Preoperative evaluation of microvascular invasion with circulating tumour DNA in operable hepatocellular carcinoma

Dong Wang, Yaping Xu, Jennifer B Goldstein, Ke Ye, Xi Hu, Liang Xiao, Lifeng Li, Lianpeng Chang, Yanfang Guan, Guo Long, Qiongzhi He, Xin Yi, Jianjun Zhang, Zhiming Wang, Xuefeng Xia, Ledu Zhou, Dong Wang, Yaping Xu, Jennifer B Goldstein, Ke Ye, Xi Hu, Liang Xiao, Lifeng Li, Lianpeng Chang, Yanfang Guan, Guo Long, Qiongzhi He, Xin Yi, Jianjun Zhang, Zhiming Wang, Xuefeng Xia, Ledu Zhou

Abstract

Background & aims: Microvascular invasion (MVI) is a critical prognostic factor for operable hepatocellular carcinoma (HCC). This study aimed to explore the performance of circulating tumour DNA (ctDNA) in evaluating MVI status preoperatively.

Methods: Seventy-three HCC patients were enrolled and randomly divided into a training cohort and a validation cohort in a 2:1 ratio, and preoperative blood and surgical tissue samples were obtained. Genomic alterations were analysed using targeted deep sequencing with a 1021-gene panel.

Results: In training cohort, 260 somatic mutations were identified in 40 blood samples (81.6%). CtDNA mutation was verified in paired tissue sample in 39 patients (97.5%). In univariate analysis, ctDNA allele frequency (AF) and largest tumour diameter were associated with the presence of MVI, but ctDNA AF was the only independent risk factor in multivariate analysis. With the cut-off value of 0.83%, ctDNA AF determined the presence of MVI with the sensitivity of 89.7% and specificity of 80.0% in the training cohort, and the sensitivity of 78.6% and the specificity of 81.8% in the validation cohort. In preoperative evaluation, ctDNA AF, AFP level and BCLC staging were associated with recurrence-free survival in both univariate and multivariate analysis.

Conclusions: CtDNA can serve as an independent risk factor of MVI for operable HCC and help determining precise treatment strategies. The integration of ctDNA in the management of operable HCC may achieve better clinical outcomes.

Keywords: ctDNA; hepatectomy; hepatocellular carcinoma; microvascular invasion; surgical management.

Conflict of interest statement

The authors declare no conflict of interest.

© 2020 The Authors. Liver International published by John Wiley & Sons Ltd.

Figures

FIGURE 1
FIGURE 1
Genomic landscape of ctDNA in the training cohort. (A) Heatmap illustrating the common mutant genes of ctDNA. Only genes that mutated in over three patients were included. Each column represents a single patient. Different colours indicate diverse mutation types. The right bars represent the frequencies of each gene. The upper bars sum the total number of mutations only exhibited in the heatmap (not the overall mutation number) for each patient. (B) The distribution of AF for the overall ctDNA mutations. AF, allele frequency
FIGURE 2
FIGURE 2
Somatic mutations found in paired ctDNA and resected tumour tissue in the training cohort. Heatmaps indicate the AF of the somatic mutations (blue, see colour key) or their absence (grey) in the 49 pairs of tumour tissue and ctDNA. T, tissue; PL, plasma
FIGURE 3
FIGURE 3
Correlation between ctDNA parameters, nomogram model and MVI status in the training cohort. The upper column chart and broken line graph indicates the distribution of the maximal AF and blood TMB respectively. The horizontal black broken line indicates the cuf‐off value of ctDNA AF in distinguishing MVI high and low risk according to ROC analysis. The nomogram model encompass AFP, HBV‐DNA, platelet count and imaging parameters to evaluate the MVI risk for HCC patients who met the Milan criteria. Patients who are inconsistent with the Milan criteria are labelled as ‘Not applicable’. AF, allele frequency; TMB, tumour mutation burden
FIGURE 4
FIGURE 4
Significantly enriched mutant genes of ctDNA (A) and tumour tissue (B) in MVI positive and negative patients in the training cohort. Significantly enriched mutant genes is determined by χ2 or Fisher's exact test. Only genes that mutated in over three patients were included in the analysis. Genes on the right and left of y‐axis demonstrate superior population frequencies in MVI positive and negative patients respectively. Gene with P < .05 is identified with statistical significance and labelled as red box
FIGURE 5
FIGURE 5
Survival analysis for patients with different clinicopathological characteristics and ctDNA AF. The patients are grouped by MVI status (A) and maximal ctDNA AF (B). The results of both training and validation cohort, as well as the total cohort, are exhibited. (C) Multivariate COX regression analysis reveals the independent risk factors for postoperative recurrence. Only variables suggesting statistical significance in univariate Kaplan‐Meier analysis are included in the COX model, and the analysis is based on the data from the total cohort. HR, hazard ratios

