GET THE APP

Circulating DNA in Cancer: An Overview
Chemotherapy: Open Access

Chemotherapy: Open Access
Open Access

ISSN: 2167-7700

+44 1223 790975

Review Article - (2015) Volume 4, Issue 4

Circulating DNA in Cancer: An Overview

Nidhi Singh and Anoop Saraya*
Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi-110029, India
*Corresponding Author: Anoop Saraya, Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi-110029, India, Tel: +91-11-26593359 Email:

Abstract

In the last decade, research has been focused on discovering markers which have high sensitivity and specificity to detect cancer. Molecular biologists have been trying to find markers which can be detected non-invasively. Circulating nucleic acids in plasma and serum of cancer patients and the mutations detected in them have gained immense importance in this field. Circulating nucleic acids (CNAs) include DNA, RNA, nucleosomal DNA, microRNA, viral DNA. This review focuses on circulating DNA, its origin and mechanism of release, its diagnostic and prognostic utility, association with any of the clinic pathological parameters and its role in monitoring treatment efficiency in cancer. However, due to lack of uniformity in laboratory techniques, variability in disease advancement, less number of samples in various studies, lack of evidences for the origin of circulating nucleic acids, the knowledge sequestered till now in this field has not been translated in to clinical practice. With more high throughput techniques, circulating nucleic acid levels and mutations in them may give rise to a new era of cancer diagnostics and therapy.

Keywords: Molecular biologists,Circulating nucleic acids,plasma,

Circulating Nucleic Acids: Need as Cancer Markers

Cells in the body contain DNA and RNA, but these nucleic acids can also be found circulating freely in serum and plasma. Solid tumors undergoing uncontrolled proliferation of cells, angiogenesis, apoptosis are known to shed circulating nucleic acids into the blood. In the past one decade, the circulating nucleic acids have gained importance in tumor biology, because they offer a non-invasive approach for diagnosis, clinical assessment and follow-up of the treatment. Tissue biopsy, on the other hand, is an invasive and time taking procedure and can’t be taken serially to monitor events of the disease.

However, there are various protein tumor markers which can be assessed using serum and plasma of cancer patients like, prostratespecific antigen (PSA) in prostate cancer, the alpha-fetoprotein in hepatocellular carcinoma, the carcino embryogenic antigen (CEA) in colon cancer, the cancer antigen (CA)15.3 in breast cancer, and the CA19.9 in pancreatobiliary tumors [1]. But these are assessed through immunoassays and might lack sensitivity in non-secretory tumors and also may not discriminate tumor from inflammatory conditions. Moreover, there are other types of cancers which do not have any blood-based tumor markers. Furthermore, detecting tumor-specific mutations in CNAs not only provide diagnostic benefit but also help in deciding and monitoring the treatment. With such a backdrop, circulating nucleic acids from plasma and serum are emerging as a promising concept for use as a diagnostic or prognostic marker or for assessing the efficiency of any treatment of cancer.

Though the concept of circulating nucleic acids is gaining importance in the present decade, it was discovered by Mandel and Metais [2] in 1948, but their association with disease was not proposed at that time. However, in 1977, Leon et al. [3], found higher levels of CNA in plasma of patients affected by lung cancer than normal healthy individuals and those patients with higher levels of circulating DNA have worse prognosis. Stroun et al. [4] further suggested that primary tumor gave rise to CNAs. Later, by studies on pancreatic neoplasm [5,6] and acute myelogenous leukemia, presence of somatic mutations in plasma confirmed the fact that this CNA originated from tumor.

Forms of Circulating Nucleic Acids

Circulating nucleic acids are present in varied forms. They can be present as cell free forms (cell–free circulating nucleic acids) which are present as the naked DNA without any cell or associated protein, as circulating tumor cells (CTCs), which are directly being used for immunoassays, viral DNA [7], mitochondrial DNA [8], circulating cell free RNA [9] and miRNA [10]. Other bodies which carry fragments of DNA are exosomes and microvesicles [11,12]. The pictorial representation of the contributors of circulating nucleic acids is shown in Figure 1. Tumor DNA contributed by all of the above mentioned sources offer a unique opportunity for serially analyzing tumor genotype in a non-invasive manner. Thus, CTCs and circulating DNAs are often referred to as ‘liquid biopsy’. This review however focuses on cell free circulating DNA or circulating tumor DNA, its characterstics and clinical significance.

chemotherapy-Circulating-nucleic-acids

Figure 1: Circulating nucleic acids represent various types of nucleic acids from different sources. DNA from tumor is present as cell free form or in the intact tumor cells which get detached from the tumor. Apoptosis and necrosis are responsible for release of DNA from normal as well as tumor cells. Exosomes and microvesicles in peripheral blood, are formed from tumors and they carry nucleic acids. Viral DNA, micro RNA and nucleosomal DNA also contribute to the pool of nucleic acids in blood. The DNA present in the blood may have various genetic and epigenetic modifications.

Origin of Circulating DNA

Various hypothesis have been given which explain the origin of circulating DNA, but the topic is still found controversial. The circulating DNA are said to originate from processes like necrosis, apoptosis, direct active release from cells [13-15]. One of the hypotheses for the origin of cell-free circulating DNA in cancer is based on ‘‘micrometastasis’’ of tumor origin which are shed into circulation. Studies have reported higher amount of circulating DNA in cancer patients as compared to the corresponding number of cancer cells present in the circulation thus negating the hypothesis of micrometastasis [16,17].

