of insertion sites were identified by next-generation sequencing (4,18,19). Interestingly, telomerase reverse transcriptase (TERT) and mixed-lineage leukemia 4 (MLL4) gene were frequently targeted by HBV in HCC tissue, and the latter may play a major role in HCC carcinogenesis (20). Since firstly discovered at the TERT gene, HBV integration breakpoints have been widely reported in some gene targets, such as the fatty acyl CoA reductase 2 (FAR2), inositol triphosphate receptor type 1 (ITPR1,IP3R1), Interleukin-1-receptor-associated kinase 2 (IRAK2), mitogen associated protein kinase 1 (MAPK1), mixed-lineage leukemia 2 (MLL2) and MLL4 genes (2,21,22). Although HBV integration event has been reported in the HCC, and its mechanism is not clear. Therefore, systematic analysis of HBV integration targets could elucidate HCC physiologies and diseases development processes, and predict novel therapeutic targets. Here, to directly detect HBV integration breakpoints at whole genome level, we constructed four small sequencing libraries and characterized the HBV integration profiles from four patients with HCC. Therefore, our research revealed HBV integration events in the HCC, and acquired insight into the pathogenesis of HCC.

Materials and methods

Human samples and DNA extractions

Four tumor samples were collected from the patients who underwent curative primary hepatectomy or liver transplantation in the Guilin No. 924 Hospital. They were have precisely diagnosed with HCC and associated with HBV infection in the department of pathology, and the hepatitis B surface antigen (HBsAg) was positively expressed and HBV-DNA quantification was greater than 103 copies/ml (Fig. 1A and B and Table I). Subsequently, total DNA was extracted from the tumor samples using QIAamp DNA Micro kit (Qiagen Ltd.) according to the manufacturer's methods. Informed consent was obtained from the participant donors, and the protocol for the research project has been approved by a Ethics Committee of the Guilin No. 924 Hospital accord with China's Guidelines.

Table I

Clinical and biochemical characteristics of the four patients prior to whole-genome sequencing.

Table I

Clinical and biochemical characteristics of the four patients prior to whole-genome sequencing.

IDAge (years)SexHBsAgAnti-HBsAnti-HBcAFPHBV (copies/ml)
138Male+-++9,337
252Male+-++6,060
342Male+-++3,504
450Male+-++2,139

[i] HBsAg, hepatitis B surface antigen; Anti-HBs, antibody to hepatitis B surface antigen; Anti-HBc, antibody to hepatitis B core antigen; AFP, α-fetoprotein; HBV, hepatitis B virus.

Hybridized Libraries construction and sequencing

DNA purification and library preparation were conducted as previously reported methods (2). Brief, integrity and quality of total DNA were evaluated by the Qubit Fluorometer and agarose gel electrophoresis (Fig. 1). Next, the genomic DNA were fragmented randomly by a Bioruptor Pico (Diagenode, B01060001) into the target DNA fragments (170 bp) from 3 µg of total DNA. And then, the target fragments were subjected to perform DNA end repair, and added a single ‘A’ nucleotide in the 3' ends of the target DNA fragments. Subsequently, PCR were performed after adapters ligating, size-selection and tailed random primers addition to obtain sufficient amplification products for libraries construction. For these constructed libraries, we used the virus probe to hybridize, and enriched the target DNA fragments. The hybridized target DNA fragments were eluted using AW2 Buffer in Elution column (QIAamp minElute Column) And then, PCR were performed to obtain sufficient amplification products for the hybridized libraries construction. PCR products were collected and preceded to 101 cycle's paired-end index sequencing in the Illumina HiSeq 2,000 sequencer according to manufacturer's instructions (Illumina Inc.).

