Circulating Tumor DNA Analysis for Liver Cancers and Its Usefulness as a Liquid Biopsy

Atsushi Ono, Akihiro Fujimoto, Yujiro Yamamoto, Sakura Akamatsu, Nobuhiko Hiraga, Michio Imamura, Tomokazu Kawaoka, Masataka Tsuge, Hiromi Abe, C Nelson Hayes, Daiki Miki, Mayuko Furuta, Tatsuhiko Tsunoda, Satoru Miyano, Michiaki Kubo, Hiroshi Aikata, Hidenori Ochi, Yoshi-Iku Kawakami, Koji Arihiro, Hideki Ohdan, Hidewaki Nakagawa, Kazuaki Chayama, Atsushi Ono, Akihiro Fujimoto, Yujiro Yamamoto, Sakura Akamatsu, Nobuhiko Hiraga, Michio Imamura, Tomokazu Kawaoka, Masataka Tsuge, Hiromi Abe, C Nelson Hayes, Daiki Miki, Mayuko Furuta, Tatsuhiko Tsunoda, Satoru Miyano, Michiaki Kubo, Hiroshi Aikata, Hidenori Ochi, Yoshi-Iku Kawakami, Koji Arihiro, Hideki Ohdan, Hidewaki Nakagawa, Kazuaki Chayama

Abstract

Background & aims: Circulating tumor DNA (ctDNA) carrying tumor-specific sequence alterations has been found in the cell-free fraction of blood. Liver cancer tumor specimens are difficult to obtain, and noninvasive methods are required to assess cancer progression and characterize underlying genomic features.

Methods: We analyzed 46 patients with hepatocellular carcinoma who underwent hepatectomy or liver transplantation and for whom whole-genome sequencing data was available. We designed personalized assays targeting somatic rearrangements of each tumor to quantify serum ctDNA. Exome sequencing was performed using cell-free DNA paired primary tumor tissue DNA from a patient with recurrent liver cancer after transcatheter arterial chemoembolization (TACE).

Results: We successfully detected ctDNA from 100 μL of serum samples in 7 of the 46 patients before surgery, increasing with disease progression. The cumulative incidence of recurrence and extrahepatic metastasis in the ctDNA-positive group were statistically significantly worse than in the ctDNA-negative group (P = .0102 and .0386, respectively). Multivariate analysis identified ctDNA (OR 6.10; 95% CI, 1.11-33.33, P = .038) as an independent predictor of microscopic vascular invasion of the portal vein (VP). We identified 45 nonsynonymous somatic mutations in cell-free DNA after TACE and 71 nonsynonymous somatic mutations in primary tumor tissue by exome sequencing. We identified 25 common mutations in both samples, and 83% of mutations identified in the primary tumor could be detected in the cell-free DNA.

Conclusions: The presence of ctDNA reflects tumor progression, and detection of ctDNA can predict VP and recurrence, especially extrahepatic metastasis within 2 years. Our study demonstrated the usefulness of ctDNA detection and sequencing analysis of cell-free DNA for personalized treatment of liver cancer.

Keywords: AFP, α-fetoprotein; ALT, alanine aminotransferase; AST, aspartate aminotransferase; Circulating Tumor DNA; DCP, des-γ-carboxy prothrombin; Exome Sequencing; HAIC, hepatic arterial infusion chemotherapy; HBV, hepatitis B virus; HCC, hepatocellular carcinoma; HCV, hepatitis C virus; Hepatocellular Carcinoma; PCR, polymerase-chain-reaction; TACE, transcatheter arterial chemoembolization; VP, microscopic vascular invasion to portal vein; Whole-Genome Sequencing; cHCC/CC, combined hepatocellular and cholangiocarcinoma; ctDNA, circulating tumor DNA.