Necrosis, is also thought to be responsible for high amounts of DNA fragments/circulating DNA in metastatic or advanced tumors [18,19]. But interestingly, it was reported that 90% of patients have decreased circulating DNA levels after radiation therapy, though radiation therapy is presumed to induce cell death/necrosis, probably due to cell proliferation arrest [3]. Thus, the hypothesis that necrosis of cells is responsible for release of DNA into plasma does not hold.

Apoptosis is also known to contribute to circulating DNA in healthy individuals as well as in cancer patients. During apoptosis, DNA is degraded in such a manner that nucleosomal DNA fragments are formed in multiples of 180-1000 kb length [19]. These fragments are further ingested by phagocytosis and digested by lysosomal enzymes and cleared from the bloodstream [20]. However, enzymatic action on these fragments is limited as this circulating DNA is assumed to be protected by some complexes and DNAase activity is also limited in plasma of cancer patients [16,17,21]. Thus, degraded DNA is not utilized by phagocytic cells as in normal cells. But on the other hand, in neoplastic diseases, the normal apoptotic mechanisms are lost, and fragments of variable sizes in blood stream are reported in literature, due to abnormal DNA degradation.

Some studies have also shown that cell free circulating DNA/nucleic acids are released actively from the cells specially lymphocytes [21-23].

Circulating DNA Concentration: Clinical Significance

High concentrations of circulating DNA have been reported in plasma and serum from cancer patients. In various cancers like colorectal [24-28], breast [29-34], liver [35-36], periampullary [28], pancreas [37], levels of circulating DNA was found to be significantly elevated in neoplastic condition than that in non-neoplastic conditions (healthy controls or/as diseased controls). Shaprio et al. [38], showed that patients with benign gastrointestinal diseases had a lower mean concentrations of plasma cell free DNA than cancer patients. In studies on hepatocellular carcinoma, circulating DNA levels can distinguish cancer patients from patients with hepatitis B and hepatitis C [35-36].

In contrast to these findings, there are few reports on lung cancer [39], ovarian cancer [40], periampullary cancer [28], which state that cell free circulating DNA cannot be used as a unique marker for cancer. Such discrepancies may arise due to variation in source of cell free DNA (serum or plasma), variation in storage and processing conditions. These variations are discussed later in the review.

Moreover, increased amount of plasma circulating DNA is observed as the tumor progresses [34,41] and high circulating DNA levels are found in patients with advanced disease [3,42,34] or metastasis [3,37], stage of the disease [43], tumor size [41]. Circulating DNA also showed high sensitivity to detect locally advanced disease [43]. In a recent study [44], more patients with advanced pancreatic, colorectal, ovarian, bladder, gastroesophageal, breast, melanoma, hepatocellular cancers were found to have detectable levels of circulating DNA while less patients with primary brain, renal, prostrate and thyroid tumors had detectable amount of circulating DNA. Cell free DNA levels were also found to correlate with shorter survival of patients [45,37].

Circulating DNA Integrity Index and its Clinical Significance

Different origins of circulating DNA contribute differently to the circulating DNA pool, high molecular weight long DNA fragments are known to be contributed by the necrosis of the tumor cells and low molecular weight small DNA fragments have been attributed to apoptosis phenomena [46]. In plasma and serum, Wang et al. [47] found that circulating DNA is present in different sizes and this difference can be used to calculate integrity index. Integrity index is the ratio between the long and short circulating DNA fragments. They suggested that integrity index can be a simple and inexpensive way to detect gynecologic and breast cancers. Briefly, integrity index is calculated by amplifying Alu - sequences [48] or by amplification of β- actin [49], GAPDH (glyceraldehyde-3-phosphate dehydrogenase) [50], Leptin [51], LINE1 (long interspersed nuclear elements) [52].

Integrity index of circulating DNA, has been found to be elevated in patients with different types of cancers like colorectal cancers [27,28], prostate cancer [53], hepatocellular cancer [35,36], acute leukemia [49], periampullary cancers [28], primary breast cancers [30], rectal cancers [54] and nasopharyngeal cancers [55]. Interestingly, in a review focused on circulating DNA integrity index, authors have highlighted the fact that DNA integrity index is more sensitive and specific than circulating DNA concentration in differentiating cancer patients from their respective non-neoplastic diseases and healthy controls [56]. Circulating DNA integrity index has been found to associate with tumor size, TNM stage, vascular invasion, lymph node involvement and distant metastasis in hepatocellular carcinoma [35,36]. Circulating DNA integrity index was found to be potential prognostic markers in patients with primary breast cancer [30].

Patients who responded to chemoradiotherapy, their pre and post treatment cell free DNA integrity indices were found to be statistically different whereas patients who did not respond, their pre and post treatment cell free DNA integrity indices did not differ [54]. Chan et al. [55] found that DNA integrity index not only got reduced in patients with nasopharyngeal carcinoma who responded successfully to radiotherapy but was also found to be associated with reduced probability of disease free survival at higher values. Another study by Gang et al. reported that integrity index of circulating DNA differ in pre and post-surgery samples of patients with renal cell carcinoma [50].

Genetic Alterations in Circulating DNA

Circulating DNA may come from tumor or non-tumor source. Moreover, this DNA has also been observed in normal individuals under physiological stress, trauma and exhaustive exercise or during inflammation [57]. Thus, the presence of tumor-specific genetic and epigenetic alterations in the circulating DNA provide evidence that the circulating DNA being analyzed have come from the tumor itself where these mutations must have occurred in a sequential manner to cause malignant transformation. Thus, methods for the detection of tumor-specific DNA variants have been developed.