Hybrid reference genome construction and alignment analysis

The advanced analysis begins with raw data generated from the Illumina platform. The sequence tags with adapter ligation or low quality or Ns and low base quality were filtered out to obtain clean tags, which were subjected to further analyze. Next, we combined the human reference genome (hg19) and the HBV genome (NC_003977.1) together to build a hybrid reference genome for alignment analysis. The clean tags were aligned to the hybrid reference genome by Burrows-Wheeler Aligner (BWA) (23), and the alignment results were saved in BAM format files. These files were further preprocessed to be the final BAM files for the HBV integration detection, such as sorting according to the alignment position, marking duplicate reads caused by PCR.

SNPs and InDels detection and annotation

Further bioinformatic analysis for the final BAM files, single Nucleotide Polymorphisms (SNPs) are detected by GATK (24). The unique genotypes were identified for each individual, which has the highest probability at a given locus, the consensus sequence tags were collected and saved as CNS format. And then, the high confidence SNP datasets were acquired by filtering the consensus sequence tags. In addition, we detect the small Insertion and Deletion (InDels) using pair-end reads for gap alignment.

HBV integration detection

We detected the HBV integrations by seeksv, and the seeksv was an in-house tool. The HBV integration positions were filtered, because of the sequences with a sum of junction read number and abnormal read pair number smaller than 2. After integration positions identification, the distribution of the HBV integrations was analyzed, which could evaluate the numbers of integrations.

Results

Analysis of sequencing data from four tumor samples

Integrated genomic DNA was captured from tumor tissues of four patients with HCC and sequenced by Illumina HiSeq 2,000. Before doing any further analysis, quality control is essential to detect whether the data is qualified. As shown in Fig. 2, the sequencing data was good, which satisfied the subsequent advanced analysis. These raw reads with adapters, low quality and unknown bases were removed. The remaining data were called as clean read data, which were submitted for further bioinformatic analysis. In total, an average of 11,413,090 raw reads were obtained for each sample. A Phred quality score is a measure that was used to evaluate the quality for sequencing data. Phred quality scores sQ were defined as the sequencing data quality from tumor tissues of four patients, and the E indicated the sequencing error rate. They had a relationship as follows: sQ=-10log10E. It's worth noting that the GC content accounted for 43.68-44.64% when the Phred score was >30.

Further statistical analysis for clean reads, we identified 11,800,974, 11,216,998, 11,026,546 and 11,607,842 clean reads for Patient 1-3 and 4, of which 92.82, 95.95, 97.21 and 97.29%, respectively, were properly aligned to the hybrid reference genome. The average sequencing depths were 0.07, 0.08, 0.05 and 0.07-fold, and 5.47, 6.17, 3.63 and 4.93 of the hybrid reference genome were covered by the clean reads, respectively. Coverage at least 20-fold were 0.01% for four tumor tissues (Table II).

Table II

Quality control results of alignment data.

Table II

Quality control results of alignment data.

CategoriesPatient 1Patient 2Patient 3Patient 4
Clean reads11,800,97411,216,99811,026,54611,607,842
Clean bases (bp)1,180,097,4001,121,699,8001,102,654,6001,160,784,200
Mapped reads10,953,11310,763,24310,718,36011,293,616
Mapped bases (bp)1,075,438,1281,058,305,7011,049,564,6761,108,297,233
Mapping rate (%)92.8295.9597.2197.29
Uniq reads10,295,38010,118,51310,040,85210,627,022
Uniq bases (bp)1,012,882,982996,468,729985,514,3901,045,019,717
Unique rate (%)94.0094.0193.6894.10
Duplicate reads8,692,6178,282,5759,152,3009,265,976
Duplicate rate (%)79.3676.9585.3982.05
Mismatch bases (bp)5,924,6514,942,5015,471,0274,874,040
Mismatch rate (%)0.550.470.520.44
Average sequencing depth0.070.080.050.07
Coverage (%)5.476.173.634.93
Coverage at least 4X (%)0.050.050.050.05
Coverage at least 10X (%)0.020.020.020.02
Coverage at least 20X (%)0.010.010.010.01
HBV integration analysis for four tumor tissues