Figures

Figure 1
Figure 1
Quantitative ranges. Quantitative ranges were from 105 copies to 5 copies in the most sensitive assay and 105 copies to 102 copies in the least sensitive assay, as shown in the upper and lower panels, respectively.
Figure 2
Figure 2
Detection of circulating tumor DNA (ctDNA) by polymerase chain reaction (PCR) targeting for somatic rearrangements. Gel electrophoresis of PCR products. The ctDNA extracted from preoperative serum samples (S) was amplified by PCR with the use of primers designed to detect breakpoints of somatic rearrangements in each of the tumors (see Supplementary Table 1). DNA extracted from cancer tissue samples was used as a positive control (C), and DNA extracted from blood cells was used as a negative control (B). (A, B) Patients with positive ctDNA. Upper panel: Circos plots of the hepatocellular carcinomas (HCCs) in cases H1–H4 (A) and H5–H7 (B) who tested positive preoperatively for serum ctDNA. Each circle plot represents validated somatic rearrangements in each of the HCCs. Lines show chromosomal translocations (green), deletions (blue), inversions (red), and tandem duplications/translocations (orange). (CE) Patients with negative ctDNA. (F, G) DNA extracted from serum of chronic hepatitis C (HCV) (N1) and hepatitis B (HBV) (N2) patients without HCC were confirmed not to be amplified by PCR using these primer. Red numbers show the amounts of DNA extracted from tumor tissue. The red arrow shows the target product.
Figure 2
Figure 2
Detection of circulating tumor DNA (ctDNA) by polymerase chain reaction (PCR) targeting for somatic rearrangements. Gel electrophoresis of PCR products. The ctDNA extracted from preoperative serum samples (S) was amplified by PCR with the use of primers designed to detect breakpoints of somatic rearrangements in each of the tumors (see Supplementary Table 1). DNA extracted from cancer tissue samples was used as a positive control (C), and DNA extracted from blood cells was used as a negative control (B). (A, B) Patients with positive ctDNA. Upper panel: Circos plots of the hepatocellular carcinomas (HCCs) in cases H1–H4 (A) and H5–H7 (B) who tested positive preoperatively for serum ctDNA. Each circle plot represents validated somatic rearrangements in each of the HCCs. Lines show chromosomal translocations (green), deletions (blue), inversions (red), and tandem duplications/translocations (orange). (CE) Patients with negative ctDNA. (F, G) DNA extracted from serum of chronic hepatitis C (HCV) (N1) and hepatitis B (HBV) (N2) patients without HCC were confirmed not to be amplified by PCR using these primer. Red numbers show the amounts of DNA extracted from tumor tissue. The red arrow shows the target product.
Figure 2
Figure 2
Detection of circulating tumor DNA (ctDNA) by polymerase chain reaction (PCR) targeting for somatic rearrangements. Gel electrophoresis of PCR products. The ctDNA extracted from preoperative serum samples (S) was amplified by PCR with the use of primers designed to detect breakpoints of somatic rearrangements in each of the tumors (see Supplementary Table 1). DNA extracted from cancer tissue samples was used as a positive control (C), and DNA extracted from blood cells was used as a negative control (B). (A, B) Patients with positive ctDNA. Upper panel: Circos plots of the hepatocellular carcinomas (HCCs) in cases H1–H4 (A) and H5–H7 (B) who tested positive preoperatively for serum ctDNA. Each circle plot represents validated somatic rearrangements in each of the HCCs. Lines show chromosomal translocations (green), deletions (blue), inversions (red), and tandem duplications/translocations (orange). (CE) Patients with negative ctDNA. (F, G) DNA extracted from serum of chronic hepatitis C (HCV) (N1) and hepatitis B (HBV) (N2) patients without HCC were confirmed not to be amplified by PCR using these primer. Red numbers show the amounts of DNA extracted from tumor tissue. The red arrow shows the target product.
Figure 2
Figure 2
Detection of circulating tumor DNA (ctDNA) by polymerase chain reaction (PCR) targeting for somatic rearrangements. Gel electrophoresis of PCR products. The ctDNA extracted from preoperative serum samples (S) was amplified by PCR with the use of primers designed to detect breakpoints of somatic rearrangements in each of the tumors (see Supplementary Table 1). DNA extracted from cancer tissue samples was used as a positive control (C), and DNA extracted from blood cells was used as a negative control (B). (A, B) Patients with positive ctDNA. Upper panel: Circos plots of the hepatocellular carcinomas (HCCs) in cases H1–H4 (A) and H5–H7 (B) who tested positive preoperatively for serum ctDNA. Each circle plot represents validated somatic rearrangements in each of the HCCs. Lines show chromosomal translocations (green), deletions (blue), inversions (red), and tandem duplications/translocations (orange). (CE) Patients with negative ctDNA. (F, G) DNA extracted from serum of chronic hepatitis C (HCV) (N1) and hepatitis B (HBV) (N2) patients without HCC were confirmed not to be amplified by PCR using these primer. Red numbers show the amounts of DNA extracted from tumor tissue. The red arrow shows the target product.
