Abstract
Genomes of diverse origins are continuously shed into human body fluids in the form of fragmented cell-free DNA (cfDNA). These molecules maintain the genetic and epigenetic codes of their originating source, and often carry additional layers of unique information in newly discovered physico-chemical features. Characterization of cfDNA thus presents the opportunity to non-invasively reconstruct major parts of the host- and metagenome in silico. Data from a single specimen can be leveraged to detect a broad range of disease-specific signatures and has already enabled the development of many pioneering diagnostic tests. Moreover, data from serial sampling may allow unparalleled mapping of the scantily explored landscape of temporal genomic changes as it relates to various changes in different physiological and pathological states of individuals. In this review, we explore how this vast dimension of biological information accessible through cfDNA analysis is being tapped towards the development of increasingly powerful molecular assays and how it is shaping emerging technologies. We also discuss how this departure from traditional paradigms of snapshot genetic testing may pave the way for an onrush of new and exciting discoveries in human biology.
Introduction
It is now widely understood that whole or partial genomes of diverse origins, including host cells, fetal cells, microbes, and viruses, are continuously shed into various human body fluids in the form of cell-free DNA (cfDNA) [1], [2], [3], [4]. While much remains to be discovered about the fundamentals of the molecular, biological and physiological factors that affect the properties of these molecules, it is clear that DNA can be released into body fluids through various pathways and subroutines related to the mechanisms of accidental cell degradation, programmed cell death, as well as regulated extrusion [1], [2], [3], [4]. CfDNA also appears to be released from all types of cells, tissues, and organs, while maintaining the unique genetic and epigenetic features of their originating source. Moreover, emerging research shows that unique bits of genetic, biological and pathological information (e.g., genomic regions, cell identity, tissue type, and mutation clusters) are often partitioned into differently structured cfDNA subtypes via the action of numerous nuclear, cytoplasmic, and extracellular mechanisms and factors that alter the physico-chemical features of specific genomic regions and different cfDNA subtypes in distinct ways.

The potential clinical scope of cfDNA profiling.
(A) Genetic material from diverse sources is continuously shed into body fluids. (B) Moreover, the characteristics of cfDNA and its release into body fluids and its subsequent stability and fluctuation are modulated by a wide range of biological, physiological, lifestyle, environmental and pathological factors. (C) Therefore, the composition of the cfDNA population in a biospecimen is highly complex, characterized by the co-occurrence of genomes from various origins and immense genetic, epigenetic, and structural diversity among different cfDNA subtypes. (D) While it is very difficult to analyze a highly heterogeneous cfDNA population in a clinical biospecimen, it is becoming increasingly easy to do so through the coalescence of many ground-breaking advances in the cfDNA research field that allow increasingly effective partitioning and high-fidelity analysis of cfDNA subtypes. These include systematic improvements in preanalytical procedures, analytical techniques, technologies, and bioinformatics, in conjunction with an improved understanding of all the factors that determine the characteristics of cfDNA in vivo and in clinical biospecimens, as well as an ever-expanding repertoire of disease-specific markers and tissue-of-origin classifiers. This may enable the development of cfDNA tests that are fit for testing in large-scale cohorts and potential clinical roll-out in the future. (E) On one hand, tests may be developed for the minimally-invasive detection and diagnosis of a wide range of diseases. (F) On the other hand, tests may be developed to map and study temporal genomic changes in individuals or populations in a variety of contexts.
Capture, profiling and bioinformatic integration of the information encoded in the sequence composition and various physico-chemical features of cfDNA therefore presents the unique opportunity for the minimally-invasive reconstruction of major parts of the host- and metagenome in silico. This information can be harnessed in two major ways: First, data collected from a single specimen can be used to detect a broad range of disease-specific signatures, and has so far enabled the development of several breakthrough medical applications, e.g., diagnostic tests for solid tumors [5], [6], [7], fetal abnormalities [8], [9], [10], allograft rejection [11], [12], [13], [14], [15], [16], and sepsis [17, 18]. Second, dynamic analysis of data obtained from longitudinal sampling may open an unprecedented window of access for mapping the scantily explored and still mostly invisible landscape of temporal genomic changes caused by a variety of factors (e.g., aging, pathology, dietary changes, and medical therapies) (Figure 1).
In this review we explore how this new dimension of biological information accessible through cfDNA analysis has already been tapped for the development of new and more powerful cfDNA analysis modalities, how it is shaping emerging technologies, and how it represents a departure from the traditional paradigm of snapshot genetic testing, which may very likely catalyze a surge of new discoveries in human biology.
The immense heterogeneity of the genetic and epigenetic features of the total cfDNA population in a given biospecimen, in combination with the complex network of interacting biological, physiological, and environmental factors that modulate its fluctuation, makes cfDNA a very powerful biomarker and interesting biological phenomenon, but at the same time significantly complicates the selection of appropriate preanalytical steps and analytical differentiation of different cfDNA subtypes. Therefore, in this review we also draw attention to the importance of high-fidelity reverse engineering of a cfDNA sample and explore the technical and analytical challenges and solutions involved in overcoming the heterogeneity of cfDNA samples.
Measurement of total cfDNA levels
Total cfDNA levels have been correlated with a plethora of diseases commonly associated with cell or tissue injury, such as solid tumors [6, 7, 19], [20], [21], autoimmune diseases [22] (e.g., systemic lupus erythematosus [23, 24], rheumatoid arthritis [25], and systemic sclerosis [26]), trauma patients [27] (e.g., brain injuries [28, 29] and burn patients [30]), cardiovascular diseases (e.g., acute myocardial infarction [31] and acute coronary syndrome [32]), viral infections (e.g., acute Puumala Hantavirus Infection [33] and Crimean–Congo hemorrhagic fever [34]), benign gastrointestinal tract disorders [35], [36], [37], kidney disease [38, 39], lung disease (e.g., chronic obstructive pulmonary disease [40] and pulmonary embolism [41]), thyroid disease [42], pregnancy disorders (e.g., preeclampsia [43] and intrahepatic cholestasis [44]), skin conditions (e.g., psoriasis [45, 46]), stroke [47, 48], and workers exposed to occupational hazards such as pesticides [49], nuclear radiation [50], and toxic paints [51].
CfDNA levels have also been correlated with, for lack of better phrasing, more obscure medical conditions or clinical scenarios such as schizophrenia [52], extra temporal lobe epilepsy [53], sperm quality in men [54, 55], and the quality of embryos [56], pregnancy rates [57], and performance of various stress reduction exercises [58] among women undergoing in vitro fertilization. Medical treatments have also been correlated with changes in cfDNA levels, such as surgery [59], radiotherapy [59] corticosteroids [60], hemodialysis [39, 61, 62], and mechanical ventilation [63, 64].
Moreover, many studies have now shown that cfDNA levels are often significantly modulated by a variety of non-pathological conditions, and physiological and lifestyle factors (reviewed in Refs. [1, 3, 6, 65, 66]), such as age [67, 68], body mass index [69, 70], the time of day at which samples are collected [71], [72], [73], [74], food intake and creatinine levels [73], [74], [75], gender [66, 75], walking [76] and acute exercise [71, 77], [78], [79], [80], [81], [82]. Conversely, no correlation has yet been demonstrated between changes in total cfDNA levels and alcohol intake [83], smoking (reviewed in Ref. [66]), a history of betel nut chewing (seeds with stimulatory effects akin to amphetamines and cocaine) [83], frequency of blood donation [84], hematocrit or cannula placement pain [85], height [78, 86], and the menstrual cycle [87, 88].
Taken together, these studies which have focused on the relationship between total cfDNA levels and disease activity, therapy, and various physiological and lifestyle factors seem to suggest that total cfDNA levels could potentially serve as a versatile biomarker for the diagnosis and monitoring of a wide range of diseases and other clinical scenarios. However, it is also clear from these reports that aberrant levels of total cfDNA is not a phenomenon unique to specific pathological states but is instead a common consequence of pathology and many other factors. Therefore, while the findings reported by all the above-mentioned studies are interesting, it should be interpreted with caution. First, measuring the correlation between total cfDNA levels and any specific disease or physiological or lifestyle factor is complicated by the potential influence of many other co-occurring factors in the same category. Second, total cfDNA levels depend significantly on many biological factors (reviewed in Refs. [1], [2], [3], [4, 19, 65]). While most early studies have focused on the analysis of cfDNA isolated from the circulatory system, cfDNA molecules have now been detected and are investigated in most body fluids. As such, the composition of the cfDNA population in any specific body fluid is modulated by a wide range of biological factors, many of which are unique to the body fluid in question. While many details remain to be discovered, it is becoming clear that the characteristics of the cfDNA population in any body fluid depends on: (i) the physical location of the fluid (e.g., release of cfDNA can be influenced by the unique vascularization, histology, perfusion, turnover activity, and cell death and proliferation rate in different tissues, while its movement may be affected by biological barriers like the blood-brain barrier); (ii) the relative contribution of different organs and cell types to the total cfDNA pool [89], [90], [91], (iii) tissue or cell-specific genetic and epigenetic features; (iv) the mechanisms by which cfDNA is generated and released (e.g., active extrusion, association with extracellular vesicles, apoptosis, necrosis, and other cell-death subroutines which are well described in the literature, but not necessarily adequately experimentally correlated with cfDNA release, such as autophagic cell death, mitotic catastrophe, regulated necrosis, and other cell death modalities such as anoikis, entosis, parthanatos, pyroptosis, and NETosis); (v) the changes that cfDNA molecules undergo before exiting cells (e.g., fragmentation, or complexing with proteins and vesicles); (vi) the conditions surrounding the movement of cfDNA from immediate extracellular space into the body fluid; (vii) the changes that cfDNA molecules undergo after exiting cells (e.g., enzymatic degradation and interaction with other extracellular components); (viii) the stability/half-life of cfDNA in the fluid (e.g., rate of degradation, clearance, or binding and reuptake by cells).
Accurate measurement of total cfDNA levels is also significantly affected by numerous factors relating to its physico-chemical properties [1], [2], [3], [4, 92], the nature of many preanalytical steps [65, 93], [94], [95], [96], and analytical decisions [6, 7, 19, 20]. Unsurprisingly, the convergence of so many variables cause greatly overlapping values between different disease types and healthy individuals both in individual studies and in interstudy comparisons. Such overlapping data for total cfDNA has to-date precluded the establishment of a cut-off value or normal reference range for any specific disease or other clinical scenario. Indeed, given the vast number of factors that affect total cfDNA levels, it is likely that certain factors have been wrongly correlated or attributed, while others have been obscured. As such, it does not seem likely that total cfDNA levels alone could serve as a biomarker in a diagnostic setting.
Crucial advancements in genetic and epigenetic profiling of cfDNA over the last two decades have, however, enabled the accelerated discovery of new connections between cfDNA and disease, and enabled the development of clinically meaningful cfDNA assays, some of which are FDA-approved and applied in routine diagnostics, particularly in the fields of oncology and prenatal testing. In the following sections we explore these advancements and show how it may reignite interest in the previously discussed diseases/other clinical scenarios, how quantitative measurements of cfDNA may become an auxiliary marker to qualitative characterization of cfDNA, and how this may inspire researchers to investigate cfDNA in some unexplored domains of biology and medicine.
Qualitative characterization of cfDNA
CfDNA in various biospecimen types have been scrutinized for the detection of sequences or mutations that are unique to specific individuals, diseases, and organisms, and has so far led to many exciting discoveries and the development of diagnostic assays in various clinical fields.
Ample research highlights cfDNA as a prime candidate surrogate marker for various indications during cancer progression, and may become a powerful clinical tool for the management of various stages of the disease (reviewed in Refs. [6, 7, 20]), including pan-cancer screening of healthy populations or at-risk individuals, indication of disease stage and prediction of clinical outcome, guiding the selection of novel targeted therapies, identification of existing and acquired resistance-conferring mutations, tracking clonal evolution, and detection and prediction of minimal residual disease or recurrence. Taken together, this information will likely inform the selection and development of more efficient therapeutic regimes.
