Skip to main content
Top
Published in: BMC Cancer 1/2017

Open Access 01-12-2017 | Research article

Precision oncology using a limited number of cells: optimization of whole genome amplification products for sequencing applications

Authors: Shonan Sho, Colin M. Court, Paul Winograd, Sangjun Lee, Shuang Hou, Thomas G. Graeber, Hsian-Rong Tseng, James S. Tomlinson

Published in: BMC Cancer | Issue 1/2017

Login to get access

Abstract

Background

Sequencing analysis of circulating tumor cells (CTCs) enables “liquid biopsy” to guide precision oncology strategies. However, this requires low-template whole genome amplification (WGA) that is prone to errors and biases from uneven amplifications. Currently, quality control (QC) methods for WGA products, as well as the number of CTCs needed for reliable downstream sequencing, remain poorly defined. We sought to define strategies for selecting and generating optimal WGA products from low-template input as it relates to their potential applications in precision oncology strategies.

Methods

Single pancreatic cancer cells (HPAF-II) were isolated using laser microdissection. WGA was performed using multiple displacement amplification (MDA), multiple annealing and looping based amplification (MALBAC) and PicoPLEX. Quality of amplified DNA products were assessed using a multiplex/RT-qPCR based method that evaluates for 8-cancer related genes and QC-scores were assigned. We utilized this scoring system to assess the impact of de novo modifications to the WGA protocol. WGA products were subjected to Sanger sequencing, array comparative genomic hybridization (aCGH) and next generation sequencing (NGS) to evaluate their performances in respective downstream analyses providing validation of the QC-score.

Results

Single-cell WGA products exhibited a significant sample-to-sample variability in amplified DNA quality as assessed by our 8-gene QC assay. Single-cell WGA products that passed the pre-analysis QC had lower amplification bias and improved aCGH/NGS performance metrics when compared to single-cell WGA products that failed the QC. Increasing the number of cellular input resulted in improved QC-scores overall, but a resultant WGA product that consistently passed the QC step required a starting cellular input of at least 20-cells. Our modified-WGA protocol effectively reduced this number, achieving reproducible high-quality WGA products from ≥5-cells as a starting template. A starting cellular input of 5 to 10-cells amplified using the modified-WGA achieved aCGH and NGS results that closely matched that of unamplified, batch genomic DNA.