Figure 3
Figure 3
Lowest limit of detection. To confirm the lowest limit of detection, custom synthesized DNA oligos that were diluted from 105 copies to 10 copies with distilled water were amplified by polymerase chain reaction. They remained detectable in any condition when at least 10–100 copies of DNA were present.
Figure 4
Figure 4
The cumulative incidence of recurrence and extrahepatic metastasis within 2 years after hepatic resection. The cumulative incidence of recurrence (left) and extrahepatic metastasis (right) of the circulating tumor DNA (ctDNA)-positive group (green line) were statistically significantly worse than that of the ctDNA-negative group (red line) (P = .0102 and .0386, respectively).
Figure 5
Figure 5
Monitoring of serial circulating tumor DNA (ctDNA) levels. The ctDNA was quantified by real-time quantitative polymerase chain reaction in sera serially sampled before and after surgery from the four patients with positive ctDNA (cases H1–H5). The figure shows the time course of serum levels of ctDNA, α-fetoprotein (AFP), and des-γ-carboxy prothrombin (DCP) with their clinical events and treatments. Levels of ctDNA are expressed as a ratio relative to levels of those obtained using DNA extracted from tumor tissue.
Figure 5
Figure 5
Monitoring of serial circulating tumor DNA (ctDNA) levels. The ctDNA was quantified by real-time quantitative polymerase chain reaction in sera serially sampled before and after surgery from the four patients with positive ctDNA (cases H1–H5). The figure shows the time course of serum levels of ctDNA, α-fetoprotein (AFP), and des-γ-carboxy prothrombin (DCP) with their clinical events and treatments. Levels of ctDNA are expressed as a ratio relative to levels of those obtained using DNA extracted from tumor tissue.
Figure 5
Figure 5
Monitoring of serial circulating tumor DNA (ctDNA) levels. The ctDNA was quantified by real-time quantitative polymerase chain reaction in sera serially sampled before and after surgery from the four patients with positive ctDNA (cases H1–H5). The figure shows the time course of serum levels of ctDNA, α-fetoprotein (AFP), and des-γ-carboxy prothrombin (DCP) with their clinical events and treatments. Levels of ctDNA are expressed as a ratio relative to levels of those obtained using DNA extracted from tumor tissue.
Figure 6
Figure 6
Circulating tumor DNA (ctDNA) dynamics after undergoing transcatheter arterial chemoembolization (TACE). (A) Serum ctDNA levels at 1, 4, and 6 days after TACE are shown as a solid line, and serum aminotransferase (AST) and alanine aminotransferase (ALT) levels are shown as dotted lines. The x-axis shows the number of days after TACE. The y-axis on the left indicates the fold change of serum ctDNA levels compared with that before TACE, and the y-axis on the right indicates serum AST, ALT, AFP (α-fetoprotein), and des-γ-carboxy prothrombin (DCP) levels. The ctDNA levels of cases 2 and 3 increased 5- and 10-fold compared with before TACE, respectively. The ctDNA levels peaked 4 days after TACE was performed. (B) The ctDNA became detectable 4 days after TACE (S2) in two of three patients (cases H8 and H9) who were negative for ctDNA before TACE (S1). DNA extracted from cancer tissue samples was used as a positive control (C), and DNA extracted from blood cells were used as a negative control (B).
Figure 7
Figure 7
Exome sequencing of primary tumor and cell-free DNA. (A) The clinical course of case C1. Case C1 had one combined hepatocellular and cholangiocarcinoma (cHCC/CC) lesion in the right lobe that was removed by curative resection. Transcatheter arterial chemoembolization (TACE) was performed for intrahepatic recurrent lesions 2 years after the first surgery. We performed exome sequencing of cell-free DNA after the TACE and the primary tumor (red star). (B) The amount of total cell-free DNA extracted from the plasma samples serially obtained after TACE. Cell-free DNA was most abundant in plasma 2 days after TACE, and was therefore used for exome sequencing analysis. (C) Common mutations in cell-free DNA and primary tumor. Somatic mutations detected by probabilistic variant detection and low frequency variant detection are indicated by the red and pink boxes, respectively.
Figure 7
Figure 7
Exome sequencing of primary tumor and cell-free DNA. (A) The clinical course of case C1. Case C1 had one combined hepatocellular and cholangiocarcinoma (cHCC/CC) lesion in the right lobe that was removed by curative resection. Transcatheter arterial chemoembolization (TACE) was performed for intrahepatic recurrent lesions 2 years after the first surgery. We performed exome sequencing of cell-free DNA after the TACE and the primary tumor (red star). (B) The amount of total cell-free DNA extracted from the plasma samples serially obtained after TACE. Cell-free DNA was most abundant in plasma 2 days after TACE, and was therefore used for exome sequencing analysis. (C) Common mutations in cell-free DNA and primary tumor. Somatic mutations detected by probabilistic variant detection and low frequency variant detection are indicated by the red and pink boxes, respectively.