Beyond oncology, sequence characterization of cfDNA has immense potential as a diagnostic, prognostic or theranostic tool for (i) investigating miscarriage [97], fetal sexing and the detection of various fetal genetic abnormalities [8, 98], [99], [100], [101], (ii) monitoring post-transplant organ rejection, dysfunction and injury [11], [12], [13], [14], [15], [16], (iii) detecting pathogenic DNA such as bacteria (Mycobacterium tuberculosis-derived DNA [102] and pneumonia pathogens [103]), parasites [104, 105], fungi [106], viruses (e.g., Crimean–Congo hemorrhagic fever [34], and cancer-causing viruses such as Epstein-Barr virus, which is associated with nasopharyngeal carcinoma [107, 108], or Human papillomavirus (HPV), which is associated with oropharyngeal squamous cell carcinoma [109, 110] and cervical cancer [111], and (iv) studying the gut microbiome as it relates to microbial diversity and its role in human health and disease [112], [113], [114], [115]. CfDNA may also be interrogated to study environmental DNA in humans, as there is evidence of the presence of meal-derived environmental nucleic acids, such as plant and bacterial DNA, in human body fluids [116, 117]. The relative contribution of such foreign DNA to the total cfDNA population is not known but is likely low. However, as these cfDNA molecules have been shown to be able to be incorporated into the human genome [118], [119], [120], [121], [122], they may have underappreciated detrimental effects. A recent study has, for example, suggested that cfDNA may facilitate the horizontal transfer of antibiotic resistance genes [117].
Other potential uses of cfDNA sequence analysis include (i) retrieval of vector-integration sites, which may prove to be a superior approach for assessing the safety and efficacy of various kinds of gene therapy products [123], (ii) biobank management, as analysis of cfDNA in cord blood plasma has shown to be useful for sample identification [124], (iii) non-invasive pre-implantation genetic diagnosis of X-linked disorders [125, 126], (iv) diagnosis of vascular malformations [127], and (v) use as evidence in forensic casework [128], [129], [130].
While the majority of cfDNA studies have to-date focused on cell-free nuclear DNA (cf-nDNA), a growing body of evidence indicates the potential clinical utility of cell-free mitochondrial DNA (cf-mtDNA), which is released into body fluids via cellular clearance or repair processes and may be present as free floating mtDNA fragments or be associated with particles such as internal and external mitochondrial membrane fragments [3, 131]. Intact respiratory-competent mitochondria have also been found to circulate in blood plasma, which may also serve as a source of cf-mtDNA [132]. Aberrant cf-mtDNA has been correlated with a wide range of diseases and other clinical scenarios, including cancers [133, 134], such as breast cancer [135], Ewing’s sarcoma [136], urological malignancies [137], oral cancer [138], squamous cell carcinoma [139, 140], and lung adenocarcinoma [141]; cardiovascular disease [142], [143], [144]; aging [145]; neurodegenerative disease [146], such as multiple sclerosis [147], Parkinson’s disease [148] and Friedrich’s ataxia [149]; diabetes [150, 151]; trauma, surgery and ICU patients [152, 153]; sepsis [154, 155]; chronic inflammation and cognitive decline in HIV patients [156], [157], [158] and type-2 diabetes patients [159]; exposure to carcinogenic pesticides [160]; adverse health effects of spaceflight on astronauts [161, 162]; and poor outcome of patients with adult community-acquired bacterial meningitis [163]. In addition, cf-mtDNA has been shown to be influenced by exercise [164, 165], can be used for non-invasive haplogroup matching [166], and may be useful in studying various psychological issues, as cf-mtDNA levels have been correlated with psychosocial and physical stress [167], acute psychological stress [168], as well as the pathophysiology underlying suicidal behavior [169] and major depressive disorder [170].
Taken together, sequence analysis of cfDNA isolated from a single biospecimen can be used to detect an ever-expanding repertoire of disease-specific signatures, which represents a breakthrough in the application of non-invasive molecular genetic tests for personal, precision diagnostics and clinical assessments. This clinical potential of cfDNA analysis is underscored by several exciting developments, including, but not limited to: (i) four cfDNA sequence-based tests have to-date been approved by the FDA for implementation in routine diagnostics [171], including the detection of PIK3CA mutations in breast cancer, EGFR mutations (exon 19 deletions and exon 21 L858R substitution mutations) in non-small cell lung cancer (NSCLC), KRAS G12C mutations in NSCLC, and BRCA1 and BRCA2 mutations in metastatic castration-resistant prostate cancer; (ii) several Clinical Laboratory Improvement Amendments (CLIA) labs worldwide offer services for the characterization of cfDNA mutational profiles in cancer patients; (iii) numerous non-invasive prenatal testing (NIPT) facilities worldwide have incorporated cfDNA-based tests into their portfolio, including tests for fetal sexing and diagnosis of various fetal genomic aberrations; and (iv) a 24-marker qPCR assay has recently been developed for the detection of sepsis well before the development of clinical signs [18].
Apart from the handful of FDA-approved assays, and despite unprecedented research efforts in the last two decades, the development and implementation of routine tests based on mutational profiling of circulating tumor DNA (ctDNA) has been advancing at a suboptimal rate. The developmental timeline of ctDNA assays (and by extension cfDNA assays that target specific sequences in other diseases) that bear the required diagnostic sensitivity and specificity for implementation in routine clinical practice is extended by various factors that challenge the analytical detection of ctDNA. The main factors include: (i) a low proportion of ctDNA molecules (or any cfDNA molecules targeted in other diseases) vs. highly abundant background DNA originating from diverse sources, especially in early disease stages where tumors shed miniscule amounts of DNA into extracellular space, (ii) dilution of ctDNA through preanalytical steps that contribute to the release of germline DNA from peripheral blood cells, (iii) an ever expanding list of selectable products, analytical techniques and technologies, many of which show varying degrees of efficiency and bias towards specific applications, sample processing procedures, and physico-chemical features of cfDNA, and lastly (iv) ctDNA profiling is complicated by the presence of cancer-specific mutations in clonal hematopoiesis (CH)-derived cfDNA in both cancer patients [172] and healthy subjects that do not have cancer at the time of measurement and may never develop cancer [173], [174], [175], [176], [177], [178]. It is currently not clear how the biological noise created by CH-derived cfDNA may be overcome to prevent the misdiagnosis of CH-derived mutations in cfDNA as malignancy. One possibility is that CH-derived cfDNA may be distinguished from ctDNA on the basis of fragment size differences, as ctDNA fragments have been shown to be shorter than CH-derived cfDNA [179]. It is worth noting here that CH mutations should not be considered merely as false-positive results in ctDNA assays, but should be evaluated for potential pathological and clinical significance as CH mutations have been correlated with increased risk of developing severe Covid-19 [180], hematological malignancies, and cardiovascular disease [181]. Moreover, cancer patients with CH are more likely to develop myeloid neoplasms after chemotherapy vs. non-carriers [181].
Concerning the low proportion of ctDNA molecules to background cfDNA, several strategies may be considered to maximize the chances of capturing ctDNA molecules, thereby increasing the sensitivity and specificity of assays (reviewed in Refs. [6, 7]). These include: (i) collecting and processing larger volumes of body fluid samples (e.g., drawing more blood); (ii) collecting body fluids that are closest to the region of interest, which usually have higher proportions of the target molecule compared to other body fluids (e.g., CSF for brain tumors, urine for bladder cancer, and stool for colorectal cancer); (iii) using optimized analytical techniques, such as performing independent assays on aliquoted replicates or interrogating multiple mutations; (iv) the use of preanalytical strategies that minimize the release of germline DNA from peripheral cells [65, 182]; (v) the use of extraction procedures that provide the highest yield of cfDNA or that are biased toward capturing target molecules; (vi) the development and use of increasingly sensitive assays (e.g., targeted sequencing); and (vii) use of molecular barcoding and integrated bioinformatics approaches that allow the construction of consensus sequences and elimination of random PCR and sequencing errors.
In addition to the implementation of these steps, a growing body of evidence indicates that the probability of capturing ctDNA molecules can be significantly increased by extending the analysis of hotspot mutations to interrogation of other disease-specific alterations in genetic and epigenetic features of cfDNA, thereby markedly increasing the sensitivity and specificity of mutation-based assays.
Beyond hotspot mutation analysis
Every year, cfDNA is characterized with higher resolution, and just in the last few years more information on the physico-chemical features of cfDNA has been mined than in the previous three decades [92, 183], [184], [185]. Through this continual unfolding of new knowledge on the composition of cfDNA has it become clear that cfDNA molecules do not merely maintain the primary genetic and epigenetic information stored in the DNA of their originating cells, but often carry extra layers of information in various secondary physico-chemical features that arise upon changes to its primary form. Here, primary genetic information refers to (i) the DNA code itself, which indicates origin, e.g., nDNA, mtDNA, metagenomic DNA, including features like gene sequences, the repetitive element landscape, GC-content, sequence motifs (e.g., transcription factor binding sites), mutations, and copy number variations, (ii) structural and numerical chromosomal abnormalities, and (iii) topological forms like extrachromosomal circular DNA [186], [187], [188]. Primary epigenetic information refers to classic DNA methylation (e.g., hypermethylation of promoter CpG islands, hypermethylation of tumor suppressor genes, hypomethylation of oncogenes, and global hypomethylation across the genome), a wide range of histone modifications, as well as nucleosome spacing patterns. Secondary physico-chemical features refer to the various changes that can occur to DNA molecules outside the context of its normal function in the nucleus, such as (i) the binding of cfDNA to DNA binding proteins. There are many DNA-binding proteins that exist in the human body and in the extracellular space (e.g., HMGB-1, fibrinogen, HDL, albumin, CRP, SAA). However, there is a lack of studies concerning the binding of cfDNA to these proteins, and its effects on cfDNA purification, its effects on downstream analysis, or its potential clinical utility; (ii) the binding of DNA to macromolecules (e.g., heparin [189], [190], [191]); (iii) packaging of DNA into or association with vesicles (e.g., micronuclei [2, 192]), extracellular vesicles [193], [194], [195], [196], [197], [198], [199], [200], [201], [202], [203], [204], [205], apoptotic bodies [206, 207], and outer membrane vesicles [208], [209], [210], [211], (iv) association with complex macromolecular structures [212], [213], [214], (v) binding to cell membranes on the outer surface [215, 216], (vi) unique fragmentation patterns [184, 217], [218], [219], [220], [221], [222], [223], [224], and (vii) unique fragment end-points and motifs [225], [226], [227]. These changes may occur during the movement of cfDNA from the nucleus to the cytoplasm, in the cytoplasm, during movement from cytoplasm to the extracellular space, and finally in the extracellular space. There are many ways by which the characterization of these various genetic and epigenetic features may be beneficial towards the development of increasingly powerful cfDNA-based clinical assays.
First, in contrast to the limited number of recurrent hotspot DNA mutations, which typically affect only small regions of the genome, many of these modifications already occur early during tumorigenesis and across a much larger portion of the genome, in turn corresponding to higher proportionality in the total cfDNA population, significantly increasing the probability of detection. For these reasons, profiling of non-classical genetic and primary and secondary epigenetic features of cfDNA is now widely considered as a potential auxiliary marker to the profiling of hotspot mutations in cfDNA. Moreover, in some cases, the characterization of epigenetic features may even outperform mutational profiling and serve as a standalone biomarker. One breakthrough in this regard includes the FDA-approved Epi proColon 2.0 CE test, which is a qualitative in vitro diagnostic test for the measurement of aberrant methylation of SEPT9, which has been associated with colorectal cancer, in patients over the age of 50. Patients with positive results may then be referred for diagnostic colonoscopy. Also of significance, a landmark study on breast cancer patients was recently conducted. Based on the unique methylation patterns observed in breast cancer tissue vs. healthy tissue, researchers used cfMeDIP-Seq to identify breast cancer methylation signatures in the cfDNA of asymptomatic individuals, which enabled minimally-invasive stratification between breast cancer cases and cancer-free subjects before clinical presentation and up to five years before clinical diagnosis using conventional approaches [228].
While not yet ready for implementation in routine clinical practice, numerous studies have demonstrated very strong correlations between epigenetic characteristics of cfDNA and various indications in different types of cancer [229], [230], [231], [232], [233], [234], [235], [236], [237], [238], [239], [240], [241], [242], [243], [244], [245], [246], [247]. It is also interesting to note that, beyond the field of oncology, epigenetic characterization of cfDNA has shown potential for the diagnosis and monitoring of cardiovascular disease [248], diabetes [249, 250], liver fibrosis [251, 252], psychosocial stress [167], aging [68], multiple sclerosis [253], psychotic episodes [254], and paracetamol overdose [255].
