Elsevier

Current Problems in Cancer

Volume 41, Issue 3, May–June 2017, Pages 212-221
Current Problems in Cancer

Circulating tumor DNA—From bench to bedside

https://doi.org/10.1016/j.currproblcancer.2017.02.005Get rights and content

Abstract

In the era of personalized medicine, tumor sampling is paramount to enable the assessment of actionable molecular aberrations to help rationalize and guide treatment decisions. Longitudinal tracking of such aberrations may also be helpful to detect emerging drug resistance and to allow for timely modifications to ongoing therapies to improve patient outcomes. Nevertheless, tumor tissue sampling involves an invasive procedure with potential risks to patients and involves logistical challenges. As such, other less invasive and safer methods such as blood sampling for molecular profiling has been gaining traction. In this article, we discuss the concept of circulating tumor DNA, the technology platforms available for its interrogation, and its current applications in the clinic. We also envision how circulating tumor DNA may be applied at multiple time points along a patient’s cancer journey to guide diagnosis, prognostication, and therapeutic decisions.

Introduction

Solid tumor sampling has been the cornerstone of cancer diagnosis for decades, and it is also the foundation upon which oncologists build treatment plans. In this era of precision medicine, armed with our improved understanding of tumor biology, including the concept of clonal evolution and intratumor heterogeneity, it has become evident that oncogenesis is a dynamic process where tumors are constantly evolving to seek compensatory escape routes under distinct selection pressures from antitumor therapeutic agents. As such, a tumor biopsy undertaken at a single time point at the time of diagnosis may not be truly representative of subsequent molecular changes that may have occurred during the course of a patient’s disease because of the potential for clonal evolution.1 A recent analysis of somatic genomic profiles with next-generation sequencing (NGS) of over 15,000 circulating tumor DNA (ctDNA) samples with matched tissue from 386 patients with advanced cancers showed concordance of more than 90% for truncal driver mutations, but only 13%-33% concordance for subclonal aberrations, such as epidermal growth factor receptor (EGFR) T790M mutations, likely reflecting new aberrations detected with the potential emergence of drug resistance.2

The acknowledgment of this phenomenon has led to growing advocation for the use of repeat tumor biopsies upon treatment progression to enable better characterization of tumors to guide treatment choices. Nonetheless, solid tumor sampling is an invasive process, which can be technically challenging and not without risks of procedural complications. Additionally, it is logistically challenging for patients to undergo frequent multiple biopsies, while issues with intratumor heterogeneity raises concerns of whether a single core biopsy is reflective of the genomic landscape of the whole tumor. The concept of a “liquid biopsy,” whereby cells and other cell products from the mononuclear cellular fractions can be analyzed from a blood draw, has become an attractive alternative to solid tumor sampling. Blood sampling is a less invasive process that will allow serial sampling, thus possibly obtaining a “real time” reflection of the tumor status of a patient and potentially minimizing issues of sampling bias. As advanced cancers shed cells and other cellular fragments into the bloodstream from both primary and metastatic sites, blood sampling may also provide a global summary of tumor heterogeneity.

The Food and Drug Administration (FDA) and European Medical Agency recently approved the first blood-based companion diagnostic assay, Cobas EGFR mutation test v2 (Roche Molecular systems) in June 2016. This real-time polymerase chain reaction (PCR) test identifies EGFR exon 19 deletions and EGFR exon 21 L858R mutations based on the detection of ctDNA in plasma derived from EDTA anticoagulated peripheral whole blood, therefore indicating tumor sensitivity to EGFR inhibitors, such as erlotinib (Tarceva, Roche). In September 2016, this FDA approval was extended to include EGFR T790M testing, a common resistance mutation emerging on anti-EGFR therapy, where a positive T790M mutation indicates sensitivity to osimertinib (Targrisso, Roche).

In this article, we focus on the use of ctDNA and discuss its clinical applications as a surrogate for conventional tumor biopsies for a range of uses, including molecular profiling, monitoring of antitumor response, and emerging resistance so as to guide treatment modification.

