Multi-purpose utility of circulating plasma DNA testing in patients with advanced cancers

Geraldine Perkins, Timothy A Yap, Lorna Pope, Amy M Cassidy, Juliet P Dukes, Ruth Riisnaes, Christophe Massard, Philippe A Cassier, Susana Miranda, Jeremy Clark, Katie A Denholm, Khin Thway, David Gonzalez De Castro, Gerhardt Attard, L Rhoda Molife, Stan B Kaye, Udai Banerji, Johann S de Bono, Geraldine Perkins, Timothy A Yap, Lorna Pope, Amy M Cassidy, Juliet P Dukes, Ruth Riisnaes, Christophe Massard, Philippe A Cassier, Susana Miranda, Jeremy Clark, Katie A Denholm, Khin Thway, David Gonzalez De Castro, Gerhardt Attard, L Rhoda Molife, Stan B Kaye, Udai Banerji, Johann S de Bono

Abstract

Tumor genomic instability and selective treatment pressures result in clonal disease evolution; molecular stratification for molecularly targeted drug administration requires repeated access to tumor DNA. We hypothesized that circulating plasma DNA (cpDNA) in advanced cancer patients is largely derived from tumor, has prognostic utility, and can be utilized for multiplex tumor mutation sequencing when repeat biopsy is not feasible. We utilized the Sequenom MassArray System and OncoCarta panel for somatic mutation profiling. Matched samples, acquired from the same patient but at different time points were evaluated; these comprised formalin-fixed paraffin-embedded (FFPE) archival tumor tissue (primary and/or metastatic) and cpDNA. The feasibility, sensitivity, and specificity of this high-throughput, multiplex mutation detection approach was tested utilizing specimens acquired from 105 patients with solid tumors referred for participation in Phase I trials of molecularly targeted drugs. The median cpDNA concentration was 17 ng/ml (range: 0.5-1600); this was 3-fold higher than in healthy volunteers. Moreover, higher cpDNA concentrations associated with worse overall survival; there was an overall survival (OS) hazard ratio of 2.4 (95% CI 1.4, 4.2) for each 10-fold increase in cpDNA concentration and in multivariate analyses, cpDNA concentration, albumin, and performance status remained independent predictors of OS. These data suggest that plasma DNA in these cancer patients is largely derived from tumor. We also observed high detection concordance for critical 'hot-spot' mutations (KRAS, BRAF, PIK3CA) in matched cpDNA and archival tumor tissue, and important differences between archival tumor and cpDNA. This multiplex sequencing assay can be utilized to detect somatic mutations from plasma in advanced cancer patients, when safe repeat tumor biopsy is not feasible and genomic analysis of archival tumor is deemed insufficient. Overall, circulating nucleic acid biomarker studies have clinically important multi-purpose utility in advanced cancer patients and further studies to pursue their incorporation into the standard of care are warranted.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. DNA concentrations (ng/mL) classified by…
Figure 1. DNA concentrations (ng/mL) classified by tumor types.
Box and whisker plots showing 25th, 50th and 75th percentiles, upper and lower adjacent values (whiskers) and Tukey outliers (•). P value is for a two-sided unpaired t-test on log10 DNA concentrations using Welch's correction for unequal variances.
Figure 2. cpDNA concentrations for mutational detection…
Figure 2. cpDNA concentrations for mutational detection by Sequenom OncoCarta panel (v1.0).
2A: Nonparametric ROC analyses were used to assess the limit of the Sequenom platform to detect OncoCarta panel mutations in cpDNA. Each dot on the graph corresponds to the sensitivity and specificity at one of the observed concentrations. Mutations were considered ‘available for detection’ if they were detected in the patient's FFPE tissue. Mutations were detected in FFPE samples from 37 patients. The concentration of cpDNA with the optimal ability to detect a mutation is 29.95 ng/ml (Likelihood ratio = 7.3043). The AUC calculated is 0.8075 (95% CI 0.6552–0.9598). Patients whose FFPE was unavailable or tested negative for mutations were excluded from the analysis. The specificity reference lines for quartiles of DNA concentrations are indicated in red dashed lines. 2B: Graph showing the types of mutations and cpDNA concentrations at which they were detected in different tumors. Mutations were detected in six oncogenes. Symbols represent different tumor types.
Figure 3. Relationship between cpDNA concentration and…
Figure 3. Relationship between cpDNA concentration and survival.
(3A) Kaplan-Meier graph showing survival curves by cpDNA concentration in 101 patients with advanced solid tumors. Patients in the unfavorable category had concentrations greater than a healthy volunteer cohort maximum of 13.3 ng/ml (logrank p = 0.0383). (3B) Survivor function estimated from univariate Cox regression showing predicted survival curves for a range of cpDNA concentrations. A hazard ratio of 2.4 (p = 0.002) is depicted between adjacent curves.
Figure 4. Relationship between cpDNA concentration and…
Figure 4. Relationship between cpDNA concentration and RMH prognostic score.
Scatterplot showing the relationship between cpDNA concentration and RMH prognostic score. There was a significant positive linear trend between log10(cpDNA) and RMH score (beta = 0.252, p<0.0001).