Second, whereas tissue biopsies and other conventional screening methods are limited to the detection of cancer in specific tissues, characterization of genome-wide methylation patterns may enable parallel interrogation of multiple cancer types. In the context of cancer screening, it is essential to identify the tissue of origin of underlying cancers. While this remains a complex issue and challenging task, much progress has been made in mapping the landscape of tissue specific methylation patterns [232, 256], [257], [258], as well as various other tissue-of-origin classifiers. For example, the tissue-of-origin of cfDNA molecules have been determined through the profiling of nucleosome-depleted regions, transcription-factor binding sites [259, 260], fragmentation profiles [226], unique fragment end-points and end-motifs [225], [226], [227], and post-translational histone modifications [261].
Third, while the existence of secondary epigenetic changes to cfDNA has been known for some time, there is a major ongoing breakthrough in the discovery that these secondary changes to DNA that are encoded into cfDNA are not merely random changes that indiscriminately affect the entire genome. Instead, these epigenetic modifications of DNA are often unique to specific scenarios, reflecting the unique mechanistic underpinnings of processes that act on specific genomic regions, in specific cell types, in specific tissues, or in specific disease states. In other words, this means that unique bits of biological and pathological information are partitioned into different cfDNA subtypes. Different cfDNA subtypes can then be analyzed to gain insight not only into the tissue-of-origin, but also be scrutinized to study other biological phenomena and pathological events. Therefore, in addition to enhancing mutation-based cfDNA assays and aiding in determining the tissue-of-origin of cfDNA molecules, it is likely that systematic mapping of these additional epigenetic cfDNA features will lead to the identification of unique, currently unknown features that exhibit more disease-specific qualities, which will increase the pool of molecules that can be grouped under disease-defining variants, thereby increasing the differentiating power of the method.
As with hotspot mutational profiling, epigenetic profiling of cfDNA has many biological, preanalytical, technical, as well as analytical issues and limitations that need to be overcome on route to the development of clinically meaningful assays. The major limitations include: (i) overlapping modifications in pathological and ordinary biological processes, (ii) biological noise induced by stochastic fluctuations in epigenetic marks or markers, and (iii) methodological biases, such as fragment-length biases of different sequencing chemistries and DNA library preparation methods (reviewed in Ref. [92]).
In addition to interrogating a wide range of genetic and epigenetic features of cfDNA, increasing evidence indicates that the sensitivity and specificity of cfDNA tests may be increased substantially by the parallel assessment of cfDNA and other non-DNA liquid biopsy markers. While the most promising approach thus far appears to be the combinatorial analysis of cfDNA and various proteins [262, 263], there may be great synergistic potential in combining cfDNA analysis with the profiling of other biomarkers [264], such as circulating tumor cells (CTCs) [265], [266], [267], [268], extracellular vesicles [269, 270]), miRNAs [271], mRNA transcripts [272], or metabolites [273].
Conclusion – new perspectives on the importance of cfDNA biology
The composition of the total cfDNA population in any body fluid, especially blood plasma, is highly complex and consists of DNA fragments from diverse origins, including host DNA originating from multiple organs and cell types as well as heterogeneous metagenomic DNA. In addition to differences in sequence information, cfDNA molecules can vary significantly in their physico-chemical features, which not only reflect the characteristics of their originating cell but include a wide range of possible alterations effected through various processes and factors encountered in the different biological compartments through which DNA molecules move following the disruption of its primary structure in the nucleus. Moreover, there is often an overlap of prominent genetic and physico-chemical features in cfDNA fragments that originate from disparate sources and processes. In line with this, the proportion of different cfDNA subtypes is determined by numerous biological, physiological, lifestyle, and environmental factors, many of which are inextricably linked. Additionally, the measured quantitative and qualitative characteristics of cfDNA depends significantly on the nature of preanalytical steps, the efficiency and biases of purification methods, and analytical decisions.
The vast biological and structural diversity of cfDNA, along with the complex network of factors that modulate its fluctuation is a double-edged sword. On one side, the richness of information accessible through cfDNA analysis represents an unparalleled treasure trove that can be mined to infer the changing physiological and pathological state of an individual.
First, data captured from a single biospecimen can be used to detect a broad range of disease-specific signatures, which represents a breakthrough in the application of non-invasive molecular tests for precision clinical assessments and diagnostics. This clinical potential of cfDNA is demonstrated by FDA-approved ctDNA assays, mutational profiling of ctDNA in CLIA labs, and the wide-spread clinical implementation of NIPT assays. It is conceivable that such routine clinical assays may in the near-future be developed for the detection of pathogens, monitoring of organ transplant procedures, and many other pathologies and clinical scenarios, likely including many that have not yet been considered in cfDNA research.
Second, data captured from the profiling of longitudinally collected cfDNA samples can significantly expand the window through which the genome can be studied, allowing a much deeper understanding of how the genome functions, reacts to the environment, and changes over time in response to changes in physiology and pathology. This may prime the ground for a significant leap in genomic sciences, as our current understanding of human genome biology is achieved mainly through snapshot analyses.
What is meant by snapshot analyses is that, through various means we procure a fraction of a whole organism (e.g., tissue biopsies and cell culture models) and in this bounded setting isolate and analyze various molecules (DNA, RNA, proteins, lipids, etc.) that serve as mere proxies for a process taking place. In other words, we isolate a specific process or sub-set of processes from a greater overarching process in order to infer or extrapolate its character and function in a whole organism. On one hand, such a reductive approach has been invaluable for studying numerous integral phenomena in molecular biology, e.g., elucidating most structural features of the genome. On the other hand it should be recognized that contextual logic is substantially traded off through the application of scientific reductionism, wherein sub-processes are inexorably sequestered from the context of the larger process(es) within which it is embedded. This obfuscates the relationships between sub-processes and its relation to the whole, which inherently precludes a proper study of the true dynamic nature of the genome. However, analysis of cfDNA may have an underappreciated role in overcoming this “static” view or interpretation of the genome. A good example of the latter, and why a dynamic view of the genome is highly desirable, is the recent successes achieved in assessing and monitoring dynamic genomic changes in tumors over time, such as changes in the mutational landscape, intratumor heterogeneity, genetic response of a tumor to therapy, and mechanisms that lead to the emergence of resistance against therapy. This approach has already significantly improved the outcome of cancer patients (reviewed in Refs. [6, 7, 20]). Conversely, accurate monitoring of dynamic tumor changes is virtually impossible with tissue biopsies in most cases.
The benefits of temporal genome analysis will not be limited to the domain of oncology, as all cells in the body seem to share the capacity to shed their genome into body fluids, while conserving the unique genetic and epigenetic traits of their originator cells. Moreover, as mentioned previously, a growing body of evidence indicates that the tissue or cellular origin of cfDNA can be identified by virtue of cell and tissue-specific epigenetic signatures, such as nucleosome spacing, fragmentation profiles, unique sequence motifs such as fragment end-points and transcription factor binding sites, histone modifications, and differentially methylated regions. While much remains to be discovered on this front, there is currently an unprecedented effort underway to map cell-specific genetic and epigenetic signatures. It therefore seems plausible that serial collection and characterization of cfDNA may in the future allow the investigation of time-dependent genomic changes over the whole body or specific regions of interest under a wide variety of conditions, whether relating to normal genomic functioning, malfunctioning processes, or the positive or negative impacts of drug and environmental effects.
Some intriguing possibilities include the study and monitoring of (i) the nature of genetic mosaicism, (ii) the safety and efficacy of gene therapies, (iii) the dynamic response of the genome to diet, (iv) correlations between genomic changes and a variety of diseases, such as aging, psychological stress, and metabolic disorders, and (v) unknown links between different diseases.
Beyond probing dynamic changes to the genetic and epigenetic code, recent studies have shown that gene expression programs could be inferred from nucleosome occupancy patterns [259, 274] and post-translational histone modifications [261], representing a new modality in the perusal of cfDNA. Further refinement of such approaches may in the future create the possibility of non-invasively identifying and studying activities/factors that cause positive vs. negative gene expression responses. Additionally, cfDNA could potentially be leveraged to monitor gene expression patterns in various poorly understood biological processes, such as the developmental transitions in growing embryos, which is currently studied through extremely costly and tedious longitudinal collection and dissection of thousands of rat organs.
Taken together, cfDNA analysis clearly shows immense potential not only as an important and versatile clinical biomarker, but also as a powerful research tool in basic molecular biology and genetics. However, on the other side of the double-edged sword mentioned earlier, there are some serious drawbacks and complications posed by the biological and structural diversity of cfDNA and the concurrent array of related preanalytical and analytical challenges. For example, it significantly limits the clinical utility of total cfDNA levels, it makes it very challenging to differentiate analytically between cfDNA subtypes, and it complicates the detection of scarce biomarkers (e.g., ctDNA hotspot mutations) in specific assays, whether for clinical or basic research interests.
Therefore, unlocking the full potential of cfDNA requires not only high-fidelity reconstruction of the quantitative, genetic and epigenetic features of cfDNA contained in biospecimens, but is dependent on the accurate elucidation and systemic mapping of all known and unknown features as it relates to various biological, physiological, lifestyle, environmental, preanalytical, and analytical variables in the widest sense possible. In other words, the importance of a deep and structured enquiry into the structure and biology of cfDNA cannot be overstated. While there is still a significant lack of knowledge, studies on the complete genetic and epigenetic features of cfDNA molecules as it relates to a variety of contexts has come rapidly to the front and will in the next couple of years likely become the principal center of research interest and be one of the major drivers of progress in the field.
As discussed above, an improved understanding of cfDNA biology will on the one hand expand the repertoire of disease-specific markers, facilitate the discovery of new links between the properties of cfDNA and various diseases and clinical scenarios, and enable the characterization of temporal genome changes and gene expression programs.
On the other hand, deeper knowledge of cfDNA biology may facilitate improvements in the sensitivity and specificity of various cfDNA assays, allowing rapid advancements in each of the former avenues of cfDNA research. First, a better understanding of the biological and physiological factors that determine the release and extracellular stability of different types of cfDNA molecules will likely inform the development of various strategies that maximize the likelihood of capturing or detecting target molecules. For example, (i) research indicates that ctDNA molecules are enriched in body fluids that are in closest proximity to the tumor in question, (ii) prior to sample collection, there may be optimal patient conditions that either favor the release of target molecules or limit the release of background molecules into the body fluids in question (the large number of factors that can affect the characteristics of cfDNA demonstrates the importance of collecting and documenting the right meta-data from patients, and documenting patient conditions prior to and during biospecimen collection). Second, when the mechanisms of release of specific cfDNA molecules are understood, it may become feasible to leverage existing or newly developed drugs or mechanical methods (e.g., temporarily opening the blood-brain barrier by focused ultrasound and microbubbles) to either limit non-specific release of cfDNA or promote the release of cfDNA molecules into the body fluids in question. Third, knowledge of the exact physical and chemical properties of specific cfDNA molecules or the structures with which they become associated will enable the selection, tailoring or development of new extraction procedures that are either biased towards the capture of specific cfDNA molecules, or eliminate non-specific DNA molecules, such as contaminating DNA originating from the lysis of peripheral blood cells. The utility of such biology-informed methods is exemplified by studies that have shown that selective capture of short cfDNA fragments, which have recently been shown to harbor increased amounts of cancer-associated mutations, substantially increases the analytical and diagnostic sensitivity and specificity for the detection of various cancer-specific mutations [220]. Similarly, cancer-specific aberrations in DNA methylation have recently been demonstrated to correlate with physico-chemical changes that are characterized by increased affinity for gold nanoparticles, which indicates the intriguing possibility for the development of extraction methods that selectively purify tumor-derived cfDNA [275]. Similarly, some studies suggest that specific extracellular vesicles (exosomes) carry specific DNA cargo, which represents another possible approach for the selective capture of specific cfDNA molecules [198], [199], [200]. Lastly, many of the different forms in which cfDNA subtypes exist are differently affected by many of the steps that precede extraction and analysis. Although these differences have not yet been thoroughly explored, knowledge of this will enable the selection of preanalytical methods that suit specific study objectives [65].
Funding source: Alexander von Humboldt Foundation
Acknowledgments
Abel Jacobus Bronkhorst was supported by the Georg Forster Research Fellowship Programme of the Alexander von Humboldt Foundation. The financial assistance of the Alexander von Humboldt Foundation is hereby acknowledged.
-
Research funding: This work was funded by Alexander von Humboldt Foundation.
-
Author contributions: All authors have accepted responsibility for the entire content of this manuscript and approved its submission.
-
Competing interests: Authors state no conflict of interest.
-
Informed consent: Not applicable.
-
Ethical approval: Not applicable.