In order to explore the HBV integration events in the HCC patients, we used the seeksv to detect the HBV integration sites, and conducted the annotation and classification by ANNOVAR. In total, the 220 HBV integration events were detected from the tumor tissues of four HCC patients, and an average of 55 breakpoints for each sample. The integration breakpoints distributions for four samples were 143, 40, 25 and 12, respectively (Fig. 3), which evaluated the numbers of integrations located in different gene regions. Our results indicated that the HBV integration breakpoints had a preference for chromosomes 2, 3 and 5 (Table III). Majority of HBV breakpoints in HCC were found near coding genes (116 of 220 breakpoints). Eight of the 220 HBV breakpoints were located in known coding genes, such as ITGA9, FAM19A4, TTBK1, POT1, FLNC, OR51V1, HSPA4, ITGA4, and these breakpoints were significantly over-represented in exon and promoter (defined as 0 to -2 kb relative to the transcriptional start site) regions. In contrast, 85 of 220 HBV breakpoints were mainly located in introns. In addition, we identified 688 potential mutations of SNPs four samples by aligning to the hybrid reference genome. Majority of SNPs (666/688) were somatic single-base mutations, while the minority (22/688) occured small insertions and deletions (InDels), which were detected by pair-end reads for gap alignment (Table SI).

Table III

Distribution of the targets in chromosomes.

Table III

Distribution of the targets in chromosomes.

chrPatient 1Patient 2Patient 3Patient 4
chr19310
chr210468
chr311360
chr44020
chr510180
chr65102
chr76030
chr83101
chr96200
chr106300
chr117010
chr123020
chr1310010
chr148000
chr158210
chr164200
chr176030
chr183200
chr193001
chr208020
chr210000
chr225000
chrX2000

[i] Chr, chromosome.

Discussion

Hepatocellular carcinoma (HCC) is one of the most lethal malignancies, and it's development was a multifactorial process because of several direct and indirect mechanisms (25). HBV DNA integration events were frequently detected in the HCC patients, which was observed more usually in the tumors than in adjacent liver tissues, and was associated with patient's survival (4,21). Exactly as our clinical examination results for four HCC patients, the hepatitis B surface antigen (HBsAg) was positively expressed, and HBV DNA integration was frequently detected in the most HCC patients (26). Linghao Zhao and his colleagues identified 4,225 HBV integration events in tumor and adjacent non-tumor samples from 426 patients with HCC, and found that they preferred rare fragile sites and functional genomic regions, such as CpG islands (27). In addition, some researchers observed massive genomic perturbations near viral integration sites, such as direct gene disruption, viral promoter-driven human transcription, viral-human transcript fusion and DNA copy number alteration (28).

Traditionally, majority of the HBV integration events were detected by PCR-based methods, such as Alu-HBV PCR, and preferred the Alu regions (2). As a result, the HBV integration that located in the Alu regions can be frequently detected (2). Several studies have shown that the whole-genome sequencing combined with HBV DNA capture for HBV DNA integration events detection was feasible and effective, with considerable savings in time and cost, and could help improve the early diagnosis of hepatocellular carcinomas, and thus the survival rate. (4,21,27). In our study, we selected the whole-genome sequencing combined with HBV DNA capture to identify the HBV integration breakpoints from four tumor samples, which learns about the biological characteristics of HBV integrated into the human genome and provides some references for targeted therapy of HCC patients in the future.

