Cell free circulating tumor nucleic acids, a revolution in personalized cancer medicine

https://doi.org/10.1016/j.critrevonc.2019.102827Get rights and content

Abstract

Innovative diagnostics are becoming an essential component in personalized cancer medicine. These diagnostics are increasingly based on cell-free nucleic acids and membrane vesicles. Isolating and sequencing cell free circulating DNA (cfDNA) in plasma may progressively substitute tumor biopsies. A small albeit now detectable fraction of cfDNA correspond to circulating tumor DNA (ctDNA). In this review, we describe the pre-analytical procedures for collecting ctDNA from plasma, since these procedures should be optimized within laboratories depending on the available infrastructures. We also provide an overview of the technological breakthrough in ctDNA Isolation for instance digital PCR methods and next generation sequencing techniques and discuss their key challenges. The clinical implementations of liquid biopsy and more specifically ctDNA in cancer management are reviewed. We predict in the near future, ctDNA will be used more routinely to guide cancer treatment and provide a new approach to personalize treatment in precision medicine.

Section snippets

The history of liquid biopsy

In 1948, Mandel and Métais detected and quantified the presence of cell-free nucleic acid (cfNA) in human blood of both healthy and diseased patients for the first time (Schwarzenbach et al., 2011). In 1966, researchers discovered high levels of cfDNA in lupus patients (Thierry et al., 2016). These studies attracted some attention in the scientific community and the presence of tumor-specific mutations in cfNA was not reported by scientists until 1994. In the plasma of patients with pancreatic

The source of ctDNA

Circulating tumor DNA or ctDNA can be released from CTCs, primary and secondary tumors into the circulation of cancer patients. They could be presented in many forms; either cfDNA, bound to complex proteins, bound to cell surfaces, or vesicles (Thierry et al., 2016). Most ctDNA is made up of apoptotic and/or necrotic tumor cells that release their divided DNA into the blood (Wang et al., 2017b, 2003) (Fig. 1). Besides, viable tumor cells can release microvesicles (or exosomes), which are

The quantity of ctDNA in bloodstream

The number of cfDNA copies in cancer patients varies extensively (Lange and Laird, 2013), which depends on the tumor type, location and cancer stage (Han et al., 2017). While the concentration of cfDNA in cancer patients' blood is higher than in healthy individuals' blood and non-malignant patients, the concentration of cfDNA varies significantly with an average of 180 ng/mL (Barbany et al., 2019). However, in some studies less than 100 ng/mL has been reported for the most of cancer patients (

Detection methods

Circulating tumor DNA containing the same molecular aberrations as the solid tumor, found in the bloodstream, refers to DNA from cancer cells and tumors (Thierry et al., 2016). Several methods can detect the existence of tumor-derived DNA in cfDNA. Nonetheless, the low amount, high degradation, and high mix of normal cfDNA with ctDNA cause serious challenges to choose the appropriate detection method. Since almost all tumors are marked by various subclonal populations, the problem would be

ctDNA as biomarkers

The detection of ctDNA in plasma could be useful for many diagnostic applications in addition to limiting the need for solid biopsies (Wang et al., 2017a). The discovery of a proportion of circulating DNA in cancer patients has created the potential for a so-called "liquid biopsy" to mark tumor genetic characteristics as an alternative to tissue biopsy (Esposito et al., 2016). CtDNAs are thought to be easily detected even in the early stages of cancer patients’ plasma (Alix-Panabieres and

Challenges for diagnostic and prognostic tests

As we move into the era of personalized cancer medicine, the need for more innovative cancer diagnostic and prognostic assays would seriously raise. Cellular nanoparticulates such as cfNAs (cfDNA, cfRNA) are now considered as important biomarkers with high performance in medicine. There is huge hope that blood-borne cfNA could replace more invasive tumor solid biopsies in order to detect mutation/methylation in cancer and monitor treatment. Usually, conventional techniques for cfDNA biomarker

Future developments in personalized medicine

CTCs and ctDNA analyses have paved new diagnostic and prognostic avenues and are nominated to date the bases of liquid biopsy. To what extent in the future they might replace tumor solid biopsies is not completely known but their contribution in molecular personalized medicine is anticipated. However, they are not expected to entirely replace tumor biopsies since they cannot address many important factors such as changes in and interactions with the tumor microenvironment (Lewis et al., 2015).

Conclusions

In the next few years, liquid biopsy especially cfDNA-based will be implemented in clinical studies and drug development and it will likely become an integral part of diagnostics in oncology but to reach that goal, a more laborious plan of devoted research with appropriate cohort studies and standardized analytical methods is necessary.

Funding

This study was supported financially by Iran National Science Foundation (INSF; proposal number: 93048371).

