Abstract
Circulating cell‐free DNA (cfDNA) has spurred much interest as a biomarker in oncology. However, inter‐ and intra‐individual cfDNA levels vary greatly. Consequently, in order to base clinical decisions on cfDNA measurements, normal reference intervals are essential to avoid that ordinary variation is confused with clinically relevant change. The lack of reference intervals may potentially explain the ambiguous results reported in the field. Our study aimed to establish reference intervals and to evaluate the association between cfDNA and demographic and clinical variables, including colorectal cancer (CRC). Plasma samples and clinical data from 2817 subjects were collected including 1930 noncancer individuals and 887 CRC patients. cfDNA was measured using droplet digital polymerase chain reaction (PCR). The large cohort combined with robust cfDNA quantification enabled establishment of reference intervals (<67 years: 775‐4860 copies/mL; ≥67 years: 807‐6561 copies/mL). A cfDNA level above the age‐stratified 90% percentile was prognostic of reduced survival in both noncancer individuals and CRC patients, with HR values of 2.56 and 2.01, respectively. Moreover, cfDNA levels increased significantly with age, elevated BMI and chronic diseases. In CRC, the cfDNA level was increased for Stage IV, but not Stage I to Stage III cancer. In summary, the use of reference intervals revealed that high cfDNA levels were predictive of shorter survival in both noncancer individuals and CRC patients, and that CRC development did not affect the cfDNA level until metastatic dissemination. Furthermore, cfDNA levels were impacted by age and chronic diseases. Conclusively, our study presents reference intervals that will help pave the way for clinical utilization of cfDNA.
Keywords: biomarker, circulating cell‐free DNA, colorectal cancer, overall survival, reference intervals
Short abstract
What's new?
Circulating cell‐free DNA (cfDNA) has spurred much interest as a potential biomarker in oncology. However, large inter‐ and intra‐individual variations have led to ambiguous study results and hampered the identification of clinically relevant changes. This study analyzes circulating cfDNA levels in a large cohort of 2817 individuals, with 887 colorectal cancer patients among them. The use of age‐stratified cfDNA reference intervals reveals that aberrant cfDNA levels are prognostic of overall survival in both cancer and non‐cancer individuals, independently of age, gender, and multimorbidity. Altogether, the results could facilitate the interpretation of cfDNA cancer research and clinical use of cfDNA biomarkers.
Abbreviations
- BMI
body‐mass index
- cfDNA
circulating cell‐free DNA
- CI
confidence interval
- CRC
colorectal cancer
- ctDNA
circulating tumor DNA
- ddPCR
droplet digital PCR
- HR
hazard ratio
- IBD
inflammatory bowel disease
- OS
overall survival
- RA
rheumatoid arthritis
1. INTRODUCTION
In the last decade, circulating cell‐free DNA (cfDNA) has been extensively studied as a diagnostic, prognostic and predictive biomarker of somatic disease, especially in oncology. 1 , 2 , 3 , 4 , 5 However, many of the results have been conflicting and ambiguous, and possibly therefore the use of cfDNA has still not been implemented in clinical practice. In order to make clinical decisions based on biological measurements, it is essential to determine a reference interval so that clinically relevant, pathological changes are not confused with ordinary variation.
The cfDNA level is an equilibrium of cfDNA release/clearance and plasma volume. The major cell types contributing to the cfDNA pool are white blood cells, erythrocyte progenitors, endothelial cells and hepatocytes. 6 , 7 In metastatic cancer, the malignant cells may also provide a substantial contribution. 8 The cfDNA concentration may vary considerably between individuals 9 , 10 and moreover, intraindividual variance has been reported during the day 11 , 12 and with physical activity. 13 , 14 cfDNA levels have further been shown to increase significantly with age, 9 , 15 , 16 but whether this is due to the accumulation of age‐related chronic diseases remains unknown. The establishment of cfDNA reference intervals in a robust cohort with a standardized analytical pipeline is thus necessary to forward cfDNA biomarker research and facilitate the implementation in daily clinical routine.
The present study aimed to establish reference intervals for cfDNA levels in healthy elderly individuals. It further investigated the association between cfDNA levels and age, demographic factors, chronic diseases and colorectal cancer (CRC) characteristics. Finally, it analyzed whether aberrantly high cfDNA levels were prognostic for decreased overall survival (OS) in both noncancer individuals and patients with CRC.
2. MATERIALS AND METHODS
2.1. Subjects
Research subjects were recruited in the period 2006 to 2019 from Danish surgical centers at Bispebjerg, Herlev, Herning, Hillerød, Horsens, Hvidovre, Randers, Aarhus and Viborg (flowchart of inclusion in Supplementary Figure 1). They were analyzed for cfDNA in various in‐house studies, 2 , 17 , 18 , 19 , 20 , 21 and further included in the present study in a cross‐sectional design. Subjects comprised both noncancer individuals and patients with CRC. The noncancer individuals were recruited through the population‐based Danish National CRC Screening Program, offered to all Danes between 50 and 75 years of age, and from subjects referred to colonoscopy due to CRC‐related bowel symptoms. Blood was collected after bowel preparation, but prior to colonoscopy. If the noncancer individuals did not undergo colonoscopy, blood was collected at the subject's convenience. The patients with CRC were recruited at diagnosis and blood was collected prior to operation. Inclusion criteria were age >18; exclusion criteria were acute illness, pregnancy and a diagnosis of cancer other than CRC <3 years before blood collection. Furthermore, all included CRC patients were treatment naïve, that is, patients treated with neoadjuvant chemotherapy or radiation therapy prior to blood sampling were excluded. The study was conducted in accordance with the Helsinki Declaration. All subjects provided written informed consent. The use of biological material and clinical data was approved by the National Committee on Health Research Ethics and the Danish National Data Protection Agency.
2.2. Clinical data collection
Data were collected from inclusion questionnaires, electronic patient files, the Danish Colorectal Cancer Group database 22 and the national Danish Personal Registry. 23 Diseases were grouped in categories. “Heart disease” included previous acute myocardial infarction, angina, chronic ischemic heart disease, atrial fibrillation/flutter, valvular heart disease, heart failure and conductive disorders. “Diabetes” included both Type 1 and Type 2. “Inflammatory bowel disease” (IBD) included ulcerative colitis and Crohn's disease. “Pulmonary disease” included chronic asthma, chronic emphysema and chronic bronchitis. Also, diagnoses of hypertension and rheumatoid arthritis (RA) were registered.
2.3. Isolation of cfDNA
In accordance with a prestudy defined standard operating procedure, whole blood was collected in BD Vacutainer K2 EDTA tubes (Becton Dickinson) and processed within 2 hours from venipuncture. The plasma was isolated by double centrifugation at 3000g for 10 minutes at 20°C, transferred to cryotubes (TTP) and stored at −80°C. Blood from CRC patients and noncancer individuals was processed identically. The plasma was transferred from local hospitals to a central freezer facility for storage until cfDNA extraction. cfDNA extraction was performed at a single facility: in brief, 4 to 8 mL of plasma was thawed at room temperature and cfDNA was extracted fusing a QIAsymphony robot (Qiagen) using the QIAsymphony DSP Circulating DNA kit (Cat No./ID: 937556, Qiagen) according to the manufacturer's instructions. Purified cfDNA was eluted in LoBind tubes or LoBind 96‐well plates (Eppendorf AG) and stored at −80°C until analysis. The time from plasma storage to cfDNA quantification was recorded as “freeze time.”
2.4. Droplet digital PCR, quality control and cfDNA quantification
Droplet digital PCR (ddPCR) was used for cfDNA quantification and quality control of the cfDNA extraction procedure throughout the study. ddPCR was conducted in accordance with the dMIQE guidelines. 24 All ddPCR analyses were performed at the Department of Molecular Medicine, Aarhus University Hospital, using the standard operating procedure, defined prior to the initiation of the first study. The reaction mix, containing all reagents (18 pmol forward primer, 18 pmol reverse primer, 5 pmol probe, 2xSupermix for Probes no UTP [Bio‐Rad]) except template DNA, was prepared in an isolated pre‐PCR room to avoid contamination. Just before droplet generation, 2 μL template DNA was added to each reaction, bringing the final volume to 22 μL. One‐nanoliter droplets were generated on the QX200 AutoDG Droplet Generator (Bio‐Rad), using the procedure described by the manufacturer. After droplet generation, samples were amplified by PCR in an S1000 Thermal Cycler (Bio‐Rad) at 95°C for 10 minutes and 45 cycles of 95°C for 30 seconds, 56°C for 1 minute and 98°C for 10 minutes and analyzed on a QX200 reader (Bio‐Rad). All data was Poisson corrected. Positive and no‐template controls were included for each assay and each batch.
To estimate cfDNA purification efficiency and assess the amount of DNA contamination from lysed lymphocytes in each sample, a duplex ddPCR assay was used as previously described. 21 In brief, a fixed amount of soybean CPP1 DNA fragments was added to each plasma sample before cfDNA extraction, and thereafter the purification efficiency was calculated as the percent recovery of CPP1 fragments. Lymphocyte DNA contamination from peripheral blood cell lysis was estimated by an assay targeting the VDJ rearranged IGH locus specific for B cells. 25 Samples with lymphocyte DNA contamination were excluded. The duplex assay details are given in Supplementary Table 1.
To estimate the cfDNA concentration, the number of cfDNA templates in plasma was quantified, using a ddPCR assay targeting a reference region on Chromosome 3. This region rarely shows copy number variation in CRC. 26 Primer and probe sequences for this assay are likewise provided in Supplementary Table 1.
2.5. Statistical methods
Descriptive statistics for non‐Gaussian distributed data were reported as medians with 10% and 90% percentiles, and for Gaussian distributions as mean with 95% confidence interval (CI). In linear regression analyses, non‐Gaussian distributions were natural log transformed to achieve a log‐Gaussian distribution and the statistical significance was assessed using t‐tests. Of potential biases, bowel preparation has been found to have a minor systematic effect on the plasma cfDNA level 27 and freeze time, defined as the time from venipuncture until cfDNA extraction and quantification, has been suggested to decrease cfDNA concentration. 28 Furthermore, the cfDNA analyses were conducted in batches, in relation to in‐house studies, in the period 2016 to 2019. Accordingly, these potential biases were adjusted for in all regression analyses.
Establishment of reference intervals for cfDNA was done a posteriori following guidelines from the Clinical and Laboratory Standards Institute. 29 The preanalytical conditions were evaluated, and the workable subject preparation as well as the strict regulations for specimen collection were standardized as previously described. 30 Reference intervals were made on log‐Gaussian data. No outliers were identified with Dixon's method. 31 The reference limits were defined as the 2.5% and 97.5% percentiles and the 95% CI values of the upper/lower limits were calculated.
Follow‐up time and OS were calculated from the time of blood collection to the time of death, other censoring or end of follow‐up (1 March 2019). The associations between survival and independent variables were analyzed by Cox regressions. For all statistical analyses, a two‐sided P value below .05 was considered significant. All assumptions for the statistical models were fulfilled. For statistical analyses, STATA V.12.1 (StataCorp LP, TX) was used.
3. RESULTS
A total of 2820 subjects were included in the cohort: 1931 noncancer individuals without cancer at time of blood sampling, and 889 patients with CRC. All CRC patients underwent colonoscopy as part of the diagnostic work‐up. Of the noncancer individuals, 1056 underwent colonoscopy. Of these, 838 had a “clean colon” and 218 had adenomas. The remaining 875 noncancer individuals were all screening participants, who tested negative, and therefore did not undergo colonoscopy. Blood samples were collected from all individuals and the plasma cfDNA level determined by ddPCR. During experimental analysis, three samples were excluded (3/2820, 0.1%): two due to cfDNA measurement failure and one due to lymphocyte DNA contamination, leaving 1930 noncancer individuals and 887 patients with CRC for analyses. The median freeze time for a plasma sample was 1060 days, ranging from 66 to 3971 days. Freeze time of ≥5 years was associated with a decrease in cfDNA concentration and was thus included as a variable in all subsequent regression analyses (Supplementary Figure 2). The mean age was 66 years and 57.9% were males (Supplementary Table 2). The median follow‐up time was 1457 days (437‐4768 days). During follow‐up, 259 individuals died (9.2%) and 5 were censored due to migration to other countries (0.2%). For 2409 of the included individuals, information about chronic diseases was available: 1419 had no diseases, and 990 had one or more diseases.
3.1. Establishment of cfDNA reference intervals
The median cfDNA level of all 1930 noncancer individuals was 2051 copies/mL plasma and increased significantly with increasing age binned by decades (Figure 1A). A reference sample group was defined, consisting of the subset of noncancer individuals without chronic disease, who did not undergo colonoscopy. The latter because the colonoscopy bowel preparation was shown to cause a minor (7.7%) increase in the cfDNA level (Supplementary Table 3). The reference sample group consisted of 501 individuals (“reference individuals”) and to estimate the impact of age on cfDNA levels in the reference individuals, the cfDNA levels were displayed in small intervals (Figure 1B). An increase in the median cfDNA level was revealed at age 67 and above, indicating a need for age‐stratified reference intervals. Accordingly, the reference group was divided into two subgroups: below 67 years and 67 years or above. The age‐stratified cfDNA levels in the two groups were significantly different (median 1839 and 2252 copies/mL, respectively, P < .001 in t‐test) and the fraction of individuals in each group that were outside the total reference interval (2.5% under 792 copies per mL for age < 67 years and 4.8% over 5477 copies per mL for age ≥ 67) was above the 4.1%, at which it is recommended to partition the reference group. 32 Consequently, age‐stratified reference intervals were established for individuals <67 and ≥67 years (Table 1). These were 775 to 4860 copies/mL and 807 to 6561 copies/mL, respectively.
FIGURE 1.

cfDNA levels in noncancer individuals. A, Violin plots showing the distribution of cfDNA levels, stratified by 10‐year intervals, in all individuals. B, Box plots illustrating the distribution of cfDNA levels in all reference individuals in 3‐year intervals. Between “64‐66” and “67‐69” years, the median increased, and therefore 67 was chosen as cut‐off for the age‐dependent substratification of reference intervals. C, Violin plots showing cfDNA levels for noncancer individuals stratified by chronic diseases number. Age stratified values presented in Table 2. D, Violin plots showing cfDNA levels in noncancer individuals by disease group. IBD, inflammatory bowel disease. Box plot shows the median (line) and the interquartile range (box), while violins represent the distribution of cfDNA. Box plot whiskers represent the minimum and maximum values. Differences in cfDNA levels were tested with t‐test, P value <.05 considered statistically significant
TABLE 1.
cfDNA reference intervals stratified by age
| n | Reference interval, cfDNA copies/mL plasma a | Lower limit 95% CI, cfDNA copies/mL plasma | Upper limit 95% CI, cfDNA copies/mL plasma | P value | cfDNA copies/mL plasma median (10%;90%) | |
|---|---|---|---|---|---|---|
| All individuals | 501 | 792 to 5477 | 707 to 886 | 4837 to 6620 | 1971 (1122;3712) | |
| By age | ||||||
| <67 | 317 | 775 to 4860 | 693 to 883 | 4070 to 6552 | — | 1839 (1047;3506) |
| ≥67 | 184 | 807 to 6561 | 608 to 985 | 5279 to 10406 | <.001 | 2252 (1294;3943) |
Abbreviations: CI, confidence interval; n, number; cfDNA, circulating cell‐free DNA; 10%;90%, percentiles.
According to Reference 32, subgroups should be made if >4,1% of the values lie outside the upper limit of the common reference interval. 4.8% of the group ≥67 years had values outside this. Cut‐off age estimated from Figure 1B. Difference in cfDNA level between groups estimated by using t‐test.
3.2. Associations between cfDNA and demographic variables and chronic diseases
Elevated cfDNA levels were significantly associated with age and increasing BMI (P < .001), while there was a tendency toward an association between increased cfDNA levels and male gender (P = .082). Smoking was associated with significantly decreased cfDNA levels (Supplementary Table 4). An association between cfDNA levels and alcohol consumption could not be shown. When compared to the reference individuals, the cfDNA levels in individuals with chronic disease were significantly elevated. Stratification for number (Figure 1C) and type of chronic disease (Figure 1D) revealed a significantly increasing cfDNA level with an accumulating number of chronic diseases and intertype variation. Univariate regression analyses confirmed these findings (Supplementary Table 4).
All significant variables were combined in a multivariable regression analysis (Table 2). Increased age and BMI remained significantly associated with higher cfDNA levels, P < .001. Likewise, the association between smoking and reduced cfDNA levels remained. Oppositely, the weaker association with male gender disappeared completely. For chronic diseases, “one disease” and “three or more diseases” remained significantly associated with higher cfDNA levels.
TABLE 2.
Multivariable regression of the association between cfDNA levels and age for noncancer individuals
| Multivariable linear regression | Regression coefficient a | P value |
|---|---|---|
| Age (continuous) | 0.01 | <.001 |
| Gender | ||
| Female | — | — |
| Male | 0.004 | .862 |
| BMI (continuous) | 0.03 | <.001 |
| Smoking | ||
| Nonsmoker | — | — |
| Smoker | −0.13 | <.001 |
| Former smoker b | −0.01 | .706 |
| Number of chronic diseases | ||
| No disease | — | — |
| One disease | 0.08 | .006 |
| Two diseases | 0.03 | .443 |
| Three or more diseases | 0.14 | .023 |
Data log transformed to achieve Gaussian distribution. Adjusted for bowel preparation and freeze time.
Former smoker: nonsmoker for >6 months; cfDNA: circulating cell‐free DNA, BMI: bodymass index.
3.3. Associations between cfDNA and tumor characteristics in patients with CRC
To investigate if characteristics of adenomas and CRCs were associated with increased cfDNA levels, data from 887 CRC patients and 218 individuals with adenomas were analyzed (Figure 2, Table 3). Reference individuals and individuals with adenomas had comparable cfDNA levels. For CRC patients, the cfDNA level was significantly higher, but when stratified for stage and age, the difference was found to be driven by Stage IV patients (Table 3). The cfDNA levels in plasma from patients with Stages I to III were similar to that of reference individuals. The association between cfDNA and other histopathological characteristics was also evaluated: cfDNA levels increased with increasing tumor size (P = .028), but no other significant associations were found. cfDNA levels for CRC patients with comorbidities are shown in Supplementary Table 5.
FIGURE 2.

cfDNA levels during CRC development. Box plot showing the cfDNA levels through CRC development in reference individuals, adenomas and CRC stages I to IV. Box plot illustrates the median (line) and the interquartile range (box). Whiskers represent the minimum and maximum values. Differences in cfDNA levels tested with t‐test, P value <.05 considered statistically significant. CRC, colorectal cancer; N.S., nonsignificant
TABLE 3.
Univariable associations between cfDNA levels, adenomas and CRC characteristics
| Univariable linear regression | n (%) | cfDNA copies per mL plasma a median (10%;90%) | Regression coefficient | P value |
|---|---|---|---|---|
| Adenomas and CRC | ||||
| Reference individuals | 501 | 1971 (1122;3712) | — | — |
| Adenomas | 218 | 2113 (1080;4240) | 0.06 | .291 |
| All CRC stages | 887 (100) | 2185 (1024;5750) | 0.12 | .015 |
| Age (continuous) b | 0.01 | <.001 | ||
| CRC stage | ||||
| Reference individuals | — | — | ||
| Stage I | 201 (22.7) | 2120 (1033;4537) | 0.09 | .137 |
| Stage II | 327 (36.9) | 2282 (990;5096) | 0.09 | .179 |
| Stage III | 302 (34.1) | 2123 (1005;5457) | 0.08 | .207 |
| Stage IV | 57 (6.4) | 3036 (1336; 31592) | 0.78 | <.001 |
| Test for trend, cfDNA increase across stages | <.001 | |||
| Tumor (pT stage) | ||||
| T1 | 124 (14.0) | 2066 (932;4826) | — | — |
| T2 | 103 (11.6) | 2211 (1140;4746) | 0.03 | .741 |
| T3 | 494 (56.0) | 2128 (948;4950) | −0.03 | .697 |
| T4 | 140 (15.8) | 2339 (1116;7618) | 0.16 | .098 |
| Missing | 26 (2.9) | |||
| Lymph node (pN stage) | ||||
| N0 | 371 (41.8) | 2153 (983;5002) | — | — |
| N1 | 198 (22.3) | 2217 (924;6089) | 0.03 | .621 |
| N2 | 126 (14.2) | 2058 (1037;6135) | −0.01 | .998 |
| Missing | 192 (21.6) | |||
| Metastasis (pM stage) | ||||
| M0 | 830 (93.6) | 2158 (1011;5049) | — | — |
| M1 | 57 (6.4) | 3036 (1336;31 592) | 0.67 | <.001 |
| Tumor location b | ||||
| Anus and rectum | 181 (20.4) | 2125 (902;4281) | — | — |
| Left colon c | 361 (40.7) | 2185 (967;5897) | 0.09 | .210 |
| Right colon d | 325 (36.6) | 2244 (1150;6162) | 0.10 | .162 |
| Missing | 20 (2.3) | |||
| Histological type b | ||||
| AC | 624 (70.4) | 2304 (1130;5750) | — | — |
| Mucinous AC | 43 (4.9) | 2260 (1278;8168) | 0.15 | .150 |
| Neuroendocrine AC | 5 (0.6) | — | — | — |
| Signet ring cell AC | 3 (0.3) | — | — | — |
| Missing | 212 (23.9) | |||
| Tumor differentiation b | ||||
| High | 3 (0.3) | 1757 (1358;2458) | — | — |
| Medium | 318 (35.9) | 2396 (1114;5997) | 0.22 | .606 |
| Low | 73 (8.2) | 1996 (1190;8002) | 0.27 | .530 |
| Missing | 493 (55.6) | |||
| Tumor diameter mm b | ||||
| <25 | 39 (4.4) | 1980 (1202;8002) | ||
| 25 to 50 | 221 (24.8) | 2185 (878;6270) | ||
| 50 to 75 | 168 (18.9) | 2283 (1114;4950) | ||
| 75 to 100 | 64 (7.2) | 2437 (1084;8688) | ||
| >100 | 36 (4.1) | 2654 (1358;7728) | ||
| Missing | 360 (40.6) | |||
| Tumor size (continuous) | 0.03 | .028 | ||
| Perineural growth b | ||||
| No | 281 (31.7) | 2359 (1089;6510) | — | — |
| Yes | 160 (18.0) | 2287 (1195;6817) | −0.05 | .511 |
| Missing | 446 (50.3) | |||
| Satellites b | ||||
| No | 370 (41.7) | 2354 (1207;6603) | — | — |
| Yes | 60 (6.8) | 2491 (1359;7700) | 0.04 | .719 |
| Missing | 457 (51.5) | |||
| Venous invasion b | ||||
| No | 320 (36.1) | 2234 (1153;5730) | — | — |
| Yes | 135 (15.2) | 2557 (1176;8893) | 0.16 | .066 |
| Missing | 432 (48.7) | |||
| Tumor budding b | ||||
| No | 345 (38.9) | 2350 (1217;6837) | — | — |
| Yes | 68 (7.7) | 2605 (1105;7508) | 0.07 | .523 |
| Missing | 474 (53.4) |
Abbreviations: AC, adenocarcinoma; cfDNA, circulating cell‐free DNA; CRC, colorectal cancer; n, number; %, percentage; 10%;90%, percentiles.
Data log transformed to achieve Gaussian distribution. All linear regressions adjusted for study origin and freeze time.
Adjusted for CRC stage.
Sigmoid, descending, splenic flexure.
Transverse, hepatic flexure, ascending, coecum.
3.4. Associations between cfDNA and OS
To evaluate if high cfDNA levels were associated with decreased OS, an “aberrant cfDNA limit” was defined as the age‐stratified 90% percentile for reference individuals (Table 1). Using this cut‐off, the noncancer individuals and patients with CRC were separated into two groups: normal or aberrant cfDNA level (below or above the limit, respectively). Cox regression analyses were used to assess the association between the two groups and OS. The analyses were adjusted for variables expected to influence OS or cfDNA levels, that is, age, gender, BMI, smoking status, alcohol consumption, chronic diseases and CRC stage (Table 4). For both noncancer individuals and CRC patients, aberrantly high cfDNA levels were significantly associated with decreased OS (HR 2.56 and 2.01, respectively). In noncancer individuals, the following variables were also associated with decreased OS: higher age, smoking and ≥3 chronic diseases. Alcohol consumption >21 units/week was borderline associated with a decreased OS (P = .051). In CRC patients, high age and Stages III and IV disease were associated with decreased OS. A trend toward shorter OS was also observed for smoking. Furthermore, a higher BMI was borderline associated with longer OS (P = .051).
TABLE 4.
Associations between cfDNA levels and OS
| Noncancer individuals | Patients with CRC | |||||
|---|---|---|---|---|---|---|
| Multivariable Cox regression, survival analysis | HR | 95% CI | P value | HR | 95% CI | P value |
| Aberrant cfDNA level a | ||||||
| No | — | — | — | — | — | — |
| Yes | 2.56 | 1.37 to 4.78 | .003 | 2.01 | 1.17 to 3.43 | .011 |
| Age | 1.13 | 1.09 to 1.17 | <.001 | 1.05 | 1.03 to 1.08 | <.001 |
| Male gender | 1.45 | 0.83 to 2.50 | .182 | 0.98 | 0.61 to 1.60 | .942 |
| Smoking | ||||||
| Nonsmoker | — | — | — | — | — | — |
| Smoker | 2.41 | 1.33 to 4.37 | .004 | 1.66 | 0.92 to 3.00 | .090 |
| Former smoker b | 1.56 | 0.78 to 3.09 | .207 | 1.24 | 0.75 to 2.04 | .401 |
| BMI (continuous) | 0.94 | 0.88 to 1.01 | .095 | 0.96 | 0.92 to 1.00 | .051 |
| Alcohol consumption | ||||||
| 0 units/week | 1.44 | 0.69 to 2.97 | .345 | 1.24 | 0.73 to 2.10 | .427 |
| 1 to 7 units/week | — | — | — | — | — | — |
| 8 to 14 units/week | 0.74 | 0.36 to 1.53 | .413 | 0.83 | 0.41 to 1.69 | .614 |
| 15 to 21 units/week | 0.80 | 0.32 to 1.97 | .622 | 1.93 | 0.93 to 3.98 | .074 |
| >21 units/week | 2.23 | 1.00 to 5.00 | .051 | 1.14 | 0.49 to 2.61 | .765 |
| Number of chronic diseases | ||||||
| No disease | — | — | — | — | — | — |
| One disease | 1.67 | 0.86 to 3.23 | .130 | 0.89 | 0.42 to 1.86 | .744 |
| Two diseases | 1.53 | 0.60 to 3.92 | .395 | 1.74 | 0.82 to 3.72 | .151 |
| Three or more diseases | 2.87 | 1.11 to 7.41 | .029 | 1.44 | 0.31 to 6.63 | .636 |
| CRC stage | ||||||
| Stage I | — | — | — | — | — | — |
| Stage II | — | — | — | 0.87 | 0.42 to 1.82 | .727 |
| Stage III | — | — | — | 2.13 | 1.03 to 4.39 | .040 |
| Stage IV | — | — | — | 35.0 | 17.2 to 71.2 | <.001 |
Aberrant cfDNA limit: >90% percentile for age‐stratified cfDNA reference level (Table 1). Adjusted for bowel preparation, study origin and freeze time.
Former smoker defined as nonsmoker >6 months; BMI, bodymass index; CI, confidence interval; cfDNA, circulating cell‐free DNA; CRC, colorectal cancer; HR, hazard ratio.
4. DISCUSSION
Clinical benefit of cfDNA as a cancer biomarker has not yet been documented, possibly due to the lack of standardized preanalytical conditions and relevant reference intervals. In our study, age‐stratified reference intervals for cfDNA levels in a population of healthy individuals were defined. The results showed that higher age, chronic diseases, and disseminated CRC were independently associated with increased cfDNA levels. Furthermore, the increase in cfDNA levels for several clinically important chronic diseases was quantified, and it was shown that aberrantly high cfDNA levels were an independent prognostic marker for shorter OS in both noncancer individuals and CRC patients.
To establish cfDNA reference values, a group of 501 healthy individuals, comprising both genders, were chosen from a population aged 50 to 80 years. Previously, it has been common practise to use, for example, healthy young males. However, such a reference group is irrelevant in most oncological settings, where the population is commonly above 50 years and of both sexes. The cfDNA measurement here was made without extensive subject preparation criteria (eg, time of day for blood collection or physical activity restriction, all of which have been shown to affect the cfDNA level 11 , 12 , 13 , 14 ) and consequently the cfDNA levels may vary more than that if strict preanalytical criteria had been applied. However, this mirrors the clinical reality well, and when the results were controlled for potential outliers, the variance of the reference values was within the recommended limits. 29 Although reference intervals are often stratified by gender, it was not done here. The present data showed no difference between genders after adjusting for smoking status, alcohol consumption and chronic diseases in the multivariable regression analysis (Table 2).
Instead, the reference intervals were stratified by age. In agreement with previous publications, 9 , 15 , 16 the present data showed that cfDNA levels increased with higher age for both noncancer individuals (Table 2) and also for patients with CRC (Table 3). Furthermore, stratifications indicated that the association was neither confounded by gender, BMI, smoking or chronic disease for noncancer individuals nor by tumor stage for patients with CRC. Why cfDNA levels increase with age is not completely understood. Older age has previously been shown to be related to increasing inflammation, so‐called “inflammaging.” 33 As white blood cells, contributing greatly to the cfDNA pool, are involved in inflammaging, their increased turnover may be part of the explanation. In contrast to the present study, a recent meta‐analysis of 892 healthy controls, pooled from eight different studies, found no significant association between cfDNA levels and age. 10 However, the included studies used different analytical methodologies for cfDNA quantification, which complicate the integration and interpretation of the result. 28 As recommended in recently published guidelines for cfDNA analysis, 34 the cfDNA levels reported in the present study were produced by absolute quantification by ddPCR.
Several small‐scale studies have shown an association between acute diseases and increased cfDNA levels. 35 , 36 , 37 , 38 , 39 The present study showed that chronic diseases, independently of age, were associated with higher cfDNA levels as well. Although cfDNA levels are only transiently affected by acute disease, they may be permanently affected by chronic disease. The results further indicated that the effect of chronic diseases on the cfDNA level was cumulative, as the level increased with an increasing number of diseases. This suggests that cfDNA from all pathologically affected tissues accumulates in the blood. The association between cfDNA levels and, for example, cardiovascular disease could be confounded by gender, smoking status or alcohol consumption. Likewise, the association with, for example, diabetes could be confounded by higher BMI, which increases inflammation and cfDNA levels. 40 , 41 Owing to the large cohort size, it was possible to adjust for this in a multivariable regression: multimorbidity remained significantly associated with higher cfDNA levels. Moreover, the results showed that smoking was associated with decreased cfDNA levels. This association has not been described previously, and the underlying mechanism is unclear. No significant association between alcohol consumption and cfDNA levels was observed in the present data, though excessive alcohol intake may cause liver cell damage. These unexpected results could relate to the fact that the smoke/alcohol variables were self‐reported, and such data can be heavily biased by the lack of memory or by misreporting due to fear of stigmatization (social desirability bias).
Previous research has shown that cancer cells contribute to the cfDNA pool, especially in disseminated disease, 8 and that cancer patients may have higher cfDNA levels compared to healthy individuals. 10 The present study included patients with CRC in all stages, as well as patients with adenomas. Comparable levels of cfDNA were observed in healthy individuals and individuals with adenomas, which is in agreement with findings from previous studies. 15 , 42 Furthermore, comparable cfDNA levels in healthy individuals and Stages I to III CRC were found; only patients with disseminated disease (Stage IV) had significantly increased cfDNA levels. This finding was supported by other previous reports. 9 , 43 Tumor characteristics associated with high cfDNA levels also included large tumor size. It has previously been shown that increased circulating tumor DNA (ctDNA) levels were associated with a tumor size above 5 cm. 43 This corroborates the present findings, as ctDNA is part of the total cfDNA quantity. Taken together, these findings indicate that CRC generally does not affect the overall cfDNA level until late in development.
To determine whether increased cfDNA levels were prognostic of OS, an “aberrant cfDNA limit” was defined prior to data analysis. The analysis revealed that aberrant cfDNA levels were associated with decreased OS independently of age, smoking, alcohol consumption, chronic diseases and CRC stage (Table 4). The significance of the result was emphasized as aberrant cfDNA levels were prognostic in both noncancer individuals and CRC patients. The association between elevated cfDNA levels and OS in CRC has been reported in previous studies including disseminated CRC 44 and CRC in all stages. 42 , 45 For noncancer individuals, increased cfDNA levels have been reported to be associated with decreased OS in individuals with acute disease. 39 , 46 Furthermore, it has been suggested that increasing cfDNA levels are a proxy marker for frailty in elderly. 47 , 48 Therefore, we hypothesize that aberrant cfDNA levels may be used as a proxy marker for overall health (regardless of CRC diagnosis) and that cfDNA levels possibly could be integrated in artificial intelligence algorithms, predicting patient outcome in future personalized oncological strategies. Whether the 90% percentile is the best clinical decision limits for the “aberrant cfDNA level” must be warranted in future studies.
The present study has some limitations. Firstly, though the Danish registries and databases are highly accurate and valid, 22 , 23 subjects registered as healthy may still have nondiagnosed chronic diseases. The risk of acute disease during blood collection is however not an issue, as acutely ill subjects were not included in the study. Secondly, even though subjects with a cancer diagnosis <3 years from blood collection were excluded, some of the participants may have occult recurrence of a former cancer or new, undiagnosed metachronous cancer. Thirdly, information on the chronic diseases treatment in the study group was not available. Hypothetically, well‐regulated RA or IBD would cause less inflammation compared to dysregulated RA/IBD, which would impact the release of cfDNA from white blood cells. Overall, these limitations might increase the cfDNA reference intervals variance. However, we find this acceptable, as it reflects the clinical reality.
Conclusively, as numerous factors clearly affect cfDNA levels, cfDNA research results should be carefully interpreted. Our results underline that especially higher age and the presence of chronic diseases are associated with increased cfDNA levels, which could influence cfDNA research interpretation. For example, when results are presented as varying allele frequencies (ctDNA in the numerator, overall cfDNA level in the denominator), then interindividual comparison would be greatly influenced by, for example, differences in age. Another important finding is that aberrant cfDNA levels are prognostic for decreased OS for both noncancer and CRC individuals. Finally, by reporting reference intervals for cfDNA levels, we hope to unlock the clinical potential of cfDNA as a biomarker, and we foresee that cfDNA could be incorporated into future predictive oncological stratification tools and prognostic artificial intelligence algorithms.
CONFLICT OF INTEREST
The authors declare that they have no competing interests.
ETHICS STATEMENT
This study was conducted in accordance with the Helsinki Declaration. All subjects provided written informed consent. The use of biological material and clinical data was approved by the National Committee on Health Research Ethics and the Danish National Data Protection Agency.
Supporting information
Appendix S1: Supplementary Information
ACKNOWLEDGEMENTS
We thank the patients and healthy individuals for their participation. The Danish Cancer Biobank is acknowledged for providing access to blood materials. We thank Søren Laurberg, Lene Hjerrild Iversen, Frank Viborg Mortensen, Kåre Andersson Gotschalck and “The Danish Collaborative Research Group on Early Detection of Colorectal Cancer” for their work with subject recruitment, blood collection and data recording. The Danish Collaborative Research Group on Early Detection of Colorectal Cancer includes the following: Lars Nannestad Jørgensen and Morten Rasmussen (Bispebjerg Hospital, Copenhagen), Jakob W. Hendel (Herlev Hospital, Herlev), Mogens R. Madsen and Anders H. Madsen (Herning Hospital, Herning), Jesper Vilandt and Thore Hillig (Hillerød Hospital, Hillerød), Søren Brandsborg (Horsens Hospital, Horsens), Linnea Ferm and Eva Rømer (Hvidovre Hospital, Hvidovre), Tobias Boest (Randers Hospital, Randers) and Ali Khalid (Viborg Hospital, Viborg).
CLA received Grant No. R133‐A8520‐00‐S41, R146‐A9466‐16‐S2, R231‐A13845 and R257‐A14700 from the Danish Cancer Society, Grant No. 1309‐00006B from the Danish Council for Strategic Research, Grant No. 4183‐00619 from the Danish Council for Independent Research, Medical Sciences and Grant No. NNF14OC0012747 and NNF17OC0025052 from the Novo Nordisk Foundation.
NØ received a grant from The Neye Foundation and from Dansk Kræftforskningsfond.
SØJ received a grant from Dansk Kræftforskningsfond.
HJN received grants from The Augustinus Foundation, The Kornerup Fund, The Axel Muusfeldt Fund, The KID Fund, The Toyota Fund, The Aage and Johanne Louis Hansen Fund, The Orient Fund, The Walter and O. Kristiane Christensen Fund, The A.P. Moeller and Chastine Mc‐Kinney Moeller Foundation, The P.M. Christiansen Family Fund, The Aase and Ejnar Danielsen Fund, The Inger Bonnén Fund, The Hans and Nora Buchard Fund, The Elna and Jørgen Fagerholdt Fund, The Sofus C.E. Friis Fund, The Eva and Henry Fraenkel Fund, The Sven and Ina Hansen Fund, The Henrik Henriksen Fund, The Jørgen Holm Fund, The Humanitarian Fund, The IMK Fund, Foundation Jochum, The Obel Family Fund and Kristaand Viggo Petersen Fund.