References
1. Aucamp, J, Bronkhorst, AJ, Badenhorst, CPS, Pretorius, PJ. The diverse origins of circulating cell-free DNA in the human body: a critical re-evaluation of the literature. Biol Rev Camb Phil Soc 2018;93:1649–83. https://doi.org/10.1111/brv.12413.Search in Google Scholar
2. Grabuschnig, S, Bronkhorst, AJ, Holdenrieder, S, Rosales Rodriguez, I, Schliep, KP, Schwendenwein, D, et al.. Putative origins of cell-free DNA in humans: a review of active and passive nucleic acid release mechanisms. Int J Mol Sci 2020;21:8062. https://doi.org/10.3390/ijms21218062.Search in Google Scholar
3. Thierry, AR, El Messaoudi, S, Gahan, PB, Anker, P, Stroun, M. Origins, structures, and functions of circulating DNA in oncology. Cancer Metastasis Rev 2016;35:347–76. https://doi.org/10.1007/s10555-016-9629-x.Search in Google Scholar
4. Bronkhorst, AJ, Ungerer, V, Diehl, F, Anker, P, Dor, Y, Fleischhacker, M, et al.. Towards systematic nomenclature for cell-free DNA. Hum Genet 2021;140:565–78. https://doi.org/10.1007/s00439-020-02227-2.Search in Google Scholar
5. Bettegowda, C, Sausen, M, Leary, RJ, Kinde, I, Wang, Y, Bartlett, B, et al.. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 2014;6:224ra24. https://doi.org/10.1093/neuonc/nou206.24.Search in Google Scholar
6. Bronkhorst, AJ, Ungerer, V, Holdenrieder, S. The emerging role of cell-free DNA as a molecular marker for cancer management. Biomol Detect Quantif 2019;17:100087. https://doi.org/10.1016/j.bdq.2019.100087.Search in Google Scholar
7. Wan, JCM, Massie, C, Garcia-Corbacho, J, James, D, Caldas, C, Pacey, S, et al.. Liquid biopsies come of age: clinical applications of circulating tumour DNA. Nat Rev Cancer 2017;17:223–38. https://doi.org/10.1038/nrc.2017.7.Search in Google Scholar
8. Lo, YD, Corbetta, N, Chamberlain, PF, Rai, V, Sargent, IL, Redman, CW, et al.. Presence of fetal DNA in maternal plasma and serum. Lancet 1997;350:485–7. https://doi.org/10.1016/s0140-6736(97)02174-0.Search in Google Scholar
9. Allyse, M, Minear, MA, Berson, E, Sridhar, S, Chandrasekharan, S, Rote, M, et al.. Non-invasive prenatal testing: a review of international implementation and challenges. Int J Womens Health 2015;7:113. https://doi.org/10.2147/ijwh.s67124.Search in Google Scholar PubMed PubMed Central
10. Dondorp, W, De Wert, G, Bombard, Y, Bianchi, DW, Bergmann, C, Borry, P, et al.. Non-invasive prenatal testing for aneuploidy and beyond: challenges of responsible innovation in prenatal screening. Eur J Hum Genet 2015;23:1438–50. https://doi.org/10.1038/ejhg.2015.57.Search in Google Scholar PubMed PubMed Central
11. Bloom, RD, Bromberg, JS, Poggio, ED, Bunnapradist, S, Langone, AJ, Sood, P, et al.. Cell-free DNA and active rejection in kidney allografts. J Am Soc Nephrol 2017;28:2221–32. https://doi.org/10.1681/asn.2016091034.Search in Google Scholar
12. De Vlaminck, I, Valantine, HA, Snyder, TM, Strehl, C, Cohen, G, Luikart, H, et al.. Circulating cell-free DNA enables noninvasive diagnosis of heart transplant rejection. Sci Transl Med 2014;6:241ra77. https://doi.org/10.1126/scitranslmed.3007803.Search in Google Scholar PubMed PubMed Central
13. Schütz, E, Fischer, A, Beck, J, Harden, M, Koch, M, Wuensch, T, et al.. Graft-derived cell-free DNA, a noninvasive early rejection and graft damage marker in liver transplantation: a prospective, observational, multicenter cohort study. PLoS Med 2017;14:e1002286. https://doi.org/10.1371/journal.pmed.1002286.Search in Google Scholar PubMed PubMed Central
14. Sharon, E, Shi, H, Kharbanda, S, Koh, W, Martin, LR, Khush, KK, et al.. Quantification of transplant-derived circulating cell-free DNA in absence of a donor genotype. PLoS Comput Biol 2017;13:e1005629. https://doi.org/10.1371/journal.pcbi.1005629.Search in Google Scholar PubMed PubMed Central
15. Sigdel, TK, Archila, FA, Constantin, T, Prins, SA, Liberto, J, Damm, I, et al.. Optimizing detection of kidney transplant injury by assessment of donor-derived cell-free DNA via massively multiplex PCR. J Clin Med 2019;8:19. https://doi.org/10.3390/jcm8010019.Search in Google Scholar PubMed PubMed Central
16. Snyder, TM, Khush, KK, Valantine, HA, Quake, SR. Universal noninvasive detection of solid organ transplant rejection. Proc Natl Acad Sci USA 2011;108:6229–34. https://doi.org/10.1073/pnas.1013924108.Search in Google Scholar PubMed PubMed Central
17. Rhodes, A, Wort, SJ, Thomas, H, Collinson, P, David, ED. Plasma DNA concentration as a predictor of mortality and sepsis in critically ill patients. Crit Care 2006;10:R60. https://doi.org/10.1186/cc4894.Search in Google Scholar PubMed PubMed Central
18. Ullrich, E, Heidinger, P, Soh, J, Villanova, L, Grabuschnig, S, Bachler, T, et al.. Evaluation of host-based molecular markers for the early detection of human sepsis. J Biotechnol 2020;310:80–8. https://doi.org/10.1016/j.jbiotec.2020.01.013.Search in Google Scholar PubMed
19. Bronkhorst, AJ, Ungerer, V, Holdenrieder, S. Early detection of cancer using circulating tumor DNA: biological, physiological and analytical considerations. Crit Rev Clin Lab Sci 2019;57:1–17. https://doi.org/10.1080/10408363.2019.1700902.Search in Google Scholar PubMed
20. Heitzer, E, Haque, IS, Roberts, CES, Speicher, MR. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat Rev Genet 2019;20:71–88. https://doi.org/10.1038/s41576-018-0071-5.Search in Google Scholar PubMed
21. Fleischhacker, M, Schmidt, B. Circulating nucleic acids (CNAs) and cancer--a survey. Biochim Biophys Acta 2007;1775:181–232. https://doi.org/10.1016/j.bbcan.2006.10.001.Search in Google Scholar PubMed
22. Duvvuri, B, Lood, C. Cell-free DNA as a biomarker in autoimmune rheumatic diseases. Front Immunol 2019;10:502. https://doi.org/10.3389/fimmu.2019.00502.Search in Google Scholar PubMed PubMed Central
23. Tan, EM, Schur, PH, Carr, RI, Kunkel, HG. Deoxybonucleic acid (DNA) and antibodies to DNA in the serum of patients with systemic lupus erythematosus. J Clin Invest 1966;45:1732–40. https://doi.org/10.1172/jci105479.Search in Google Scholar
24. Rumore, PM, Steinman, CR. Endogenous circulating DNA in systemic lupus erythematosus. occurrence as multimeric complexes bound to histone. J Clin Invest 1990;86:69–74. https://doi.org/10.1172/jci114716.Search in Google Scholar
25. Hashimoto, T, Yoshida, K, Hashimoto, N, Nakai, A, Kaneshiro, K, Suzuki, K, et al.. Circulating cell free DNA: a marker to predict the therapeutic response for biological DMARDs in rheumatoid arthritis. Int J Rheumatic Dis 2017;20:722–30. https://doi.org/10.1111/1756-185x.12959.Search in Google Scholar PubMed
26. Mosca, M, Giuliano, T, Cuomo, G, Doveri, M, Tani, C, Curcio, M, et al.. Cell-free DNA in the plasma of patients with systemic sclerosis. Clin Rheumatol 2009;28:1437–40. https://doi.org/10.1007/s10067-009-1245-5.Search in Google Scholar PubMed
27. Gögenur, M, Burcharth, J, Gögenur, 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. https://doi.org/10.1186/s13054-016-1578-9.Search in Google Scholar PubMed PubMed Central
28. Regner, A, Meirelles, LDS, Ikuta, N, Cecchini, A, Simon, D. Prognostic utility of circulating nucleic acids in acute brain injuries. Expert Rev Mol Diagn 2018;18:925–38. https://doi.org/10.1080/14737159.2018.1535904.Search in Google Scholar PubMed
29. Macher, H, Egea-Guerrero, JJ, Revuelto-Rey, J, Gordillo-Escobar, E, Enamorado-Enamorado, J, Boza, A, et al.. Role of early cell-free DNA levels decrease as a predictive marker of fatal outcome after severe traumatic brain injury. Clin Chim Acta 2012;414:12–7. https://doi.org/10.1016/j.cca.2012.08.001.Search in Google Scholar PubMed
30. Chiu, TW, Young, R, Chan, LYS, Burd, A, Lo, DYM. Plasma cell-free DNA as an indicator of severity of injury in burn patients. Clin Chem Lab Med 2006;44:13–7. https://doi.org/10.1515/cclm.2006.003.Search in Google Scholar
31. Lippi, G, Sanchis-Gomar, F, Cervellin, G. Cell-free DNA for diagnosing myocardial infarction: not ready for prime time. Clin Chem Lab Med 2015;53:1895–901. https://doi.org/10.1515/cclm-2015-0252.Search in Google Scholar
32. Balta, S, Demirkol, S, Cakar, M, Karaman, M, Ay, SA, Arslan, Z. Cell-free circulating DNA as a novel biomarker in patients with the acute coronary syndrome. Cardiology 2013;126:122–3. https://doi.org/10.1159/000353266.Search in Google Scholar
33. Outinen, TK, Kuparinen, T, Jylhava, J, Leppanen, S, Mustonen, J, Makela, S, et al.. Plasma cell-free DNA levels are elevated in acute Puumala hantavirus infection. PLoS One 2012;7:e31455. https://doi.org/10.1371/journal.pone.0031455.Search in Google Scholar
34. Bakir, M, Engin, A, Kuskucu, MA, Bakir, S, Gundag, O, Midilli, K. Relationship of plasma cell-free DNA level with mortality and prognosis in patients with Crimean-Congo hemorrhagic fever. J Med Virol 2016;88:1152–8. https://doi.org/10.1002/jmv.24446.Search in Google Scholar
35. Shapiro, B, Chakrabarty, M, Cohn, EM, Leon, SA. Determination of circulating DNA levels in patients with benign or malignant gastrointestinal disease. Cancer 1983;51:2116–20. https://doi.org/10.1002/1097-0142(19830601)51:11<2116::aid-cncr2820511127>3.0.co;2-s.10.1002/1097-0142(19830601)51:11<2116::AID-CNCR2820511127>3.0.CO;2-SSearch in Google Scholar
36. Netz, U, Perry, Z, Mizrahi, S, Kirshtein, B, Czeiger, D, Sebbag, G, et al.. Cell-free deoxyribonucleic acid as a prognostic marker of bowel ischemia in patients with small bowel obstruction. Surgery 2017;162:1063–70. https://doi.org/10.1016/j.surg.2017.06.015.Search in Google Scholar
37. Arnalich, F, Maldifassi, MC, Ciria, E, Quesada, A, Codoceo, R, Herruzo, R, et al.. Association of cell-free plasma DNA with perioperative mortality in patients with suspected acute mesenteric ischemia. Clin Chim Acta 2010;411:1269–74. https://doi.org/10.1016/j.cca.2010.05.017.Search in Google Scholar
38. Celec, P, Vlkova, B, Laukova, L, Babickova, J, Boor, P. Cell-free DNA: the role in pathophysiology and as a biomarker in kidney diseases. Expet Rev Mol Med 2018;20:e1. https://doi.org/10.1017/erm.2017.12.Search in Google Scholar
39. Korabecna, M, Opatrna, S, Wirth, J, Rulcova, K, Eiselt, J, Sefrna, F, et al.. Cell-free plasma DNA during peritoneal dialysis and hemodialysis and in patients with chronic kidney disease. Ann N Y Acad Sci 2008;1137:296–301. https://doi.org/10.1196/annals.1448.014.Search in Google Scholar
