Cell-free DNA (cfDNA): Clinical Significance and Utility in Cancer Shaped By Emerging Technologies

Precision oncology is predicated upon the ability to detect specific actionable genomic alterations and to monitor their adaptive evolution during treatment to counter resistance. Because of spatial and temporal heterogeneity and comorbidities associated with obtaining tumor tissues, especially in the case of metastatic disease, traditional methods for tumor sampling are impractical for this application. Known to be present in the blood of cancer patients for decades, cell-free DNA (cfDNA) is beginning to inform on tumor genetics, tumor burden, and mechanisms of progression and drug resistance. This substrate is amenable for inexpensive noninvasive testing and thus presents a viable approach to serial sampling for screening and monitoring tumor progression. The fragmentation, low yield, and variable admixture of normal DNA present formidable technical challenges for realization of this potential. This review summarizes the history of cfDNA discovery, its biological properties, and explores emerging technologies for clinically relevant sequence-based analysis of cfDNA in cancer patients. Molecular barcoding (or Unique Molecular Identifier, UMI)-based methods currently appear to offer an optimal balance between sensitivity, flexibility, and cost and constitute a promising approach for clinically relevant assays for near real-time monitoring of treatment-induced mutational adaptations to guide evidence-based precision oncology. Mol Cancer Res; 14(10); 898–908. ©2016 AACR.

[1]  Ash A. Alizadeh,et al.  Integrated digital error suppression for improved detection of circulating tumor DNA , 2016, Nature Biotechnology.

[2]  Matthew W. Snyder,et al.  Cell-free DNA Comprises an In Vivo Nucleosome Footprint that Informs Its Tissues-Of-Origin , 2016, Cell.

[3]  Hanlee P. Ji,et al.  Pan-cancer analysis of the extent and consequences of intratumor heterogeneity , 2015, Nature Medicine.

[4]  C. Collins,et al.  Functional analysis of androgen receptor mutations that confer anti-androgen resistance identified in circulating cell-free DNA from prostate cancer patients , 2016, Genome Biology.

[5]  A. Bronkhorst,et al.  Cell-free DNA: Preanalytical variables. , 2015, Clinica chimica acta; international journal of clinical chemistry.

[6]  Adrian V. Lee,et al.  Sensitive Detection of Mono- and Polyclonal ESR1 Mutations in Primary Tumors, Metastatic Lesions, and Cell-Free DNA of Breast Cancer Patients , 2015, Clinical Cancer Research.

[7]  B. Kermani,et al.  Analytical and Clinical Validation of a Digital Sequencing Panel for Quantitative, Highly Accurate Evaluation of Cell-Free Circulating Tumor DNA , 2015, PloS one.

[8]  J. Gummert,et al.  Donor-Derived Cell-Free DNA Is a Novel Universal Biomarker for Allograft Rejection in Solid Organ Transplantation. , 2015, Transplantation proceedings.

[9]  Nicholas J. Wang,et al.  Exome Sequencing of Cell-Free DNA from Metastatic Cancer Patients Identifies Clinically Actionable Mutations Distinct from Primary Disease , 2015, PloS one.

[10]  D. Dwivedi,et al.  Delayed but not Early Treatment with DNase Reduces Organ Damage and Improves Outcome in a Murine Model of Sepsis , 2015, Shock.

[11]  T. Gould,et al.  Extracellular DNA and histones: double‐edged swords in immunothrombosis , 2015, Journal of thrombosis and haemostasis : JTH.

[12]  Lawrence D True,et al.  Sequencing small genomic targets with high efficiency and extreme accuracy , 2015, Nature Methods.

[13]  Michael P. Schroeder,et al.  In silico prescription of anticancer drugs to cohorts of 28 tumor types reveals targeting opportunities. , 2015, Cancer cell.

[14]  M. Nykter,et al.  The Evolutionary History of Lethal Metastatic Prostate Cancer , 2015, Nature.

[15]  Martin E. Gleave,et al.  Androgen Receptor Gene Aberrations in Circulating Cell-Free DNA: Biomarkers of Therapeutic Resistance in Castration-Resistant Prostate Cancer , 2015, Clinical Cancer Research.

