Somatic copy number profiling from hepatocellular carcinoma circulating tumor cells

Colin M Court, Shuang Hou, Lian Liu, Paul Winograd, Benjamin J DiPardo, Sean X Liu, Pin-Jung Chen, Yazhen Zhu, Matthew Smalley, Ryan Zhang, Saeed Sadeghi, Richard S Finn, Fady M Kaldas, Ronald W Busuttil, Xianghong J Zhou, Hsian-Rong Tseng, James S Tomlinson, Thomas G Graeber, Vatche G Agopian, Colin M Court, Shuang Hou, Lian Liu, Paul Winograd, Benjamin J DiPardo, Sean X Liu, Pin-Jung Chen, Yazhen Zhu, Matthew Smalley, Ryan Zhang, Saeed Sadeghi, Richard S Finn, Fady M Kaldas, Ronald W Busuttil, Xianghong J Zhou, Hsian-Rong Tseng, James S Tomlinson, Thomas G Graeber, Vatche G Agopian

Abstract

Somatic copy number alterations (SCNAs) are important genetic drivers of many cancers. We investigated the feasibility of obtaining SCNA profiles from circulating tumor cells (CTCs) as a molecular liquid biopsy for hepatocellular carcinoma (HCC). CTCs from ten HCC patients underwent SCNA profiling. The Cancer Genome Atlas (TCGA) SCNA data were used to develop a cancer origin classification model, which was then evaluated for classifying 44 CTCs from multiple cancer types. Sequencing of 18 CTC samples (median: 4 CTCs/sample) from 10 HCC patients using a low-resolution whole-genome sequencing strategy (median: 0.88 million reads/sample) revealed frequent SCNAs in previously reported HCC regions such as 8q amplifications and 17p deletions. SCNA profiling revealed that CTCs share a median of 80% concordance with the primary tumor. CTCs had SCNAs not seen in the primary tumor, some with prognostic implications. Using a SCNA profiling model, the tissue of origin was correctly identified for 32/44 (73%) CTCs from 12/16 (75%) patients with different cancer types.

Keywords: Molecular medicine; Prognostic markers.

Conflict of interest statement

Competing interestsH.R.T has ownership in the intellectual property used to isolate circulating tumor cells in this study (NanoVelcro CTC Assay), which has been licensed to CytoLumina Technologies Corp. H.R.T. and S.X.L. have financial interests in CytoLumina Technologies Corp. given their role in the company. All other authors report no conflicts of interest.

© This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply 2020.