K-ras is the most frequently mutated oncogene, reported in colon, pancreas, lung and thyroid tumors and can be used as a useful marker [58,59]. Kopreski et al. [59] detected K-ras mutations in plasma from 83% of patients whose tumors had such mutations. However, review of studies showed that the concordance with K-ras mutations as compared to the primary tumor was only 50%. Also, plasma DNA was found to have some other mutations also which were not present in the primary tumor, may be due to heterogenous tumor clones [61]. In a study, on metastatic colorectal cancer, Bettegowda et al. [44] showed that the circulatory tumor DNA had high sensitivity and specificity for detection of clinically relevant KRAS gene mutations. In a study on pancreatic cancer, the K-ras mutation rates in tissue and circulating DNA were 74.7% and 62.6%, respectively, and the concordance rate between them was 58 of 75 samples (77.3%) [62]. They also found that survival did not appear to differ by the presence of K-ras mutations in tissue DNA, but the survival of patients with K-ras mutations in circulating DNA was significantly shorter than that of patients without mutations. However in some studies, no correlation exists between Kras mutational status in circulating DNA and clinicopathological parameters or survival [37].

In addition, most frequent mutations in colorectal cancers like APC, K-ras, TP53 , PIK3CA and BRAF , were found to be more useful than using CEA and CA19-9 levels. [63]. In patients with carcinoma of breast, presence of amplified HER2 in circulating cell free DNA during follow-up, have become a marker for prognosis and for response to treatment with monoclonal antibodies such as trastuzumab [64]. Many chemotherapeutic agents and targeted therapy are targeted on molecules which are an important part of certain pathways like KRAS, BRAF, EGFR or p53 . Analysis of circulating DNA key mutations in these in plasma DNA offers a non-invasive and quick way for predicting the response to treatment and monitoring the disease [65-66].

In addition, most frequent mutations in colorectal cancers like APC, K-ras, TP53 , PIK3CA and BRAF , were found to be more useful than using CEA and CA19-9 levels. [63]. In patients with carcinoma of breast, presence of amplified HER2 in circulating cell free DNA during follow-up, have become a marker for prognosis and for response to treatment with monoclonal antibodies such as trastuzumab [64]. Many chemotherapeutic agents and targeted therapy are targeted on molecules which are an important part of certain pathways like KRAS, BRAF, EGFR or p53 . Analysis of circulating DNA key mutations in these in plasma DNA offers a non-invasive and quick way for predicting the response to treatment and monitoring the disease [65-66].

Hypermethylation of the promoters of various genes result in loss of transcription of the gene, leading to loss of gene expression. Examining these epigenetic changes in circulating DNA, may offer a possible method for early detection of cancers or analyzing the effect of the treatment. Some of the examples that link hypermethylated genes to cancer and detected in circulating DNA are-promoter hypermethylation of ITIH5, DKK3, RASSF1A tumor suppressor genes in breast cancer [70], NPTX2 in pancreatic cancer [71], UCHL1 (ubiquitin carboxyl-terminal hydrolase L1) in hepatocellular and esophageal carcinoma [72,73], GSTP1 (Glutathione- S- transferase) gene in prostate cancer [74], SEPT9 (Septin) gene and hMLH1 gene in colorectal cancer [75,76]. Some of the recent studies showing association of mutations in circulating DNA with disease progression, survival and follow-up of the patients undergoing treatment and assessing the acquired resistance are mentioned in Table 1.

Study group Year Cancer type Finding
Gonzalez-Cao M [86] 2015 Melanoma BRAF V600E alleles in ct DNA can predict treatment outcome in melanoma patients
Takeshita T [87] 2015 Breast cancer PIK3CA mutation found in ct DNA can predict relapse free survival and breast cancer specific survival in triple negative breast cancer patients
Bronte G [88] 2015 Colorectal cancer Ct DNA can be used as tool to study primary and acquired resistance to anti-EGFR monoclonal antibodies
Garcia-Murillas I[89] 2015 breast cancer Mutation tracking by sequencing analysis in circulating tumor DNA predicts relapse in early breast cancer and to monitor minimal residual disease
Powrozek T [90] 2015 Lung cancer Methylation of DCLK1 promoter region in cell free DNA was found to be prognostic indicator.
Delmonico L [91] 2015 Breast cancer Hypermethylation of p16 INK4A gene was found to discriminate breast cancer from impalpable breast lesions.
Sun W [92] 2015 Non-Small Cell Lung Carcinoma Ct DNA has a high degree of specificity to detect EGFR mutations and also capable of monitoring disease progression during EGFR-TKI treatment.
Sorenson BS et al. [42] 2014 Non-Small Cell Lung Carcinoma Observed acquired T790 mutation (in plasma DNA) after treatment with erlotinib
Salkeni MA et al [93] 2013 Glioblastoma Mutated EGFR  reported to have  a poor prognosis and associated with chemoresistance and radioresistance
Schwarzenbach H et al. [94] 2012 Breast cancer LOH at D12S1725 mapping to cyclin D2 gene locus, correlated significantly with shorter overall survival
Yoon KA et al. [76] 2009 Lung cancer Found that circulating DNA levels can differentiate between lung cancer patients from healthy controls
Ligget T et al. [95] 2010 Pancreatic  cancer Differential methylation of cell free circulating DNA can differentiate between pancreatic cancer patients from chronic pancreatitis
Reinert T et al. [96] 2015 Colorectal cancer Observed that circulating DNA analysis can be used to monitor disease burden following colorectal surgery
LoNigro C et al. [97] 2013 Melanoma Methylated tissue factor pathway inhibitor2 (TFPI2) inserum is a biomarker of metastatic melanoma
Ponomaryova et al. [98] 2013 Lung cancer Methylation in RARB2 and RASSF1 gene found in circulating DNA can be an important tool for diagnosis and post treatment follow-up
Mussolin et al. [99] 2013 Pediatric Lymphoma Cell free plasma DNA was significantly was higher in lymphoma cases than in controls and these higher levels correlated with stage and survival in different sub types of lymphoma
Mouliere et al. [100] 2013 Colorectal cancer High KRAS and BRAF mutation load was found in cancer patients than controls