40. Avriel, A, Rozenberg, D, Raviv, Y, Heimer, D, Bar-Shai, A, Gavish, R, et al.. Prognostic utility of admission cell-free DNA levels in patients with chronic obstructive pulmonary disease exacerbations. Int J Chronic Obstr Pulm Dis 2016;11:3153–61. https://doi.org/10.2147/copd.s113256.Search in Google Scholar
41. Arnalich, F, Maldifassi, MC, Ciria, E, Codoceo, R, Renart, J, Fernández-Capitán, C, et al.. Plasma levels of mitochondrial and nuclear DNA in patients with massive pulmonary embolism in the emergency department: a prospective cohort study. Crit Care 2013;17:R90. https://doi.org/10.1186/cc12735.Search in Google Scholar PubMed PubMed Central
42. Caglar, O, Cilgin, B, Eroglu, M, Cayir, A. Evaluation of circulating cell free DNA in plasma as a biomarker of different thyroid diseases. Braz J Otorhinolaryngol 2020;86:321–6. https://doi.org/10.1016/j.bjorl.2018.12.008.Search in Google Scholar PubMed PubMed Central
43. Taglauer, ES, Wilkins-Haug, L, Bianchi, DW. Review: cell-free fetal DNA in the maternal circulation as an indication of placental health and disease. Placenta 2014;35:S64–8. https://doi.org/10.1016/j.placenta.2013.11.014.Search in Google Scholar PubMed PubMed Central
44. Vlkova, B, Kalousova, M, Germanova, A, Parizek, A, Hajek, Z, Zima, T, et al.. Cell-free DNA is higher and more fragmented in intrahepatic cholestasis of pregnancy. Prenat Diagn 2016;36:1156–8. https://doi.org/10.1002/pd.4952.Search in Google Scholar PubMed
45. Kashiwada-Nakamura, K, Myangat, TM, Kajihara, I, Kanemaru, H, Sawamura, S, Makino, K, et al.. Circulating janus kinase family DNA levels in psoriasis: elevated JAK2 DNA copy number in cell-free DNA. J Dermatol 2022;49:e106–7.10.1111/1346-8138.16245Search in Google Scholar PubMed
46. Beranek, M, Fiala, Z, Kremlacek, J, Andrys, C, Krejsek, J, Hamakova, K, et al.. Changes in circulating cell-free DNA and nucleosomes in patients with exacerbated psoriasis. Arch Dermatol Res 2017;309:815–21. https://doi.org/10.1007/s00403-017-1785-5.Search in Google Scholar PubMed
47. Glebova, KV, Veiko, NN, Nikonov, AA, Porokhovnik, LN, Kostuyk, SV. Cell-free DNA as a biomarker in stroke: current status, problems and perspectives. Crit Rev Clin Lab Sci 2018;55:55–70. https://doi.org/10.1080/10408363.2017.1420032.Search in Google Scholar PubMed
48. Lam, NYL, Rainer, TH, Wong, LKS, Lam, W, Lo, YMD. Plasma DNA as a prognostic marker for stroke patients with negative neuroimaging within the first 24 h of symptom onset. Resuscitation 2006;68:71–8. https://doi.org/10.1016/j.resuscitation.2005.05.021.Search in Google Scholar PubMed
49. Cayir, A, Coskun, M, Coskun, M, Cobanoglu, H. DNA damage and circulating cell free DNA in greenhouse workers exposed to pesticides. Environ Mol Mutagen 2018;59:161–9. https://doi.org/10.1002/em.22148.Search in Google Scholar PubMed
50. Korzeneva, IB, Kostuyk, SV, Ershova, LS, Osipov, AN, Zhuravleva, VF, Pankratova, GV, et al.. Human circulating plasma DNA significantly decreases while lymphocyte DNA damage increases under chronic occupational exposure to low-dose gamma-neutron and tritium β-radiation. Mutat Res 2015;779:1–15. https://doi.org/10.1016/j.mrfmmm.2015.05.004.Search in Google Scholar PubMed
51. Villalba-Campos, M, Ramirez-Clavijo, SR, Sanchez-Corredor, MC, Rondon-Lagos, M, Ibanez-Pinilla, M, Palma, RM, et al.. Quantification of cell-free DNA for evaluating genotoxic damage from occupational exposure to car paints. J Occup Med Toxicol 2016;11:33. https://doi.org/10.1186/s12995-016-0123-8.Search in Google Scholar PubMed PubMed Central
52. Jiang, J, Chen, X, Sun, L, Qing, Y, Yang, X, Hu, X, et al.. Analysis of the concentrations and size distributions of cell-free DNA in schizophrenia using fluorescence correlation spectroscopy. Transl Psychiatry 2018;8:104. https://doi.org/10.1038/s41398-018-0153-3.Search in Google Scholar PubMed PubMed Central
53. Alapirtti, T, Jylhava, J, Raitanen, J, Makinen, R, Peltola, J, Hurme, MA, et al.. The concentration of cell-free DNA in video-EEG patients is dependent on the epilepsy syndrome and duration of epilepsy. Neurol Res 2016;38:45–50. https://doi.org/10.1080/01616412.2015.1127004.Search in Google Scholar PubMed
54. Li, HG, Huang, SY, Zhou, H, Liao, AH, Xiong, CL. Quick recovery and characterization of cell-free DNA in seminal plasma of normozoospermia and azoospermia: implications for non-invasive genetic utilities. Asian J Androl 2009;11:703–9. https://doi.org/10.1038/aja.2009.65.Search in Google Scholar PubMed PubMed Central
55. Boissiere, A, Gala, A, Ferrieres-Hoa, A, Mullet, T, Baillet, S, Petiton, A, et al.. Cell-free and intracellular nucleic acids: new non-invasive biomarkers to explore male infertility. Basic Clin Androl 2017;27:7. https://doi.org/10.1186/s12610-017-0052-0.Search in Google Scholar PubMed PubMed Central
56. Rule, KN, Chosed, RJ, Chang, TA, Robinson, RD, Wininger, JD, Roudebush, W. Blastocoel cell-free DNA, a marker of embryonic quality. Fertil Steril 2017;108:E106. https://doi.org/10.1016/j.fertnstert.2017.07.324.Search in Google Scholar
57. Czamanski-Cohen, J, Sarid, O, Cwikel, J, Lunenfeld, E, Douvdevani, A, Levitas, E, et al.. Increased plasma cell-free DNA is associated with low pregnancy rates among women undergoing IVF-embryo transfer. Reprod Biomed Online 2013;26:36–41. https://doi.org/10.1016/j.rbmo.2012.09.018.Search in Google Scholar PubMed
58. Czamanski-Cohen, J, Sarid, O, Cwikel, J, Levitas, E, Lunenfeld, E, Douvdevani, A, et al.. Decrease in cell free DNA levels following participation in stress reduction techniques among women undergoing infertility treatment. Arch Womens Ment Health 2014;17:251–3. https://doi.org/10.1007/s00737-013-0407-2.Search in Google Scholar PubMed
59. Davis, GLJr, Davis Iv, JS. Detection of circulating DNA by counterimmunoelectrophoresis (CIE). Arthritis Rheum 1973;16:52–8. https://doi.org/10.1002/art.1780160108.Search in Google Scholar PubMed
60. Hughes, GR, Cohen, SA, Lightfoot, RWJr, Meltzer, JI, Christian, CL. The release of DNA into serum and synovial fluid. Arthritis Rheum 1971;14:259–66. https://doi.org/10.1002/art.1780140211.Search in Google Scholar PubMed
61. Atamaniuk, J, Kopecky, C, Skoupy, S, Saemann, MD, Weichhart, T. Apoptotic cell-free DNA promotes inflammation in haemodialysis patients. Nephrol Dial Transplant 2012;27:902–5. https://doi.org/10.1093/ndt/gfr695.Search in Google Scholar PubMed
62. Tovbin, D, Novack, V, Wiessman, MP, Abd Elkadir, A, Zlotnik, M, Douvdevani, A. Circulating cell-free DNA in hemodialysis patients predicts mortality. Nephrol Dial Transplant 2012;27:3929–35. https://doi.org/10.1093/ndt/gfs255.Search in Google Scholar PubMed
63. Wijeratne, S, Butt, A, Burns, S, Sherwood, K, Boyd, O, Swaminathan, R. Cell-free plasma DNA as a prognostic marker in intensive treatment unit patients. Ann N Y Acad Sci 2004;1022:232–8. https://doi.org/10.1196/annals.1318.036.Search in Google Scholar PubMed
64. Okkonen, M, Lakkisto, P, Korhonen, A-M, Parviai-Nen, I, Reinikainen, M, Varpula, T, et al.. Plasma cell-free DNA in patients needing mechanical ventilation. Crit Care 2011;15:R196. https://doi.org/10.1186/cc10357.Search in Google Scholar PubMed PubMed Central
65. Ungerer, V, Bronkhorst, AJ, Holdenrieder, S. Preanalytical variables that affect the outcome of cell-free DNA measurements. Crit Rev Clin Lab Sci 2020;57:484–507. https://doi.org/10.1080/10408363.2020.1750558.Search in Google Scholar PubMed
66. Yuwono, NL, Warton, K, Ford, CE. The influence of biological and lifestyle factors on circulating cell-free DNA in blood plasma. Elife 2021;10:e69679. https://doi.org/10.7554/elife.69679.Search in Google Scholar
67. 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;132:20–6. https://doi.org/10.1016/j.mad.2010.11.001.Search in Google Scholar PubMed
68. Teo, YV, Capri, M, Morsiani, C, Pizza, G, Faria, AMC, Franceschi, C, et al.. Cell-free DNA as a biomarker of aging. Aging Cell 2019;18:e12890. https://doi.org/10.1111/acel.12890.Search in Google Scholar PubMed PubMed Central
69. Haghiac, M, Vora, NL, Basu, S, Johnson, KL, Presley, L, Bianchi, DW, et al.. Increased death of adipose cells, a path to release cell-free DNA into systemic circulation of obese women. Obesity 2012;20:2213–9. https://doi.org/10.1038/oby.2012.138.Search in Google Scholar PubMed PubMed Central
70. Livergood, MC, LeChien, KA, Trudell, AS. Obesity and cell-free DNA “no calls”: is there an optimal gestational age at time of sampling? Am J Obstet Gynecol 2017;216:413. e1–9. https://doi.org/10.1016/j.ajog.2017.01.011.Search in Google Scholar PubMed
71. Fatouros, IG, Jamurtas, AZ, Nikolaidis, MG, Destouni, A, Michailidis, Y, Vrettou, C, et al.. Time of sampling is crucial for measurement of cell-free plasma DNA following acute aseptic inflammation induced by exercise. Clin Biochem 2010;43:1368–70. https://doi.org/10.1016/j.clinbiochem.2010.08.020.Search in Google Scholar PubMed
72. Madsen, AT, Hojbjerg, JA, Sorensen, BS, Winther-Larsen, A. Day-to-day and within-day biological variation of cell-free DNA. EBioMedicine 2019;49:284–90. https://doi.org/10.1016/j.ebiom.2019.10.008.Search in Google Scholar PubMed PubMed Central
73. Korabecna, M, Horinek, A, Bila, N, Opatrna, S. Circadian rhythmicity and clearance of cell-free DNA in human plasma. Dordrecht: Springer; 2010:195–8 pp.10.1007/978-90-481-9382-0_26Search in Google Scholar
74. Meddeb, R, Dache, ZAA, Thezenas, S, Otandault, A, Tanos, R, Pastor, B, et al.. Quantifying circulating cell-free DNA in humans. Sci Rep 2019;9:5220. https://doi.org/10.1038/s41598-019-41593-4.Search in Google Scholar PubMed PubMed Central
75. Poulet, G, Hulot, J-S, Blanchard, A, Bergerot, D, Xiao, W, Ginot, F, et al.. Circadian rhythm and circulating cell-free DNA release on healthy subjects. 2022 [Preprint].10.21203/rs.3.rs-1270021/v1Search in Google Scholar
76. Neuberger, EW, Brahmer, A, Ehlert, T, Kluge, K, Philippi, KF, Boedecker, SC, et al.. Validating quantitative PCR assays for cfDNA detection without DNA extraction in exercising SLE patients. Sci Rep 2021;11:13581. https://doi.org/10.1038/s41598-021-92826-4.Search in Google Scholar PubMed PubMed Central
77. Schmidt, S, Taenny, P, Petry, J, Haller, N, Simon, P, Helmig, S. Circulating, cell-free DNA as a marker for exercise load in intermittent sports. PLoS One 2018;13:e0191915. https://doi.org/10.1371/journal.pone.0191915.Search in Google Scholar PubMed PubMed Central
78. Breitbach, S, Sterzing, B, Magallanes, C, Tug, S, Simon, P. Direct measurement of cell-free DNA from serially collected capillary plasma during incremental exercise. J Appl Physiol 2014;117:119–30. https://doi.org/10.1152/japplphysiol.00002.2014.Search in Google Scholar PubMed
79. Breitbach, S, Tug, S, Simon, P. Circulating cell-free DNA. Sports Med 2012;42:565–86. https://doi.org/10.2165/11631380-000000000-00000.Search in Google Scholar PubMed
80. Atamaniuk, J, Vidotto, C, Kinzlbauer, M, Bachl, N, Tiran, B, Tschan, H. Cell-free plasma DNA and purine nucleotide degradation markers following weightlifting exercise. Eur J Appl Physiol 2010;110:695–701. https://doi.org/10.1007/s00421-010-1532-5.Search in Google Scholar PubMed
81. Atamaniuk, J, Stuhlmeier, KM, Vidotto, C, Tschan, H, Dossenbach-Glaninger, A, Mueller, MM. Effects of ultra-marathon on circulating DNA and mRNA expression of pro- and anti-apoptotic genes in mononuclear cells. Eur J Appl Physiol 2008;104:711–7. https://doi.org/10.1007/s00421-008-0827-2.Search in Google Scholar PubMed
82. 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;57:633–6. https://doi.org/10.1373/clinchem.2010.158030.Search in Google Scholar PubMed
83. Hsieh, CC, Hsu, HS, Chang, SC, Chen, YJ. Circulating cell-free DNA levels could predict oncological outcomes of patients undergoing esophagectomy for esophageal squamous cell carcinoma. Int J Mol Sci 2016;17:2131. https://doi.org/10.3390/ijms17122131.Search in Google Scholar PubMed PubMed Central
84. Zhong, XY, Hahn, S, Kiefer, V, Holzgreve, W. Is the quantity of circulatory cell-free DNA in human plasma and serum samples associated with gender, age and frequency of blood donations? Ann Hematol 2007;86:139–43. https://doi.org/10.1007/s00277-006-0182-5.Search in Google Scholar PubMed
85. Brodbeck, K, Schick, S, Bayer, B, Anslinger, K, Krüger, K, Mayer, Z, et al.. Biological variability of cell-free DNA in healthy females at rest within a short time course. Int J Leg Med 2020;134:911–9. https://doi.org/10.1007/s00414-019-02240-9.Search in Google Scholar PubMed
86. Breitbach, S, Tug, S, Helmig, S, Zahn, D, Kubiak, T, Michal, M, et al.. Direct quantification of cell-free, circulating DNA from unpurified plasma. PLoS One 2014;9:e87838. https://doi.org/10.1371/journal.pone.0087838.Search in Google Scholar PubMed PubMed Central
87. Yuwono, NL, Henry, CE, Ford, CE, Warton, K. Total and endothelial cell-derived cell-free DNA in blood plasma does not change during menstruation. PLoS One 2021;16:e0250561. https://doi.org/10.1371/journal.pone.0250561.Search in Google Scholar PubMed PubMed Central
88. Pölcher, M, Ellinger, J, Willems, S, El-Maarri, O, Höller, T, Amann, C, et al.. Impact of the menstrual cycle on circulating cell-free DNA. Anticancer Res 2010;30:2235–40.Search in Google Scholar
89. Neuberger, EW, Sontag, S, Brahmer, A, Philippi, KF, Radsak, MP, Wagner, W, et al.. Physical activity specifically evokes release of cell-free DNA from granulocytes thereby affecting liquid biopsy. Clin Epigenet 2022;14:29. https://doi.org/10.1186/s13148-022-01245-3.Search in Google Scholar PubMed PubMed Central
90. Brahmer, A, Neuberger, E, Esch-Heisser, L, Haller, N, Jorgensen, MM, Baek, R, et al.. Platelets, endothelial cells and leukocytes contribute to the exercise-triggered release of extracellular vesicles into the circulation. J Extracell Vesicles 2019;8:1615820. https://doi.org/10.1080/20013078.2019.1615820.Search in Google Scholar PubMed PubMed Central
91. Moss, J, Magenheim, J, Neiman, D, Zemmour, H, Loyfer, N, Korach, A, et al.. Comprehensive human cell-type methylation atlas reveals origins of circulating cell-free DNA in health and disease. Nat Commun 2018;9:5068. https://doi.org/10.1038/s41467-018-07466-6.Search in Google Scholar PubMed PubMed Central
92. 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;36:350–68. https://doi.org/10.1016/j.ccell.2019.09.003.Search in Google Scholar PubMed
93. Bronkhorst, AJ, Aucamp, J, Pretorius, PJ. Cell-free DNA: preanalytical variables. Clin Chim Acta 2015;450:243–53. https://doi.org/10.1016/j.cca.2015.08.028.Search in Google Scholar PubMed
94. Bronkhorst, AJ, Ungerer, V, Holdenrieder, S. Comparison of methods for the isolation of cell-free DNA from cell culture supernatant. Tumor Biol 2020;42:1010428320916314. https://doi.org/10.1177/1010428320916314.Search in Google Scholar PubMed
95. Greytak, SR, Engel, KB, Parpart-Li, S, Murtaza, M, Bronkhorst, AJ, Pertile, MD, et al.. Harmonizing cell-free DNA collection and processing practices through evidence-based guidance. Clin Cancer Res 2020;26:3104–9. https://doi.org/10.1158/1078-0432.ccr-19-3015.Search in Google Scholar
96. Meddeb, R, Pisareva, E, Thierry, AR. Guidelines for the preanalytical conditions for analyzing circulating cell-free DNA. Clin Chem 2019;65:623–33. https://doi.org/10.1373/clinchem.2018.298323.Search in Google Scholar PubMed
97. Colley, E, Devall, AJ, Williams, H, Hamilton, S, Smith, P, Morgan, NV, et al.. Cell-free DNA in the investigation of miscarriage. J Clin Med 2020;9:3428. https://doi.org/10.3390/jcm9113428.Search in Google Scholar PubMed PubMed Central
98. Hui, L, Bianchi, DW. Fetal fraction and noninvasive prenatal testing: what clinicians need to know. Prenat Diagn 2020;40:155–63. https://doi.org/10.1002/pd.5620.Search in Google Scholar PubMed
99. Hui, L. Noninvasive approaches to prenatal diagnosis: historical perspective and future directions. Methods Mol Biol 2019;1885:45–58.10.1007/978-1-4939-8889-1_3Search in Google Scholar PubMed
100. Chitty, LS, Lo, YD. Noninvasive prenatal screening for genetic diseases using massively parallel sequencing of maternal plasma DNA. Cold Spring Harb Perspect Med 2015;5:a023085. https://doi.org/10.1101/cshperspect.a023085.Search in Google Scholar PubMed PubMed Central
101. Lo, YMD, Tein, MSC, Lau, TK, Haines, CJ, Leung, TN, Poon, PMK, et al.. Quantitative analysis of fetal DNA in maternal plasma and serum: implications for noninvasive prenatal diagnosis. Am J Hum Genet 1998;62:768–75. https://doi.org/10.1086/301800.Search in Google Scholar PubMed PubMed Central
102. Yamamoto, M, Ushio, R, Watanabe, H, Tachibana, T, Tanaka, M, Yokose, T, et al.. Detection of mycobacterium tuberculosis-derived DNA in circulating cell-free DNA from a patient with disseminated infection using digital PCR. Int J Infect Dis 2018;66:80–2. https://doi.org/10.1016/j.ijid.2017.11.018.Search in Google Scholar PubMed
103. Langelier, C, Fung, M, Caldera, S, Deiss, T, Lyden, A, Prince, BC, et al.. Detection of pneumonia pathogens from plasma cell-free DNA. Am J Respir Crit Care Med 2020;201:491–5. https://doi.org/10.1164/rccm.201904-0905le.Search in Google Scholar
104. Weerakoon, KG, McManus, DP. Cell-free DNA as a diagnostic tool for human parasitic infections. Trends Parasitol 2016;32:378–91. https://doi.org/10.1016/j.pt.2016.01.006.Search in Google Scholar PubMed
105. Vera, IM, Kessler, A, Ting, LM, Harawa, V, Keller, T, Allen, D, et al.. Plasma cell-free DNA predicts pediatric cerebral malaria severity. JCI Insight 2020;5:e136279. https://doi.org/10.1172/jci.insight.136279.Search in Google Scholar PubMed PubMed Central
106. Armstrong, AE, Rossoff, J, Hollemon, D, Hong, DK, Muller, WJ, Chaudhury, S. Cell-free DNA next-generation sequencing successfully detects infectious pathogens in pediatric oncology and hematopoietic stem cell transplant patients at risk for invasive fungal disease. Pediatr Blood Cancer 2019;66:e27734. https://doi.org/10.1002/pbc.27734.Search in Google Scholar PubMed
107. Chan, KCA, Woo, JKS, King, A, Zee, BCY, Lam, WKJ, Chan, SL, et al.. Analysis of plasma epstein–barr virus DNA to screen for nasopharyngeal cancer. N Engl J Med 2017;377:513–22. https://doi.org/10.1056/nejmoa1701717.Search in Google Scholar
108. Chan, KCA, Leung, SF, Yeung, SW, Chan, ATC, Lo, YMD. Quantitative analysis of the transrenal excretion of circulating EBV DNA in nasopharyngeal carcinoma patients. Clin Cancer Res 2008;14:4809–13. https://doi.org/10.1158/1078-0432.ccr-08-1112.Search in Google Scholar
109. Wuerdemann, N, Jain, R, Adams, A, Speel, EM, Wagner, S, Joosse, SA, et al.. Cell-free HPV-DNA as a biomarker for oropharyngeal squamous cell carcinoma – a step towards personalized medicine? Cancers 2020;12:2997. https://doi.org/10.3390/cancers12102997.Search in Google Scholar PubMed PubMed Central
110. Hanna, GJ, Supplee, JG, Kuang, Y, Mahmood, U, Lau, CJ, Haddad, RI, et al.. Plasma HPV cell-free DNA monitoring in advanced HPV-associated oropharyngeal cancer. Ann Oncol 2018;29:1980–6. https://doi.org/10.1093/annonc/mdy251.Search in Google Scholar PubMed
111. Kang, Z, Stevanovic, S, Hinrichs, CS, Cao, L. Circulating cell-free DNA for metastatic cervical cancer detection, genotyping, and monitoring. Clin Cancer Res 2017;23:6856–62. https://doi.org/10.1158/1078-0432.ccr-17-1553.Search in Google Scholar
112. Burnham, P, Kim, MS, Agbor-Enoh, S, Luikart, H, Valantine, HA, Khush, KK, et al.. Single-stranded DNA library preparation uncovers the origin and diversity of ultrashort cell-free DNA in plasma. Sci Rep 2016;6:27859. https://doi.org/10.1038/srep27859.Search in Google Scholar PubMed PubMed Central
113. Chiu, KP, Alice, LY. Application of cell-free DNA sequencing in characterization of bloodborne microbes and the study of microbe-disease interactions. PeerJ 2019;7:e7426. https://doi.org/10.7717/peerj.7426.Search in Google Scholar PubMed PubMed Central
114. Kowarsky, M, Camunas-Soler, J, Ma, K. Numerous uncharacterized and highly divergent microbes which colonize humans are revealed by circulating cell-free DNA. Proc Natl Acad Sci USA 2017;114:9623–8. https://doi.org/10.1073/pnas.1707009114.Search in Google Scholar PubMed PubMed Central
115. Stroun, M, Anker, P. Bacterial nucleic acid synthesis in plants following bacterial contact. Mol Gen Genet 1971;113:92–8. https://doi.org/10.1007/bf00335008.Search in Google Scholar
116. Spisák, S, Solymosi, N, Ittzés, P, Bodor, A, Kondor, D, Vattay, G, et al.. Complete genes may pass from food to human blood. PLoS One 2013;8:e69805. https://doi.org/10.1371/journal.pone.0069805.Search in Google Scholar PubMed PubMed Central
117. Woegerbauer, M, Bellanger, X, Merlin, C. Cell-free DNA: an underestimated source of antibiotic resistance gene dissemination at the interface between human activities and downstream environments in the context of wastewater reuse. Front Microbiol 2020;11. Available from: https://doi.org/10.3389/fmicb.2020.00671.Search in Google Scholar PubMed PubMed Central