References

    1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394‐424.
    1. Dutkowski P, Linecker M, DeOliveira ML, Mullhaupt B, Clavien PA. Challenges to liver transplantation and strategies to improve outcomes. Gastroenterology. 2015;148(2):307‐323.
    1. Roayaie S, Jibara G, Tabrizian P, et al. The role of hepatic resection in the treatment of hepatocellular cancer. Hepatology. 2015;62(2):440‐451.
    1. Zhu Q, Wang Z, Hu Y, et al. miR‐21 promotes migration and invasion by the miR‐21‐PDCD4‐AP‐1 feedback loop in human hepatocellular carcinoma. Oncol Rep. 2012;27(5):1660‐1668.
    1. Zhou L, Huang Y, Li J, Wang Z. The mTOR pathway is associated with the poor prognosis of human hepatocellular carcinoma. Med Oncol. 2010;27(2):255‐261.
    1. Roayaie S, Blume IN, Thung SN, et al. A system of classifying microvascular invasion to predict outcome after resection in patients with hepatocellular carcinoma. Gastroenterology. 2009;137(3):850‐855.
    1. Villanueva A, Hoshida Y, Battiston C, et al. Combining clinical, pathology, and gene expression data to predict recurrence of hepatocellular carcinoma. Gastroenterology. 2011;140(5):1501‐1512 e1502.
    1. Cong W‐M, Bu H, Chen J, et al. Practice guidelines for the pathological diagnosis of primary liver cancer: 2015 update. World J Gastroenterol. 2016;22(42):9279‐9287.
    1. Banerjee S, Wang DS, Kim HJ, et al. A computed tomography radiogenomic biomarker predicts microvascular invasion and clinical outcomes in hepatocellular carcinoma. Hepatology. 2015;62(3):792‐800.
    1. Sasaki YO, Yamada T, Tanaka H, et al. Risk of recurrence in a long‐term follow‐up after surgery in 417 patients with hepatitis B‐ or hepatitis C‐related hepatocellular carcinoma. Ann Surg. 2006;244(5):771‐780.
    1. Shindoh J, Makuuchi M, Matsuyama Y, et al. Complete removal of the tumor‐bearing portal territory decreases local tumor recurrence and improves disease‐specific survival of patients with hepatocellular carcinoma. J Hepatol. 2016;64(3):594‐600.
    1. Lei Z, Li J, Wu D, et al. Nomogram for preoperative estimation of microvascular invasion risk in hepatitis B virus‐related hepatocellular carcinoma within the milan criteria. JAMA Surg. 2016;151(4):356‐363.
    1. Mínguez B, Hoshida Y, Villanueva A, et al. Gene‐expression signature of vascular invasion in hepatocellular carcinoma. J Hepatol. 2011;55(6):1325‐1331.
    1. Wang W‐T, Yang LI, Yang Z‐X, et al. Assessment of microvascular invasion of hepatocellular carcinoma with diffusion Kurtosis imaging. Radiology. 2018;286(2):571‐580.
    1. Xu R‐H, Wei W, Krawczyk M, et al. Circulating tumour DNA methylation markers for diagnosis and prognosis of hepatocellular carcinoma. Nat Mater. 2017;16(11):1155‐1161.
    1. Hu Z‐Y, Xie N, Tian C, et al. Identifying circulating tumor DNA mutation profiles in metastatic breast cancer patients with multiline resistance. EBioMedicine. 2018;32:111‐118.
    1. Nong J, Gong Y, Guan Y, et al. Circulating tumor DNA analysis depicts subclonal architecture and genomic evolution of small cell lung cancer. Nat Commun. 2018;9(1):3114.
    1. Xie M, Lu C, Wang J, et al. Age‐related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med. 2014;20(12):1472‐1478.