Table 1: Summary of studies showing types of mutations detected in circulating DNA and their association with disease progression and treatment pattern.

Apart from differential diagnosis and association with clinicopathological parameters and survival, circulating DNA have been found to be useful in number of other applications also. The mutations detected in circulating DNA can be of help in deciding the type of therapy, predicting the response of treatment, for finding the reason for acquired resistance to therapy.

Circulating DNA: Challenges Faced

Despite the numerous studies on this subject, there is no consensus about the correlation between circulating DNA concentration and tumor stage, location and size [41,42,77]. And despite various advancement in the techniques used to detect circulating DNA, it is not detectable in all cases. This may be due to different anatomical region of tumor, differences in the approach of tumor to the blood vessel, differing severity of the disease.

Both the levels of circulating nucleic acids and the presence of underlying mutation in this shed DNA have the potential to behave not only as diagnostic and prognostic marker but also as a tool to monitor the effect of any treatment given. Though, there are technical limitations regarding extraction of cell free DNA and subsequent processing, analysis of circulating nucleic acids/DNA may become easier with better high throughput techniques.

Such discrepancies may arise firstly due to difference in the use of serum and plasma as the starting material. Several anticoagulants (EDTA, heparin and lithium-heparin) which have been used may be responsible for variability in yield of circulating DNA. Secondly, variation in sample processing at the level of centrifugation steps, delay in sample processing and duration and condition of storage, all contribute to the disparity between various studies [61]. Also, some studies use quantitative methods and some use qualitative methods, which can again lead to contradicting results.

Whether plasma or serum DNA is the ideal candidate for studying circulating DNA has been a topic of debate. The concentration of DNA in serum has been reported to be 4-6 fold higher in serum as compared to that in plasma [78-81]. But the increased levels of DNA in serum may be so because of lysis of blood cells during storage and processing/ separation of serum [56]. Studies reported that time delay and the storage temperature of blood before centrifugation had a significant impact on DNA concentration in serum and thus serum DNA can be of diagnostic value only if serum preparation is done at ambient temperature and minimizing the time period between blood collection and centrifugation. However, plasma was shown to be a better source of tumor derived circulating cell-free DNA than serum for the detection of mutations and it reflects probably the same concentration of circulating DNA as in circulation while serum DNA can get elevated not because of tumor DNA but because of clotting process [80,82-84].

With these contraindications in view, various projects are being run to set guidelines for sample collection, sampe processing and storage in order to analyze circulating DNA levels, its integrity and tumor associated mutations. One such project is SPIDIA (Standardization and improvement of generic pre-analytical tools and procedures for invitro diagnostics) funded by European Commission. This project focused on External Quality Assessmet (EQA) of pre-analytical steps of DNA/RNA analyses. As a part of this project, blood samples were sent to different laboratories where DNA/RNA is extracted and returned. The External Quality Assessment organizer assess the quality of DNA retreived from various laboratories. On the basis of these surveys, evidence-based guidelines will be set for the pre-analytical steps involved in DNA/RNA analysis [85].

With well standardized pre-analytical phase, and with more high throughput techniques to detect mutations, circulating cell free DNA concentration are finding relevance in personalized medicine. Circulating cell-free DNA and the molecular alterations associated with it may not only find importance as non-invasive diagnostic or prognostic marker but also as markers to monitor tumor burden during treatment.

