Mutation profiling of tumor DNA from plasma and tumor tissue of colorectal cancer patients with a novel, high-sensitivity multiplexed mutation detection platform

Evelyn Kidess, Kyra Heirich, Matthew Wiggin, Valentina Vysotskaia, Brendan C Visser, Andre Marziali, Bertram Wiedenmann, Jeffrey A Norton, Mark Lee, Stefanie S Jeffrey, George A Poultsides, Evelyn Kidess, Kyra Heirich, Matthew Wiggin, Valentina Vysotskaia, Brendan C Visser, Andre Marziali, Bertram Wiedenmann, Jeffrey A Norton, Mark Lee, Stefanie S Jeffrey, George A Poultsides

Abstract

Background: Circulating tumor DNA (ctDNA) holds promise as a non-invasive means for tumor monitoring in solid malignancies. Assays with high sensitivity and multiplexed analysis of mutations are needed to enable broad application.

Methods: We developed a new assay based on sequence-specific synchronous coefficient of drag alteration (SCODA) technology, which enriches for mutant DNA to achieve high sensitivity and specificity. This assay was applied to plasma and tumor tissue from non-metastatic and metastatic colorectal cancer (CRC) patients, including patients undergoing surgical resection for CRC liver metastases.

Results: Across multiple characterization experiments, the assay demonstrated a limit of detection of 0.001% (1 molecule in 100,000) for the majority of the 46 mutations in the panel. In CRC patient samples (n=38), detected mutations were concordant in tissue and plasma for 93% of metastatic patients versus 54% of non-metastatic patients. For three patients, ctDNA identified additional mutations not detected in tumor tissue. In patients undergoing liver metastatectomy, ctDNA anticipated tumor recurrence earlier than carcinoembryonic antigen (CEA) value or imaging.

Conclusions: The multiplexed SCODA mutation enrichment and detection method can be applied to mutation profiling and quantitation of ctDNA, and is likely to have particular utility in the metastatic setting, including patients undergoing metastatectomy.

Figures

Figure 1. Multiplexed SCODA mutation enrichment and…
Figure 1. Multiplexed SCODA mutation enrichment and detection assay
A) Method. Following plasma preparation (1), plasma DNA is extracted (2a) and spiked with randomly barcoded internal controls (2b) representing every mutation in the panel, and amplified with a limited number of cycles in a multiplex PCR reaction (3). PCR primers are designed to amplify regions of the genome containing the 46 mutations of interest, and carry universal linker sequences and sample barcodes to enable subsequent sample processing and multiplexing. Amplified DNA and internal controls are enriched for the 46 mutations of interest by SCODA cyclic electrophoresis (4) in a gel containing hybridization probes against all mutations of interest. DNA enriched for mutant sequences (5a) is amplified through additional PCR as per conventional Illumina library construction, pooled with additional samples, and sequenced (6). B) Assay characterization. Each graph denotes results from samples created by titrating synthetic DNA carrying the specified mutation into reference wild-type (WT) DNA. Horizontal axes denote estimated input copies per sample, while the vertical axes denote the number of copies detected by the assay. Limit of Detection (LOD) is calculated as the greater of a single copy of mutant DNA or three standard deviations above the average background detected for each mutation in wild-type DNA samples. Input copies are limited to 10 and greater to limit the effect of sampling fluctuation in the titration. Each sample is 300 ng of DNA, such that 10 copies are equivalent to an allele fraction of ~ 0.01%.
Figure 1. Multiplexed SCODA mutation enrichment and…
Figure 1. Multiplexed SCODA mutation enrichment and detection assay
A) Method. Following plasma preparation (1), plasma DNA is extracted (2a) and spiked with randomly barcoded internal controls (2b) representing every mutation in the panel, and amplified with a limited number of cycles in a multiplex PCR reaction (3). PCR primers are designed to amplify regions of the genome containing the 46 mutations of interest, and carry universal linker sequences and sample barcodes to enable subsequent sample processing and multiplexing. Amplified DNA and internal controls are enriched for the 46 mutations of interest by SCODA cyclic electrophoresis (4) in a gel containing hybridization probes against all mutations of interest. DNA enriched for mutant sequences (5a) is amplified through additional PCR as per conventional Illumina library construction, pooled with additional samples, and sequenced (6). B) Assay characterization. Each graph denotes results from samples created by titrating synthetic DNA carrying the specified mutation into reference wild-type (WT) DNA. Horizontal axes denote estimated input copies per sample, while the vertical axes denote the number of copies detected by the assay. Limit of Detection (LOD) is calculated as the greater of a single copy of mutant DNA or three standard deviations above the average background detected for each mutation in wild-type DNA samples. Input copies are limited to 10 and greater to limit the effect of sampling fluctuation in the titration. Each sample is 300 ng of DNA, such that 10 copies are equivalent to an allele fraction of ~ 0.01%.
Figure 2. Mutations detected in tissue and…
Figure 2. Mutations detected in tissue and plasma with the multiplexed SCODA mutation enrichment and detection assay
In highlighted cells, the detected mutation allele (or alleles) is specified, and for plasma, the number of detected copies (cp) of mutant DNA is also provided, normalized for a 5 mL input volume of plasma. ND, not detected.
Figure 3. Perioperative dynamics of plasma mutation…
Figure 3. Perioperative dynamics of plasma mutation levels in patients with stage IV colorectal cancer
Panels A-D show plasma levels of detected mutations in the circulating tumor DNA (ctDNA, left y-axis) as well as CEA values (right y-axis) in patients that underwent surgery with the intent of complete metastasis resection. The value at time point 0 represents the preoperative ctDNA level. ctDNA levels (cp, copies) are normalized for 5 mL input volume of plasma. After blood draw, surgery was performed at day 0. Arrows indicate imaging assessments by computed tomography (CT). In panels B, C and D, colored shading indicates the administration of chemotherapy. PD, progressive disease; NED, no evidence of disease. CEA normal range is 0-5 ng/mL.