Notably, we successfully identified the HBV integration breakpoints at the single base level, which can effectively validate confidence of HBV capture sequencing (2). Our data indicate that a large proportion of SNPs (666/688) were identified, which suggested the main tendency of HBV integration, and the InDels occasionally occurred in the HBV integration events. Subsequently, we analyzed the HBV integration breakpoints that distributed in distinct genomic elements, and found that majority of HBV integration breakpoints in the four tumor samples located in the coding region (116 of 220 HBV integration breakpoints). Of the 220 HBV integration breakpoints, 9 were located in known coding genes that significantly over-represented in exon and promoter regions. In contrast, 85 of 220 HBV integration breakpoints were mainly located in the introns, which suggested the preference among the HBV integration breakpoints. It is intriguing that virus genes inserted into host genomic DNA in totally random ways in the previously studies (29). However, some recent studies indicated that virus genes integration events had preference among the different regions (4,9,19). Our HBV integration profiles also demonstrated that HBV integration breakpoints preferentially landed in transcription units and specific chromosomes. Ding et al (9) found the HBV integration preference located in chromosomes 11 and 17. However, a preference for chromosome 3 has been reported in chronic hepatitis tissues without HCC by Alu-PCR (21). Our results indicated that the HBV integration breakpoints had a preference for chromosomes 2, 3 and 5. There are some potential reasons for the preferences, including the great HCC heterogeneity, different subclasses of HCC, sub-genotypes with different integration capabilities, etc. Therefore, we need a large number of samples to verify the results of this study, further explore the function of these target genes in the pathological mechanism of HCC and the characteristics they are in the TCGA database, which will provide more references for our future to discussion of ‘therapeutics in HCC patients. Additionally, future analysis for the potential relationship between HBV subtypes and their integration frequencies was needed in the future.

In summary, our results strengthened understanding that the HBV DNA integration events are implicated in HCC physiologies and diseases, and further demonstrated that the HBV insertional sequence capturing may be a useful tool to study the related human diseases.

Supplementary Material

Summary of single nucleotide polymorphisms and InDels from thefour tumor samples.

Acknowledgements

Not applicable.

Funding

The current study was supported by the Scientific Research and Technology Development Planning Project of Guilin (grant no. 2016012702-1), the Science and Technology Planning Project of Guangdong Province, China (grant no. 2017B020209001) and the Science and Technology Planning Project of Guangdong Province, China (grant no. 2016A020215027).

Availability of data and materials

The datasets used and/or analyzed are available from the corresponding author on reasonable request.

Authors' contributions

WS, MY and YD conceived and designed the experiments. GY, FL, MO, CL, JC, HL, YZ, WX, YW and YX performed the experiments. GY, FL, MO, CL and WX analyzed the data. GY, WS and MY drafted the manuscript. GY and YD revised the manuscript critically for important intellectual content. MY and YD obtained the funding. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was performed in accordance with the Helsinki Declaration and approved by the Ethics Committee of the Guilin no. 924 Hospital. Informed consent was obtained from the participant donors.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ringelhan M, Pfister D, O'Connor T, Pikarsky E and Heikenwalder M: The immunology of hepatocellular carcinoma. Nat Immunol. 19:222–232. 2018.PubMed/NCBI View Article : Google Scholar

2 

Li W, Zeng X, Lee NP, Liu X, Chen S, Guo B, Yi S, Zhuang X, Chen F, Wang G, et al: HIVID: An efficient method to detect HBV integration using low coverage sequencing. Genomics. 102:338–344. 2013.PubMed/NCBI View Article : Google Scholar

3 

Wu G, Ding H and Zeng C: Overview of HBV whole genome data in public repositories and the Chinese HBV reference sequences. Prog Nat Sci. 18:13–20. 2008.

4 

Sung WK, Zheng H, Li S, Chen R, Liu X, Li Y, Lee NP, Lee WH, Ariyaratne PN, Tennakoon C, et al: Genome-wide survey of recurrent HBV integration in hepatocellular carcinoma. Nat Genet. 44:765–769. 2012.PubMed/NCBI View Article : Google Scholar

5 

Gehring AJ, Ho ZZ, Tan AT, Aung MO, Lee KH, Tan KC, Lim SG and Bertoletti A: Profile of tumor antigen-specific CD8 T cells in patients with hepatitis B virus-related hepatocellular carcinoma. Gastroenterology. 137:682–690. 2009.PubMed/NCBI View Article : Google Scholar

6 

Lau CC, Sun T, Ching AK, He M, Li JW, Wong AM, Co NN, Chan AW, Li PS, Lung RW, et al: Viral-human chimeric transcript predisposes risk to liver cancer development and progression. Cancer Cell. 25:335–349. 2014.PubMed/NCBI View Article : Google Scholar