Authors’ contributions

MAK and AP designed the study. MAK and AP drafted the manuscript. MAK, and JPT coordinated, edited, and finalized the drafting of the manuscript. All authors read and approved the final manuscript.

Declaration of Competing Interest

The authors declare that they have no competing interests.

Acknowledgements

Our sincere thanks go to Mrs. Afsaneh Mojtabanezhad Shariatpanahi, and Ms Marjan Azghandi, who provided good comments and feedback, Ms. Mona Nematy who designed and drew the figures and Mr. Ebrahim Pouladin for his support in cancer research programs.

References (120)

  • L. Hamzehzadeh et al.

    Common KRAS and NRAS gene mutations in sporadic colorectal cancer in Northeastern Iranian patients

    Curr. Probl. Cancer

    (2018)
  • X. Han et al.

    Circulating tumor DNA as biomarkers for Cancer detection

    Genomics Proteomics Bioinformatics

    (2017)
  • S.A. Joosse et al.

    Tumor-educated platelets as liquid biopsy in Cancer patients

    Cancer Cell

    (2015)
  • M.A. Kerachian et al.

    Long interspersed nucleotide element-1 (LINE-1) methylation in colorectal cancer

    Clin. Chim. Acta

    (2019)
  • Y.M. Lo et al.

    Presence of fetal DNA in maternal plasma and serum

    Lancet

    (1997)
  • Y.M. Lo et al.

    Rapid clearance of fetal DNA from maternal plasma

    Am. J. Hum. Genet.

    (1999)
  • N. Normanno et al.

    The liquid biopsy in the management of colorectal cancer patients: current applications and future scenarios

    Cancer Treat. Rev.

    (2018)
  • Z.D. Smith et al.

    High-throughput bisulfite sequencing in mammalian genomes

    Methods

    (2009)
  • C. Alix-Panabieres et al.

    Clinical applications of circulating tumor cells and circulating tumor DNA as liquid biopsy

    Cancer Discov.

    (2016)
  • N. Andor et al.

    Pan-cancer analysis of the extent and consequences of intratumor heterogeneity

    Nat. Med.

    (2016)
  • P. Angsuwatcharakon et al.

    Detection of pancreas site-specific methylation from circulating DNA of patients with pancreatic Cancer; a potential biomarker for liquid biopsy

    Gastroenterology

    (2017)
  • A.A. Azad et al.

    Androgen receptor gene aberrations in circulating cell-free DNA: biomarkers of therapeutic resistance in castration-resistant prostate Cancer

    Clin. Cancer Res.

    (2015)
  • G. Barbany et al.

    Cell-free tumour DNA testing for early detection of cancer - a potential future tool

    J. Intern. Med.

    (2019)
  • C. Bettegowda et al.

    Detection of circulating tumor DNA in early- and late-stage human malignancies

    Sci. Transl. Med.

    (2014)
  • S. Breitbach et al.

    Direct quantification of cell-free, circulating DNA from unpurified plasma

    PLoS One

    (2014)
  • D. Cappellen et al.

    Frequent activating mutations of FGFR3 in human bladder and cervix carcinomas

    Nat. Genet.

    (1999)
  • K.C. Chan et al.

    Cancer genome scanning in plasma: detection of tumor-associated copy number aberrations, single-nucleotide variants, and tumoral heterogeneity by massively parallel sequencing

    Clin. Chem.

    (2013)
  • T.R. Church et al.

    Prospective evaluation of methylated SEPT9 in plasma for detection of asymptomatic colorectal cancer

    Gut

    (2014)
  • V. Combaret et al.

    Circulating MYCN DNA as a tumor-specific marker in neuroblastoma patients

    Cancer Res.

    (2002)
  • S.J. Dawson et al.

    Analysis of circulating tumor DNA to monitor metastatic breast cancer

    N. Engl. J. Med.

    (2013)
  • 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)
  • 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. Elazezy et al.

    Techniques of using circulating tumor DNA as a liquid biopsy component in cancer management

    Comput. Struct. Biotechnol. J.

    (2018)
  • F. Farace et al.

    A direct comparison of CellSearch and ISET for circulating tumour-cell detection in patients with metastatic carcinomas

    Br. J. Cancer

    (2011)
  • M.R. Fernando et al.

    New evidence that a large proportion of human blood plasma cell-free DNA is localized in exosomes

    PLoS One

    (2017)
  • T. Forshew et al.

    Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA

    Sci. Transl. Med.

    (2012)
  • D.C. Garcia-Olmo et al.

    Cell-free nucleic acids circulating in the plasma of colorectal cancer patients induce the oncogenic transformation of susceptible cultured cells

    Cancer Res.