Figures

Fig. 1. Experimental design of the study.
Fig. 1. Experimental design of the study.
a Workflow used in the study for CTC isolation and SCNA profiling using the NanoVelcro assay. Following gradient centrifugation, HCC CTCs are selectively captured on the NanoVelcro surface for identification and subsequent isolation by LMD. CTC samples undergo WGA and QC prior to low-resolution whole-genome sequencing for SCNA profiling. b A total of four specimens are obtained from each patient. From peripheral blood, germline genomic DNA is obtained from the bulk buffy coat layer. Circulating tumor cells are obtained and sequenced as described in (a). From the surgically resected specimen, a sample of both the primary tumor and the peritumoral normal liver are obtained and sequenced.
Fig. 2. Global copy number profiles for…
Fig. 2. Global copy number profiles for blood, peritumoral normal liver, CTCs, and primary tumor (n = 8) or solitary metastasis (n = 1, H167) demonstrates the recapitulation of SCNAs from the tumor in CTC samples, supporting the potential of CTCs to act as a liquid biopsy of the tumor.
Patient H169’s CTC (shown in Supplementary Fig. 1) STR analysis did not match that of the tumor or blood sample, suggesting contamination.
Fig. 3. Heatmap showing copy number profiles…
Fig. 3. Heatmap showing copy number profiles at 59 loci frequently amplified or lost in HCC. Gain (red) or loss (blue) at each of the 59 cytobands (identified on the right of the heatmap) in both CTC and tumor samples from all nine patients that passed STR analysis are shown.
Unsupervised hierarchical clustering demonstrates clustering of CTC–tumor pairs for all nine patients. CTC—circulating tumor cell, 1°—Primary tumor.
Fig. 4. Comparison of low-resolution whole-genome copy…
Fig. 4. Comparison of low-resolution whole-genome copy number profiles for primary tumor, CTCs 1 and 2, normal liver, and whole blood reveals reproduction of the majority of the SCNAs from the primary tumor in both CTC samples.
a Whole genome SCNA profiles for the primary tumor as well as the two CTC samples. b Chromosome 17p loss is seen in the primary tumor and both CTC samples, but not the normal liver or whole blood. The location of the tumor suppressor TP53 gene, the most frequently mutated or lost gene in HCC, is indicated by the blue triangle. c Chromosome 20 amplification was seen in both CTC samples but not in the primary tumor. Chromosome 20 amplifications are a recurrent somatic copy number alteration in HCC associated with 2 important oncogenes. Overexpression of AIB1 (orange triangle) is frequently described in HCC, and has previously been associated with invasiveness. In addition, recent research has demonstrated that the oncogenic effects of MYC dysregulation, a common occurrence in HCC, require overexpression of AURKA (green triangle) for stabilization. Furthermore, in p53-altered HCC patients, the MYC-AURKA complex is an actionable drug target based on preclinical studies.
Fig. 5. Copy number alterations at all…
Fig. 5. Copy number alterations at all genes for the 32 cancer types were transformed using t-SNE into a 2-dimensional space.
The mean of all samples for each cancer type is plotted as well as the individual samples for the best clustering (TGCT—Blue) and worst clustering (ESCA—Pink) cancer types. All individual samples are plotted by cancer type in Supplementary Fig. 5.
Fig. 6. Cancer class prediction based on…
Fig. 6. Cancer class prediction based on SCNA profile for each of the 15 HCC CTC samples using the classification model.
9/15 (60%) of CTCs were correctly classified as being HCC, while 6/15 (40%) were classified as being from a GI source. 5/9 (56%) patients had at least one CTC classified as being from a HCC tumor.