References

    1. Paez J.G., Janne P.A., Lee J.C. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004;304:1497–1500.
    1. Lynch T.J., Bell D.W., Sordella R. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004;350:2129–2139.
    1. Karapetis C.S., Khambata-Ford S., Jonker D.J. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med. 2008;359:1757–1765.
    1. Heinrich M.C., Corless C.L., Demetri G.D. Kinase mutations and imatinib response in patients with metastatic gastrointestinal stromal tumor. J Clin Oncol. 2003;21:4342–4349.
    1. Dawson S.J., Tsui D.W., Murtaza M. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med. 2013;368:1199–1209.
    1. Murtaza M., Dawson S.J., Tsui D.W. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497:108–112.
    1. Bruix J., Sherman M. American Association for the Study of Liver D. Management of hepatocellular carcinoma: an update. Hepatology. 2011;53:1020–1022.
    1. Bruix J., Sherman M. Practice Guidelines Committee AAftSoLD. Management of hepatocellular carcinoma. Hepatology. 2005;42:1208–1236.
    1. Schwarzenbach H., Hoon D.S., Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 2011;11:426–437.
    1. Gormally E., Caboux E., Vineis P. Circulating free DNA in plasma or serum as biomarker of carcinogenesis: practical aspects and biological significance. Mutat Res. 2007;635:105–117.
    1. Castells A., Puig P., Mora J. K-ras mutations in DNA extracted from the plasma of patients with pancreatic carcinoma: diagnostic utility and prognostic significance. J Clin Oncol. 1999;17:578–584.
    1. Sorenson G.D. Detection of mutated KRAS2 sequences as tumor markers in plasma/serum of patients with gastrointestinal cancer. Clin Cancer Res. 2000;6:2129–2137.
    1. Diaz L.A., Jr., Williams R.T., Wu J. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature. 2012;486:537–540.
    1. Diehl F., Schmidt K., Choti M.A. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14:985–990.
    1. Yung T.K., Chan K.C., Mok T.S. Single-molecule detection of epidermal growth factor receptor mutations in plasma by microfluidics digital PCR in non-small cell lung cancer patients. Clin Cancer Res. 2009;15:2076–2084.
    1. Chan K.C., Jiang P., Zheng Y.W. Cancer genome scanning in plasma: detection of tumor-associated copy number aberrations, single-nucleotide variants, and tumoral heterogeneity by massively parallel sequencing. Clin Chem. 2013;59:211–224.
    1. Leary R.J., Sausen M., Kinde I. Detection of chromosomal alterations in the circulation of cancer patients with whole-genome sequencing. Sci Transl Med. 2012;4:162ra154.
    1. Li H., Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760.
    1. Fujimoto A., Totoki Y., Abe T. Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat Genet. 2012;44:760–764.
    1. Krzywinski M., Schein J., Birol I. Circos: an information aesthetic for comparative genomics. Genome Res. 2009;19:1639–1645.