[16]  V. Wong,et al.  Lengthening and shortening of plasma DNA in hepatocellular carcinoma patients , 2015, Proceedings of the National Academy of Sciences.

[17]  D. Kim,et al.  Concomitant ALK translocation and EGFR mutation in lung cancer: a comparison of direct sequencing and sensitive assays and the impact on responsiveness to tyrosine kinase inhibitor. , 2015, Annals of oncology : official journal of the European Society for Medical Oncology.

[18]  F. Demichelis,et al.  Tumor clone dynamics in lethal prostate cancer , 2014, Science Translational Medicine.

[19]  A. Schneeweiss,et al.  Plasma DNA integrity as a biomarker for primary and metastatic breast cancer and potential marker for early diagnosis , 2014, Breast Cancer Research and Treatment.

[20]  M. Erlander,et al.  BRAF V600E mutations in urine and plasma cell-free DNA from patients with Erdheim-Chester disease , 2014, Oncotarget.

[21]  E. Phimister,et al.  Screening for trisomies in circulating DNA. , 2014, The New England journal of medicine.

[22]  Ash A. Alizadeh,et al.  An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage , 2013, Nature Medicine.

[23]  L. Migliore,et al.  Mutation Research / Fundamental and Molecular Mechanisms of Mutagenesis , 2014 .

[24]  Kang Zhang,et al.  DNA sequencing versus standard prenatal aneuploidy screening. , 2014, The New England journal of medicine.

[25]  Jeffrey A. Hussmann,et al.  High-throughput DNA sequencing errors are reduced by orders of magnitude using circle sequencing , 2013, Proceedings of the National Academy of Sciences.

[26]  Nathalie Zahra,et al.  Influence of Plasma Processing on Recovery and Analysis of Circulating Nucleic Acids , 2013, PloS one.

[27]  F. Mouliere,et al.  Circulating cell free DNA: Preanalytical considerations. , 2013, Clinica chimica acta; international journal of clinical chemistry.

[28]  N. Rosenfeld,et al.  Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA , 2013, Nature.

[29]  Alan Ashworth,et al.  Noninvasive Detection of HER2 Amplification with Plasma DNA Digital PCR , 2013, Clinical Cancer Research.

[30]  Carlos Caldas,et al.  Analysis of circulating tumor DNA to monitor metastatic breast cancer. , 2013, The New England journal of medicine.

[31]  A. Hidalgo-Miranda,et al.  Cancer Progression Mediated by Horizontal Gene Transfer in an In Vivo Model , 2012, PloS one.

[32]  G. Parmigiani,et al.  Detection of Chromosomal Alterations in the Circulation of Cancer Patients with Whole-Genome Sequencing , 2012, Science Translational Medicine.

[33]  M. Radic,et al.  Neutrophil Extracellular Traps: Double-Edged Swords of Innate Immunity , 2012, Journal of Immunology.

[34]  Jesse J. Salk,et al.  Detection of ultra-rare mutations by next-generation sequencing , 2012, Proceedings of the National Academy of Sciences.

[35]  Johannes G. Reiter,et al.  The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers , 2012, Nature.

[36]  N. Rosenfeld,et al.  Noninvasive Identification and Monitoring of Cancer Mutations by Targeted Deep Sequencing of Plasma DNA , 2012, Science Translational Medicine.

[37]  A. Sivachenko,et al.  Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer , 2012, Nature Genetics.

[38]  I. Mittra,et al.  Nucleic acids in circulation: Are they harmful to the host? , 2012, Journal of Biosciences.

[39]  Michael Gundry,et al.  Direct mutation analysis by high-throughput sequencing: from germline to low-abundant, somatic variants. , 2012, Mutation research.

[40]  Jeff Mellen,et al.  High-Throughput Droplet Digital PCR System for Absolute Quantitation of DNA Copy Number , 2011, Analytical chemistry.

[41]  A. Maitra,et al.  Recurrent GNAS Mutations Define an Unexpected Pathway for Pancreatic Cyst Development , 2011, Science Translational Medicine.