    1. Ortiz‐Cuaran S, Villar S, Gouas D, et al. Association between HBX status, aflatoxin‐induced R249S TP53 mutation and risk of hepatocellular carcinoma in a case‐control study from Thailand. Cancer Lett. 2013;331(1):46‐51.
    1. Villar S, Le Roux‐Goglin E, Gouas DA, et al. Seasonal variation in TP53 R249S‐mutated serum DNA with aflatoxin exposure and hepatitis B virus infection. Environ Health Perspect. 2011;119(11):1635‐1640.
    1. Zehir A, Benayed R, Shah RH, et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med. 2017;23(6):703‐713.
    1. Huang X, Lu S. A Meta‐analysis comparing the effect of anatomical resection vs non‐anatomical resection on the long‐term outcomes for patients undergoing hepatic resection for hepatocellular carcinoma. HPB (Oxford). 2017;19(10):843‐849.
    1. Moris D, Tsilimigras DI, Kostakis ID, et al. Anatomic versus non‐anatomic resection for hepatocellular carcinoma: a systematic review and meta‐analysis. Eur J Surg Oncol. 2018;44(7):927‐938.
    1. Vitale A, Cucchetti A, Qiao GL, et al. Is resectable hepatocellular carcinoma a contraindication to liver transplantation? A novel decision model based on "number of patients needed to transplant" as measure of transplant benefit. J Hepatol. 2014;60(6):1165‐1171.
    1. Dawson S‐J, Tsui DWY, Murtaza M, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med. 2013;368(13):1199‐1209.
    1. Li J, Han XU, Yu X, et al. Clinical applications of liquid biopsy as prognostic and predictive biomarkers in hepatocellular carcinoma: circulating tumor cells and circulating tumor DNA. J Exp Clin Cancer Res. 2018;37(1):213.
    1. Zhang Y, Zhou L, Qin L. High‐throughput 3D cell invasion chip enables accurate cancer metastatic assays. J Am Chem Soc. 2014;136(43):15257‐15262.
    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7‐30.
    1. Marrero JA, Kulik LM, Sirlin CB, et al. Diagnosis, staging, and management of hepatocellular carcinoma: 2018 Practice Guidance by the American Association for the Study of Liver Diseases. Hepatology. 2018;68(2):723‐750.
    1. Omata M, Cheng A‐L, Kokudo N, et al. Asia‐Pacific clinical practice guidelines on the management of hepatocellular carcinoma: a 2017 update. Hepatol Int. 2017;11(4):317‐370.
    1. Singal AG, Pillai A, Tiro J. Early detection, curative treatment, and survival rates for hepatocellular carcinoma surveillance in patients with cirrhosis: a meta‐analysis. PLoS Med. 2014;11(4):e1001624.
    1. Mohamed NA, Swify EM, Amin NF, Soliman MM, Tag‐Eldin LM, Elsherbiny NM. Is serum level of methylated RASSF1A valuable in diagnosing hepatocellular carcinoma in patients with chronic viral hepatitis C? Arab J Gastroenterol. 2012;13(3):111‐115.
    1. Qu C, Wang Y, Wang P, et al. Detection of early‐stage hepatocellular carcinoma in asymptomatic HBsAg‐seropositive individuals by liquid biopsy. Proc Natl Acad Sci U S A. 2019;116(13):6308‐6312.
    1. Ng CKY, Di Costanzo GG, Tosti N, et al. Genetic profiling using plasma‐derived cell‐free DNA in therapy‐naive hepatocellular carcinoma patients: a pilot study. Ann Oncol. 2018;29(5):1286‐1291.

Source: PubMed

3
Iratkozz fel