References

    1. Swarup V, Rajeswari MR. Circulating (cell-free) nucleic acids--a promising, non-invasive tool for early detection of several human diseases. FEBS Lett. 2007;581:795–799.
    1. Schwarzenbach H, Hoon DS, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 2011;11:426–437.
    1. Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, Thornton K, Agrawal N, Sokoll L, Szabo SA, Kinzler KW, Vogelstein B, Diaz LA., Jr Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14:985–990.
    1. Thierry AR, Mouliere F, El Messaoudi S, Mollevi C, Lopez-Crapez E, Rolet F, Gillet B, Gongora C, Dechelotte P, Robert B, Del Rio M, Lamy PJ, Bibeau F, et al. Clinical validation of the detection of KRAS and BRAF mutations from circulating tumor DNA. Nat Med. 2014;20:430–435.
    1. Burrell RA, McGranahan N, Bartek J, Swanton C. The causes and consequences of genetic heterogeneity in cancer evolution. Nature. 2013;501:338–345.
    1. Greaves M, Maley CC. Clonal evolution in cancer. Nature. 2012;481:306–313.
    1. Siegel R, Desantis C, Jemal A. Colorectal cancer statistics, 2014. CA Cancer J Clin. 2014;64:104–117.
    1. Van Cutsem E, Kohne CH, Lang I, Folprecht G, Nowacki MP, Cascinu S, Shchepotin I, Maurel J, Cunningham D, Tejpar S, Schlichting M, Zubel A, Celik I, et al. Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treatment for metastatic colorectal cancer: updated analysis of overall survival according to tumor KRAS and BRAF mutation status. J Clin Oncol. 2011;29:2011–2019.
    1. Bokemeyer C, Bondarenko I, Hartmann JT, de Braud F, Schuch G, Zubel A, Celik I, Schlichting M, Koralewski P. Efficacy according to biomarker status of cetuximab plus FOLFOX-4 as first-line treatment for metastatic colorectal cancer: the OPUS study. Ann Oncol. 2011;22:1535–1546.
    1. Loewenstein MS, Zamcheck N. Carcinoembryonic antigen (CEA) levels in benign gastrointestinal disease states. Cancer. 1978;42:1412–1418.
    1. Tanaka T, Tanaka M, Tanaka T, Ishigamori R. Biomarkers for colorectal cancer. Int J Mol Sci. 2010;11:3209–3225.
    1. Litvka A, Cercek A, Segal N, Reidy-Lagunes D, Stadler ZK, Yaeger RD, Kemeny NE, Weiser MR, Pessin MS, Saltz L. False-positive elevations of carcinoembryonic antigen in patients with a history of resected colorectal cancer. J Natl Compr Canc Netw. 2014;12:907–913.
    1. Selzner M, Hany TF, Wildbrett P, McCormack L, Kadry Z, Clavien PA. Does the novel PET/CT imaging modality impact on the treatment of patients with metastatic colorectal cancer of the liver? Ann Surg. 2004;240:1027–1034. discussion 1035-1026.
    1. Metser U, You J, McSweeney S, Freeman M, Hendler A. Assessment of tumor recurrence in patients with colorectal cancer and elevated carcinoembryonic antigen level: FDG PET/CT versus contrast-enhanced 64-MDCT of the chest and abdomen. AJR Am J Roentgenol. 2010;194:766–771.
    1. Jones RP, Stattner S, Sutton P, Dunne DF, McWhirter D, Fenwick SW, Malik HZ, Poston GJ. Controversies in the oncosurgical management of liver limited stage IV colorectal cancer. Surg Oncol. 2014;23:53–60.
    1. Lam VW, Spiro C, Laurence JM, Johnston E, Hollands MJ, Pleass HC, Richardson AJ. A systematic review of clinical response and survival outcomes of downsizing systemic chemotherapy and rescue liver surgery in patients with initially unresectable colorectal liver metastases. Ann Surg Oncol. 2012;19:1292–1301.