Ørntoft M‐BW, Jensen SØ, Øgaard N, et al. Age‐stratified reference intervals unlock the clinical potential of circulating cell‐free DNA as a biomarker of poor outcome for healthy individuals and patients with colorectal cancer. Int. J. Cancer. 2021;148:1665–1675. 10.1002/ijc.33434
Mai‐Britt Worm Ørntoft and Sarah Østrup Jensen contributed equally to the manuscript
†Deceased.
Funding information Kristaand Viggo Petersen Fund; The Obel Family Fund; Foundation Jochum; The IMK Fund; The Humanitarian Fund; The Jørgen Holm Fund; The Henrik Henriksen Fund; The Sven and Ina Hansen Fund; The Eva and Henry Fraenkel Fund; The Sofus C.E. Friis Fund; The Elna and Jørgen Fagerholdt Fund; The Hans and Nora Buchard Fund; The Inger Bonnén Fund; The Aase and Ejnar Danielsen Fund; The P.M. Christiansen Family Fund; The A.P. Moeller and Chastine Mc‐Kinney Moeller Foundation; The Walter and O. Kristiane Christensen Fund; The Orient Fund; The Aage and Johanne LouisHansen Fund; The Toyota Fund; The KID Fund; The Axel Muusfeldt Fund; The Kornerup Fund; Augustinus Foundation; Dansk Kræftforskningsfond; The Neye Foundation; Novo Nordisk Foundation, Grant/Award Numbers: NNF17OC0025052, NNF14OC0012747; Danish Council for Independent Research, Medical Sciences, Grant/Award Number: 4183‐00619; Danish Council for Strategic Research, Grant/Award Number: 1309‐00006B; Danish Cancer Society, Grant/Award Numbers: R231‐A13845, R146‐A9466‐16‐S2, R133‐A8520‐00‐S41
DATA AVAILABILITY STATEMENT
The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.
REFERENCES
- 1. Wan JCM, Massie C, Garcia‐Corbacho J, et al. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat Rev Cancer. 2017;17:223‐238. [DOI] [PubMed] [Google Scholar]
- 2. Scholer LV, Reinert T, Orntoft MW, et al. Clinical implications of monitoring circulating tumor DNA in patients with colorectal cancer. Clin Cancer Res. 2017;23:5437‐5445. [DOI] [PubMed] [Google Scholar]
- 3. Heitzer E, Ulz P, Geigl JB. Circulating tumor DNA as a liquid biopsy for cancer. Clin Chem. 2015;61:112‐123. [DOI] [PubMed] [Google Scholar]
- 4. Merker JD, Oxnard GR, Compton C, et al. Circulating tumor DNA analysis in patients with cancer: American Society of Clinical Oncology and College of American Pathologists Joint Review. J Clin Oncol. 2018;36:1631‐1641. [DOI] [PubMed] [Google Scholar]
- 5. Abbosh C, Birkbak NJ, Wilson GA, et al. Phylogenetic ctDNA analysis depicts early‐stage lung cancer evolution. Nature. 2017;545:446‐451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Snyder MW, Kircher M, Hill AJ, Daza RM, Shendure J. Cell‐free DNA comprises an in vivo nucleosome footprint that informs its tissues‐of‐origin. Cell. 2016;164:57‐68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Moss J, Magenheim J, Neiman D, et al. Comprehensive human cell‐type methylation atlas reveals origins of circulating cell‐free DNA in health and disease. Nat Commun. 2018;9:5068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Bettegowda C, Sausen M, Leary RJ, et al. Detection of circulating tumor DNA in early‐ and late‐stage human malignancies. Sci Transl Med. 2014;6:224ra24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Meddeb R, Dache ZAA, Thezenas S, et al. Quantifying circulating cell‐free DNA in humans. Sci Rep. 2019;9:5220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. van der Pol Y, Mouliere F. Toward the early detection of cancer by decoding the epigenetic and environmental fingerprints of cell‐free DNA. Cancer Cell. 2019;36:350‐368. [DOI] [PubMed] [Google Scholar]
- 11. Toth K, Patai AV, Kalmar A, et al. Circadian rhythm of methylated Septin 9, cell‐free DNA amount and tumor markers in colorectal cancer patients. Pathol Oncol Res. 2017;23:699‐706. [DOI] [PubMed] [Google Scholar]
- 12. Madsen AT, Hojbjerg JA, Sorensen BS, Winther‐Larsen A. Day‐to‐day and within‐day biological variation of cell‐free DNA. EBioMedicine. 2019;49:284‐290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Beiter T, Fragasso A, Hudemann J, Niess AM, Simon P. Short‐term treadmill running as a model for studying cell‐free DNA kinetics in vivo. Clin Chem. 2011;57:633‐636. [DOI] [PubMed] [Google Scholar]
- 14. Hummel EM, Hessas E, Muller S, et al. Cell‐free DNA release under psychosocial and physical stress conditions. Transl Psychiat. 2018;8:236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Hao TB, Shi W, Shen XJ, et al. Circulating cell‐free DNA in serum as a biomarker for diagnosis and prognostic prediction of colorectal cancer. Br J Cancer. 2014;111:1482‐1489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Jylhava J, Kotipelto T, Raitala A, Jylha M, Hervonen A, Hurme M. Aging is associated with quantitative and qualitative changes in circulating cell‐free DNA: the vitality 90+ study. Mech Ageing Dev. 2011;132:20‐26. [DOI] [PubMed] [Google Scholar]
- 17. Henriksen TV, Reinert T, Christensen E, et al. The effect of surgical trauma on circulating free DNA levels in cancer patients‐implications for studies of circulating tumor DNA. Mol Oncol. 2020;14:1670‐1679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Jensen SO, Ogaard N, Orntoft MW, et al. Novel DNA methylation biomarkers show high sensitivity and specificity for blood‐based detection of colorectal cancer‐a clinical biomarker discovery and validation study. Clin Epigenet. 2019;11:158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Orntoft MB, Nielsen HJ, Orntoft TF, Andersen CL, Danish study group on early detection of colorectal cancer . Performance of the colorectal cancer screening marker Sept9 is influenced by age, diabetes and arthritis: a nested case‐control study. BMC Cancer. 2015;15:819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Reinert T, Henriksen TV, Christensen E, et al. Analysis of plasma cell‐free DNA by ultradeep sequencing in patients with stages I to III colorectal cancer. JAMA Oncol. 2019;65(4):625‐634. https://www.ncbi.nlm.nih.gov/pubmed/25654990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Reinert T, Scholer LV, Thomsen R, et al. Analysis of circulating tumour DNA to monitor disease burden following colorectal cancer surgery. Gut. 2016;65:625‐634. [DOI] [PubMed] [Google Scholar]