Conclusion

The modified-WGA protocol coupled with the 8-gene QC serve as an effective strategy to enhance the quality of low-template WGA reactions. Furthermore, a threshold number of 5–10 cells are likely needed for a reliable WGA reaction and product with high fidelity to the original starting template.
Appendix
Available only for authorised users
Literature
2.
go back to reference Alix-Panabières C, Pantel K. Clinical applications of circulating tumor cells and circulating tumor DNA as liquid biopsy. Cancer Discovery. 2016;6(5):479.CrossRefPubMed Alix-Panabières C, Pantel K. Clinical applications of circulating tumor cells and circulating tumor DNA as liquid biopsy. Cancer Discovery. 2016;6(5):479.CrossRefPubMed
5.
go back to reference Ignatiadis M, Dawson SJ. Circulating tumor cells and circulating tumor DNA for precision medicine: dream or reality? Ann Oncol. 2014;25(12):2304–13.CrossRefPubMed Ignatiadis M, Dawson SJ. Circulating tumor cells and circulating tumor DNA for precision medicine: dream or reality? Ann Oncol. 2014;25(12):2304–13.CrossRefPubMed
6.
go back to reference Wills QF, Mead AJ. Application of single-cell genomics in cancer: promise and challenges. Human Molecular Genetics. 2015;24(R1):R74-R84. doi:10.1093/hmg/ddv235. PubMed. Wills QF, Mead AJ. Application of single-cell genomics in cancer: promise and challenges. Human Molecular Genetics. 2015;24(R1):R74-R84. doi:10.​1093/​hmg/​ddv235. PubMed.
7.
go back to reference Hong B, Zu Y. Detecting Circulating Tumor Cells: Current Challenges and New Trends. Theranostics. 2013;3(6):377–94. doi:10.7150/thno.5195. PubMed. Hong B, Zu Y. Detecting Circulating Tumor Cells: Current Challenges and New Trends. Theranostics. 2013;3(6):377–94. doi:10.​7150/​thno.​5195. PubMed.
9.
go back to reference Court CM, Ankeny JS, Sho S, Tomlinson JS. Circulating tumor cells in gastrointestinal cancer: current practices and future directions. In: Bentrem D, Benson AB, editors. Gastrointestinal Malignancies. Cham: Springer International Publishing; 2016. p. 345–76.CrossRef Court CM, Ankeny JS, Sho S, Tomlinson JS. Circulating tumor cells in gastrointestinal cancer: current practices and future directions. In: Bentrem D, Benson AB, editors. Gastrointestinal Malignancies. Cham: Springer International Publishing; 2016. p. 345–76.CrossRef
20.
go back to reference van Beers EH, Joosse SA, Ligtenberg MJ, Fles R, Hogervorst FBL, Verhoef S, et al. A multiplex PCR predictor for aCGH success of FFPE samples. Br J Cancer. 2005;94(2):333–7.CrossRef van Beers EH, Joosse SA, Ligtenberg MJ, Fles R, Hogervorst FBL, Verhoef S, et al. A multiplex PCR predictor for aCGH success of FFPE samples. Br J Cancer. 2005;94(2):333–7.CrossRef
21.
go back to reference Garvin T, Aboukhalil R, Kendall J, Baslan T, Atwal GS, Hicks J, et al. Interactive analysis and assessment of single-cell copy-number variations. Nat Meth. 2015;12(11):1058–60. doi:10.1038/nmeth.3578.CrossRef Garvin T, Aboukhalil R, Kendall J, Baslan T, Atwal GS, Hicks J, et al. Interactive analysis and assessment of single-cell copy-number variations. Nat Meth. 2015;12(11):1058–60. doi:10.​1038/​nmeth.​3578.CrossRef
24.
go back to reference Hayes DF, Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Miller MC, et al. Circulating tumor cells at each follow-up time point during therapy of metastatic breast cancer patients predict progression-free and overall survival. Clin Cancer Res. 2006;12(14):4218.CrossRefPubMed Hayes DF, Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Miller MC, et al. Circulating tumor cells at each follow-up time point during therapy of metastatic breast cancer patients predict progression-free and overall survival. Clin Cancer Res. 2006;12(14):4218.CrossRefPubMed
26.
go back to reference Barker DL, Hansen MS, Faruqi AF, Giannola D, Irsula OR, Lasken RS, et al. Two methods of whole-genome amplification enable accurate genotyping across a 2320-SNP linkage panel. Genome Res. 2004;14 doi:10.1101/gr.1949704. Barker DL, Hansen MS, Faruqi AF, Giannola D, Irsula OR, Lasken RS, et al. Two methods of whole-genome amplification enable accurate genotyping across a 2320-SNP linkage panel. Genome Res. 2004;14 doi:10.​1101/​gr.​1949704.
27.
go back to reference Polzer B, Medoro G, Pasch S, Fontana F, Zorzino L, Pestka A, et al. Molecular profiling of single circulating tumor cells with diagnostic intention. EMBO Molecular Medicine. 2014;6(11):1371–86. Polzer B, Medoro G, Pasch S, Fontana F, Zorzino L, Pestka A, et al. Molecular profiling of single circulating tumor cells with diagnostic intention. EMBO Molecular Medicine. 2014;6(11):1371–86.
29.
33.
go back to reference Nilsen G, Liestol K, Loo P, Moen Vollan HK, Eide MB, Rueda OM, et al. Copynumber: efficient algorithms for single- and multi-track copy number segmentation. BMC Genomics. 2012;13 doi:10.1186/1471-2164-13-591. Nilsen G, Liestol K, Loo P, Moen Vollan HK, Eide MB, Rueda OM, et al. Copynumber: efficient algorithms for single- and multi-track copy number segmentation. BMC Genomics. 2012;13 doi:10.​1186/​1471-2164-13-591.
34.
go back to reference Geigl JB, Obenauf AC, Waldispuehl-Geigl J, Hoffmann EM, Auer M, Hörmann M, et al. Identification of small gains and losses in single cells after whole genome amplification on tiling oligo arrays. Nucleic Acids Research. 2009;37(15):e105-e. doi:10.1093/nar/gkp526. Geigl JB, Obenauf AC, Waldispuehl-Geigl J, Hoffmann EM, Auer M, Hörmann M, et al. Identification of small gains and losses in single cells after whole genome amplification on tiling oligo arrays. Nucleic Acids Research. 2009;37(15):e105-e. doi:10.​1093/​nar/​gkp526.
36.
Metadata
Title
Precision oncology using a limited number of cells: optimization of whole genome amplification products for sequencing applications
Authors
Shonan Sho
Colin M. Court
Paul Winograd
Sangjun Lee
Shuang Hou
Thomas G. Graeber
Hsian-Rong Tseng
James S. Tomlinson
Publication date
01-12-2017
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2017
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-017-3447-6

Other articles of this Issue 1/2017

BMC Cancer 1/2017 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine