Skip to main content
Erschienen in: BMC Cancer 1/2017

Open Access 01.12.2017 | Research article

The evidence base for circulating tumour DNA blood-based biomarkers for the early detection of cancer: a systematic mapping review

verfasst von: Ian A. Cree, Lesley Uttley, Helen Buckley Woods, Hugh Kikuchi, Anne Reiman, Susan Harnan, Becky L. Whiteman, Sian Taylor Philips, Michael Messenger, Angela Cox, Dawn Teare, Orla Sheils, Jacqui Shaw, For the UK Early Cancer Detection Consortium

Erschienen in: BMC Cancer | Ausgabe 1/2017

Abstract

Background

The presence of circulating cell-free DNA from tumours in blood (ctDNA) is of major importance to those interested in early cancer detection, as well as to those wishing to monitor tumour progression or diagnose the presence of activating mutations to guide treatment. In 2014, the UK Early Cancer Detection Consortium undertook a systematic mapping review of the literature to identify blood-based biomarkers with potential for the development of a non-invasive blood test for cancer screening, and which identified this as a major area of interest. This review builds on the mapping review to expand the ctDNA dataset to examine the best options for the detection of multiple cancer types.

Methods

The original mapping review was based on comprehensive searches of the electronic databases Medline, Embase, CINAHL, the Cochrane library, and Biosis to obtain relevant literature on blood-based biomarkers for cancer detection in humans (PROSPERO no. CRD42014010827). The abstracts for each paper were reviewed to determine whether validation data were reported, and then examined in full. Publications concentrating on monitoring of disease burden or mutations were excluded.

Results

The search identified 94 ctDNA studies meeting the criteria for review. All but 5 studies examined one cancer type, with breast, colorectal and lung cancers representing 60% of studies. The size and design of the studies varied widely. Controls were included in 77% of publications. The largest study included 640 patients, but the median study size was 65 cases and 35 controls, and the bulk of studies (71%) included less than 100 patients. Studies either estimated cfDNA levels non-specifically or tested for cancer-specific mutations or methylation changes (the majority using PCR-based methods).

Conclusion

We have systematically reviewed ctDNA blood biomarkers for the early detection of cancer. Pre-analytical, analytical, and post-analytical considerations were identified which need to be addressed before such biomarkers enter clinical practice. The value of small studies with no comparison between methods, or even the inclusion of controls is highly questionable, and larger validation studies will be required before such methods can be considered for early cancer detection.
Abkürzungen
14–3-3 s
14–3-3 sigma or tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein theta
ADAM
metallopeptidase with thrombospondin type 1 motif, 1
AIM1
absent in melanoma 1
ALU
Alu repeat/element 9e
APC
Adenomatous Polyposis Coli
ARF
alternate reading frame
BIN1
bridging integrator 1
BLU
zinc finger MYND-type containing 10
BM
biomarker
BNC1
basonuclin 1
bp
base pair
BRAF
B-Raf proto-oncogene, serine/threonine kinase
BRCA1
breast cancer 1, DNA repair associated
BRINP3
BMP/Retinoic Acid Inducible Neural Specific 3
CALCA
calcitonin related polypeptide alpha
CDH1
cadherin 1
CDH13
cadherin 13
CDO1
cysteine dioxygenase type 1
cfDNA
circulating cell-free DNA
CHD1
chromodomain helicase DNA binding protein 1
CHRM2
cholinergic receptor muscarinic 2
CINAHL
Cumulative Index to Nursing and Allied Health Literature
CLSI
Clinical & Laboratory Standards Institute
CRC
colorectal carcinoma
CST6
cystatin 6
ctDNA
circulating tumour DNA
CYCD2
cyclin D2
DAPK1
death-associated protein kinase 1
DCC
DCC Netrin 1 receptor
DCLK1
doublecortin like kinase 1
ddPCR
digital droplet polymerase chain reaction
DKK3
Dickkopf WNT signaling pathway inhibitor 3
DLEC1
deleted in lung and esophageal cancer 1
DNA
dexoxyribonucleic acid
ECDC
UK Early Cancer Detection Consortium
EGFR
epidermal growth factor receptor (HER1)
EP300
E1A binding protein P300
ERBB2
erb-B2 receptor tyrosine kinase 2 (HER2)
ESR
estrogen receptor 1
FAM5C
BMP/retinoic acid inducible neural specific 3 (BRINP3)
FDA
US Food and Drug Adminstration
FHIT
fragile histidine triad
FIT
faecal immunohistochemical testing
FoBT
faecal occult blood testing
GAPDH
glyceraldehyde-3-phosphate dehydrogenase
gCYC
cyclophilin A
GNA11
G protein subunit alpha 11
GNAQ
G protein subunit alpha Q
GPC3
glypican 3
GSTP1
glutathione S-transferase pi 1
HCC
hepatocellular carcinoma
HER1
human epidermal growth factor receptor 1
HER2
human epidermal growth factor receptor 2
HIC1
HIC ZBTB transcriptional repressor 1
HNSCC
head and neck squamous cell carcinoma
HOXA7
Homeobox A7
HOXA9
Homeobox A9
HOXD13
Homeobox D13
hTERT
human telomerase reverse transcriptase DNA
IgH
immunoglobulin heavy locus
INK4A
cyclin dependent kinase inhibitor 2A (CDKN2A/P16)
ISO
International Standards Organization
ITIH5
inter-alpha-trypsin inhibitor heavy chain family member 5
KLK10
kallikrein related peptidase 10
KRAS
KRAS Proto-Oncogene, GTPase
LCH
Langerhans cell histocytosis
LINE1
long interspersed nuclear element 1
LoH
loss of heterozygosity
Max
maximum
MDG1
microvascular endothelial differentiation gene 1
MGMT
O(6)-methyl-guanine-DNA methyltransferase
Min
minimum
MLH1
MutL Homolog 1
mtDNA
mitochondrial DNA
MYC
MYC proto-oncogene
MYF3
myogenic differentiation 1 (MYOD1)
MYLK
myosin light chain kinase
NGS
next generation sequencing
NICE
UK National Institute for Health and Care Excellence
NOS
not otherwise specified
OPCML
opioid binding protein/cell adhesion molecule like
P14
P14 ARF tumor suppressor protein gene
P16
P16 cyclin-dependent kinase inhibitor 2A (CDKN2A)
P21
cyclin dependent kinase inhibitor 1A
P53
tumor protein P53
PCDH10
Protocadherin 10
PCDHGB7
protocadherin gamma subfamily B7
PCR
polymerase chain reaction
PIK3CA
phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha
PPIA
Peptidylprolyl isomerase A
PTGS2
Prostaglandin-endoperoxid synthase 2
qPCR
quantitative polymerase chain reaction
RARbeta2
Retinoid-acid-receptor-beta gene
RASSF1A
Ras association domain family member 1
RUNX3
runt related transcription factor 3
SFN
Stratifin
SFRP5
secreted frizzled related protein 5
SHOX2
short stature homeobox 2
SLC26A4
solute carrier family 26 member 4
SLC5A8
solute carrier family 5 member 8
SOX17
SRY-Box 17
SRBC
serum deprivation response factor-related gene
STARD
Standards for Reporting of Diagnostic Accuracy Studies
TAC1
tachykinin precursor 1
TFPI2
tissue factor pathway inhibitor 2
THBD-M
thrombomodulin
TIMP3
tissue inhibitor of metalloproteinase 3
TMS
tumor differentially expressed protein 1
UCHL1
Ubiquitin C-Terminal Hydrolase L1
V600E
Mutation resulting in an amino acid substitution at position 600 in BRAF, from a valine (V) to a glutamic acid (E)
VHL
Von Hippel Lindau gene
ZFP42
ZFP42 Zinc Finger Protein

Background

The early detection of cancers before they metastasise to other organs allows definitive local treatment, resulting in excellent survival rates. This is particularly true for breast cancer, but also others, including lung and colorectal cancer [1]. Early detection and diagnosis has therefore been a major goal of cancer research for many years, and the concept of early detection from a blood sample has been the focus of considerable effort. However, to date no blood biomarkers have had sufficient sensitivity and specificity to warrant their clinical use for early cancer detection, and their potential remains unrealised [2]. Hanahan and Weinberg [3] identified the major biological attributes of cancer, and it is apparent that most if not all of these biological processes give rise to biomarkers present in blood [4]. Circulating cell free DNA produced from cancers is known as circulating tumour DNA (ctDNA), and represents a subset of the circulating DNA (cfDNA) normally present at low levels in the blood of healthy individuals.
Since the first description of circulating cfDNA in blood [5, 6], it has become clear that total ctDNA levels rise in a number of disorders in addition to cancer including myocardial infarction [7], serious infections, and inflammatory conditions [8], as well as pregnancy where it can be used for prenatal diagnosis [9]. The source of this DNA appears to be mainly the result of cell death – either by necrosis or apoptosis [5, 911]. A raised ctDNA level is therefore non-specific, but may indicate the presence of serious disease. In blood, ctDNA is always present as small fragments, which makes assay design challenging [12]. Nevertheless, many analytical methods are available to measure ctDNA, and the field is rapidly maturing to the point where it may be clinically relevant to many patients.
In 2014, the UK Early Cancer Detection Consortium (ECDC) conducted a rapid mapping review of blood biomarkers of potential interest for cancer screening [13], and identified 814 biomarkers, including 39 ctDNA biomarkers. This paper uses the list generated from the mapping review, updated with relevant publications published since its completion to discuss the candidacy of ctDNA markers for early detection of cancer.

Methods

Our mapping review [13] conducted comprehensive searches of the electronic databases Medline, Embase, CINAHL, the Cochrane library, and Biosis to obtain relevant literature on blood-based biomarkers for cancer detection in humans (PROSPERO no. CRD42014010827). The search period finished in July 2014, therefore the searches have been updated to December 2016 using the same search terms. The abstracts of the publications retrieved were reviewed to identify those with validation data (usually indicated by case-control design) and to determine what ctDNA biomarkers had been measured in serum or plasma. Full details of the methods used are published elsewhere [13], and described briefly here. English language publications of any sample size were eligible and the full eligibility criteria used are provided in Table 1.
Table 1
Search criteria for ctDNA publications
Inclusion Criteria
Exclusion Criteria
English language studies
Studies published in non-English language
Studies within last seven years (2010–2016)
Studies published in 2009 or earlier
Controlled studies
Citation titles without abstracts
Validation Studies (comparison with controls)
Parallel publications and reviews based on the same or overlapping patient populationsa
Cancer detection/ diagnosis/screening
Prognosis or prediction (treatment response) associated markers
Biomarkers measured in blood plasma or serum
(markers or biomarkers)
Tissue, blood cells, or other bodily fluid samples
DNA (including cfDNA and ctDNA)
Abstracts of panels which do not state which biomarkers are studied
Human DNA
Viral and microbial DNA
aReviews and meta-analyses are cited, but not considered as evidence, but studies were included if they appeared to contain new data
The search strategy was deliberately inclusive, using keywords and subject headings as follows, to provide a comprehensive list of those ctDNA candidate biomarkers that had been used to identify cancers from blood samples. The search terms included ‘cancer’ ‘diagnosis’, ‘markers’, ‘blood’, and ‘screening’ with ‘DNA’, ‘cfDNA’, or ‘ctDNA’. Keywords and subject headings were determined by members of the ECDC working with the review team at the University of Sheffield. The results of the searches were collated in an Endnote database and results tabulated, with references, size of study, and methods used. To avoid bias, two reviewers conducted screening; references identified by either as relevant were included for further inspection. Those featuring ctDNA with data related to diagnosis or detection of three or more types of cancer were identified and retained for closer scrutiny to determine their potential utility.

Results

Following the updated searches and study selection, a total of 84 ctDNA markers were identified from 94 individual publications (Table 2 and Fig. 1).
Table 2
Individually identified markers with detection ability in ctDNA
No
Biomarker
Acronym
Cancer
DNA alteration
Assay type (qPCR, ddPCR, BEAMing, NGS, Other)
Size Cases (controls)
Plasma or Serum
Refs
1
14–3-3 sigma
14–3-3 s
Breast
Methylation
qPCR
106 (74)
Serum
[48]
2
absent in melanoma 1
AIM1; Beta/gamma crystallin domain-containing protein 1
Lung
Methylation
qPCR
76 (30)
Serum
[62]
3
ADAM: metallopeptidase with thrombospondin type 1 motif, 1
ADAMTS1
Pancreatic
Methylation
qPCR
42
Serum
[63]
4
Adenomatous Polyposis Coli
APC
Lung
Methylation
qPCR
76 (30)
Serum
[62]
CRC
Mutation
qPCR
33 (10)
Plasma
[64]
Testicular
Methylation
qPCR
73 (35)
Serum
[47]
CRC
Mutation
qPCR
191
Plasma
[65]
CRC
Methylation
qPCR
33
Serum
[53]
CRC
Mutation
PCR
104
Serum
[66]
Ovarian
Methylation
qPCR
87 (62)
Serum
[67]
Renal
Methylation
PCR
35 (54)
Serum
[68]
Breast
Methylation
qPCR
36 (30)
Plasma
[69]
Lung
Methylation
qPCR
110 (50)
Plasma
[70]
Renal
Methylation
qPCR
27 (15)
Plasma
[71]
CRC
Methylation
PCR
60 (100)
Plasma
[72]
5
ALU repeat
Alu 115 bp
Breast
NA
qPCR
39 (49)
Plasma
[22]
Alu 247 bp
Pancreatic
NA
qPCR
73 (43)
Plasma
[73]
CRC
NA
qPCR
50 (35)
Plasma
[20]
Breast
NA
qPCR
293 (100)
Plasma
[19]
Thyroid
NA
qPCR
176 (19)
Plasma
[24]
CRC
NA
qPCR
104 (173)
Serum
[23]
6
basonuclin 1
BNC1
Pancreatic
Methylation
qPCR
42
Serum
[63]
7
BIN1
BIN1
Breast
Methylation
qPCR
76 (30)
Serum
[62]
8
BLU
BLU
Lung
Methylation
qPCR
63 (36)
Plasma
[74]
9
BRAF
BRAF (V600E)
Melanoma
Mutation
qPCR
221
Both
[17]
Lung
Mutation
NGS
68 (107)
Plasma
[75]
LCH
Mutation
qPCR
30
Plasma
[76]
CRC
Mutation
qPCR
106
Plasma
[77]
Thyroid
Mutation
qPCR
77
Plasma
[78]
CRC
Mutation
BEAMing
503
Plasma
[21]
CRC
Mutation
qPCR
191
Plasma
[65]
10
BRCA1
BRCA1
Breast
Methylation
qPCR
89
Serum
[79]
Breast
Methylation
qPCR
36 (30)
Plasma
[69]
Ovarian
Methylation
PCR
50
Serum
[80]
Ovarian
Methylation
PCR
33 (33)
Plasma
[81]
11
CALCA
CALCA
Ovarian
Methylation
PCR
30 (30)
Plasma
[82]
12
CDH1
CDH1
Ovarian
Methylation
qPCR
87 (62)
Serum
[67]
13
CDH13
CDH13
Lung
Methylation
qPCR
63 (36)
Plasma
[74]
Lung
Methylation
qPCR
110 (50)
Plasma
[70]
14
CDO1
CDO1
Various
Methylation
qPCR
150 (60)
Plasma
[83]
15
CHD1
CHD1
Lung
Methylation
qPCR
76 (30)
Serum
[62]
16
CST6
CST6
Breast
Methylation
qPCR
196 (37)
Plasma
[84]
Breast
Methylation
qPCR
36 (30)
Plasma
[69]
17
CHRM2
CHRM2
Gastric
Methylation
qPCR
58 (30)
Serum
[85]
18
CYCD2
CYCD2
CRC
Methylation
qPCR
30 (30)
Plasma
[86]
19
DAPK1
DAPK1
HNSCC
Methylation
PCR
40 (41)
Serum
[87]
20
DCC
DCC
Lung
Methylation
qPCR
76 (30)
Serum
[62]
21
DCLK1
DCLK1
Lung
Methylation
qPCR
65 (95)
Plasma
[88]
Lung
Methylation
qPCR
32 (8)
Plasma
[89]
22
DKK3
DKK3
Breast
Methylation
qPCR
604 (59)
Serum
[90]
23
DLEC1
DLEC1
Lung
Methylation
qPCR
110 (50)
Plasma
[70]
HNSCC
Methylation
PCR
40 (41)
Serum
[87]
24
DNA (NOS)
DNA
Lung
NA
qPCR v Seq
30 (26)
Plasma
[91]
Various
No
NGS
77 (35)
Plasma
[45]
Various
No
NGS
640
Plasma
[16]
Lung
No
qPCR
65 (44)
Plasma
[92]
Ovarian
No
bDNA
36 (41)
Serum
[93]
25
e-cadherin
e-cadherin
Colorectal
Methylation
PCR
60 (100)
Plasma
[72]
26
EGFR
EGFR
Lung
Mutation
NGS
68 (107)
Plasma
[75]
27
EP300
EP300
Ovarian
Methylation
PCR
30 (30)
Plasma
[82]
28
ERBB2
HER2
Lung
Mutation
NGS
68 (107)
Plasma
[75]
Breast
Amplification
qPCR
120 (98)
Plasma
[14]
Oesphageal
Amplification
qPCR
41 (34)
Plasma
[94]
29
ESR
ESR
Breast
Methylation
qPCR
106 (74)
Serum
[48]
Breast
Methylation
qPCR
36 (30)
Plasma
[69]
30
FAM5C
FAM5C
Gastric
Methylation
qPCR
58 (30)
Serum
[85]
31
FHIT
FHIT
Lung
Methylation
qPCR
63 (36)
Plasma
[74]
Renal
Methylation
qPCR
27 (15)
Plasma
[71]
32
Glyceraldehyde-3-phosphate dehydrogenase
GAPDH
Breast
NA
qPCR
200 (100)
Serum
[26]
Breast
NA
qPCR
33 (50)
Serum
[27]
Breast
NA
qPCR
27 (32)
Serum
[28]
Breast
NA
qPCR
33 (32)
Serum
[29]
33
GNA11
GNA11
Uveal Melanoma
Mutation
NGS
28
Plasma
[34]
34
GNAQ
GNAQ
Uveal Melanoma
Mutation
NGS
28
Plasma
[34]
35
GPC3
GPC3
Pancreatic
Methylation
qPCR
30 (30)
Plasma
[86]
36
GSTP1
GSTP1
Breast
Methylation
qPCR
89
Serum
[79]
Breast
Methylation
qPCR
36 (30)
Plasma
[69]
Prostate
Methylation
PCR
12 (10)
Plasma
[95]
Prostate
Methylation
qPCR
31 (44)
Plasma
[96]
Testicular
Methylation
qPCR
73 (35)
Serum
[47]
Renal
Methylation
PCR
35 (54)
Serum
[68]
Prostate
Methylation
PCR
31 (34)
Serum
[97]
37
HIC1
HIC1
CRC
Methylation
PCR
30 (30)
Plasma
[98]
CRC
Methylation
qPCR
30 (30)
Plasma
[86]
38
HOXA7
HOXA7
Various
Methylation
qPCR
150 (60)
Plasma
[83]
39
HOXA9
HOXA9
Various
Methylation
qPCR
150 (60)
Plasma
[83]
40
HOXD13
HOXD13
Breast
Methylation
qPCR
253 (434)
Serum
[99]
41
IgH
FR3A/VLJH
Lymphoma
Clonality
NGS
75
Plasma
[43]
42
ITIH5
 
Breast
Methylation
qPCR
604 (59)
Serum
[90]
43
INK4A
INK4A
HCC
Methylation
Seq
66 (43)
Plasma
[100]
44
KLK10
KLK10
Lung
Methylation
qPCR
110 (50)
Plasma
[70]
45
KRAS
KRAS
Lung
Mutation
NGS
68 (107)
Plasma
[75]
CRC
Mutation
qPCR
52
Plasma
[101]
CRC
Mutation
qPCR
35 (135)
Plasma
[30]
CRC
Mutation
qPCR
229 (100)
Plasma
[102]
CRC
Mutation
qPCR
106
Plasma
[77]
Lung
Mutation
qPCR
82 (11)
Plasma
[103]
CRC
Mutation
BEAMing
503
Plasma
[21]
CRC
Mutation
qPCR
191
Plasma
[65]
CRC
Mutation
PCR
104
Serum
[66]
46
LINE1 Repeat
LINE1 79 bp
CRC
NA
qPCR
50 (35)
Plasma
[20]
LINE1 300 bp
CRC
NA
qPCR
503
Plasma
[21]
Breast
NA
qPCR
293 (100)
Plasma
[19]
47
MDG1
MDG1
CRC
Methylation
PCR
30 (30)
Plasma
[98]
48
Microsatellite alterations
FHIT LoH
Lung
NA
PCR
87 (14)
Plasma
[104]
FHIT LoH
Lung
NA
PCR
32 (10)
Serum
[105]
LoH
Oesophageal
NA
PCR
18 (22)
Plasma
[106]
LoH
CRC
NA
qPCR
33
Serum
[53]
3p LoH
Lung
NA
qPCR
64
Plasma
[107]
49
mitochondrial DNA
mtDNA
Breast
NA
qPCR
60 (51)
Plasma
[108]
50
MLH1
hMLH1
Breast
Methylation
qPCR
253 (434)
Serum
[99]
51
MYC
MYC
Neuroblastoma
Amplification
ddPCR
44
Plasma
[42]
52
MYF3
MYF3
Pancreatic
Methylation
qPCR
30 (30)
Plasma
[86]
53
MYLK
MYLK
Gastric
Methylation
qPCR
58 (30)
Serum
[85]
54
O(6)-methyl-guanine-DNA methyltransferase
MGMT
Lung
Methylation
qPCR
76
Serum
[62]
CRC
Methylation
qPCR
33
Serum
[53]
Breast
Methylation
qPCR
89
Serum
[79]
55
OPCML
OPCML
Ovarian
Methylation
qPCR
87 (62)
Serum
[67]
56
P14 ARF tumor suppressor protein gene
P14
Testicular
Methylation
qPCR
73 (35)
Serum
[47]
Renal
Methylation
PCR
35 (54)
Serum
[68]
57
P16 cyclin-dependent kinase inhibitor 2A
P16, CDKN2A
Testicular
Methylation
qPCR
73 (35)
Serum
[47]
Renal
Methylation
PCR
35 (54)
Serum
[68]
Breast
Methylation
qPCR
36 (30)
Plasma
[69]
Lung
Methylation
qPCR
63 (36)
Plasma
[74]
Breast
Methylation
qPCR
253 (434)
Serum
[99]
HNSCC
Methylation
qPCR
40 (41)
Serum
[87]
58
P21
P21
Breast
Methylation
qPCR
36 (30)
Plasma
[69]
59
P53
 
Various
Mutation
qPCR
20 (16)
Plasma
[109]
Various
NA
qPCR
120 (120)
Plasma
[110]
CRC
Mutation
qPCR
191
Plasma
[65]
CRC
Mutation
PCR
104
Serum
[66]
SCLC
Mutation
qPCR
51 (123)
Plasma
[55]
60
PCDHGB7
PCDHGB7
Breast
Methylation
qPCR
253 (434)
Serum
[99]
61
Peptidylprolyl isomerase A
cyclophilin A, gCYC, PPIA
CRC
NA
qPCR
229 (100)
Plasma
[102]
62
PIK3CA
PIK3CA
Breast
Mutation
qPCR
76
Both
[18]
Lung
Mutation
NGS
68 (107)
Plasma
[75]
CRC
Mutation
BEAMing
503
Plasma
[21]
CRC
Mutation
qPCR
191
Plasma
[65]
63
Prostaglandin-endoperoxid synthase 2
PTGS2
Renal
Methylation
PCR
35 (54)
Serum
[68]
Testicular
Methylation
qPCR
73 (35)
Serum
[47]
64
Protocadherin 10
PCDH10
CRC
Methylation
qPCR
67
Plasma
[111]
65
Retinoid-acid-receptor-beta gene
RARbeta2
Breast
Methylation
PCR
20 (25)
Plasma
[112]
CRC
Methylation
qPCR
33
Serum
[53]
Renal
Methylation
PCR
35 (54)
Serum
[68]
Lung
Methylation
qPCR
63 (36)
Plasma
[74]
66
RASSF1A
RASSF1A
Breast
Methylation
PCR
93 (76)
Plasma
[113]
Breast
Methylation
PCR
20 (25)
Plasma
[112]
Breast
Methylation
qPCR
39 (49)
Plasma
[22]
Breast
Methylation
qPCR
604 (59)
Serum
[90]
Melanoma
Methylation
qPCR
84 (68)
Plasma
[114]
Lung
Methylation
qPCR
76 (30)
Serum
[62]
Testicular
Methylation
qPCR
73 (35)
Serum
[47]
CRC
Methylation
qPCR
33
Serum
[53]
Ovarian
Methylation
qPCR
87 (62)
Serum
[67]
Renal
Methylation
PCR
35 (54)
Serum
[68]
Lung
Methylation
qPCR
63 (36)
Plasma
[74]
Lung
Methylation
qPCR
110 (50)
Plasma
[70]
HCC
Methylation
PCR
40 (20)
Serum
[115, 116]
HCC
Methylation
PCR
50 (50)
Serum
[117]
Renal
Methylation
PCR
27 (15)
Plasma
[71]
Breast
Methylation
qPCR
253 (434)
Serum
[99]
CRC
Methylation
PCR
30 (30)
Plasma
[98]
Renal
Methylation
qPCR
157 (43)
Serum
[118]
Ovarian
Methylation
PCR
50
Serum
[80]
Ovarian
Methylation
PCR
30 (30)
Plasma
[82]
67
RUNX3
RUNX3
Ovarian
Methylation
PCR
87 (62)
Serum
[67]
68
Septin 9
Septin 9
CRC
Methylation
qPCR
97 (172)
Plasma
[119]
CRC
Methylation
qPCR
378 (285)
Plasma
[120]
CRC
Methylation
qPCR
60 (24)
Plasma
[121]
CRC
Methylation
qPCR
55 (1457)
Plasma
[58]
Lung
Methylation
qPCR
70 (100)
Plasma
[122]
CRC
Methylation
qPCR
135 (341)
Plasma
[123]
CRC
Methylation
qPCR
50 (94)
Plasma
[124]
CRC
Methylation
qPCR
44 (444)
Plasma
[59]
69
SFN
SFN
Breast
Methylation
qPCR
253 (434)
Serum
[99]
70
SFRP5
SFRP5
Ovarian
Methylation
qPCR
87 (62)
Serum
[67]
71
SHOX2
SHOX2
Lung
Methylation
qPCR
188 (155)
Plasma
[125]
Lung
Methylation
qPCR
118 (212
Plasma
[126]
72
SOX17
SOX17
Breast
Methylation
qPCR
114 (60)
Plasma
[127]
Various
Methylation
qPCR
150(60)
Plasma
[83]
73
SLC26A4
SLC26A4
Thyroid
Methylation
qPCR
176 (19)
Plasma
[24]
74
SLC5A8
SLC5A8 SLC26A4
Thyroid
Methylation
qPCR
176 (19)
Plasma
[24]
75
SRBC
SRBC
Pancreatic
Methylation
qPCR
30 (30)
Plasma
[86]
76
TAC1
TAC1
Various
Methylation
qPCR
150 (60)
Plasma
[83]
77
human telomerase reverse transcriptase DNA
hTERT
CRC
NA
qPCR
35 (135)
Plasma
[30]
HCC
NA
qPCR
70 (30)
Plasma
[31]
HCC
NA
qPCR
60 (29)
Plasma
[32]
HNSCC
NA
qPCR
200
Plasma
[33]
78
TFPI2
TFPI2
Ovarian
Methylation
PCR
87 (62)
Serum
[67]
79
THBD-M
THBD-M
CRC
Methylation
qPCR
107 (98)
Plasma & Serum
[128]
80
TIMP3
TIMP3
Renal
Methylation
PCR
35 (54)
Serum
[68]
Breast
Methylation
qPCR
36 (30)
Plasma
[69]
81
TMS
TMS
Pancreatic
Methylation
qPCR
30 (30)
Plasma
[86]
82
UCHL1
UCHL1
HNSCC
Methylation
PCR
40 (41)
Serum
[87]
83
Von Hippel Lindau gene
VHL
CRC
Methylation
qPCR
30 (30)
Plasma
[86]
Pancreatic
Methylation
qPCR
30 (30)
Plasma
[86]
Renal
Methylation
qPCR
157 (43)
Serum
[118]
84
ZFP42
ZFP42
Various
Methylation
qPCR
150 (60)
Plasma
[83]
CRC colorectal cancer, HNSCC head and neck squamous cell carcinoma, HCC hepatocellular carcinoma, LCH Langerhans cell histocytosis, SCLC small cell lung cancer
The ctDNA biomarkers divided naturally into two groups:
I.
those with potential specificity for neoplasia (ctDNA - usually mutations or DNA alterations such as methylation), and
 
II.
those designed to measure DNA levels, which may not be specific to neoplasia.
 
Figure 2 shows the distribution of studies by cancer type, including two publications on amplification [12, 14], and one on clonality [15]. One of the amplification papers looked at HER2 [14], while the other examined multiple targets by NGS [12].
Of the 94 publications included, 72 publications (77%) were case-control design diagnostic validation studies, and 22 were case series. The size and design of the studies varied widely. The largest study included 640 cancer patients [16]. The median study size was 65 cases, with a mean of 98 cases (range 12–640 cancer patients), indicating that the bulk of studies (67/94, 71%) included <100 patients (Fig. 3).
Most publications were focussed on ctDNA in plasma (n = 67) rather than serum (n = 25) with 2 comparing both. Plasma was used for 38 markers, and serum for 28 markers, and either for 18 markers (Fig. 4). Two comparative studies of serum and plasma were conducted: one for BRAF mutations, and the other for PIK3CA mutations [17, 18].
The target of ctDNA studies and the methods used to measure these targets varied considerably (Figs. 5 and 6 respectively). Non-specific total ctDNA levels (quantitation) were usually estimated by size distribution assays based on repeats: LINE1, and ALU were used in 3 [1921] and 6 publications respectively [2025]. However, some single genes were also used to measure DNA levels – particularly GAPDH in a series of 4 publications on breast cancer [2629], and hTERT in 4 publications [3033]. The majority of publications examined gene methylation markers (n = 49), though most examined methylation of multiple target genes for a particular tumour type (Fig. 5). Genes commonly mutated in cancer were also markers of interest, namely APC, BRAF, EGFR, HER2, GNAQ, GNA11, KRAS, P53, and PIK3CA. Only one gene, APC, was studied for both methylation and mutation. Few markers were used to identify particular tumour types, but some are particularly likely to occur in certain tumour types. GNAQ and GNA11 mutations have been identified in the plasma of uveal melanoma patients and are rare in other tumour types [34]. Other mutations are not tumour type-specific, and mutations in 6 of the 9 genes listed above were reported in multiple tumour types.

Discussion

The number of publications on ctDNA is increasing rapidly [35, 36], and a recent review emphasises the potential of the field [37]. Most (71%) are small case control studies with less than 100 patients, and in our view very few studies meet the requirements of analytical validation allowing their use within accredited (ISO:15,189) clinical laboratories, though some may have unpublished commercially-held analytical validation data. The stage and size of the tumours included is variable, and few studies are large enough to give robust subgroup assessments. Larger tumours produce more ctDNA, though tumour type also has an impact [16]. The value of small studies with no comparison between methods, or even the inclusion of controls is highly questionable. Most include a statement that ‘larger studies are required’, but larger trials rarely result due to the necessary cost implications. Unless well-designed prospective studies based on sample size calculations are performed, there is little likelihood of such methods reaching clinical practice for the detection of cancer at an early stage. There is also a likelihood of bias in that negative results for these markers are rarely if ever reported, and unlike clinical trials, there is no requirement for the registration of diagnostic validation studies. The use of ctDNA for early cancer detection comes under existing molecular pathology guidance, which emphasises the requirements for careful pre-analytical preparation, analysis, and reporting of results [38]. It is important that studies adhere to the Standards for Reporting of Diagnostic Accuracy Studies (STARD) guidance [39], and regional guidance (e.g. US Food and Drug Adminstration (FDA); UK National Institute for Health and Care Excellence (NICE); Clinical & Laboratory Standards Institute (CLSI)). It is hardly surprising then that, to date, no ctDNA markers have made it into screening programmes, due in part to the economic feasibility of completing the necessary stages of validation [40]. Nevertheless, there is encouraging evidence that ctDNA can be used to detect cancers of many types [16], and the poor quality of many studies should not detract from this fact.
A plethora of methods are available for ctDNA measurement, which have been well reviewed elsewhere [41]. BEAMing, PCR clamping methods, and deep sequencing using NGS are now the most commonly used [42, 43] and are widely regarded as the most sensitive methods currently available. A recent report of copy number variation (CNV) in breast cancer is not surprising given the ability of this method to detect such changes in pregnancy [15]. However, it should be noted that many of these methods are expensive. The development of highly sensitive NGS methods for ctDNA may prove necessary to obtain the best results [44], but large blood samples (> 10 ml may be needed as the number of DNA molecules present in small samples is often low) [45]. This may be at odds with the key requirement of cost effectiveness for screening programmes, and in our view this represents a real challenge for ctDNA. The problem is probably not insuperable if automation allows the integration of such methods into large blood sciences laboratories, but this is not as yet the case.
As ctDNA is composed largely of short fragments, short amplicons are required for maximum sensitivity of PCR reactions, particularly if mutations are being detected [46]. This is compounded by DNA loss in some reactions, particularly bisulphite modification of DNA, and it may be preferable to use nuclease protection assays [47, 48]. Methylation of key genes involved in carcinogenesis can be found in ctDNA, and has been studied by many groups, but it should be noted that substantial numbers of normal controls also have methylation of ctDNA for these genes [49].
It is clear that high sensitivity methods will be needed if ctDNA is to be used for early cancer detection. Several factors affect the sensitivity of ctDNA measurement. The first is the extraction method, and there are as yet too few studies which have compared the different options available, which now include automated instruments as well as manual extraction systems [50, 51]. The proportion of tumour derived DNA (ctDNA) in total cfDNA is greater in plasma than serum, and the higher ctDNA levels in serum are due to leakage from leukocytes during clotting [17]. The dilution effect for ctDNA in serum results in a reduced ability to detect mutations, particularly by methods with low analytical sensitivity [50]. Most groups working in the field realise this, and the majority of publications now look at plasma rather than serum.
Several publications were noteworthy, including one influential study which did not include healthy controls [16]. However, the comparison of DNA levels and multiple mutations in plasma from many different tumours types is helpful [44], and makes it clear that some tumours (e.g. gliomas) do not have high ctDNA levels in plasma, as previously found when comparing CSF with plasma [52]. This is also one of several publications that examines early stage disease, and shows that patients with localised disease have lower ctDNA levels [16]. Few publications have examined the ability of ctDNA to detect smaller tumours, though all agree that ctDNA levels increase as tumours enlarge [42].
Choice of target also influences results: the use of LINE1 and ALU repeats allows quantitative size distribution of DNA to be measured. Several publications suggest that this can distinguish cancer, and even pre-cancerous conditions from controls [30]. The size distribution of CRC appears to be different from other tumours due to first pass hepatic metabolism [20, 53]. Absolute quantitation by single gene methods such as GAPDH or hTERT will result in lower estimates of DNA content, and it is likely that this is due to the higher sensitivity of the ALU and LINE1 assays [30].
The use of mutations common within cancers is attractive, and the use of ctDNA to provide companion diagnostic information in patients in whom biopsy material is not available is now entering practice [54]. However, it should be noted that such mutations in P53 can occur in the blood of healthy controls, and could give rise to substantial numbers of false positive results [55].
Septin 9 methylation is often regarded as a model for future work [56, 57], and it is notable that there are some large studies [58] within the evidence base for the use of this marker in colorectal cancer, often used in addition to other markers, such as faecal occult blood testing (FoBT) or faecal immunohistochemical testing (FIT). Pre-analytical factors have been examined for this marker [59], including diurnal variation [60]. Plasma methylation of Septin 9 is now available as a commercial test (Epi proColon 2.0; Epigenomics AG, Berlin, Germany) which has recently obtained FDA approval for colorectal cancer screening (April 2016). This is the first blood test to be approved for cancer screening, and represents an encouraging milestone.
Other methylation targets have been studied in depth and show considerable promise. These include APC for colorectal cancer, with a large number of studies (Table 2), and SHOX3, for which a recent meta-analysis suggests that it could have an important role in the diagnosis of lung cancer [61].
There is an encouraging trend towards larger, more ambitious studies, supported by the commercial sector (e.g. (https://​clinicaltrials.​gov/​ct2/​show/​NCT02889978, and https://​clinicaltrials.​gov/​ct2/​show/​NCT03085888). Case control studies (particular retrospective ones) can give biased results, and prospective studies in at-risk cohorts would be more useful in examining the predictive capability of these markers. Such prospective studies should include controls proven not to have cancer. The comparison of new with existing methods (e.g. tumour markers, radiology), and competing technologies, is recommended, and often required by regulators. This has cost implications for funding bodies, but is essential if the field is to progress rapidly.

Conclusions

While ctDNA analysis may provide a viable option for the early detection of cancers, not all cancers are detectable using current methods. However, improvements in technology are rapidly overcoming some of the issues of analytical sensitivity, and it is likely that mutation and methylation analysis of ctDNA will improve specificity for the diagnosis of cancer.

Acknowledgements

We are grateful to the wider Early Cancer Detection Consortium for their assistance in putting together this paper, and for the many discussions which underpin it. Patient and Public representatives were involved in this work.

Funding

This work was conducted on behalf of the Early Cancer Detection Consortium, within the programme of work for work packages & 2. The Early Cancer Detection Consortium is funded by Cancer Research UK under grant number: C50028/A18554. It was subsequently supported by an unrestricted educational grant from PinPoint Cancer Ltd. (www.​pinpointcancer.​co.​uk), following cessation of the grant in 2016. Neither of the two funding bodies had any input or influence over the design, study, collection, analysis, or interpretation of the data.

Availability of data and materials

The papers quoted are publically available from the publishers, and many are now open access.

Authors’ information

IC is a pathologist and has recently moved to a post with the International Agency for Research on Cancer of the World Health Organisation in Lyon. LU, and SH are Research Fellows in systematic review and HBW is an Information Specialist working at the University of Sheffield, UK. HK is a scientist and PhD student working on early cancer detection. AR is a Lecturer in Biomedical Science working at Coventry University, UK. STP is an associate professor with a NIHR Career Development Fellowship using quantitative research methods to assess new screening programmes. MM is a healthcare scientist at the University of Leeds with expertise in biomarker and in vitro diagnostic (IVD) development, validation and clinical evaluation. AC is Professor of Cancer Genetic Epidemiology at the University of Sheffield, UK. DT is Reader in Epidemiology and Biostatistics at the University of Sheffield, UK. OS is Director of the Trinity Translational Medicine Institute (TTMI) and Professor in Molecular Pathology at Trinity College Dublin, Eire. JS is Professor of Translational Cancer Genetics at Leicester University, UK, with a particular interest in cfDNA.
Not applicable.
Not applicable.

Competing interests

The ECDC has grant funding for early cancer biomarker research from Cancer Research UK who funded this work. The ECDC involves several companies as follows: GE Healthcare, Life Technologies, NALIA Systems Ltd., and Perkin-Elmer. Individual ECDC members have declared their interests to the ECDC secretariat. IC was formerly chairman and CEO of PinPoint Cancer Ltd., a spin-out company from ECDC which in part funded the completion of this work though provision of staff time (IC). MM is supported by the National Institute for Health Research Diagnostic Evidence Co-operative Leeds. The views expressed are those of the author(s) and not necessarily those of the NHS, the NIHR or the UK Department of Health.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat McPhail S, et al. Stage at diagnosis and early mortality from cancer in England. Br J Cancer. 2015;112 Suppl 1:S108–15.PubMedCrossRef McPhail S, et al. Stage at diagnosis and early mortality from cancer in England. Br J Cancer. 2015;112 Suppl 1:S108–15.PubMedCrossRef
2.
Zurück zum Zitat Duffy MJ. Tumor markers in clinical practice: a review focusing on common solid cancers. Med Princ Pract. 2013;22(1):4–11.PubMedCrossRef Duffy MJ. Tumor markers in clinical practice: a review focusing on common solid cancers. Med Princ Pract. 2013;22(1):4–11.PubMedCrossRef
3.
Zurück zum Zitat Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.PubMedCrossRef Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.PubMedCrossRef
5.
Zurück zum Zitat Lo YM, et al. Presence of fetal DNA in maternal plasma and serum. Lancet. 1997;350(9076):485–7.PubMedCrossRef Lo YM, et al. Presence of fetal DNA in maternal plasma and serum. Lancet. 1997;350(9076):485–7.PubMedCrossRef
6.
Zurück zum Zitat Johnson PJ, Lo YM. Plasma nucleic acids in the diagnosis and management of malignant disease. Clin Chem. 2002;48(8):1186–93.PubMed Johnson PJ, Lo YM. Plasma nucleic acids in the diagnosis and management of malignant disease. Clin Chem. 2002;48(8):1186–93.PubMed
7.
Zurück zum Zitat Lou X, et al. A novel Alu-based real-time PCR method for the quantitative detection of plasma circulating cell-free DNA: sensitivity and specificity for the diagnosis of myocardial infarction. Int J Mol Med. 2015;35(1):72–80.PubMed Lou X, et al. A novel Alu-based real-time PCR method for the quantitative detection of plasma circulating cell-free DNA: sensitivity and specificity for the diagnosis of myocardial infarction. Int J Mol Med. 2015;35(1):72–80.PubMed
8.
Zurück zum Zitat Swarup V, Rajeswari MR. Circulating (cell-free) nucleic acids--a promising, non-invasive tool for early detection of several human diseases. FEBS Lett. 2007;581(5):795–9.PubMedCrossRef Swarup V, Rajeswari MR. Circulating (cell-free) nucleic acids--a promising, non-invasive tool for early detection of several human diseases. FEBS Lett. 2007;581(5):795–9.PubMedCrossRef
9.
10.
Zurück zum Zitat Zonta E, Nizard P, Taly V. Assessment of DNA integrity, applications for cancer research. Adv Clin Chem. 2015;70:197–246.PubMedCrossRef Zonta E, Nizard P, Taly V. Assessment of DNA integrity, applications for cancer research. Adv Clin Chem. 2015;70:197–246.PubMedCrossRef
11.
Zurück zum Zitat Lo YM, et al. Maternal plasma DNA sequencing reveals the genome-wide genetic and mutational profile of the fetus. Sci Transl Med. 2010;2(61):61ra91.PubMedCrossRef Lo YM, et al. Maternal plasma DNA sequencing reveals the genome-wide genetic and mutational profile of the fetus. Sci Transl Med. 2010;2(61):61ra91.PubMedCrossRef
12.
Zurück zum Zitat Heitzer E, et al. Establishment of tumor-specific copy number alterations from plasma DNA of patients with cancer. Int J Cancer. 2013;133(2):346–56.PubMedPubMedCentralCrossRef Heitzer E, et al. Establishment of tumor-specific copy number alterations from plasma DNA of patients with cancer. Int J Cancer. 2013;133(2):346–56.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Uttley L, et al. Building the evidence base of blood-based biomarkers for early detection of cancer: a rapid systematic mapping review. EBioMedicine. 2016;10:164–73.PubMedPubMedCentralCrossRef Uttley L, et al. Building the evidence base of blood-based biomarkers for early detection of cancer: a rapid systematic mapping review. EBioMedicine. 2016;10:164–73.PubMedPubMedCentralCrossRef
14.
15.
Zurück zum Zitat Kirkizlar E, et al. Detection of clonal and subclonal copy-number variants in cell-free DNA from patients with breast cancer using a massively multiplexed PCR methodology. Transl Oncol. 2015;8(5):407–16.PubMedPubMedCentralCrossRef Kirkizlar E, et al. Detection of clonal and subclonal copy-number variants in cell-free DNA from patients with breast cancer using a massively multiplexed PCR methodology. Transl Oncol. 2015;8(5):407–16.PubMedPubMedCentralCrossRef
16.
17.
Zurück zum Zitat Aung KL, et al. Analytical validation of BRAF mutation testing from circulating free DNA using the amplification refractory mutation testing system. J Mol Diagn. 2014;16(3):343–9.PubMedCrossRef Aung KL, et al. Analytical validation of BRAF mutation testing from circulating free DNA using the amplification refractory mutation testing system. J Mol Diagn. 2014;16(3):343–9.PubMedCrossRef
18.
Zurück zum Zitat Board, R.E., et al., Detection of PIK3CA mutations in circulating free DNA in patients with breast cancer. Breast Cancer Res Treat, 2010. 120(2): p. 461–467. Board, R.E., et al., Detection of PIK3CA mutations in circulating free DNA in patients with breast cancer. Breast Cancer Res Treat, 2010. 120(2): p. 461–467.
19.
Zurück zum Zitat Madhavan D, et al. Plasma DNA integrity as a biomarker for primary and metastatic breast cancer and potential marker for early diagnosis. Breast Cancer Res Treat. 2014;146(1):163–74.PubMedCrossRef Madhavan D, et al. Plasma DNA integrity as a biomarker for primary and metastatic breast cancer and potential marker for early diagnosis. Breast Cancer Res Treat. 2014;146(1):163–74.PubMedCrossRef
20.
21.
Zurück zum Zitat Tabernero J, et al. Analysis of circulating DNA and protein biomarkers to predict the clinical activity of regorafenib and assess prognosis in patients with metastatic colorectal cancer: a retrospective, exploratory analysis of the CORRECT trial. Lancet Oncol. 2015;16(8):937–48.PubMedCrossRef Tabernero J, et al. Analysis of circulating DNA and protein biomarkers to predict the clinical activity of regorafenib and assess prognosis in patients with metastatic colorectal cancer: a retrospective, exploratory analysis of the CORRECT trial. Lancet Oncol. 2015;16(8):937–48.PubMedCrossRef
22.
Zurück zum Zitat Agostini M, et al. Circulating cell-free DNA: a promising marker of regional lymphonode metastasis in breast cancer patients. Cancer Biomark. 2012;11(2–3):89–98.PubMedCrossRef Agostini M, et al. Circulating cell-free DNA: a promising marker of regional lymphonode metastasis in breast cancer patients. Cancer Biomark. 2012;11(2–3):89–98.PubMedCrossRef
23.
Zurück zum Zitat Hao TB, et al. Circulating cell-free DNA in serum as a biomarker for diagnosis and prognostic prediction of colorectal cancer. Br J Cancer. 2014;111(8):1482–9.PubMedPubMedCentralCrossRef Hao TB, et al. Circulating cell-free DNA in serum as a biomarker for diagnosis and prognostic prediction of colorectal cancer. Br J Cancer. 2014;111(8):1482–9.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Zane M, et al. Circulating cell-free DNA, SLC5A8 and SLC26A4 hypermethylation, BRAF(V600E): a non-invasive tool panel for early detection of thyroid cancer. Biomed Pharmacother. 2013;67(8):723–30.PubMedCrossRef Zane M, et al. Circulating cell-free DNA, SLC5A8 and SLC26A4 hypermethylation, BRAF(V600E): a non-invasive tool panel for early detection of thyroid cancer. Biomed Pharmacother. 2013;67(8):723–30.PubMedCrossRef
25.
Zurück zum Zitat Sikora K, et al. Evaluation of cell-free DNA as a biomarker for pancreatic malignancies. Int J Biol Markers. 2014:e136–41. Sikora K, et al. Evaluation of cell-free DNA as a biomarker for pancreatic malignancies. Int J Biol Markers. 2014:e136–41.
26.
Zurück zum Zitat Gong B, et al. Cell-free DNA in blood is a potential diagnostic biomarker of breast cancer. Oncol Lett. 2012;3(4):897–900.PubMedPubMedCentral Gong B, et al. Cell-free DNA in blood is a potential diagnostic biomarker of breast cancer. Oncol Lett. 2012;3(4):897–900.PubMedPubMedCentral
27.
Zurück zum Zitat Zhong XY, et al. Elevated level of cell-free plasma DNA is associated with breast cancer. Arch Gynecol Obstet. 2007;276(4):327–31.PubMedCrossRef Zhong XY, et al. Elevated level of cell-free plasma DNA is associated with breast cancer. Arch Gynecol Obstet. 2007;276(4):327–31.PubMedCrossRef
28.
Zurück zum Zitat Seefeld M, et al. Parallel assessment of circulatory cell-free DNA by PCR and nucleosomes by ELISA in breast tumors. Int J Biol Markers. 2008;23(2):69–73.PubMedCrossRef Seefeld M, et al. Parallel assessment of circulatory cell-free DNA by PCR and nucleosomes by ELISA in breast tumors. Int J Biol Markers. 2008;23(2):69–73.PubMedCrossRef
29.
Zurück zum Zitat Zanetti-Dallenbach RA, et al. Levels of circulating cell-free serum DNA in benign and malignant breast lesions. Int J Biol Markers. 2007;22(2):95–9.PubMedCrossRef Zanetti-Dallenbach RA, et al. Levels of circulating cell-free serum DNA in benign and malignant breast lesions. Int J Biol Markers. 2007;22(2):95–9.PubMedCrossRef
30.
Zurück zum Zitat Perrone F, et al. Circulating free DNA in a screening program for early colorectal cancer detection. Tumori. 2014;100(2):115–21.PubMed Perrone F, et al. Circulating free DNA in a screening program for early colorectal cancer detection. Tumori. 2014;100(2):115–21.PubMed
31.
Zurück zum Zitat Divella R, et al. PAI-1, t-PA and circulating hTERT DNA as related to virus infection in liver carcinogenesis. Anticancer Res. 2008;28(1A):223–8.PubMed Divella R, et al. PAI-1, t-PA and circulating hTERT DNA as related to virus infection in liver carcinogenesis. Anticancer Res. 2008;28(1A):223–8.PubMed
32.
Zurück zum Zitat Yang YJ, et al. Quantification of plasma hTERT DNA in hepatocellular carcinoma patients by quantitative fluorescent polymerase chain reaction. Clin Invest Med. 2011;34(4):E238.PubMedCrossRef Yang YJ, et al. Quantification of plasma hTERT DNA in hepatocellular carcinoma patients by quantitative fluorescent polymerase chain reaction. Clin Invest Med. 2011;34(4):E238.PubMedCrossRef
33.
Zurück zum Zitat Mazurek AM, et al. Assessment of the total cfDNA and HPV16/18 detection in plasma samples of head and neck squamous cell carcinoma patients. Oral Oncol. 2016;54:36–41.PubMedCrossRef Mazurek AM, et al. Assessment of the total cfDNA and HPV16/18 detection in plasma samples of head and neck squamous cell carcinoma patients. Oral Oncol. 2016;54:36–41.PubMedCrossRef
34.
Zurück zum Zitat Metz CH, et al. Ultradeep sequencing detects GNAQ and GNA11 mutations in cell-free DNA from plasma of patients with uveal melanoma. Cancer Med. 2013;2(2):208–15.PubMedPubMedCentralCrossRef Metz CH, et al. Ultradeep sequencing detects GNAQ and GNA11 mutations in cell-free DNA from plasma of patients with uveal melanoma. Cancer Med. 2013;2(2):208–15.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Benesova L, et al. Mutation-based detection and monitoring of cell-free tumor DNA in peripheral blood of cancer patients. Anal Biochem. 2013;433(2):227–34.PubMedCrossRef Benesova L, et al. Mutation-based detection and monitoring of cell-free tumor DNA in peripheral blood of cancer patients. Anal Biochem. 2013;433(2):227–34.PubMedCrossRef
36.
Zurück zum Zitat Crowley E, et al. Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol. 2013;10(8):472–84.PubMedCrossRef Crowley E, et al. Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol. 2013;10(8):472–84.PubMedCrossRef
39.
Zurück zum Zitat Bossuyt PM, et al. STARD 2015: an updated list of essential items for reporting diagnostic accuracy studies. Clin Chem. 2015;61(12):1446–52.PubMedCrossRef Bossuyt PM, et al. STARD 2015: an updated list of essential items for reporting diagnostic accuracy studies. Clin Chem. 2015;61(12):1446–52.PubMedCrossRef
40.
Zurück zum Zitat Ladabaum U, et al. Colorectal cancer screening with blood-based biomarkers: cost-effectiveness of methylated septin 9 DNA versus current strategies. Cancer Epidemiol Biomark Prev. 2013;22(9):1567–76.CrossRef Ladabaum U, et al. Colorectal cancer screening with blood-based biomarkers: cost-effectiveness of methylated septin 9 DNA versus current strategies. Cancer Epidemiol Biomark Prev. 2013;22(9):1567–76.CrossRef
41.
Zurück zum Zitat Heitzer E, Ulz P, Geigl JB. Circulating tumor DNA as a liquid biopsy for cancer. Clin Chem. 2015;61(1):112–23.PubMedCrossRef Heitzer E, Ulz P, Geigl JB. Circulating tumor DNA as a liquid biopsy for cancer. Clin Chem. 2015;61(1):112–23.PubMedCrossRef
42.
Zurück zum Zitat Kurihara S, et al. Circulating free DNA as non-invasive diagnostic biomarker for childhood solid tumors. J Pediatr Surg. 2015;50(12):2094–7.PubMedCrossRef Kurihara S, et al. Circulating free DNA as non-invasive diagnostic biomarker for childhood solid tumors. J Pediatr Surg. 2015;50(12):2094–7.PubMedCrossRef
43.
Zurück zum Zitat Kurtz DM, et al. Noninvasive monitoring of diffuse large B-cell lymphoma by immunoglobulin high-throughput sequencing. Blood. 2015;125(24):3679–87.PubMedPubMedCentralCrossRef Kurtz DM, et al. Noninvasive monitoring of diffuse large B-cell lymphoma by immunoglobulin high-throughput sequencing. Blood. 2015;125(24):3679–87.PubMedPubMedCentralCrossRef
44.
45.
Zurück zum Zitat Belic J, et al. Rapid identification of plasma DNA samples with increased ctDNA levels by a modified FAST-SeqS approach. Clin Chem. 2015;61(6):838–49.PubMedCrossRef Belic J, et al. Rapid identification of plasma DNA samples with increased ctDNA levels by a modified FAST-SeqS approach. Clin Chem. 2015;61(6):838–49.PubMedCrossRef
46.
Zurück zum Zitat Andersen RF, et al. Improved sensitivity of circulating tumor DNA measurement using short PCR amplicons. Clin Chim Acta. 2015;439:97–101.PubMedCrossRef Andersen RF, et al. Improved sensitivity of circulating tumor DNA measurement using short PCR amplicons. Clin Chim Acta. 2015;439:97–101.PubMedCrossRef
47.
Zurück zum Zitat Ellinger J, et al. CpG island hypermethylation of cell-free circulating serum DNA in patients with testicular cancer. J Urol. 2009;182(1):324–9.PubMedCrossRef Ellinger J, et al. CpG island hypermethylation of cell-free circulating serum DNA in patients with testicular cancer. J Urol. 2009;182(1):324–9.PubMedCrossRef
48.
Zurück zum Zitat Martinez-Galan J, et al. Quantitative detection of methylated ESR1 and 14-3-3-sigma gene promoters in serum as candidate biomarkers for diagnosis of breast cancer and evaluation of treatment efficacy. Cancer Biol Ther. 2008;7(6):958–65.PubMedCrossRef Martinez-Galan J, et al. Quantitative detection of methylated ESR1 and 14-3-3-sigma gene promoters in serum as candidate biomarkers for diagnosis of breast cancer and evaluation of treatment efficacy. Cancer Biol Ther. 2008;7(6):958–65.PubMedCrossRef
49.
Zurück zum Zitat Kristensen LS, et al. Methylation profiling of normal individuals reveals mosaic promoter methylation of cancer-associated genes. Oncotarget. 2012;3(4):450–61.PubMedPubMedCentralCrossRef Kristensen LS, et al. Methylation profiling of normal individuals reveals mosaic promoter methylation of cancer-associated genes. Oncotarget. 2012;3(4):450–61.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Sorber L, et al. A comparison of cell-free DNA isolation kits: isolation and quantification of cell-free DNA in plasma. J Mol Diagn. 2017;19(1):162–8.PubMedCrossRef Sorber L, et al. A comparison of cell-free DNA isolation kits: isolation and quantification of cell-free DNA in plasma. J Mol Diagn. 2017;19(1):162–8.PubMedCrossRef
52.
Zurück zum Zitat De Mattos-Arruda L, et al. Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nat Commun. 2015;6:8839.PubMedPubMedCentralCrossRef De Mattos-Arruda L, et al. Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nat Commun. 2015;6:8839.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Taback B, Saha S, Hoon DS. Comparative analysis of mesenteric and peripheral blood circulating tumor DNA in colorectal cancer patients. Ann N Y Acad Sci. 2006;1075:197–203.PubMedCrossRef Taback B, Saha S, Hoon DS. Comparative analysis of mesenteric and peripheral blood circulating tumor DNA in colorectal cancer patients. Ann N Y Acad Sci. 2006;1075:197–203.PubMedCrossRef
54.
Zurück zum Zitat Uchida J, et al. Diagnostic accuracy of noninvasive genotyping of EGFR in lung cancer patients by deep sequencing of plasma cell-free DNA. Clin Chem. 2015;61(9):1191–6.PubMedCrossRef Uchida J, et al. Diagnostic accuracy of noninvasive genotyping of EGFR in lung cancer patients by deep sequencing of plasma cell-free DNA. Clin Chem. 2015;61(9):1191–6.PubMedCrossRef
55.
Zurück zum Zitat Fernandez-Cuesta L, et al. Identification of circulating tumor DNA for the early detection of small-cell lung cancer. EBioMedicine. 2016;10:117–23.PubMedPubMedCentralCrossRef Fernandez-Cuesta L, et al. Identification of circulating tumor DNA for the early detection of small-cell lung cancer. EBioMedicine. 2016;10:117–23.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Payne SR. From discovery to the clinic: the novel DNA methylation biomarker (m)SEPT9 for the detection of colorectal cancer in blood. Epigenomics. 2010;2(4):575–85.PubMedCrossRef Payne SR. From discovery to the clinic: the novel DNA methylation biomarker (m)SEPT9 for the detection of colorectal cancer in blood. Epigenomics. 2010;2(4):575–85.PubMedCrossRef
58.
Zurück zum Zitat Church TR, et al. Prospective evaluation of methylated SEPT9 in plasma for detection of asymptomatic colorectal cancer. Gut. 2014;63(2):317–25.PubMedCrossRef Church TR, et al. Prospective evaluation of methylated SEPT9 in plasma for detection of asymptomatic colorectal cancer. Gut. 2014;63(2):317–25.PubMedCrossRef
59.
Zurück zum Zitat Potter NT, et al. Validation of a real-time PCR-based qualitative assay for the detection of methylated SEPT9 DNA in human plasma. Clin Chem. 2014;60(9):1183–91.PubMedCrossRef Potter NT, et al. Validation of a real-time PCR-based qualitative assay for the detection of methylated SEPT9 DNA in human plasma. Clin Chem. 2014;60(9):1183–91.PubMedCrossRef
60.
Zurück zum Zitat Toth K, et al. Circadian rhythm of methylated Septin 9, cell-free DNA amount and tumor markers in colorectal cancer patients. Pathol Oncol Res. 2016; Toth K, et al. Circadian rhythm of methylated Septin 9, cell-free DNA amount and tumor markers in colorectal cancer patients. Pathol Oncol Res. 2016;
61.
Zurück zum Zitat Zhao QT, et al. Diagnostic value of SHOX2 DNA methylation in lung cancer: a meta-analysis. Onco Targets Ther. 2015;8:3433–9.PubMedPubMedCentral Zhao QT, et al. Diagnostic value of SHOX2 DNA methylation in lung cancer: a meta-analysis. Onco Targets Ther. 2015;8:3433–9.PubMedPubMedCentral
62.
63.
64.
Zurück zum Zitat Diehl F, et al. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proc Natl Acad Sci U S A. 2005;102(45):16368–73.PubMedPubMedCentralCrossRef Diehl F, et al. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proc Natl Acad Sci U S A. 2005;102(45):16368–73.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Lin JK, et al. Clinical relevance of alterations in quantity and quality of plasma DNA in colorectal cancer patients: based on the mutation spectra detected in primary tumors. Ann Surg Oncol, 2014 21 Suppl. 4:S680–6. Lin JK, et al. Clinical relevance of alterations in quantity and quality of plasma DNA in colorectal cancer patients: based on the mutation spectra detected in primary tumors. Ann Surg Oncol, 2014 21 Suppl. 4:S680–6.
66.
Zurück zum Zitat Wang JY, et al. Molecular detection of APC, K- ras, and p53 mutations in the serum of colorectal cancer patients as circulating biomarkers. World J Surg. 2004;28(7):721–6.PubMedCrossRef Wang JY, et al. Molecular detection of APC, K- ras, and p53 mutations in the serum of colorectal cancer patients as circulating biomarkers. World J Surg. 2004;28(7):721–6.PubMedCrossRef
67.
Zurück zum Zitat Zhang Q, et al. A multiplex methylation-specific PCR assay for the detection of early-stage ovarian cancer using cell-free serum DNA. Gynecol Oncol. 2013;130(1):132–9.PubMedCrossRef Zhang Q, et al. A multiplex methylation-specific PCR assay for the detection of early-stage ovarian cancer using cell-free serum DNA. Gynecol Oncol. 2013;130(1):132–9.PubMedCrossRef
68.
Zurück zum Zitat Hauser S, et al. Serum DNA hypermethylation in patients with kidney cancer: results of a prospective study. Anticancer Res. 2013;33(10):4651–6.PubMed Hauser S, et al. Serum DNA hypermethylation in patients with kidney cancer: results of a prospective study. Anticancer Res. 2013;33(10):4651–6.PubMed
69.
Zurück zum Zitat Radpour R, et al. Hypermethylation of tumor suppressor genes involved in critical regulatory pathways for developing a blood-based test in breast cancer. PLoS One. 2011;6(1):e16080.PubMedPubMedCentralCrossRef Radpour R, et al. Hypermethylation of tumor suppressor genes involved in critical regulatory pathways for developing a blood-based test in breast cancer. PLoS One. 2011;6(1):e16080.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Zhang Y, et al. Methylation of multiple genes as a candidate biomarker in non-small cell lung cancer. Cancer Lett. 2011;303(1):21–8.PubMedCrossRef Zhang Y, et al. Methylation of multiple genes as a candidate biomarker in non-small cell lung cancer. Cancer Lett. 2011;303(1):21–8.PubMedCrossRef
71.
Zurück zum Zitat Skrypkina I, et al. Concentration and methylation of cell-free DNA from blood plasma as diagnostic markers of renal cancer. Dis Markers. 2016;2016:3693096.PubMedPubMedCentralCrossRef Skrypkina I, et al. Concentration and methylation of cell-free DNA from blood plasma as diagnostic markers of renal cancer. Dis Markers. 2016;2016:3693096.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Pack SC, et al. Usefulness of plasma epigenetic changes of five major genes involved in the pathogenesis of colorectal cancer. Int J Color Dis. 2013;28(1):139–47.CrossRef Pack SC, et al. Usefulness of plasma epigenetic changes of five major genes involved in the pathogenesis of colorectal cancer. Int J Color Dis. 2013;28(1):139–47.CrossRef
73.
Zurück zum Zitat Sikora K, et al. Evaluation of cell-free DNA as a biomarker for pancreatic malignancies. Int J Biol Markers. 2015;30(1):e136–41.PubMedCrossRef Sikora K, et al. Evaluation of cell-free DNA as a biomarker for pancreatic malignancies. Int J Biol Markers. 2015;30(1):e136–41.PubMedCrossRef
74.
Zurück zum Zitat Hsu HS, et al. Characterization of a multiple epigenetic marker panel for lung cancer detection and risk assessment in plasma. Cancer. 2007;110(9):2019–26.PubMedCrossRef Hsu HS, et al. Characterization of a multiple epigenetic marker panel for lung cancer detection and risk assessment in plasma. Cancer. 2007;110(9):2019–26.PubMedCrossRef
75.
Zurück zum Zitat Couraud S, et al. Noninvasive diagnosis of actionable mutations by deep sequencing of circulating free DNA in lung cancer from never-smokers: a proof-of-concept study from BioCAST/IFCT-1002. Clin Cancer Res. 2014;20(17):4613–24.PubMedCrossRef Couraud S, et al. Noninvasive diagnosis of actionable mutations by deep sequencing of circulating free DNA in lung cancer from never-smokers: a proof-of-concept study from BioCAST/IFCT-1002. Clin Cancer Res. 2014;20(17):4613–24.PubMedCrossRef
76.
Zurück zum Zitat Hyman DM, et al. Prospective blinded study of BRAFV600E mutation detection in cell-free DNA of patients with systemic histiocytic disorders. Cancer Discov. 2015;5(1):64–71.PubMedCrossRef Hyman DM, et al. Prospective blinded study of BRAFV600E mutation detection in cell-free DNA of patients with systemic histiocytic disorders. Cancer Discov. 2015;5(1):64–71.PubMedCrossRef
77.
Zurück zum Zitat Thierry AR, et al. Clinical validation of the detection of KRAS and BRAF mutations from circulating tumor DNA. Nat Med. 2014;20(4):430–5.PubMedCrossRef Thierry AR, et al. Clinical validation of the detection of KRAS and BRAF mutations from circulating tumor DNA. Nat Med. 2014;20(4):430–5.PubMedCrossRef
78.
Zurück zum Zitat Kim BH, et al. Detection of plasma BRAF(V600E) mutation is associated with lung metastasis in papillary thyroid carcinomas. Yonsei Med J. 2015;56(3):634–40.PubMedPubMedCentralCrossRef Kim BH, et al. Detection of plasma BRAF(V600E) mutation is associated with lung metastasis in papillary thyroid carcinomas. Yonsei Med J. 2015;56(3):634–40.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Sharma G, et al. Clinical significance of promoter hypermethylation of DNA repair genes in tumor and serum DNA in invasive ductal breast carcinoma patients. Life Sci. 2010;87(3–4):83–91.PubMedCrossRef Sharma G, et al. Clinical significance of promoter hypermethylation of DNA repair genes in tumor and serum DNA in invasive ductal breast carcinoma patients. Life Sci. 2010;87(3–4):83–91.PubMedCrossRef
80.
Zurück zum Zitat Ibanez de Caceres I, et al. Tumor cell-specific BRCA1 and RASSF1A hypermethylation in serum, plasma, and peritoneal fluid from ovarian cancer patients. Cancer Res. 2004;64(18):6476–81.PubMedCrossRef Ibanez de Caceres I, et al. Tumor cell-specific BRCA1 and RASSF1A hypermethylation in serum, plasma, and peritoneal fluid from ovarian cancer patients. Cancer Res. 2004;64(18):6476–81.PubMedCrossRef
82.
Zurück zum Zitat Liggett, T.E., et al., Distinctive DNA methylation patterns of cell-free plasma DNA in women with malignant ovarian tumors. Gynecol Oncol, 2011. 120(1): p. 113–20. Liggett, T.E., et al., Distinctive DNA methylation patterns of cell-free plasma DNA in women with malignant ovarian tumors. Gynecol Oncol, 2011. 120(1): p. 113–20.
83.
Zurück zum Zitat Hulbert A, et al. Early detection of lung cancer using DNA promoter Hypermethylation in plasma and sputum. Clin Cancer Res. 2016; Hulbert A, et al. Early detection of lung cancer using DNA promoter Hypermethylation in plasma and sputum. Clin Cancer Res. 2016;
84.
Zurück zum Zitat Chimonidou M, et al. CST6 promoter methylation in circulating cell-free DNA of breast cancer patients. Clin Biochem. 2013;46(3):235–40.PubMedCrossRef Chimonidou M, et al. CST6 promoter methylation in circulating cell-free DNA of breast cancer patients. Clin Biochem. 2013;46(3):235–40.PubMedCrossRef
85.
Zurück zum Zitat Chen L, et al. Hypermethylated FAM5C and MYLK in serum as diagnosis and pre-warning markers for gastric cancer. Dis Markers. 2012;32(3):195–202.PubMedPubMedCentralCrossRef Chen L, et al. Hypermethylated FAM5C and MYLK in serum as diagnosis and pre-warning markers for gastric cancer. Dis Markers. 2012;32(3):195–202.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Melson J, et al. Commonality and differences of methylation signatures in the plasma of patients with pancreatic cancer and colorectal cancer. Int J Cancer. 2014;134(11):2656–62.PubMedCrossRef Melson J, et al. Commonality and differences of methylation signatures in the plasma of patients with pancreatic cancer and colorectal cancer. Int J Cancer. 2014;134(11):2656–62.PubMedCrossRef
87.
Zurück zum Zitat Tian F, et al. Promoter hypermethylation of tumor suppressor genes in serum as potential biomarker for the diagnosis of nasopharyngeal carcinoma. Cancer Epidemiol. 2013;37(5):708–13.PubMedCrossRef Tian F, et al. Promoter hypermethylation of tumor suppressor genes in serum as potential biomarker for the diagnosis of nasopharyngeal carcinoma. Cancer Epidemiol. 2013;37(5):708–13.PubMedCrossRef
88.
Zurück zum Zitat Powrozek T, et al. Methylation of the DCLK1 promoter region in circulating free DNA and its prognostic value in lung cancer patients. Clin Transl Oncol. 2015; Powrozek T, et al. Methylation of the DCLK1 promoter region in circulating free DNA and its prognostic value in lung cancer patients. Clin Transl Oncol. 2015;
89.
Zurück zum Zitat Powrozek T, et al. Methylation of the DCLK1 promoter region in circulating free DNA and its prognostic value in lung cancer patients. Clin Transl Oncol. 2016;18(4):398–404.PubMedCrossRef Powrozek T, et al. Methylation of the DCLK1 promoter region in circulating free DNA and its prognostic value in lung cancer patients. Clin Transl Oncol. 2016;18(4):398–404.PubMedCrossRef
90.
Zurück zum Zitat Kloten V, et al. Promoter hypermethylation of the tumor-suppressor genes ITIH5, DKK3, and RASSF1A as novel biomarkers for blood-based breast cancer screening. Breast Cancer Res. 2013;15(1):R4.PubMedPubMedCentralCrossRef Kloten V, et al. Promoter hypermethylation of the tumor-suppressor genes ITIH5, DKK3, and RASSF1A as novel biomarkers for blood-based breast cancer screening. Breast Cancer Res. 2013;15(1):R4.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Chiappetta C, et al. Use of a new generation of capillary electrophoresis to quantify circulating free DNA in non-small cell lung cancer. Clin Chim Acta. 2013;425:93–6.PubMedCrossRef Chiappetta C, et al. Use of a new generation of capillary electrophoresis to quantify circulating free DNA in non-small cell lung cancer. Clin Chim Acta. 2013;425:93–6.PubMedCrossRef
92.
Zurück zum Zitat Szpechcinski A, et al. Plasma cell-free DNA levels and integrity in patients with chest radiological findings: NSCLC versus benign lung nodules. Cancer Lett. 2016;374(2):202–7.PubMedCrossRef Szpechcinski A, et al. Plasma cell-free DNA levels and integrity in patients with chest radiological findings: NSCLC versus benign lung nodules. Cancer Lett. 2016;374(2):202–7.PubMedCrossRef
93.
94.
95.
Zurück zum Zitat Papadopoulou E, et al. Cell-free DNA and RNA in plasma as a new molecular marker for prostate and breast cancer. Ann N Y Acad Sci. 2006;1075:235–43.PubMedCrossRef Papadopoulou E, et al. Cell-free DNA and RNA in plasma as a new molecular marker for prostate and breast cancer. Ann N Y Acad Sci. 2006;1075:235–43.PubMedCrossRef
96.
Zurück zum Zitat Dumache R, et al. Prostate cancer molecular detection in plasma samples by glutathione S-transferase P1 (GSTP1) methylation analysis. Clin Lab. 2014;60(5):847–52.PubMed Dumache R, et al. Prostate cancer molecular detection in plasma samples by glutathione S-transferase P1 (GSTP1) methylation analysis. Clin Lab. 2014;60(5):847–52.PubMed
97.
Zurück zum Zitat Minciu R, et al. Molecular diagnostic of prostate cancer from body fluids using methylation-specific PCR (MS-PCR) method. Clin Lab. 2016;62(6):1183–6.PubMed Minciu R, et al. Molecular diagnostic of prostate cancer from body fluids using methylation-specific PCR (MS-PCR) method. Clin Lab. 2016;62(6):1183–6.PubMed
98.
Zurück zum Zitat Cassinotti E, et al. DNA methylation patterns in blood of patients with colorectal cancer and adenomatous colorectal polyps. Int J Cancer. 2012;131(5):1153–7.PubMedCrossRef Cassinotti E, et al. DNA methylation patterns in blood of patients with colorectal cancer and adenomatous colorectal polyps. Int J Cancer. 2012;131(5):1153–7.PubMedCrossRef
99.