References

  1. Jacobs EL, Haskell CM (1991) Clinical use of tumor markers in oncology. Curr Probl Cancer 15: 299-360.
  2. MANDEL P, METAIS P (1948) Les acides nucléiques du plasma sanguin chez l'homme. C R Seances Soc Biol Fil 142: 241-243.
  3. Leon SA, Shapiro B, Sklaroff DM, Yaros MJ (1977) Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res 37: 646-650.
  4. Stroun M, Anker P, Maurice P, Lyautey J, Lederrey C, et al. (1989) Neoplastic characteristics of the DNA found in the plasma of cancer patients. Oncology 46: 318-322.
  5. Sorenson GD, Pribish DM, Valone FH, Memoli VA, Bzik DJ, et al. (1994) Soluble normal and mutated DNA sequences from single-copy genes in human blood. Cancer Epidemiol Biomarkers Prev 3: 67-71.
  6. Vasioukhin V, Anker P, Maurice P, Lyautey J, Lederrey C, et al. (1994) Point mutations of the N-ras gene in the blood plasma DNA of patients with myelodysplastic syndrome or acute myelogenous leukaemia. Br J Haematol 86: 774-779.
  7. Lo YM, Chan WY, Ng EK, Chan LY, Lai PB, et al. (2001) Circulating Epstein-Barr virus DNA in the serum of patients with gastric carcinoma. Clin Cancer Res 7: 1856-1859.
  8. Fliss MS, Usadel H, Caballero OL, Wu L, Buta MR, et al. (2000) Facile detection of mitochondrial DNA mutations in tumors and bodily fluids. Science 287: 2017-2019.
  9. Wieczorek AJ, Sitaramam V, Machleidt W, Rhyner K, Perruchoud AP, et al. (1987) Diagnostic and prognostic value of RNA-proteolipid in sera of patients with malignant disorders following therapy: first clinical evaluation of a novel tumor marker. Cancer Res 47: 6407-6412.
  10. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, et al. (2008) Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci U S A 105: 10513-10518.
  11. González E, Falcón-Pérez JM (2015) Cell-derived extracellular vesicles as a platform to identify low-invasive disease biomarkers. Expert Rev Mol Diagn 15: 907-923.
  12. Kahlert C, Melo SA, Protopopov A, Tang J, Seth S, et al. (2014) Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J Biol Chem 289: 3869-3875.
  13. Gormally E, Caboux E, Vineis P, Hainaut P (2007) Circulating free DNA in plasma or serum as biomarker of carcinogenesis: practical aspects and biological significance. Mutat Res 635: 105-117.
  14. Anker P, Mulcahy H, Chen XQ, Stroun M (1999) Detection of circulating tumour DNA in the blood (plasma/serum) of cancer patients. Cancer Metastasis Rev 18: 65-73.
  15. Stroun M, Lyautey J, Lederrey C, Olson-Sand A, Anker P (2001) About the possible origin and mechanism of circulating DNA apoptosis and active DNA release. Clin Chim Acta 313: 139-142.
  16. Sorenson GD, Porter DM, Barth RJ, Memoli VA, Rhodes CH, et al. (1997) Detection of mutatedKRAS2 sequences in plasma from patients with pancreatic carcinoma in comparison with the CA19-9 assay. J Int Soc Oncodev Biol Med18: 66.
  17. Chen X, Bonnefoi H, Diebold-Berger S, Lyautey J, Lederrey C, et al. (1999) Detecting tumor-related alterations in plasma or serum DNA of patients diagnosed with breast cancer. Clin Cancer Res 5: 2297-2303.
  18. Nawroz H, Koch W, Anker P, Stroun M, Sidransky D (1996) Microsatellite alterations in serum DNA of head and neck cancer patients. Nat Med 2: 1035-1037.
  19. Mead R, Duku M, Bhandari P, Cree IA (2011) Circulating tumour markers can define patients with normal colons, benign polyps, and cancers. Br J Cancer 105: 239-245.
  20. Nagata S, Nagase H, Kawane K, Mukae N, Fukuyama H (2003) Degradation of chromosomal DNA during apoptosis. Cell Death Differ 10: 108-116.
  21. Viorritto IC, Nikolov NP, Siegel RM (2007) Autoimmunity versus tolerance: can dying cells tip the balance? Clin Immunol 122: 125-134.
  22. Chen X, Bonnefoi H, Diebold-Berger S, Lyautey J, Lederrey C, et al. (1999) Detecting tumor-related alterations in plasma or serum DNA of patients diagnosed with breast cancer. Clin Cancer Res 5: 2297-2303.
  23. Gahan PB, Anker P, Stroun M (2008) Metabolic DNA as the origin of spontaneously released DNA? Ann N Y Acad Sci 1137: 7-17.
  24. Stroun M, Maurice P, Vasioukhin V, Lyautey J, Lederrey C, et al. (2000) The origin and mechanism of circulating DNA. Ann N Y Acad Sci 906: 161-168.
  25. Thijssen MA, Swinkels DW, Ruers TJ, de Kok JB (2002) Difference between free circulating plasma and serum DNA in patients with colorectal liver metastases. Anticancer Res 22: 421-425.
  26. Frattini M, Gallino G, Signoroni S, Balestra D, Battaglia L, et al. (2006) Quantitative analysis of plasma DNA in colorectal cancer patients: a novel prognostic tool. Ann N Y Acad Sci 1075: 185-190.
  27. Hao TB, Shi W, Shen XJ, Qi J, Wu XH, et al. (2014) Circulating cell-free DNA in serum as a biomarker for diagnosis and prognostic prediction of colorectal cancer. Br J Cancer 111: 1482-1489.