- 22. Ingeholm P, Gogenur I, Iversen LH. Danish colorectal cancer group database. Clin Epidemiol. 2016;8:465‐468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Lynge E, Sandegaard JL, Rebolj M. The Danish National Patient Register. Scand J Public Health. 2011;39:30‐33. [DOI] [PubMed] [Google Scholar]
- 24. Huggett JF, Foy CA, Benes V, et al. The digital MIQE guidelines: minimum information for publication of quantitative digital PCR experiments. Clin Chem. 2013;59:892‐902. [DOI] [PubMed] [Google Scholar]
- 25. Pallisgaard N, Spindler KL, Andersen RF, Brandslund I, Jakobsen A. Controls to validate plasma samples for cell free DNA quantification. Clin Chim Acta. 2015;446:141‐146. [DOI] [PubMed] [Google Scholar]
- 26. Andersen CL, Lamy P, Thorsen K, et al. Frequent genomic loss at chr16p13.2 is associated with poor prognosis in colorectal cancer. Int J Cancer. 2011;129:1848‐1858. [DOI] [PubMed] [Google Scholar]
- 27. Lech Pedersen N, Mertz Petersen M, Ladd JJ, et al. Development of blood‐based biomarker tests for early detection of colorectal neoplasia: influence of blood collection timing and handling procedures. Clin Chim Acta. 2020;507:39‐53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. El Messaoudi S, Rolet F, Mouliere F, Thierry AR. Circulating cell free DNA: Preanalytical considerations. Clin Chim Acta. 2013;424:222‐230. [DOI] [PubMed] [Google Scholar]
- 29. Gary L, Horowitz SA, Boyd JC, et al. Defining, Establishing, and Verifying Reference Intervals in the Clinical Laboratory; Approved Guideline—Third Edition. Wayne, Pennsylvania: CLSI EP28; 2008;1‐76. [Google Scholar]
- 30. Rasmussen L, Wilhelmsen M, Christensen IJ, et al. Protocol outlines for parts 1 and 2 of the prospective endoscopy III study for the early detection of colorectal cancer: validation of a concept based on blood biomarkers. JMIR Res Protoc. 2016;5:e182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Dixon WJ. Processing data for outliers. Biometrics. 1953;9:74‐89. [Google Scholar]
- 32. Lahti A, Hyltoft Petersen P, Boyd JC, Fraser CG, Jorgensen N. Objective criteria for partitioning Gaussian‐distributed reference values into subgroups. Clin Chem. 2002;48:338‐352. [PubMed] [Google Scholar]
- 33. Franceschi C, Bonafe M, Valensin S, et al. Inflamm‐aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci. 2000;908:244‐254. [DOI] [PubMed] [Google Scholar]
- 34. Meddeb R, Pisareva E, Thierry AR. Guidelines for the preanalytical conditions for analyzing circulating cell‐free DNA. Clin Chem. 2019;65:623‐633. [DOI] [PubMed] [Google Scholar]
- 35. Xie J, Yang J, Hu P. Correlations of circulating cell‐free DNA with clinical manifestations in acute myocardial infarction. Am J Med Sci. 2018;356:121‐129. [DOI] [PubMed] [Google Scholar]
- 36. Rannikko J, Seiskari T, Huttunen R, et al. Plasma cell‐free DNA and qSOFA score predict 7‐day mortality in 481 emergency department bacteraemia patients. J Intern Med. 2018;284:418‐426. [DOI] [PubMed] [Google Scholar]
- 37. O'Connell GC, Petrone AB, Tennant CS, et al. Circulating extracellular DNA levels are acutely elevated in ischaemic stroke and associated with innate immune system activation. Brain Inj. 2017;31:1369‐1375. [DOI] [PubMed] [Google Scholar]
- 38. Duvvuri B, Lood C. Cell‐free DNA as a biomarker in autoimmune rheumatic diseases. Front Immunol. 2019;10:502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Gogenur M, Burcharth J, Gogenur I. The role of total cell‐free DNA in predicting outcomes among trauma patients in the intensive care unit: a systematic review. Crit Care. 2017;21:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Panagiotakos DB, Pitsavos C, Yannakoulia M, Chrysohoou C, Stefanadis C. The implication of obesity and central fat on markers of chronic inflammation: the ATTICA study. Atherosclerosis. 2005;183:308‐315. [DOI] [PubMed] [Google Scholar]
- 41. Jylhava J, Lehtimaki T, Jula A, et al. Circulating cell‐free DNA is associated with cardiometabolic risk factors: the health 2000 survey. Atherosclerosis. 2014;233:268‐271. [DOI] [PubMed] [Google Scholar]
- 42. Bedin C, Enzo MV, Del Bianco P, Pucciarelli S, Nitti D, Agostini M. Diagnostic and prognostic role of cell‐free DNA testing for colorectal cancer patients. Int J Cancer. 2017;140:1888‐1898. [DOI] [PubMed] [Google Scholar]
- 43. He N, Song L, Kang Q, et al. The pathological features of colorectal cancer determine the detection performance on blood ctDNA. Technol Cancer Res Treat. 2018;17:1533033818791794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Spindler KG, Boysen AK, Pallisgard N, et al. Cell‐free DNA in metastatic colorectal cancer: a systematic review and meta‐analysis. Oncologist. 2017;22:1049‐1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Basnet S, Zhang ZY, Liao WQ, Li SH, Li PS, Ge HY. The prognostic value of circulating cell‐free DNA in colorectal cancer: a meta‐analysis. J Cancer. 2016;7:1105‐1113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Jackson Chornenki NL, Coke R, Kwong AC, et al. Comparison of the source and prognostic utility of cfDNA in trauma and sepsis. Intensive Care Med Exp. 2019;7:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Flicker L. Should geriatric medicine remain a specialty? Yes BMJ. 2008;337:a516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Jylhava J, Nevalainen T, Marttila S, Jylha M, Hervonen A, Hurme M. Characterization of the role of distinct plasma cell‐free DNA species in age‐associated inflammation and frailty. Aging Cell. 2013;12:388‐397. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Appendix S1: Supplementary Information
Data Availability Statement
The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.