    1. Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, Bartlett BR, Wang H, Luber B, Alani RM, Antonarakis ES, Azad NS, Bardelli A, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6:224ra224.
    1. Lecomte T, Berger A, Zinzindohoue F, Micard S, Landi B, Blons H, Beaune P, Cugnenc PH, Laurent-Puig P. Detection of free-circulating tumor-associated DNA in plasma of colorectal cancer patients and its association with prognosis. Int J Cancer. 2002;100:542–548.
    1. Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, Valtorta E, Schiavo R, Buscarino M, Siravegna G, Bencardino K, Cercek A, Chen CT, et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature. 2012;486:532–536.
    1. Diaz LA, Jr, Williams RT, Wu J, Kinde I, Hecht JR, Berlin J, Allen B, Bozic I, Reiter JG, Nowak MA, Kinzler KW, Oliner KS, Vogelstein B. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature. 2012;486:537–540.
    1. Marzese DM, Hirose H, Hoon DS. Diagnostic and prognostic value of circulating tumor-related DNA in cancer patients. Expert Rev Mol Diagn. 2013;13:827–844.
    1. Thompson JD, Shibahara G, Rajan S, Pel J, Marziali A. Winnowing DNA for rare sequences: highly specific sequence and methylation based enrichment. PLoS One. 2012;7:e31597.
    1. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012;487:330–337.
    1. Rizzo S, Bronte G, Fanale D, Corsini L, Silvestris N, Santini D, Gulotta G, Bazan V, Gebbia N, Fulfaro F, Russo A. Prognostic vs predictive molecular biomarkers in colorectal cancer: is KRAS and BRAF wild type status required for anti-EGFR therapy? Cancer Treat Rev. 2010;36(Suppl 3):S56–61.
    1. Liao X, Lochhead P, Nishihara R, Morikawa T, Kuchiba A, Yamauchi M, Imamura Y, Qian ZR, Baba Y, Shima K, Sun R, Nosho K, Meyerhardt JA, et al. Aspirin Use, Tumor PIK3CA Mutation, and Colorectal-Cancer Survival. N Engl J Med. 2012;367:1596–1606.
    1. Ogino S, Meyerhardt JA, Irahara N, Niedzwiecki D, Hollis D, Saltz LB, Mayer RJ, Schaefer P, Whittom R, Hantel A, Benson AB, 3rd, Goldberg RM, Bertagnolli MM, Fuchs CS. KRAS mutation in stage III colon cancer and clinical outcome following intergroup trial CALGB 89803. Clin Cancer Res. 2009;15:7322–7329.
    1. De Roock W, Claes B, Bernasconi D, De Schutter J, Biesmans B, Fountzilas G, Kalogeras KT, Kotoula V, Papamichael D, Laurent-Puig P, Penault-Llorca F, Rougier P, Vincenzi B, et al. Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol. 2010;11:753–762.
    1. Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014;511:543–550.
    1. Sorensen BS, Wu L, Wei W, Tsai J, Weber B, Nexo E, Meldgaard P. Monitoring of epidermal growth factor receptor tyrosine kinase inhibitor-sensitizing and resistance mutations in the plasma DNA of patients with advanced non-small cell lung cancer during treatment with erlotinib. Cancer. 2014;120:3896–3901.
    1. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, Harris PL, Haserlat SM, Supko JG, Haluska FG, Louis DN, Christiani DC, Settleman J, Haber DA. 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. Sun Y, Ren Y, Fang Z, Li C, Fang R, Gao B, Han X, Tian W, Pao W, Chen H, Ji H. Lung adenocarcinoma from East Asian never-smokers is a disease largely defined by targetable oncogenic mutant kinases. J Clin Oncol. 2010;28:4616–4620.
    1. Zuo Z, Chen SS, Chandra PK, Galbincea JM, Soape M, Doan S, Barkoh BA, Koeppen H, Medeiros LJ, Luthra R. Application of COLD-PCR for improved detection of KRAS mutations in clinical samples. Mod Pathol. 2009;22:1023–1031.