Zurück zum Zitat Shan M, et al. Detection of aberrant methylation of a six-gene panel in serum DNA for diagnosis of breast cancer. Oncotarget. 2016;7(14):18485–94.PubMedPubMedCentralCrossRef Shan M, et al. Detection of aberrant methylation of a six-gene panel in serum DNA for diagnosis of breast cancer. Oncotarget. 2016;7(14):18485–94.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Huang G, et al. Evaluation of INK4A promoter methylation using pyrosequencing and circulating cell-free DNA from patients with hepatocellular carcinoma. Clin Chem Lab Med. 2014;52(6):899–909.PubMedPubMedCentralCrossRef Huang G, et al. Evaluation of INK4A promoter methylation using pyrosequencing and circulating cell-free DNA from patients with hepatocellular carcinoma. Clin Chem Lab Med. 2014;52(6):899–909.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Kuo YB, et al. Comparison of KRAS mutation analysis of primary tumors and matched circulating cell-free DNA in plasmas of patients with colorectal cancer. Clin Chim Acta. 2014;433:284–9.PubMedCrossRef Kuo YB, et al. Comparison of KRAS mutation analysis of primary tumors and matched circulating cell-free DNA in plasmas of patients with colorectal cancer. Clin Chim Acta. 2014;433:284–9.PubMedCrossRef
102.
Zurück zum Zitat Spindler KL, et al. Circulating free DNA as biomarker and source for mutation detection in metastatic colorectal cancer. PLoS One. 2015;10(4):e0108247.PubMedPubMedCentralCrossRef Spindler KL, et al. Circulating free DNA as biomarker and source for mutation detection in metastatic colorectal cancer. PLoS One. 2015;10(4):e0108247.PubMedPubMedCentralCrossRef
103.
Zurück zum Zitat Freidin MB, et al. Circulating tumor DNA outperforms circulating tumor cells for KRAS mutation detection in thoracic malignancies. Clin Chem. 2015;61(10):1299–304.PubMedCrossRef Freidin MB, et al. Circulating tumor DNA outperforms circulating tumor cells for KRAS mutation detection in thoracic malignancies. Clin Chem. 2015;61(10):1299–304.PubMedCrossRef
104.
Zurück zum Zitat Sozzi G, et al. Detection of microsatellite alterations in plasma DNA of non-small cell lung cancer patients: a prospect for early diagnosis. Clin Cancer Res. 1999;5(10):2689–92.PubMed Sozzi G, et al. Detection of microsatellite alterations in plasma DNA of non-small cell lung cancer patients: a prospect for early diagnosis. Clin Cancer Res. 1999;5(10):2689–92.PubMed
105.
Zurück zum Zitat Eisenberger CF, et al. The detection of oesophageal adenocarcinoma by serum microsatellite analysis. Eur J Surg Oncol. 2006;32(9):954–60.PubMedCrossRef Eisenberger CF, et al. The detection of oesophageal adenocarcinoma by serum microsatellite analysis. Eur J Surg Oncol. 2006;32(9):954–60.PubMedCrossRef
106.
Zurück zum Zitat Castagnaro A, et al. Microsatellite analysis of induced sputum DNA in patients with lung cancer in heavy smokers and in healthy subjects. Exp Lung Res. 2007;33(6):289–301.PubMedCrossRef Castagnaro A, et al. Microsatellite analysis of induced sputum DNA in patients with lung cancer in heavy smokers and in healthy subjects. Exp Lung Res. 2007;33(6):289–301.PubMedCrossRef
107.
Zurück zum Zitat Andriani F, et al. Detecting lung cancer in plasma with the use of multiple genetic markers. Int J Cancer. 2004;108(1):91–6.PubMedCrossRef Andriani F, et al. Detecting lung cancer in plasma with the use of multiple genetic markers. Int J Cancer. 2004;108(1):91–6.PubMedCrossRef
108.
109.
Zurück zum Zitat Kadam SK, Farmen M, Brandt JT. Quantitative measurement of cell-free plasma DNA and applications for detecting tumor genetic variation and promoter methylation in a clinical setting. J Mol Diagn. 2012;14(4):346–56.PubMedCrossRef Kadam SK, Farmen M, Brandt JT. Quantitative measurement of cell-free plasma DNA and applications for detecting tumor genetic variation and promoter methylation in a clinical setting. J Mol Diagn. 2012;14(4):346–56.PubMedCrossRef
110.
Zurück zum Zitat Zaher ER, et al. Cell-free DNA concentration and integrity as a screening tool for cancer. Indian J Cancer. 2013;50(3):175–83.PubMedCrossRef Zaher ER, et al. Cell-free DNA concentration and integrity as a screening tool for cancer. Indian J Cancer. 2013;50(3):175–83.PubMedCrossRef
111.
Zurück zum Zitat Danese E, et al. Epigenetic alteration: new insights moving from tissue to plasma - the example of PCDH10 promoter methylation in colorectal cancer. Br J Cancer. 2013;109(3):807–13.PubMedPubMedCentralCrossRef Danese E, et al. Epigenetic alteration: new insights moving from tissue to plasma - the example of PCDH10 promoter methylation in colorectal cancer. Br J Cancer. 2013;109(3):807–13.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Skvortsova TE, et al. Cell-free and cell-bound circulating DNA in breast tumours: DNA quantification and analysis of tumour-related gene methylation. Br J Cancer. 2006;94(10):1492–5.PubMedPubMedCentralCrossRef Skvortsova TE, et al. Cell-free and cell-bound circulating DNA in breast tumours: DNA quantification and analysis of tumour-related gene methylation. Br J Cancer. 2006;94(10):1492–5.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Hoque MO, et al. Detection of aberrant methylation of four genes in plasma DNA for the detection of breast cancer. J Clin Oncol. 2006;24(26):4262–9.PubMedCrossRef Hoque MO, et al. Detection of aberrant methylation of four genes in plasma DNA for the detection of breast cancer. J Clin Oncol. 2006;24(26):4262–9.PubMedCrossRef
114.
Zurück zum Zitat Salvianti F, et al. Tumor-related methylated cell-free DNA and circulating tumor cells in melanoma. Front Mol Biosci. 2015;2:76.PubMed Salvianti F, et al. Tumor-related methylated cell-free DNA and circulating tumor cells in melanoma. Front Mol Biosci. 2015;2:76.PubMed
115.
Zurück zum Zitat Rykova EY, et al. Investigation of tumor-derived extracellular DNA in blood of cancer patients by methylation-specific PCR. Nucleosides Nucleotides Nucleic Acids. 2004;23(6–7):855–9.PubMedCrossRef Rykova EY, et al. Investigation of tumor-derived extracellular DNA in blood of cancer patients by methylation-specific PCR. Nucleosides Nucleotides Nucleic Acids. 2004;23(6–7):855–9.PubMedCrossRef
116.
Zurück zum Zitat Mohamed NA, et al. Is serum level of methylated RASSF1A valuable in diagnosing hepatocellular carcinoma in patients with chronic viral hepatitis C? Arab J Gastroenterol. 2012;13(3):111–5.PubMedCrossRef Mohamed NA, et al. Is serum level of methylated RASSF1A valuable in diagnosing hepatocellular carcinoma in patients with chronic viral hepatitis C? Arab J Gastroenterol. 2012;13(3):111–5.PubMedCrossRef
117.
Zurück zum Zitat Zhang YJ, et al. Predicting hepatocellular carcinoma by detection of aberrant promoter methylation in serum DNA. Clin Cancer Res. 2007;13(8):2378–84.PubMedCrossRef Zhang YJ, et al. Predicting hepatocellular carcinoma by detection of aberrant promoter methylation in serum DNA. Clin Cancer Res. 2007;13(8):2378–84.PubMedCrossRef
118.
Zurück zum Zitat de Martino M, et al. Serum cell-free DNA in renal cell carcinoma: a diagnostic and prognostic marker. Cancer. 2012;118(1):82–90.PubMedCrossRef de Martino M, et al. Serum cell-free DNA in renal cell carcinoma: a diagnostic and prognostic marker. Cancer. 2012;118(1):82–90.PubMedCrossRef
119.
Zurück zum Zitat deVos T, et al. Circulating methylated SEPT9 DNA in plasma is a biomarker for colorectal cancer. Clin Chem. 2009;55(7):1337–46.PubMedCrossRef deVos T, et al. Circulating methylated SEPT9 DNA in plasma is a biomarker for colorectal cancer. Clin Chem. 2009;55(7):1337–46.PubMedCrossRef
120.
121.
Zurück zum Zitat Toth K, et al. Detection of methylated septin 9 in tissue and plasma of colorectal patients with neoplasia and the relationship to the amount of circulating cell-free DNA. PLoS One. 2014;9(12):e115415.PubMedPubMedCentralCrossRef Toth K, et al. Detection of methylated septin 9 in tissue and plasma of colorectal patients with neoplasia and the relationship to the amount of circulating cell-free DNA. PLoS One. 2014;9(12):e115415.PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Powrozek T, et al. Septin 9 promoter region methylation in free circulating DNA-potential role in noninvasive diagnosis of lung cancer: preliminary report. Med Oncol. 2014;31(4):917.PubMedPubMedCentralCrossRef Powrozek T, et al. Septin 9 promoter region methylation in free circulating DNA-potential role in noninvasive diagnosis of lung cancer: preliminary report. Med Oncol. 2014;31(4):917.PubMedPubMedCentralCrossRef
123.
Zurück zum Zitat Jin P, et al. Performance of a second-generation methylated SEPT9 test in detecting colorectal neoplasm. J Gastroenterol Hepatol. 2015;30(5):830–3.PubMedCrossRef Jin P, et al. Performance of a second-generation methylated SEPT9 test in detecting colorectal neoplasm. J Gastroenterol Hepatol. 2015;30(5):830–3.PubMedCrossRef
125.
Zurück zum Zitat Kneip C, et al. SHOX2 DNA methylation is a biomarker for the diagnosis of lung cancer in plasma. J Thorac Oncol. 2011;6(10):1632–8.PubMedCrossRef Kneip C, et al. SHOX2 DNA methylation is a biomarker for the diagnosis of lung cancer in plasma. J Thorac Oncol. 2011;6(10):1632–8.PubMedCrossRef
126.
Zurück zum Zitat Weiss G, et al. Validation of the SHOX2/PTGER4 DNA methylation marker panel for plasma-based discrimination between patients with malignant and nonmalignant lung disease. J Thorac Oncol. 2017;12(1):77–84.PubMedPubMedCentralCrossRef Weiss G, et al. Validation of the SHOX2/PTGER4 DNA methylation marker panel for plasma-based discrimination between patients with malignant and nonmalignant lung disease. J Thorac Oncol. 2017;12(1):77–84.PubMedPubMedCentralCrossRef
127.
Zurück zum Zitat Chimonidou M, et al. SOX17 promoter methylation in circulating tumor cells and matched cell-free DNA isolated from plasma of patients with breast cancer. Clin Chem. 2013;59(1):270–9.PubMedCrossRef Chimonidou M, et al. SOX17 promoter methylation in circulating tumor cells and matched cell-free DNA isolated from plasma of patients with breast cancer. Clin Chem. 2013;59(1):270–9.PubMedCrossRef
128.
Zurück zum Zitat Lange CP, et al. Genome-scale discovery of DNA-methylation biomarkers for blood-based detection of colorectal cancer. PLoS One. 2012;7(11):e50266.PubMedPubMedCentralCrossRef Lange CP, et al. Genome-scale discovery of DNA-methylation biomarkers for blood-based detection of colorectal cancer. PLoS One. 2012;7(11):e50266.PubMedPubMedCentralCrossRef
Metadaten
Titel
The evidence base for circulating tumour DNA blood-based biomarkers for the early detection of cancer: a systematic mapping review
verfasst von
Ian A. Cree
Lesley Uttley
Helen Buckley Woods
Hugh Kikuchi
Anne Reiman
Susan Harnan
Becky L. Whiteman
Sian Taylor Philips
Michael Messenger
Angela Cox
Dawn Teare
Orla Sheils
Jacqui Shaw
For the UK Early Cancer Detection Consortium
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2017
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-017-3693-7

Weitere Artikel der Ausgabe 1/2017

BMC Cancer 1/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.