[42]  L. Mazutis,et al.  Quantitative and sensitive detection of rare mutations using droplet-based microfluidics. , 2011, Lab on a chip.

[43]  K. Kinzler,et al.  Detection and quantification of rare mutations with massively parallel sequencing , 2011, Proceedings of the National Academy of Sciences.

[44]  A. Jamurtas,et al.  Time of sampling is crucial for measurement of cell-free plasma DNA following acute aseptic inflammation induced by exercise. , 2010, Clinical biochemistry.

[45]  M. Kermekchiev,et al.  Direct DNA amplification from crude clinical samples using a PCR enhancer cocktail and novel mutants of Taq. , 2010, The Journal of molecular diagnostics : JMD.

[46]  Derek Y. Chiang,et al.  The landscape of somatic copy-number alteration across human cancers , 2010, Nature.

[47]  J. García-Verdugo,et al.  Molecular and Cellular Pathobiology Cancer Research Cell-Free Nucleic Acids Circulating in the Plasma of Colorectal Cancer Patients Induce the Oncogenic Transformation of Susceptible Cultured Cells , 2010 .

[48]  A. Puisieux,et al.  Influence of neuroblastoma stage on serum‐based detection of MYCN amplification , 2009, Pediatric blood & cancer.

[49]  S. Goodman,et al.  Sensitive digital quantification of DNA methylation in clinical samples , 2009, Nature Biotechnology.

[50]  T. Mok,et al.  Single-Molecule Detection of Epidermal Growth Factor Receptor Mutations in Plasma by Microfluidics Digital PCR in Non–Small Cell Lung Cancer Patients , 2009, Clinical Cancer Research.

[51]  J. Maguire,et al.  Solution Hybrid Selection with Ultra-long Oligonucleotides for Massively Parallel Targeted Sequencing , 2009, Nature Biotechnology.

[52]  Emily H Turner,et al.  Targeted Capture and Massively Parallel Sequencing of Twelve Human Exomes , 2009, Nature.

[53]  R. Swaminathan,et al.  Overview of Circulating Nucleic Acids in Plasma/Serum , 2008, Annals of the New York Academy of Sciences.

[54]  M. Fleischhacker,et al.  Circulating nucleic acids (CNAs) and cancer--a survey. , 2007, Biochimica et biophysica acta.

[55]  K. Nishio,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 , 2006, Clinical Cancer Research.

[56]  木村 英晴 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 , 2006 .

[57]  M. Kochman Detection and Quantification of Mutations in the Plasma of Patients With Colorectal Tumors , 2006 .

[58]  Frank Diehl,et al.  Detection and quantification of mutations in the plasma of patients with colorectal tumors. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[59]  S. Wallenstein,et al.  Molecular haplotyping by linking emulsion PCR: analysis of paraoxonase 1 haplotypes and phenotypes , 2005, Nucleic acids research.

[60]  A. Zychlinsky,et al.  Neutrophil Extracellular Traps Kill Bacteria , 2004, Science.

[61]  D. Chan,et al.  Increased plasma DNA integrity in cancer patients. , 2003, Cancer research.

[62]  D. Dressman,et al.  Transforming single DNA molecules into fluorescent magnetic particles for detection and enumeration of genetic variations , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[63]  A. Puisieux,et al.  Circulating MYCN DNA as a tumor-specific marker in neuroblastoma patients. , 2002, Cancer research.

[64]  C. Lam,et al.  Predominant hematopoietic origin of cell-free DNA in plasma and serum after sex-mismatched bone marrow transplantation. , 2002, Clinical chemistry.

[65]  L. Poon,et al.  Clinical Chemistry 47:9 1607–1613 (2001) Molecular Diagnostics and Genetics Effects of Blood-Processing Protocols on Fetal and Total DNA Quantification in Maternal Plasma , 2001 .

[66]  G. Evan,et al.  Proliferation, cell cycle and apoptosis in cancer , 2001, Nature.

[67]  S. Niruthisard,et al.  Human papillomavirus DNA in plasma of patients with cervical cancer , 2001, BMC Cancer.

[68]  F. O. Fackelmayer,et al.  DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. , 2001, Cancer research.