118. Mittra, I, Samant, U, Sharma, S, Raghuram, GV, Saha, T, Tidke, P, et al.. Cell-free chromatin from dying cancer cells integrate into genomes of bystander healthy cells to induce DNA damage and inflammation. Cell Death Dis 2017;3:17015. https://doi.org/10.1038/cddiscovery.2017.15.Search in Google Scholar PubMed PubMed Central
119. Mittra, I, Khare, NK, Raghuram, GV, Chaubal, R, Khambatti, F, Gupta, D, et al.. Circulating nucleic acids damage DNA of healthy cells by integrating into their genomes. J Biosci 2015;40:91–111. https://doi.org/10.1007/s12038-015-9508-6.Search in Google Scholar PubMed PubMed Central
120. Basak, R, Nair, NK, Mittra, I. Evidence for cell-free nucleic acids as continuously arising endogenous DNA mutagens. Mutat Res 2016;793–794:15–21. https://doi.org/10.1016/j.mrfmmm.2016.10.002.Search in Google Scholar PubMed
121. Raghuram, GV, Gupta, D, Subramaniam, S, Gaikwad, A, Khare, NK, Nobre, M, et al.. Physical shearing imparts biological activity to DNA and ability to transmit itself horizontally across species and kingdom boundaries. BMC Mol Biol 2017;18:21. https://doi.org/10.1186/s12867-017-0098-8.Search in Google Scholar PubMed PubMed Central
122. Gahan, P, Wyndaele, R, Mantell, S, Boggetti, B. Evidence that direct DNA uptake through cut shoots leads to genetic transformation of solanum aviculare forst. Cell Biochem Funct 2003;21:11–7. https://doi.org/10.1002/cbf.986.Search in Google Scholar PubMed
123. Cesana, D, Calabria, A, Rudilosso, L, Gallina, P, Benedicenti, F, Spinozzi, G, et al.. Retrieval of vector integration sites from cell-free DNA. Nat Med 2021;27:1458–70. https://doi.org/10.1038/s41591-021-01389-4.Search in Google Scholar PubMed
124. Albano, MS, Scaradavou, A, Stevens, CE, Rubinstein, P. Extracellular DNA in cord blood plasma and applications in cord blood banking for sample identification. Transfusion 2009;49:1685–91. https://doi.org/10.1111/j.1537-2995.2009.02168.x.Search in Google Scholar PubMed
125. Assou, S, At-Ahmed, O. Non-invasive pre-implantation genetic diagnosis of X-linked disorders. Med Hypotheses 2014;83:506–8. https://doi.org/10.1016/j.mehy.2014.08.019.Search in Google Scholar PubMed
126. Feichtinger, M, Vaccari, E, Carli, L, Wallner, E, Mädel, U, Figl, K, et al.. Non-invasive preimplantation genetic screening using array comparative genomic hybridization on spent culture media: a proof-of-concept pilot study. Reprod Biomed Online 2017;34:583–9. https://doi.org/10.1016/j.rbmo.2017.03.015.Search in Google Scholar PubMed
127. Zenner, K, Jensen, DM, Cook, TT, Dmyterko, V, Bly, RA, Ganti, S, et al.. Cell-free DNA as a diagnostic analyte for molecular diagnosis of vascular malformations. Genet Med 2021;23:123–30. https://doi.org/10.1038/s41436-020-00943-8.Search in Google Scholar PubMed PubMed Central
128. Quinones, I, Daniel, B. Cell free DNA as a component of forensic evidence recovered from touched surfaces. Forensic Sci Int Genet 2012;6:26–30. https://doi.org/10.1016/j.fsigen.2011.01.004.Search in Google Scholar PubMed
129. Sewell, J, Quinones, I, Ames, C, Multaney, B, Curtis, S, Seeboruth, H, et al.. Recovery of DNA and fingerprints from touched documents. Forensic Sci Int Genet 2008;2:281–5. https://doi.org/10.1016/j.fsigen.2008.03.006.Search in Google Scholar PubMed
130. Vandewoestyne, M, Van Hoofstat, D, Franssen, A, Van Nieuwerburgh, F, Deforce, D. Presence and potential of cell free DNA in different types of forensic samples. Forensic Sci Int Genet 2013;7:316–20. https://doi.org/10.1016/j.fsigen.2012.12.005.Search in Google Scholar PubMed
131. Chiu, RWK, Chan, LYS, Lam, NYL, Tsui, NBY, Ng, EKO, Rainer, TH, et al.. Quantitative analysis of circulating mitochondrial DNA in plasma. Clin Chem 2003;49:719–26. https://doi.org/10.1373/49.5.719.Search in Google Scholar PubMed
132. Al Amir Dache, Z, Otandault, A, Tanos, R, Pastor, B, Meddeb, R, Sanchez, C, et al.. Blood contains circulating cell-free respiratory competent mitochondria. Faseb J 2020;34:3616–30. https://doi.org/10.1096/fj.201901917rr.Search in Google Scholar PubMed
133. Yu, M. Circulating cell-free mitochondrial DNA as a novel cancer biomarker: opportunities and challenges. Mitochondrial DNA 2012;23:329–32. https://doi.org/10.3109/19401736.2012.696625.Search in Google Scholar PubMed
134. An, Q, Hu, Y, Li, Q, Chen, X, Huang, J, Pellegrini, M, et al.. The size of cell-free mitochondrial DNA in blood is inversely correlated with tumor burden in cancer patients. Precis Clin Med 2019;2:131–9. https://doi.org/10.1093/pcmedi/pbz014.Search in Google Scholar PubMed PubMed Central
135. Kohler, C, Radpour, R, Barekati, Z, Asadollahi, R, Bitzer, J, Wight, E, et al.. Levels of plasma circulating cell free nuclear and mitochondrial DNA as potential biomarkers for breast tumors. Mol Cancer 2009;8:105. https://doi.org/10.1186/1476-4598-8-105.Search in Google Scholar PubMed PubMed Central
136. Yu, M, Wan, YF, Zou, QH. Cell-free circulating mitochondrial DNA in the serum: a potential non-invasive biomarker for ewing’s sarcoma. Arch Med Res 2012;43:389–94. https://doi.org/10.1016/j.arcmed.2012.06.007.Search in Google Scholar PubMed
137. Ellinger, J, Müller, DC, Müller, SC, Hauser, S, Heukamp, LC, von Ruecker, A, et al.., editors. Circulating mitochondrial DNA in serum: a universal diagnostic biomarker for patients with urological malignancies. Urol Oncol 2012;30:509–15.10.1016/j.urolonc.2010.03.004Search in Google Scholar PubMed
138. Kandel, ES. Mutations in circulating mitochondrial DNA: cassandra of oral cancer? Oncotarget 2012;3:664. https://doi.org/10.18632/oncotarget.567.Search in Google Scholar PubMed PubMed Central
139. Uzawa, K, Baba, T, Uchida, F, Yamatoji, M, Kasamatsu, A, Sakamoto, Y, et al.. Circulating tumor-derived mutant mitochondrial DNA: a predictive biomarker of clinical prognosis in human squamous cell carcinoma. Oncotarget 2012;3:670. https://doi.org/10.18632/oncotarget.523.Search in Google Scholar PubMed PubMed Central
140. Kumar, M, Srivastava, S, Singh, SA, Das, AK, Das, GC, Dhar, B, et al.. Cell-free mitochondrial DNA copy number variation in head and neck squamous cell carcinoma: a study of non-invasive biomarker from Northeast India. Tumor Biol 2017;39:1010428317736643. https://doi.org/10.1177/1010428317736643.Search in Google Scholar PubMed
141. Huang, CY, Chen, YM, Wu, CH, Tsai, CM, Lee, YC, Perng, RP, et al.. Circulating free mitochondrial DNA concentration and its association with erlotinib treatment in patients with adenocarcinoma of the lung. Oncol Lett 2014;7:2180–4. https://doi.org/10.3892/ol.2014.2006.Search in Google Scholar PubMed PubMed Central
142. Berezin, AE. The cell-free mitochondrial DNA: a novel biomarker of cardiovascular risk? Transl Biomed 2016;7. doi: https://doi.org/10.21767/2172-0479.100068.Search in Google Scholar
143. Berezin, AE. Circulating cell-free mitochondrial DNA as biomarker of cardiovascular risk: new challenges of old findings. Angiology 2015;03:161. https://doi.org/10.4172/2329-9495.1000161.Search in Google Scholar
144. Arnalich, F, Codoceo, R, Lopez-Collazo, E, Montiel, C. Circulating cell-free mitochondrial DNA: a better early prognostic marker in patients with out-of-hospital cardiac arrest. Resuscitation 2012;83:e162–3. https://doi.org/10.1016/j.resuscitation.2012.03.032.Search in Google Scholar PubMed
145. Pinti, M, Cevenini, E, Nasi, M, De Biasi, S, Salvioli, S, Monti, D, et al.. Circulating mitochondrial DNA increases with age and is a familiar trait: implications for “inflamm-aging”. Eur J Immunol 2014;44:1552–62. https://doi.org/10.1002/eji.201343921.Search in Google Scholar PubMed
146. Lowes, H, Kurzawa-Akanbi, M, Pyle, A, Hudson, G. Post-mortem ventricular cerebrospinal fluid cell-free-mtDNA in neurodegenerative disease. Sci Rep 2020;10:15253. https://doi.org/10.1038/s41598-020-72190-5.Search in Google Scholar PubMed PubMed Central
147. Varhaug, KN, Vedeler, CA, Myhr, KM, Aarseth, JH, Tzoulis, C, Bindoff, LA. Increased levels of cell-free mitochondrial DNA in the cerebrospinal fluid of patients with multiple sclerosis. Mitochondrion 2017;34:32–5. https://doi.org/10.1016/j.mito.2016.12.003.Search in Google Scholar PubMed
148. Lowes, H, Pyle, A, Santibanez-Koref, M, Hudson, G. Circulating cell-free mitochondrial DNA levels in Parkinson’s disease are influenced by treatment. Mol Neurodegener 2020;15:10. https://doi.org/10.1186/s13024-020-00362-y.Search in Google Scholar PubMed PubMed Central
149. Dantham, S, Srivastava, AK, Gulati, S, Rajeswari, MR. Plasma circulating cell-free mitochondrial DNA in the assessment of Friedreich’s ataxia. J Neurol Sci 2016;365:82–8. https://doi.org/10.1016/j.jns.2016.04.016.Search in Google Scholar PubMed
150. Zhong, S, Ng, MC, Lo, YM, Chan, JC, Johnson, PJ. Presence of mitochondrial tRNA(Leu(UUR)) A to G 3243 mutation in DNA extracted from serum and plasma of patients with type 2 diabetes mellitus. J Clin Pathol 2000;53:466–9. https://doi.org/10.1136/jcp.53.6.466.Search in Google Scholar PubMed PubMed Central
151. Silzer, T, Barber, R, Sun, J, Pathak, G, Johnson, L, O’Bryant, S, et al.. Circulating mitochondrial DNA: new indices of type 2 diabetes-related cognitive impairment in Mexican Americans. PLoS One 2019;14:e0213527. https://doi.org/10.1371/journal.pone.0213527.Search in Google Scholar PubMed PubMed Central
152. Nakahira, K, Kyung, SY, Rogers, AJ, Gazourian, L, Youn, S, Massaro, AF, et al.. Circulating mitochondrial DNA in patients in the ICU as a marker of mortality: derivation and validation. PLoS Med 2013;10:e1001577. https://doi.org/10.1371/journal.pmed.1001577.Search in Google Scholar PubMed PubMed Central
153. Thurairajah, K, Briggs, GD, Balogh, ZJ. The source of cell-free mitochondrial DNA in trauma and potential therapeutic strategies. Eur J Trauma Emerg Surg 2018;44:325–34. https://doi.org/10.1007/s00068-018-0954-3.Search in Google Scholar PubMed PubMed Central
154. Timmermans, K, Kox, M, Scheffer, GJ, Pickkers, P. Plasma nuclear and mitochondrial DNA levels, and markers of inflammation, shock, and organ damage in patients with septic shock. Shock 2016;45:607–12. https://doi.org/10.1097/shk.0000000000000549.Search in Google Scholar
155. Yan, HP, Li, M, Lu, XL, Zhu, YM, Ou-Yang, WX, Xiao, ZH, et al.. Use of plasma mitochondrial DNA levels for determining disease severity and prognosis in pediatric sepsis: a case control study. BMC Pediatr 2018;18:267. https://doi.org/10.1186/s12887-018-1239-z.Search in Google Scholar PubMed PubMed Central