References

    1. Macconaill LE, Garraway LA (2010) Clinical implications of the cancer genome. J Clin Oncol 28: 5219–5228.
    1. de Bono JS, Ashworth A (2010) Translating cancer research into targeted therapeutics. Nature 467: 543–549.
    1. Yap TA, Sandhu SK, Workman P, de Bono JS (2010) Envisioning the future of early anticancer drug development. Nat Rev Cancer 10: 514–523.
    1. Lievre A, Bachet JB, Boige V, Cayre A, Le Corre D, et al. (2008) KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab. J Clin Oncol 26: 374–379.
    1. Amado RG, Wolf M, Peeters M, Van Cutsem E, Siena S, et al. (2008) Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol 26: 1626–1634.
    1. Verweij J, Casali PG, Zalcberg J, LeCesne A, Reichardt P, et al. (2004) Progression-free survival in gastrointestinal stromal tumours with high-dose imatinib: randomised trial. Lancet 364: 1127–1134.
    1. Von Hoff DD, Stephenson JJ Jr, Rosen P, Loesch DM, Borad MJ, et al. (2010) Pilot study using molecular profiling of patients' tumors to find potential targets and select treatments for their refractory cancers. J Clin Oncol 28: 4877–4883.
    1. van Puijenbroek M, Dierssen JW, Stanssens P, van Eijk R, Cleton-Jansen AM, et al. (2005) Mass spectrometry-based loss of heterozygosity analysis of single-nucleotide polymorphism loci in paraffin embedded tumors using the MassEXTEND assay: single-nucleotide polymorphism loss of heterozygosity analysis of the protein tyrosine phosphatase receptor type J in familial colorectal cancer. J Mol Diagn 7: 623–630.
    1. Fumagalli D, Gavin PG, Taniyama Y, Kim SI, Choi HJ, et al. (2010) A rapid, sensitive, reproducible and cost-effective method for mutation profiling of colon cancer and metastatic lymph nodes. BMC Cancer 10: 101.
    1. MacConaill LE, Campbell CD, Kehoe SM, Bass AJ, Hatton C, et al. (2009) Profiling critical cancer gene mutations in clinical tumor samples. PLoS One 4: e7887.
    1. Schwarzenbach H, Hoon DS, Pantel K (2011) Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer 11: 426–437.
    1. Thierry AR, Mouliere F, Gongora C, Ollier J, Robert B, et al. (2010) Origin and quantification of circulating DNA in mice with human colorectal cancer xenografts. Nucleic Acids Res 38: 6159–6175.
    1. Goebel G, Zitt M, Muller HM (2005) Circulating nucleic acids in plasma or serum (CNAPS) as prognostic and predictive markers in patients with solid neoplasias. Dis Markers 21: 105–120.
    1. Board RE, Ellison G, Orr MC, Kemsley KR, McWalter G, et al. (2009) Detection of BRAF mutations in the tumour and serum of patients enrolled in the AZD6244 (ARRY-142886) advanced melanoma phase II study. Br J Cancer 101: 1724–1730.
    1. Lefebure B, Charbonnier F, Di Fiore F, Tuech JJ, Le Pessot F, et al. (2010) Prognostic value of circulating mutant DNA in unresectable metastatic colorectal cancer. Ann Surg 251: 275–280.
    1. Board RE, Wardley AM, Dixon JM, Armstrong AC, Howell S, et al. (2010) Detection of PIK3CA mutations in circulating free DNA in patients with breast cancer. Breast Cancer Res Treat 120: 461–467.
    1. Chen H, Tu H, Meng ZQ, Chen Z, Wang P, et al. (2010) K-ras mutational status predicts poor prognosis in unresectable pancreatic cancer. Eur J Surg Oncol 36: 657–662.
    1. Maheswaran S, Sequist LV, Nagrath S, Ulkus L, Brannigan B, et al. (2008) Detection of mutations in EGFR in circulating lung-cancer cells. N Engl J Med 359: 366–377.
    1. Board RE, Thelwell NJ, Ravetto PF, Little S, Ranson M, et al. (2008) Multiplexed assays for detection of mutations in PIK3CA. Clin Chem 54: 757–760.
    1. Arkenau HT, Barriuso J, Olmos D, Ang JE, de Bono J, et al. (2009) Prospective validation of a prognostic score to improve patient selection for oncology phase I trials. J Clin Oncol 27: 2692–2696.
    1. Carpten JD, Faber AL, Horn C, Donoho GP, Briggs SL, et al. (2007) A transforming mutation in the pleckstrin homology domain of AKT1 in cancer. Nature 448: 439–444.
    1. Schwarzenbach H, Muller V, Milde-Langosch K, Steinbach B, Pantel K (2011) Evaluation of cell-free tumour DNA and RNA in patients with breast cancer and benign breast disease. Mol Biosyst 7: 2848–2854.
    1. Kim ES, Herbst RS, Wistuba II, Lee JJ, Blumenschein J, et al. (2011) The BATTLE Trial: Personalizing Therapy for Lung Cancer. Cancer Discovery 1: 44–53.
    1. Coumans FA, Doggen CJ, Attard G, de Bono JS, Terstappen LW (2010) All circulating EpCAM+CK+CD45− objects predict overall survival in castration-resistant prostate cancer. Ann Oncol 21: 1851–1857.
    1. Arcila M, Lau C, Nafa K, Ladanyi M (2011) Detection of KRAS and BRAF mutations in colorectal carcinoma roles for high-sensitivity locked nucleic acid-PCR sequencing and broad-spectrum mass spectrometry genotyping. J Mol Diagn 13: 64–73.
    1. Wang S, An T, Wang J, Zhao J, Wang Z, et al. (2010) Potential clinical significance of a plasma-based KRAS mutation analysis in patients with advanced non-small cell lung cancer. Clin Cancer Res 16: 1324–1330.
    1. Diehl F, Schmidt K, Durkee KH, Moore KJ, Goodman SN, et al. (2008) Analysis of mutations in DNA isolated from plasma and stool of colorectal cancer patients. Gastroenterology 135: 489–498.
    1. Navin N, Kendall J, Troge J, Andrews P, Rodgers L, et al. (2011) Tumour evolution inferred by single-cell sequencing. Nature 472: 90–94.

Source: PubMed

3
Abonnere