[69]  D. Sidransky,et al.  Detection and quantitation of human papillomavirus (HPV) DNA in the sera of patients with HPV-associated head and neck squamous cell carcinoma. , 2000, Clinical cancer research : an official journal of the American Association for Cancer Research.

[70]  M. Vallejo,et al.  Tumor DNA circulating in the plasma might play a role in metastasis. The hypothesis of the genometastasis. , 1999, Histology and histopathology.

[71]  R. Kornberg,et al.  Twenty-Five Years of the Nucleosome, Fundamental Particle of the Eukaryote Chromosome , 1999, Cell.

[72]  K. Kinzler,et al.  Digital PCR. , 1999, Proceedings of the National Academy of Sciences of the United States of America.

[73]  D. Whitcombe,et al.  Detection of PCR products using self-probing amplicons and fluorescence , 1999, Nature Biotechnology.

[74]  D. Beer,et al.  Detection of erbB-2 amplifications in tumors and sera from esophageal carcinoma patients. , 1999, Clinical cancer research : an official journal of the American Association for Cancer Research.

[75]  M. Provencio,et al.  Aberrant DNA methylation of the p16INK4a gene in plasma DNA of breast cancer patients , 1999, British Journal of Cancer.

[76]  S. Leung,et al.  Quantitative analysis of cell-free Epstein-Barr virus DNA in plasma of patients with nasopharyngeal carcinoma. , 1999, Cancer research.

[77]  Y. Lo,et al.  Rapid clearance of fetal DNA from maternal plasma. , 1999, American journal of human genetics.

[78]  J. Herman,et al.  Detection of aberrant promoter hypermethylation of tumor suppressor genes in serum DNA from non-small cell lung cancer patients. , 1999, Cancer research.

[79]  W. Lau,et al.  Detection of aberrant p16 methylation in the plasma and serum of liver cancer patients. , 1999, Cancer research.

[80]  Dan S. Tawfik,et al.  Man-made cell-like compartments for molecular evolution , 1998, Nature Biotechnology.

[81]  A. Theamboonlers,et al.  Epstein-Barr viral DNA in serum of patients with nasopharyngeal carcinoma. , 1998, Clinical cancer research : an official journal of the American Association for Cancer Research.

[82]  I. Sargent,et al.  Presence of fetal DNA in maternal plasma and serum , 1997, The Lancet.

[83]  A. Kurt,et al.  Microsatellite alterations in plasma DNA of small cell lung cancer patients , 1996, Nature Medicine.

[84]  David Sidransky,et al.  Microsatellite alterations in serum DNA of head and neck cancer patients , 1996, Nature Medicine.

[85]  Valeri Vasioukhin,et al.  Point mutations of the N‐ras gene in the blood plasma DNA of patients with myelodysplastic syndrome or acute myelogenous leukaemia , 1994, British journal of haematology.

[86]  V A Memoli,et al.  Soluble normal and mutated DNA sequences from single-copy genes in human blood. , 1994, Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology.

[87]  S H Neoh,et al.  Quantitation of targets for PCR by use of limiting dilution. , 1992, BioTechniques.

[88]  K. Fleming,et al.  PRENATAL SEX DETERMINATION BY DNA AMPLIFICATION FROM MATERNAL PERIPHERAL BLOOD , 1989, The Lancet.

[89]  M. Stroun,et al.  Neoplastic characteristics of the DNA found in the plasma of cancer patients. , 1989, Oncology.

[90]  B. Shapiro,et al.  Free DNA in the serum of cancer patients and the effect of therapy. , 1977, Cancer research.

[91]  P. Schur,et al.  Deoxybonucleic acid (DNA) and antibodies to DNA in the serum of patients with systemic lupus erythematosus. , 1966, The Journal of clinical investigation.

[92]  T. Wilczok,et al.  Circulating DNA as a Possible Factor in Oncogenesis , 1965, Science.

[93]  T. Tsumita,et al.  Fate of Injected Deoxyribonucleic Acid in Mice , 1963, Nature.

[94]  P Mandel,et al.  Les acides nucleiques du plasma sanguin chez l' homme , 1948 .