    (2010)
  • V.J. Gauthier et al.

    Blood clearance kinetics and liver uptake of mononucleosomes in mice

    J. Immunol.

    (1996)
  • G. Genovese et al.

    Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence

    N. Engl. J. Med.

    (2014)
  • H. Gevensleben et al.

    Noninvasive detection of HER2 amplification with plasma DNA digital PCR

    Clin. Cancer Res.

    (2013)
  • E. Gormally et al.

    TP53 and KRAS2 mutations in plasma DNA of healthy subjects and subsequent cancer occurrence: a prospective study

    Cancer Res.

    (2006)
  • T.J. Gould et al.

    Extracellular DNA and histones: double-edged swords in immunothrombosis

    J. Thromb. Haemost.

    (2015)
  • T.Y. Ha

    MicroRNAs in human diseases: from Cancer to cardiovascular disease

    Immune Netw.

    (2011)
  • E. Heitzer et al.

    Tumor-associated copy number changes in the circulation of patients with prostate cancer identified through whole-genome sequencing

    Genome Med.

    (2013)
  • M. Houshmand et al.

    Assessment of bone morphogenetic protein 3 methylation in Iranian patients with colorectal Cancer

    Middle East J. Dig. Dis.

    (2017)
  • H.C. Hsu et al.

    Mutations of KRAS/NRAS/BRAF predict cetuximab resistance in metastatic colorectal cancer patients

    Oncotarget

    (2016)
  • W. Hu et al.

    Post surgery circulating free tumor DNA is a predictive biomarker for relapse of lung cancer

    Cancer Med.

    (2017)
  • X. Huang et al.

    Characterization of human plasma-derived exosomal RNAs by deep sequencing

    BMC Genomics

    (2013)
  • P. Ilse et al.

    Analysis of SHOX2 methylation as an aid to cytology in lung cancer diagnosis

    Cancer Genomics Proteomics

    (2014)
  • Cited by (24)

    • Validation of a highly sensitive Sanger sequencing in detecting EGFR mutations from circulating tumor DNA in patients with lung cancers

      2022, Clinica Chimica Acta
      Citation Excerpt :

      These methods differ significantly in sensitivity, specificity, and coverage of EGFR mutations [5]. Although amplification refractory mutation system (ARMS), droplet digital PCR (ddPCR), and BEAMing (beads, emulsion, amplification, and magnetics) have good sensitivity and detection capabilities to identify various stages of cancer, their clinical applicability is limited because these methods can only identify known mutations [6–8]. Conversely, NGS represents a broader approach that covers even non-hotspot regions of the EGFR gene [9], but has prolonged turnaround times compared with PCR-based assay.

    • The clinical value of circulating free tumor DNA in testicular germ cell tumor patients

      2022, Urologic Oncology: Seminars and Original Investigations
      Citation Excerpt :

      Circulating free DNA (cfDNA) are DNA fragments mostly of the length of 150 to 200 bp released to the peripheral blood (PB) from both malignant and normal cells due to apoptosis, necrosis or secretion. It is detectable in the blood serum or plasma, and appears to be a valuable source of genetic material [8]. The presence of tumor cfDNA has been identified in a number of malignancies, however, the total amounts and proportions of tumor cfDNA among all cfDNA may vary greatly depending on the tumor type and stage.

    • Noncoding RNAs in patients with colorectal cancer

      2022, Clinical Applications of Noncoding RNAs in Cancer
    • Fusion transcript discovery using RNA sequencing in formalin-fixed paraffin-embedded specimen

      2021, Critical Reviews in Oncology/Hematology
      Citation Excerpt :

      These changes overall reduce effective RNA input and the sequencing fidelity. Quantitative real-time PCR (qRT-PCR) and digital droplet PCR (ddPCR) have been used to evaluate the effects of fragmented nucleic acids (Kerachian et al., 2019) and the chemical changes on FFPE samples (Millier et al., 2017). However, a few studies (Adiconis et al., 2013) have specifically contrasted the efficacy of these approaches with RIN, DV, and PERM in the evaluation of FFPE sample sequencing outcomes.

    • A DNA methylation panel for high performance detection of colorectal cancer

      2021, Cancer Genetics
      Citation Excerpt :

      A full protocol has been developed, allowing for a minimally invasive blood-based diagnostic test (Epi proColon Ⓡ) for CRC [13], which is currently available in Europe and the US for clinical use [14]. The test detects methylated SEPT9 (mSept9) in plasma circulating DNA that is thought to be derived from the tumor's shed apoptotic and necrotic cells [15,16]. The clinical quality of the SEPT9 marker has been confirmed with more than 5000 subjects in multiple studies.

    View all citing articles on Scopus
    View full text