7 

Neuveut C, Wei Y and Buendia MA: Mechanisms of HBV-related hepatocarcinogenesis. J Hepatol. 52:594–604. 2010.PubMed/NCBI View Article : Google Scholar

8 

Tokino T and Matsubara K: Chromosomal sites for hepatitis B virus integration in human hepatocellular carcinoma. J Virol. 65:6761–6764. 1991.PubMed/NCBI View Article : Google Scholar

9 

Ding D, Lou X, Hua D, Yu W, Li L, Wang J, Gao F, Zhao N, Ren G, Li L and Lin B: Recurrent targeted genes of hepatitis B virus in the liver cancer genomes identified by a next-generation sequencing-based approach. PLoS Genet. 8(e1003065)2012.PubMed/NCBI View Article : Google Scholar

10 

Feitelson MA and Lee J: Hepatitis B virus integration, fragile sites, and hepatocarcinogenesis. Cancer Lett. 252:157–170. 2007.PubMed/NCBI View Article : Google Scholar

11 

Taha SE, El-Hady SA, Ahmed TM and Ahmed IZ: Detection of occult HBV infection by nested PCR assay among chronic hepatitis C patients with and without hepatocellular carcinoma. Egypt J Med Hum Genet. 14:353–360. 2013.

12 

Li X, Zhang J, Yang Z, Kang J, Jiang S, Zhang T, Chen T, Li M, Lv Q, Chen X, et al: The function of targeted host genes determines the oncogenicity of HBV integration in hepatocellular carcinoma. J Hepatol. 60:975–984. 2014.PubMed/NCBI View Article : Google Scholar

13 

Guerrieri F, Belloni L, Pediconi N and Levrero M: Molecular mechanisms of HBV-associated hepatocarcinogenesis. Semin Liver Dis. 33:147–156. 2013.PubMed/NCBI View Article : Google Scholar

14 

Rey-Cuille MA, Njouom R, Bekondi C, Seck A, Gody C, Bata P, Garin B, Maylin S, Chartier L, Simon F and Vray M: Hepatitis B virus exposure during childhood in Cameroon, Central African Republic and Senegal after the integration of HBV vaccine in the expanded program on immunization. Pediatr Infect Dis J. 32:1110–1115. 2013.PubMed/NCBI View Article : Google Scholar

15 

Bonilla Guerrero R and Roberts LR: The role of hepatitis B virus integrations in the pathogenesis of human hepatocellular carcinoma. J Hepatol. 42:760–777. 2005.PubMed/NCBI View Article : Google Scholar

16 

Bok J, Kim KJ, Park MH, Cho SH, Lee HJ, Lee EJ, Park C and Lee JY: Identification and extensive analysis of inverted-duplicated HBV integration in a human hepatocellular carcinoma cell line. BMB Rep. 45:365–370. 2012.PubMed/NCBI View Article : Google Scholar

17 

Arzumanyan A, Reis HM and Feitelson MA: Pathogenic mechanisms in HBV- and HCV-associated hepatocellular carcinoma. Nat Rev Cancer. 13:123–135. 2013.PubMed/NCBI View Article : Google Scholar

18 

Fujimoto A, Totoki Y, Abe T, Boroevich KA, Hosoda F, Nguyen HH, Aoki M, Hosono N, Kubo M, Miya F, et al: Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat Genet. 44:760–764. 2012.PubMed/NCBI View Article : Google Scholar

19 

Jiang Z, Jhunjhunwala S, Liu J, Haverty PM, Kennemer MI, Guan Y, Lee W, Carnevali P, Stinson J, Johnson S, et al: The effects of hepatitis B virus integration into the genomes of hepatocellular carcinoma patients. Genome Res. 22:593–601. 2012.PubMed/NCBI View Article : Google Scholar