  28. Umetani N, Kim J, Hiramatsu S, Reber HA, Hines OJ, et al. (2006) Increased integrity of free circulating DNA in sera of patients with colorectal or periampullary cancer: direct quantitative PCR for ALU repeats. Clin Chem 52: 1062-1069.
  29. Schwarzenbach H, Müller 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.
  30. Iqbal S, Vishnubhatla S, Raina V, Sharma S, Gogia A, et al. (2015) Circulating cell-free DNA and its integrity as a prognostic marker for breast cancer. Springerplus 4: 265.
  31. Huang ZH, Li LH, Hua D (2006) Quantitative analysis of plasma circulating DNA at diagnosis and during follow-up of breast cancer patients. Cancer Lett 243: 64-70.
  32. Kohler C, Radpour R, Barekati Z, Asadollahi R, Bitzer J, et al. (2009) Levels of plasma circulating cell free nuclear and mitochondrial DNA as potential biomarkers for breast tumors. Mol Cancer 8: 105.
  33. Dawson SJ, Tsui DW, Murtaza M, Biggs H, Rueda OM, et al. (2013) Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med 368: 1199-1209.
  34. Sirera R, Bremnes RM, Cabrera A, Jantus-Lewintre E, Sanmartín E, et al. (2011) Circulating DNA is a useful prognostic factor in patients with advanced non-small cell lung cancer. J Thorac Oncol 6: 286-290.
  35. Shazly SF, Eid MA, Sourogy HA, Attia GF, Ezzat SA (2010) Evaluation of serum DNA integrity as a screening and prognostic tool in patients with hepatitis C virus-related hepatocellular carcinoma. Int J Biol Markers 25: 79-86.
  36. Chen H, Sun LY, Zheng HQ, Zhang QF, Jin XM (2012) Total serum DNA and DNA integrity: diagnostic value in patients with hepatitis B virus-related hepatocellular carcinoma. Pathology 44: 318-324.
  37. Nidhi Singh, Surabhi Gupta RM, Pandey, Shyam S Chauhan, Anoop Saraya (2015). High Levels of Cell-free Circulating Nucleic Acids in Pancreatic Cancer are Associated with Vascular Encasement, Metastasis and Poor Survival. Cancer Investigation 33: 78-85.
  38. Shapiro B, Chakrabarty M, Cohn EM, Leon SA (1983) Determination of circulating DNA levels in patients with benign or malignant gastrointestinal disease. Cancer 51: 2116-2120.
  39. Zhang R, Shao F, Wu X, Ying K (2010) Value of quantitative analysis of circulating cell free DNA as a screening tool for lung cancer: a meta-analysis. Lung Cancer 69: 225-231.
  40. Chang HW, Lee SM, Goodman SN, Singer G, Cho SK, et al. (2002) Assessment of plasma DNA levels, allelic imbalance, and CA 125 as diagnostic tests for cancer. J Natl Cancer Inst 94: 1697-1703.
  41. Kohler C, Radpour R, Barekati Z, Asodollahi R, Bitzar J, et al. (2009) Levels of plasma ct cf nuclear DNA can differentiate between breast cancer cases and healthy individuals and also found to correlate with tuor size. Mol Cancer 8:105.
  42. Sorenson BS, Wu L, Wei W, Tsai J, Weber B, et al (2014) Monitoring of epidermal growth factor receptor tyrosine kinase receptor-sensitizing and resistance mutations in the plasma DNA of patients with advanced non-small cell lung cancer during treatment with erlotinib. Cancer 120: 3896-3901.
  43. Agassi R, Czeiger D, Shaked G, Avriel A, Sheynin J, et al. (2015) Measurement of circulating cell-free DNA levels by a simple fluorescent test in patients with breast cancer. Am J Clin Pathol 143: 18-24.
  44. Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, et al. (2014) Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 6: 224ra24.
  45. Nygaard AD, Holdgaard PC, Spindler KL, Pallisgaard N, Jakobsen A (2014) The correlation between cell-free DNA and tumour burden was estimated by PET/CT in patients with advanced NSCLC. Br J Cancer 110: 363-368.
  46. Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, et al. (2001) DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res 61: 1659-1665.
  47. Wang BG, Huang HY, Chen YC, Bristow RE, Kassauei K, et al. (2003) Increased plasma DNA integrity in cancer patients. Cancer Res 63: 3966-3968.
  48. Umetani N, Giuliano AE, Hiramatsu SH, Amersi F, Nakagawa T, et al. (2006) Prediction of breast tumor progression by integrity of free circulating DNA in serum. J Clin Oncol 24: 4270-4276.
  49. Gao YJ, He YJ, Yang ZL, Shao HY, Zuo Y, et al. (2010) Increased integrity of circulating cell-free DNA in plasma of patients with acute leukemia. Clin Chem Lab Med 48: 1651-1656.
  50. Gang F, Guorong L, An Z, Anne GP, Christian G, et al. (2010) Prediction of clear cell renal cell carcinoma by integrity of cell-free DNA in serum. Urology 75: 262-265.
  51. Boddy JL, Gal S, Malone PR, Shaida N, Wainscoat JS, et al. (2006) The role of cell-free DNA size distribution in the management of prostate cancer. Oncol Res 16: 35-41.
  52. Sunami E, Vu AT, Nguyen SL, Giuliano AE, Hoon DS (2008) Quantification of LINE1 in circulating DNA as a molecular biomarker of breast cancer. Ann N Y Acad Sci 1137: 171-174.