    1. Taly V, Pekin D, Benhaim L, Kotsopoulos SK, Le Corre D, Li X, Atochin I, Link DR, Griffiths AD, Pallier K, Blons H, Bouche O, Landi B, et al. Multiplex picodroplet digital PCR to detect KRAS mutations in circulating DNA from the plasma of colorectal cancer patients. Clin Chem. 2013;59:1722–1731.
    1. Diehl F, Li M, He Y, Kinzler KW, Vogelstein B, Dressman D. BEAMing: single-molecule PCR on microparticles in water-in-oil emulsions. Nat Methods. 2006;3:551–559.
    1. Newman AM, Bratman SV, To J, Wynne JF, Eclov NC, Modlin LA, Liu CL, Neal JW, Wakelee HA, Merritt RE, Shrager JB, Loo BW, Jr., Alizadeh AA, et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med. 2014;20:548–554.
    1. Diehl F, Li M, Dressman D, He Y, Shen D, Szabo S, Diaz LA, Goodman SN, David KA, Juhl H, Kinzler KW, Vogelstein B. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proc Natl Acad Sci U S A. 2005;102:16368–16373.
    1. Kinde I, Wu J, Papadopoulos N, Kinzler KW, Vogelstein B. Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci U S A. 2011;108:9530–9535.
    1. Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC, Reuben JM, Doyle GV, Allard WJ, Terstappen LW, Hayes DF. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med. 2004;351:781–791.
    1. Lucci A, Hall CS, Lodhi AK, Bhattacharyya A, Anderson AE, Xiao L, Bedrosian I, Kuerer HM, Krishnamurthy S. Circulating tumour cells in non-metastatic breast cancer: a prospective study. Lancet Oncol. 2012;13:688–695.
    1. Nagrath S, Sequist LV, Maheswaran S, Bell DW, Irimia D, Ulkus L, Smith MR, Kwak EL, Digumarthy S, Muzikansky A, Ryan P, Balis UJ, Tompkins RG, et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature. 2007;450:1235–1239.
    1. Sastre J, Maestro ML, Puente J, Veganzones S, Alfonso R, Rafael S, García-Saenz JA, Vidaurreta M, Martín M, Arroyo M, Sanz-Casla MT, Díaz-Rubio E. Circulating tumor cells in colorectal cancer: correlation with clinical and pathological variables. Annals of Oncology. 2008;19:935–938.
    1. Iinuma H, Okinaga K, Egami H, Mimori K, Hayashi N, Nishida K, Adachi M, Mori M, Sasako M. Usefulness and clinical significance of quantitative real-time RT-PCR to detect isolated tumor cells in the peripheral blood and tumor drainage blood of patients with colorectal cancer. Int J Oncol. 2006;28:297–306.
    1. Diaz LA, Jr., Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol. 2014;32:579–586.
    1. Leary RJ, Sausen M, Kinde I, Papadopoulos N, Carpten JD, Craig D, O'Shaughnessy J, Kinzler KW, Parmigiani G, Vogelstein B, Diaz LA, Jr., Velculescu VE. Detection of chromosomal alterations in the circulation of cancer patients with whole-genome sequencing. Sci Transl Med. 2012;4:162ra154.
    1. Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell. 1990;61:759–767.
    1. Baldus SE, Schaefer KL, Engers R, Hartleb D, Stoecklein NH, Gabbert HE. Prevalence and heterogeneity of KRAS, BRAF, and PIK3CA mutations in primary colorectal adenocarcinomas and their corresponding metastases. Clin Cancer Res. 2010;16:790–799.
    1. Norton SE, Luna KK, Lechner JM, Qin J, Fernando MR. A new blood collection device minimizes cellular DNA release during sample storage and shipping when compared to a standard device. J Clin Lab Anal. 2013;27:305–311.

Source: PubMed

3
Sottoscrivi