References

    1. Zack TI, et al. Pan-cancer patterns of somatic copy number alteration. Nat. Genet. 2013;45:1134–1140. doi: 10.1038/ng.2760.
    1. Hoadley KA, et al. Cell-of-origin patterns dominate the molecular classification of 10,000 tumors from 33 types of cancer. Cell. 2018;173:291–304 e6. doi: 10.1016/j.cell.2018.03.022.
    1. Beroukhim R, et al. The landscape of somatic copy-number alteration across human cancers. Nature. 2010;463:899–905. doi: 10.1038/nature08822.
    1. Baslan T, et al. Optimizing sparse sequencing of single cells for highly multiplex copy number profiling. Genome Res. 2015;25:714–724. doi: 10.1101/gr.188060.114.
    1. Court, C. M., Ankeny, J. S., Sho, S. & Tomlinson, J. S. Circulating tumor cells in gastrointestinal cancer: current practices and future directions. In Gastrointestinal Malignancies 345–376 (Springer, 2016).
    1. Alix-Panabieres C, Pantel K. Challenges in circulating tumour cell research. Nat. Rev. Cancer. 2014;14:623–631. doi: 10.1038/nrc3820.
    1. Aceto N, et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell. 2014;158:1110–1122. doi: 10.1016/j.cell.2014.07.013.
    1. Miyamoto DT, Ting DT, Toner M, Maheswaran S, Haber DA. Single-cell analysis of circulating tumor cells as a window into tumor heterogeneity. Cold Spring Harb. Symp. Quant. Biol. 2016;81:269–274. doi: 10.1101/sqb.2016.81.031120.
    1. Hata AN, et al. Tumor cells can follow distinct evolutionary paths to become resistant to epidermal growth factor receptor inhibition. Nat. Med. 2016;22:262–269. doi: 10.1038/nm.4040.
    1. Ni X, et al. Reproducible copy number variation patterns among single circulating tumor cells of lung cancer patients. Proc. Natl Acad. Sci. USA. 2013;110:21083–21088. doi: 10.1073/pnas.1320659110.
    1. Heitzer E, Ulz P, Geigl JB, Speicher MR. Non-invasive detection of genome-wide somatic copy number alterations by liquid biopsies. Mol. Oncol. 2016;10:494–502. doi: 10.1016/j.molonc.2015.12.004.
    1. Heitzer E, et al. Complex tumor genomes inferred from single circulating tumor cells by array-CGH and next-generation sequencing. Cancer Res. 2013;73:2965–2975. doi: 10.1158/0008-5472.CAN-12-4140.
    1. Carter L, et al. Molecular analysis of circulating tumor cells identifies distinct copy-number profiles in patients with chemosensitive and chemorefractory small-cell lung cancer. Nat. Med. 2017;23:114–119. doi: 10.1038/nm.4239.
    1. Ankeny JS, et al. Circulating tumour cells as a biomarker for diagnosis and staging in pancreatic cancer. Br. J. Cancer. 2016;114:1367–1375. doi: 10.1038/bjc.2016.121.
    1. Court CM, et al. A novel multimarker assay for the phenotypic profiling of circulating tumor cells in hepatocellular carcinoma. Liver Transpl. 2018;24:946–960. doi: 10.1002/lt.25062.
    1. Garvin T, et al. Interactive analysis and assessment of single-cell copy-number variations. Nat. Methods. 2015;12:1058–1060. doi: 10.1038/nmeth.3578.
    1. Guichard C, et al. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat. Genet. 2012;44:694–698. doi: 10.1038/ng.2256.
    1. Kan Z, et al. Whole-genome sequencing identifies recurrent mutations in hepatocellular carcinoma. Genome Res. 2013;23:1422–1433. doi: 10.1101/gr.154492.113.
    1. Chiang DY, et al. Focal gains of VEGFA and molecular classification of hepatocellular carcinoma. Cancer Res. 2008;68:6779–6788. doi: 10.1158/0008-5472.CAN-08-0742.
    1. Zhang J, et al. [Association of chromosome 17q copy number variation with overall survival of patients with hepatocellular carcinoma and screening of potential target genes] Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 2015;32:615–619.
    1. Kwon SM, et al. Genomic copy number alterations with transcriptional deregulation at 6p identify an aggressive HCC phenotype. Carcinogenesis. 2013;34:1543–1550. doi: 10.1093/carcin/bgt095.
    1. Roessler S, et al. Integrative genomic identification of genes on 8p associated with hepatocellular carcinoma progression and patient survival. Gastroenterology. 2012;142:957–966. e12. doi: 10.1053/j.gastro.2011.12.039.
    1. Woo HG, et al. Identification of potential driver genes in human liver carcinoma by genomewide screening. Cancer Res. 2009;69:4059–4066. doi: 10.1158/0008-5472.CAN-09-0164.
    1. Schulze K, Nault JC, Villanueva A. Genetic profiling of hepatocellular carcinoma using next-generation sequencing. J. Hepatol. 2016;65:1031–1042. doi: 10.1016/j.jhep.2016.05.035.