  53. Hanley R, Rieger-Christ KM, Canes D, Emara NR, Shuber AP, et al. (2006) DNA integrity assay: a plasma-based screening tool for the detection of prostate cancer. Clin Cancer Res 12: 4569-4574.
  54. Agostini M, Pucciarelli S, Enzo MV, Del Bianco P, Briarava M,et al.(2011) Circulating cell-free DNA: a promising marker of pathologic tumor response in rectal cancer patients receiving preoperative chemoradiotherapy. Ann Surg Oncol 18:2461-2468.
  55. Chan KC, Leung SF, Yeung SW, Chan AT, Lo YM (2008) Persistent aberrations in circulating DNA integrity after radiotherapy are associated with poor prognosis in nasopharyngeal carcinoma patients. Clin Cancer Res 14: 4141-4145.
  56. Yu J, Gu G, Ju S (2014) Recent advances in clinical applications of circulating cell free DNA integrity. Lab Med. 45 : 6-12.
  57. Fleischhacker M, Schmidt B (2007) Circulating nucleic acids(CNAs) and cancer–a survey. Biochim Biophys Acta 1775:181-232.
  58. Jung K, Fleischhacker M, Rabien A (2010) Cell-free DNA in the blood as a solid tumor biomarker--a critical appraisal of the literature. Clin Chim Acta 411: 1611-1624.
  59. Lecomte T, Ceze N, Dorval E, Laurent-Puig P (2010) Circulating free tumor DNA and colorectal cancer. Gastroenterol Clin Biol 34: 662-681.
  60. Kopreski MS, Benko FA, Borys DJ, Khan A, McGarrity TJ, et al. (2000) Somatic mutation screening: identification of individuals harboring K-ras mutations with the use of plasma DNA. J Natl Cancer Inst 92: 918-923.
  61. Garcia JM, Silva JM, Dominguez G, Silva J, Bonilla F (2001) Heterogeneous tumor clones as an explanation of discordance between plasma DNA and tumor DNA alterations. Genes Chromosomes Cancer 31: 300-301.
  62. Kinugasa H, Nouso K, Miyahara K, Morimoto Y, Dohi C, et al. (2015) Detection of K-ras gene mutation by liquid biopsy in patients with pancreatic cancer. Cancer .
  63. Levy M, Benesova L, Lipska L, Belsanova B, Minarikova P, et al. (2012) Utility of cell-free tumour DNA for post-surgical follow-up of colorectal cancer patients. Anticancer Res 32: 1621-1626.
  64. Page K, Hava N, Ward B, Brown J, Guttery DS, et al. (2011) Detection of HER2 amplification in circulating free DNA in patients with breast cancer. Br J Cancer 104: 1342-1348.
  65. Gadgeel SM, Cote ML, Schwartz AG, Matherly LH, Wozniak A, et al. (2010) Parameters for individualizing systemic therapy in non-small cell lung cancer. Drug Resist Updat 13: 196-204.
  66. Rosell R, Vergnenegre A, Liu B, Cobo M, Massuti B, et al. (2010) Biomarkers in lung oncology. Pulm Pharmacol Ther 23: 508-514.
  67. Perkins G, Yap TA, Pope L, Cassidy AM, Dukes JP, et al. (2012) Multi-purpose utility of circulating plasma DNA testing in patients with advanced cancers. PLoS One 7: e47020.
  68. Gormally E, Caboux E, Vineis P, Hainaut P (2007) Circulating free DNA in plasma or serum as biomarker of carcinogenesis: practical aspects and biological significance. Mutat Res 635: 105-117.
  69. Holdenrieder S, Nagel D, Schalhorn A, Heinemann V, Wilkowski R, et al. (2008) Clinical relevance of circulating nucleosomes in cancer. Ann N Y Acad Sci 1137: 180-189.
  70. Kloten V, Becker B, Winner K, Schrauder MG, Fasching PA, et al (2013) Promoter hypermethylation of the tumor suppressor genes ITIH3, DKK3 and RASSF1A as a novel biomarkers for blood-based breast cancer screening. Breast Cancer Res 15:R4.
  71. Yao F, Sun M, Dong M, Jing F, Chen B, et al. (2013) NPTX2 hypermethylation in pure pancreatic juice predicts pancreatic neoplasms. Am J Med Sci 346: 175-180.
  72. Yu J, Tao Q, Cheung KF, Jin H, Poon FF, et al. (2008) Epigenetic identification of ubiquitin carboxyl-terminal hydrolase L1 as a functional tumor suppressor and biomarker for hepatocellular carcinoma and other digestive tumors. Hepatology 48: 508-518.
  73. Yan WJ, Guo MZ, Yang YS (2012) The role of hypermethylation in promoter region of ubiquitin carboxyl-terminal hydrolase L1 in human esophageal cancer. Zhonghua Nei Ke Za Zhi 51: 390-393.
  74. Bryzgunova OE, Morozkin ES, Yarmoschuk ES, Yarmoschuk SV, Vlassov VV, et al. (2008) Methylation-specific sequencing of GSTP1 gene promoter in circulating/extracellular DNA from blood and urine of healthy donors and prostrate cancer patients. Ann N Y Acad Sci 1137:222-225.
  75. deVos T, Tetzner R, Model F, Weiss G, Schuster M, et al. (2009) Circulating methylated SEPT9 DNA in plasma is a biomarker for colorectal cancer. Clin Chem 55: 1337-1346.
  76. Wallner M, Herbst A, Behrens A, Crispin A, Stieber P, et al. (2006) Methylation of serum DNA is an independent prognostic marker in colorectal cancer. Clin Cancer Res 12: 7347-7352.
  77. Yoon KA, Park S, Lee SH, Kim JH, Lee JS (2009) Comparison of circulating plasma DNA levels between lung cancer patients and healthy controls. J Mol Diagn 11: 182-185.