Section snippets

Development and technical aspects of ctDNA assay

Although the detection of circulating free DNA (cfDNA) was first described by Mandel and Metais,3 it was not until almost 4 decades later when Stroun et al4 showed evidence of the presence of ctDNA in cfDNA fragments. The presence of cfDNA is largely a result of cell death, with highly fragmented, double-stranded DNA of approximately 150 bp in size being released into the bloodstream. Currently, the most successful application of cfDNA in the clinics is arguably its use in prenatal testing for

Clinical applications of ctDNA

The identification and analysis of ctDNA holds promise for a variety of clinical applications in terms of diagnostic, prognostic, and therapeutic implications (Fig 1). Currently, its use to aid therapeutic decisions in treatment choice has been most widely implemented, especially with the FDA and European Medical Agency approval of the Cobas EGFR mutation test to guide treatment decisions in NSCLC. Nonetheless, its other clinical applications are also under intense research, and with improving

Other blood-based biomarkers

Although a detailed discussion is beyond the scope of this article, other blood-based biomarkers have also been studied as potential surrogates for tumor biopsies, of which circulating tumor cells are the most intensively investigated.33 Other nucleic acid fragments studied include microRNA (miRNA)—small noncoding RNA that regulate gene expression at a posttranscriptional level—which have also been shown to be present in serum samples and may therefore act as potential biomarkers for disease.47

Future perspectives

Although advances in assay techniques have improved detection rates of cfDNA, it remains challenging to differentiate tumor-specific ctDNA from normal cfDNA, with a wide variability of ctDNA-to-cfDNA ratio observed in different studies. Analysis of cfDNA in 52 patients with metastatic breast cancer showed a median mutant allele fraction of 4% (interquartile range: 1-14).30 Studies in other tumor types including prostate and colorectal cancers have shown mutation frequencies of 0.09%-17.75% and

References (50)

  • F. Diehl et al.

    Circulating mutant DNA to assess tumor dynamics

    Nat Med

    (2008)
  • F. Diehl et al.

    Detection and quantification of mutations in the plasma of patients with colorectal tumors

    Proc Natl Acad Sci U S A.

    (2005)
  • M. Jung et al.

    Changes in concentration of DNA in serum and plasma during storage of blood samples

    Clin Chem

    (2003)
  • C.R. Newton et al.

    Analysis of any point mutation in DNA. The amplification refractory mutation system (ARMS)

    Nucleic Acids Res

    (1989)
  • H. Kimura et al.

    Detection of epidermal growth factor receptor mutations in serum as a predictor of the response to gefitinib in patients with non-small-cell lung cancer

    Clin Cancer Res

    (2006)
  • R.E. Board et al.

    Detection of PIK3CA mutations in circulating free DNA in patients with breast cancer

    Breast Cancer Res Treat

    (2010)
  • K.L. Spindler et al.

    Quantitative cell-free DNA, KRAS, and BRAF mutations in plasma from patients with metastatic colorectal cancer during treatment with cetuximab and irinotecan

    Clin Cancer Res

    (2012)
  • D. Dressman et al.

    Transforming single DNA molecules into fluorescent magnetic particles for detection and enumeration of genetic variations

    Proc Natl Acad Sci U S A

    (2003)
  • M.J. Higgins et al.

    Detection of tumor PIK3CA status in metastatic breast cancer using peripheral blood

    Clin Cancer Res

    (2012)
  • L.A. Diaz et al.

    The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers

    Nature

    (2012)
  • S. Misale et al.

    Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer

    Nature

    (2012)
  • B.J. Hindson et al.

    High-throughput droplet digital PCR system for absolute quantitation of DNA copy number

    Anal Chem

    (2011)
  • A.S. Whale et al.

    An international inter-laboratory digital pcr study demonstrates high reproducibility for the measurement of a rare sequence variant

    bioRxiv

    (2016)
  • A.G. Sacher et al.

    Prospective validation of rapid plasma genotyping for the detection of EGFR and KRAS mutations in advanced lung cancer

    JAMA Oncol

    (2016)
  • J.A. Beaver et al.

    Detection of cancer DNA in plasma of patients with early-stage breast cancer

    Clin Cancer Res

    (2014)
  • View full text