    1. Xu Y, et al. Overexpression of transcriptional coactivator AIB1 promotes hepatocellular carcinoma progression by enhancing cell proliferation and invasiveness. Oncogene. 2010;29:3386–3397. doi: 10.1038/onc.2010.90.
    1. Dauch D, et al. A MYC-aurora kinase A protein complex represents an actionable drug target in p53-altered liver cancer. Nat. Med. 2016;22:744–753. doi: 10.1038/nm.4107.
    1. Tong Z, et al. Steroid receptor coactivator 1 promotes human hepatocellular carcinoma progression by enhancing Wnt/beta-catenin signaling. J. Biol. Chem. 2015;290:18596–18608. doi: 10.1074/jbc.M115.640490.
    1. Lu L, et al. Aurora kinase A mediates c-Myc’s oncogenic effects in hepatocellular carcinoma. Mol. Carcinog. 2015;54:1467–1479. doi: 10.1002/mc.22223.
    1. Li M, et al. Downregulation of amplified in breast cancer 1 contributes to the anti-tumor effects of sorafenib on human hepatocellular carcinoma. Oncotarget. 2016;7:29605–29619. doi: 10.18632/oncotarget.8812.
    1. Deng M, Bragelmann J, Kryukov I, Saraiva-Agostinho N, Perner S. FirebrowseR: an R client to the Broad Instituteas Firehose Pipeline. Database. 2017;2017:baw160. doi: 10.1093/database/baw160.
    1. van der Maaten L. Accelerating t-SNE using tree-based algorithms. J. Mach. Learn. Res. 2014;15:3221–3245.
    1. Kumar-Sinha C, Chinnaiyan AM. Precision oncology in the age of integrative genomics. Nat. Biotechnol. 2018;36:46–60. doi: 10.1038/nbt.4017.
    1. Webb S. The cancer bloodhounds. Nat. Biotechnol. 2016;34:1090–1094. doi: 10.1038/nbt.3717.
    1. Sundaresan TK, et al. Detection of T790M, the acquired resistance EGFR mutation, by tumor biopsy versus noninvasive blood-based analyses. Clin Cancer Res. 2016;22(5):1103–1110. doi: 10.1158/1078-0432.CCR-15-1031.
    1. Park SM, et al. Molecular profiling of single circulating tumor cells from lung cancer patients. Proc. Natl Acad. Sci. USA. 2016;113:E8379–E8386. doi: 10.1073/pnas.1608461113.
    1. Gao Y, et al. Single-cell sequencing deciphers a convergent evolution of copy number alterations from primary to circulating tumor cells. Genome Res. 2017;27:1312–1322. doi: 10.1101/gr.216788.116.
    1. Ciriello G, et al. Emerging landscape of oncogenic signatures across human cancers. Nat. Genet. 2013;45:1127–1133. doi: 10.1038/ng.2762.
    1. Hieronymus H, et al. Tumor copy number alteration burden is a pan-cancer prognostic factor associated with recurrence and death. Elife. 2018;7:e37294. doi: 10.7554/eLife.37294.
    1. Xie L, et al. FGFR2 gene amplification in gastric cancer predicts sensitivity to the selective FGFR inhibitor AZD4547. Clin. Cancer Res. 2013;19:2572–2583. doi: 10.1158/1078-0432.CCR-12-3898.
    1. Bhan I, et al. Detection and analysis of circulating epithelial cells in liquid biopsies from patients with liver disease. Gastroenterology. 2018;155:2016–2018. e11. doi: 10.1053/j.gastro.2018.09.020.
    1. de Bourcy CF, et al. A quantitative comparison of single-cell whole genome amplification methods. PLoS ONE. 2014;9:e105585. doi: 10.1371/journal.pone.0105585.
    1. Gerlinger M, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 2012;366:883–892. doi: 10.1056/NEJMoa1113205.
    1. Court CM, et al. Determination of hepatocellular carcinoma grade by needle biopsy is unreliable for liver transplant candidate selection. Liver Transplant. 2017;23(9):1123–1132. doi: 10.1002/lt.24811.
    1. Molparia B, Nichani E, Torkamani A. Assessment of circulating copy number variant detection for cancer screening. PLoS ONE. 2017;12:e0180647. doi: 10.1371/journal.pone.0180647.
    1. Court CM, et al. Reality of single circulating tumor cell sequencing for molecular diagnostics in pancreatic cancer. J. Mol. Diagn. 2016;18:688–696. doi: 10.1016/j.jmoldx.2016.03.006.
    1. Lin M, et al. Nanostructure embedded microchips for detection, isolation, and characterization of circulating tumor cells. Acc. Chem. Res. 2014;47:2941–2950. doi: 10.1021/ar5001617.
    1. Durinck S, Spellman PT, Birney E, Huber W. Mapping identifiers for the integration of genomic datasets with the R/bioconductor package biomaRt. Nat. Protoc. 2009;4:1184–1191. doi: 10.1038/nprot.2009.97.
    1. Krijthe, J. H. Rtsne: T-distributed stochastic neighbor embedding using a Barnes-Hut implementation. (2015).
    1. Warnes, G. R. gplots: Various R Programming Tools for Plotting Data. (2011).

Source: PubMed

3
Subscribe