    1. Vallee A., Marcq M., Bizieux A. Plasma is a better source of tumor-derived circulating cell-free DNA than serum for the detection of EGFR alterations in lung tumor patients. Lung Cancer. 2013;82:373–374.
    1. Metz C.H., Scheulen M., Bornfeld N. Ultradeep sequencing detects GNAQ and GNA11 mutations in cell-free DNA from plasma of patients with uveal melanoma. Cancer Med. 2013;2:208–215.
    1. Newman A.M., Bratman S.V., To J. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med. 2014;20:548–554.
    1. Wilson C.H., McIntyre R.E., Arends M.J. The activating mutation R201C in GNAS promotes intestinal tumourigenesis in ApcMin/+ mice through activation of Wnt and ERK1/2 MAPK pathways. Oncogene. 2010;29:4567–4575.
    1. Cheng A.L., Kang Y.K., Chen Z. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2009;10:25–34.
    1. Llovet J.M., Ricci S., Mazzaferro V. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378–390.
    1. Chan S.L., Yeo W. Targeted therapy of hepatocellular carcinoma: present and future. J Gastroenterol Hepatol. 2012;27:862–872.
    1. Wei Z., Doria C., Liu Y. Targeted therapies in the treatment of advanced hepatocellular carcinoma. Clin Med Insights Oncol. 2013;7:87–102.
    1. Santoro A., Rimassa L., Borbath I. Tivantinib for second-line treatment of advanced hepatocellular carcinoma: a randomised, placebo-controlled phase 2 study. Lancet Oncol. 2013;14:55–63.
    1. Faivre S., Raymond E., Boucher E. Safety and efficacy of sunitinib in patients with advanced hepatocellular carcinoma: an open-label, multicentre, phase II study. Lancet Oncol. 2009;10:794–800.
    1. Thomas M.B., Morris J.S., Chadha R. Phase II trial of the combination of bevacizumab and erlotinib in patients who have advanced hepatocellular carcinoma. J Clin Oncol. 2009;27:843–850.
    1. Siegel A.B., Cohen E.I., Ocean A. Phase II trial evaluating the clinical and biologic effects of bevacizumab in unresectable hepatocellular carcinoma. J Clin Oncol. 2008;26:2992–2998.
    1. Zhu A.X., Blaszkowsky L.S., Ryan D.P. Phase II study of gemcitabine and oxaliplatin in combination with bevacizumab in patients with advanced hepatocellular carcinoma. J Clin Oncol. 2006;24:1898–1903.
    1. Kanai F., Yoshida H., Tateishi R. A phase I/II trial of the oral antiangiogenic agent TSU-68 in patients with advanced hepatocellular carcinoma. Cancer Chemother Pharmacol. 2011;67:315–324.
    1. Philip P.A., Mahoney M.R., Holen K.D. Phase 2 study of bevacizumab plus erlotinib in patients with advanced hepatocellular cancer. Cancer. 2012;118:2424–2430.
    1. Zhu A.X., Sahani D.V., Duda D.G. Efficacy, safety, and potential biomarkers of sunitinib monotherapy in advanced hepatocellular carcinoma: a phase II study. J Clin Oncol. 2009;27:3027–3035.
    1. Hsu C.H., Yang T.S., Hsu C. Efficacy and tolerability of bevacizumab plus capecitabine as first-line therapy in patients with advanced hepatocellular carcinoma. Br J Cancer. 2010;102:981–986.

Source: PubMed

3
Se inscrever