20 

Amaddeo G, Cao Q, Ladeiro Y, Imbeaud S, Nault JC, Jaoui D, Gaston Mathe Y, Laurent C, Laurent A, Bioulac-Sage P, et al: Integration of tumour and viral genomic characterisations in HBV-related hepatocellular carcinomas. Gut. 64:820–829. 2015.PubMed/NCBI View Article : Google Scholar

21 

Murakami Y, Saigo K, Takashima H, Minami M, Okanoue T, Bréchot C and Paterlini-Bréchot P: Large scaled analysis of hepatitis B virus (HBV) DNA integration in HBV related hepatocellular carcinomas. Gut. 54:1162–1168. 2005.PubMed/NCBI View Article : Google Scholar

22 

Saigo K, Yoshida K, Ikeda R, Sakamoto Y, Murakami Y, Urashima T, Asano T, Kenmochi T and Inoue I: 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. 29:703–708. 2008.PubMed/NCBI View Article : Google Scholar

23 

Li H and Durbin R: Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 25:1754–1760. 2009.PubMed/NCBI View Article : Google Scholar

24 

Bauer DC: Variant calling comparison CASAVA1.8 and GATK. Nat Prec, 2011.

25 

Lim L, Tran BM, Vincan E, Locarnini S and Warner N: HBV-related hepatocellular carcinoma: The role of integration, viral proteins and miRNA. Future Virol. 7:1237–1249. 2012.

26 

Tamori A, Nishiguchi S, Kubo S, Narimatsu T, Habu D, Takeda T, Hirohashi K and Shiomi S: HBV DNA integration and HBV-transcript expression in non-B, non-C hepatocellular carcinoma in Japan. J Med Virol. 71:492–498. 2003.PubMed/NCBI View Article : Google Scholar

27 

Zhao LH, Liu X, Yan HX, Li WY, Zeng X, Yang Y, Zhao J, Liu SP, Zhuang XH, Lin C, et al: Genomic and oncogenic preference of HBV integration in hepatocellular carcinoma. Nat Commun. 7(12992)2016.PubMed/NCBI View Article : Google Scholar

28 

Zhang Z: Abstract LB-400: The effects of hepatitis B virus integration into the genomes of hepatocellular carcinoma patients. Cancer Res. 72 (Suppl 8):LB–400. 2012.PubMed/NCBI View Article : Google Scholar

29 

Bréchot C, Gozuacik D, Murakami Y and Paterlini-Bréchot P: Molecular bases for the development of hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC). Semin Cancer Biol. 10:211–231. 2000.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

December-2020
Volume 13 Issue 6

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang M, Yang G, Li F, Ou M, Li C, Chen J, Lin H, Zhang Y, Xue W, Wu Y, Wu Y, et al: HBV integrated genomic characterization revealed hepatocyte genomic alterations in HBV‑related hepatocellular carcinomas. Mol Clin Oncol 13: 79, 2020
APA
Yang, M., Yang, G., Li, F., Ou, M., Li, C., Chen, J. ... Dai, Y. (2020). HBV integrated genomic characterization revealed hepatocyte genomic alterations in HBV‑related hepatocellular carcinomas. Molecular and Clinical Oncology, 13, 79. https://doi.org/10.3892/mco.2020.2149
MLA
Yang, M., Yang, G., Li, F., Ou, M., Li, C., Chen, J., Lin, H., Zhang, Y., Xue, W., Wu, Y., Xu, Y., Sui, W., Dai, Y."HBV integrated genomic characterization revealed hepatocyte genomic alterations in HBV‑related hepatocellular carcinomas". Molecular and Clinical Oncology 13.6 (2020): 79.
Chicago
Yang, M., Yang, G., Li, F., Ou, M., Li, C., Chen, J., Lin, H., Zhang, Y., Xue, W., Wu, Y., Xu, Y., Sui, W., Dai, Y."HBV integrated genomic characterization revealed hepatocyte genomic alterations in HBV‑related hepatocellular carcinomas". Molecular and Clinical Oncology 13, no. 6 (2020): 79. https://doi.org/10.3892/mco.2020.2149