  78. Taback B, O'Day SJ, Hoon DS (2004) Quantification of circulating DNA in the plasma and serum of cancer patients. Ann N Y Acad Sci 1022: 17-24.
  79. Holdenrieder S, Stieber P, Chan LY, Geiger S, Kremer A, et al. (2005) Cell-free DNA in serum and plasma: comparison of ELISA and quantitative PCR. Clin Chem 51: 1544-1546.
  80. Lee TH, Montalvo L, Chrebtow V, Busch MP (2001) Quantitation of genomic DNA in plasma and serum samples: higher concentrations of genomic DNA found in serum than in plasma. Transfusion 41: 276-282.
  81. Thijssen MA, Swinkels DW, Ruers TJ, de Kok JB (2002) Difference between free circulating plasma and serum DNA in patients with colorectal liver metastases. Anticancer Res 22: 421-425.
  82. Jung M, Klotzek S, Lewandowski M, Fleischhacker M, Jung K (2003) Changes in concentration of DNA in serum and plasma during storage of blood samples. Clin Chem 49: 1028-1029.
  83. Sozzi G, Roz L, Conte D, Mariani L, Andriani F, et al. (2005) Effects of prolonged storage of whole plasma or isolated plasma DNA on the results of circulating DNA quantification assays. J Natl Cancer Inst 97: 1848-1850.
  84. Vallée A, Marcq M, Bizieux A, Kouri CE, Lacroix H, et al. (2013) Plasma is a better source of tumor-derived circulating cell-free DNA than serum for the detection of EGFR alterations in lung tumor patients. Lung Cancer 82: 373-374.
  85. Kristensen GB, Aakre KM, Kristoffersen AH, Sandberg S (2014) How to conduct External Quality Assessment Schemes for the pre-analytical phase? Biochem Med (Zagreb) 24: 114-122.
  86. Gonzalez-Cao M, Mayo-de-Las-Casas C, Molina-Vila MA, De Mattos-Arruda L, Muñoz-Couselo E, et al. (2015) BRAF mutation analysis in circulating free tumor DNA of melanoma patients treated with BRAF inhibitors. Melanoma Res .
  87. Takeshita T, Yamamoto Y, Yamamoto-Ibusuki M, Inao T, Sueta A, et al. (2015) Prognostic role of PIK3CA mutations of cell-free DNA in early-stage triple negative breast cancer. Cancer Sci .
  88. Bronte G, Silvestris N, Castiglia M, Galvano A, Passiglia F, et al. (2015) New findings on primary and acquired resistance to anti-EGFR therapy in metastatic colorectal cancer: do all roads lead to RAS? Oncotarget 6: 24780-24796.
  89. Garcia-Murillas I, Schiavon G, Weigelt B, Ng C, Hrebien S, et al. (2015) Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Sci Transl Med 7: 302ra133.
  90. Powrózek T, Krawczyk P, Nicoś M, Kuźnar-Kaminska B, Batura-Gabryel H, et al. (2015) Methylation of the DCLK1 promoter region in circulating free DNA and its prognostic value in lung cancer patients. Clin Transl Oncol .
  91. Delmonico L, Moreira Ados S, Franco MF, Esteves EB, Scherrer L, et al. (2015) CDKN2A (p14(ARF)/p16(INK4a)) and ATM promoter methylation in patients with impalpable breast lesions. Hum Pathol 46: 1540-1547.
  92. Sun W, Yuan X, Tian Y, Wu H, Xu H, et al. (2015) Non-invasive approaches to monitor EGFR-TKI treatment in non-small-cell lung cancer. J Hematol Oncol 8: 95.
  93. Salkeni MA, Zarzour A, Ansay TY, McPherson CM, Warnick RE, et al. (2013) Detection of EGFRvIII mutant DNA in the peripheral blood of brain tumor patients. J Neurooncol 115: 27-35.
  94. Schwarzenbach H, Eichelser C, Kropidlowski J, Janni WE, Rack B, et al. (2012) LOH at 8 marker sites detected in plasma DNA in breast cancer patients correlated with tumor stage, tumor size and lymph node metastasis positive PR and HER2 receptor status. Clin Cancer Res 18:5919-5930.
  95. Liggett T, Melnikov A, Yi QL, Replogle C, Brand R, et al. (2010) Differential methylation of cell-free circulating DNA among patients with pancreatic cancer versus chronic pancreatitis. Cancer 116: 1674-1680.
  96. Reinert T, Schøler LV, Thomsen R, Tobiasen H, Vang S, et al. (2015) Analysis of circulating tumour DNA to monitor disease burden following colorectal cancer surgery. Gut .
  97. Lo Nigro C, Wang H, McHugh A, Lattanzio L, Matin R, et al. (2013) Methylated tissue factor pathway inhibitor 2 (TFPI2) DNA in serum is a biomarker of metastatic melanoma. J Invest Dermatol 133: 1278-1285.
  98. Ponomaryova AA, Rykova EY, Cherdyntseva NV, Skvortsova TE, Dobrodeev AY, et al. (2013) Potentialities of aberrantly methylated circulating DNA for diagnostics and post-treatment follow-up of lung cancer patients. Lung Cancer 81: 397-403.
  99. Mussolin L, Burnelli R, Pillon M, Carraro E, Farruggia P, et al. (2013) Plasma cell-free DNA in paediatric lymphomas. J Cancer 4: 323-329.
  100. Mouliere F, Messaoudi S, Pang D, Dritschilo A, Thierry AR (2014) Multi-marker analysis of circulating cell-free DNA toward personalized medicine for colorectal cancer. Mol Oncol 8: 927-941.
Citation: Singh N, Sarya A (2015) Circulating DNA in Cancer: An Overview. Chemo Open Access 4:172.

Copyright: © 2015 Saraya A, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top