156. Arshad, O, Gadawska, I, Sattha, B, Cote, HCF, Hsieh, AYY, Canadian Institutes of Health Research Team on Cellular A, et al.. Elevated cell-free mitochondrial DNA in filtered plasma is associated with HIV infection and inflammation. J Acquir Immune Defic Syndr 2018;78:111–8. https://doi.org/10.1097/qai.0000000000001650.Search in Google Scholar
157. Mehta, SR, Perez-Santiago, J, Hulgan, T, Day, TR, Barnholtz-Sloan, J, Gittleman, H, et al.. Cerebrospinal fluid cell-free mitochondrial DNA is associated with HIV replication, iron transport, and mild HIV-associated neurocognitive impairment. J Neuroinflammation 2017;14:72. https://doi.org/10.1186/s12974-017-0848-z.Search in Google Scholar PubMed PubMed Central
158. Perez-Santiago, J, Schrier, RD, de Oliveira, MF, Gianella, S, Var, SR, Day, TR, et al.. Cell-free mitochondrial DNA in CSF is associated with early viral rebound, inflammation, and severity of neurocognitive deficits in HIV infection. J Neurovirol 2016;22:191–200. https://doi.org/10.1007/s13365-015-0384-5.Search in Google Scholar PubMed PubMed Central
159. Bae, JH, Jo, SI, Kim, SJ, Lee, JM, Jeong, JH, Kang, JS, et al.. Circulating cell-free mtDNA contributes to AIM2 inflammasome-mediated chronic inflammation in patients with type 2 diabetes. Cells 2019;8:328. https://doi.org/10.3390/cells8040328.Search in Google Scholar PubMed PubMed Central
160. Budnik, LT, Kloth, S, Baur, X, Preisser, AM, Schwarzenbach, H. Circulating mitochondrial DNA as biomarker linking environmental chemical exposure to early preclinical lesions elevation of mtDNA in human serum after exposure to carcinogenic halo-alkane-based pesticides. PLoS One 2013;8:e64413. https://doi.org/10.1371/journal.pone.0064413.Search in Google Scholar PubMed PubMed Central
161. Bisserier, M, Shanmughapriya, S, Rai, AK, Gonzalez, C, Brojakowska, A, Garikipati, VNS, et al.. Cell-free mitochondrial DNA as a potential biomarker for astronauts’ health. J Am Heart Assoc 2021;10:e022055. https://doi.org/10.1161/jaha.121.022055.Search in Google Scholar PubMed PubMed Central
162. Bezdan, D, Grigorev, K, Meydan, C, Pelissier Vatter, FA, Cioffi, M, Rao, V, et al.. Cell-free DNA (cfDNA) and exosome profiling from a year-long human spaceflight reveals circulating biomarkers. iScience 2020;23:101844. https://doi.org/10.1016/j.isci.2020.101844.Search in Google Scholar PubMed PubMed Central
163. Lu, CH, Chang, WN, Tsai, NW, Chuang, YC, Huang, CR, Wang, HC. The value of serial plasma nuclear and mitochondrial DNA levels in adult community-acquired bacterial meningitis. QJM 2010;103:169–75. https://doi.org/10.1093/qjmed/hcp201.Search in Google Scholar PubMed
164. Ohlsson, L, Hall, A, Lindahl, H, Danielsson, R, Gustafsson, A, Lavant, E, et al.. Increased level of circulating cell-free mitochondrial DNA due to a single bout of strenuous physical exercise. Eur J Appl Physiol 2020;120:897–905. https://doi.org/10.1007/s00421-020-04330-8.Search in Google Scholar PubMed PubMed Central
165. Shockett, PE, Khanal, J, Sitaula, A, Oglesby, C, Meachum, WA, Castracane, VD, et al.. Plasma cell-free mitochondrial DNA declines in response to prolonged moderate aerobic exercise. Phys Rep 2016;4:e12672. https://doi.org/10.14814/phy2.12672.Search in Google Scholar PubMed PubMed Central
166. Newell, C, Hume, S, Greenway, SC, Podemski, L, Shearer, J, Khan, A. Plasma-derived cell-free mitochondrial DNA: a novel non-invasive methodology to identify mitochondrial DNA haplogroups in humans. Mol Genet Metabol 2018;125:332–7. https://doi.org/10.1016/j.ymgme.2018.10.002.Search in Google Scholar PubMed
167. Hummel, E, Hessas, E, Müller, S, Beiter, T, Fisch, M, Eibl, A, et al.. Cell-free DNA release under psychosocial and physical stress conditions. Transl Psychiatry 2018;8:236. https://doi.org/10.1038/s41398-018-0264-x.Search in Google Scholar PubMed PubMed Central
168. Trumpff, C, Marsland, AL, Basualto-Alarcón, C, Martin, JL, Carroll, JE, Sturm, G, et al.. Acute psychological stress increases serum circulating cell-free mitochondrial DNA. Psychoneuroendocrinology 2019;106:268–76. https://doi.org/10.1016/j.psyneuen.2019.03.026.Search in Google Scholar PubMed PubMed Central
169. Lindqvist, D, Fernstrom, J, Grudet, C, Ljunggren, L, Traskman-Bendz, L, Ohlsson, L, et al.. Increased plasma levels of circulating cell-free mitochondrial DNA in suicide attempters: associations with HPA-axis hyperactivity. Transl Psychiatry 2016;6:e971. https://doi.org/10.1038/tp.2016.236.Search in Google Scholar PubMed PubMed Central
170. Lindqvist, D, Wolkowitz, OM, Picard, M, Ohlsson, L, Bersani, FS, Fernstrom, J, et al.. Circulating cell-free mitochondrial DNA, but not leukocyte mitochondrial DNA copy number, is elevated in major depressive disorder. Neuropsychopharmacology 2018;43:1557–64. https://doi.org/10.1038/s41386-017-0001-9.Search in Google Scholar PubMed PubMed Central
171. Cisneros-Villanueva, M, Hidalgo-Pérez, L, Rios-Romero, M, Cedro-Tanda, A, Ruiz-Villavicencio, C, Page, K, et al.. Cell-free DNA analysis in current cancer clinical trials: a review. Br J Cancer 2022;126:391–400. https://doi.org/10.1038/s41416-021-01696-0.Search in Google Scholar PubMed PubMed Central
172. Hu, Y, Ulrich, BC, Supplee, J, Kuang, Y, Lizotte, PH, Feeney, NB, et al.. False-positive plasma genotyping due to clonal hematopoiesis. Clin Cancer Res 2018;24:4437–43. https://doi.org/10.1158/1078-0432.ccr-18-0143.Search in Google Scholar
173. Anglesio, MS, Papadopoulos, N, Ayhan, A, Nazeran, TM, Noe, M, Horlings, HM, et al.. Cancer-associated mutations in endometriosis without cancer. N Engl J Med 2017;376:1835–48. https://doi.org/10.1056/nejmoa1614814.Search in Google Scholar
174. Fernandez-Cuesta, L, Perdomo, S, Avogbe, PH, Leblay, N, Delhomme, TM, Gaborieau, V, et al.. Identification of circulating tumor DNA for the early detection of small-cell lung cancer. EBioMedicine 2016;10:117–23. https://doi.org/10.1016/j.ebiom.2016.06.032.Search in Google Scholar PubMed PubMed Central
175. Genovese, G, Kahler, AK, Handsaker, RE, Lindberg, J, Rose, SA, Bakhoum, SF, et al.. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med 2014;371:2477–87. https://doi.org/10.1056/nejmoa1409405.Search in Google Scholar PubMed PubMed Central
176. Gormally, E, Vineis, P, Matullo, G, Veglia, F, Caboux, E, Le Roux, E, et al.. TP53 and KRAS2 mutations in plasma DNA of healthy subjects and subsequent cancer occurrence: a prospective study. Cancer Res 2006;66:6871–6. https://doi.org/10.1158/0008-5472.can-05-4556.Search in Google Scholar
177. Scherer, F, Kurtz, DM, Newman, AM, Stehr, H, Craig, AFM, Esfahani, MS, et al.. Distinct biological subtypes and patterns of genome evolution in lymphoma revealed by circulating tumor DNA. Sci Transl Med 2016;8:364ra155. https://doi.org/10.1126/scitranslmed.aai8545.Search in Google Scholar PubMed PubMed Central
178. Newman, AM, Lovejoy, AF, Klass, DM, Kurtz, DM, Chabon, JJ, Scherer, F, et al.. Integrated digital error suppression for improved detection of circulating tumor DNA. Nat Biotechnol 2016;34:547–55. https://doi.org/10.1038/nbt.3520.Search in Google Scholar PubMed PubMed Central
179. Marass, F, Stephens, D, Ptashkin, R, Zehir, A, Berger, MF, Solit, DB, et al.. Fragment size analysis may distinguish clonal hematopoiesis from tumor-derived mutations in cell-free DNA. Clin Chem 2020;66:616–8. https://doi.org/10.1093/clinchem/hvaa026.Search in Google Scholar PubMed PubMed Central
180. Bolton, KL, Koh, Y, Foote, MB, Im, H, Jee, J, Sun, CH, et al.. Clonal hematopoiesis is associated with risk of severe Covid-19. Nat Commun 2021;12:5975. https://doi.org/10.1038/s41467-021-26138-6.Search in Google Scholar PubMed PubMed Central
181. Chan, HT, Chin, YM, Nakamura, Y, Low, SK. Clonal hematopoiesis in liquid biopsy: from biological noise to valuable clinical implications. Cancers 2020;12:2277. https://doi.org/10.3390/cancers12082277.Search in Google Scholar PubMed PubMed Central
182. Haselmann, V, Ahmad-Nejad, P, Geilenkeuser, WJ, Duda, A, Gabor, M, Eichner, R, et al.. Results of the first external quality assessment scheme (EQA) for isolation and analysis of circulating tumour DNA (ctDNA). Clin Chem Lab Med 2018;56:220–8. https://doi.org/10.1515/cclm-2017-0283.Search in Google Scholar PubMed
183. Angeles, AK, Janke, F, Bauer, S, Christopoulos, P, Riediger, AL, Sültmann, H. Liquid biopsies beyond mutation calling: genomic and epigenomic features of cell-free DNA in cancer. Cancers 2021;13:5615. https://doi.org/10.3390/cancers13225615.Search in Google Scholar PubMed PubMed Central
184. Lo, YD, Han, DS, Jiang, P, Chiu, RW. Epigenetics, fragmentomics, and topology of cell-free DNA in liquid biopsies. Science 2021;372:eaaw3616. https://doi.org/10.1126/science.aaw3616.Search in Google Scholar PubMed
185. Gai, W, Sun, K. Epigenetic biomarkers in cell-free DNA and applications in liquid biopsy. Genes 2019;10:32. https://doi.org/10.3390/genes10010032.Search in Google Scholar PubMed PubMed Central
186. Sin, ST, Jiang, P, Deng, J, Ji, L, Cheng, SH, Dutta, A, et al.. Identification and characterization of extrachromosomal circular DNA in maternal plasma. Proc Natl Acad Sci USA 2020;117:1658–65. https://doi.org/10.1073/pnas.1914949117.Search in Google Scholar PubMed PubMed Central
187. Zhu, J, Zhang, F, Du, M, Zhang, P, Fu, S, Wang, L. Molecular characterization of cell-free eccDNAs in human plasma. Sci Rep 2017;7:10968. https://doi.org/10.1038/s41598-017-11368-w.Search in Google Scholar PubMed PubMed Central
188. Kumar, P, Dillon, LW, Shibata, Y, Jazaeri, AA, Jones, DR, Dutta, A. Normal and cancerous tissues release extrachromosomal circular DNA (eccDNA) into the circulation. Mol Cancer Res 2017;15:1197–205. https://doi.org/10.1158/1541-7786.mcr-17-0095.Search in Google Scholar PubMed PubMed Central
189. de Vries, JC, Barendrecht, AD, Clark, CC, Urbanus, RT, Boross, P, de Maat, S, et al.. Heparin forms polymers with cell-free DNA which elongate under shear in flowing blood. Sci Rep 2019;9:18316. https://doi.org/10.1038/s41598-019-54818-3.Search in Google Scholar PubMed PubMed Central
190. Bendas, G, Borsig, L. Cancer cell adhesion and metastasis: selectins, integrins, and the inhibitory potential of heparins. Int J Cell Biol 2012;2012:676731. https://doi.org/10.1155/2012/676731.Search in Google Scholar PubMed PubMed Central
191. Gockel, LM, Heyes, M, Li, H, Al Nahain, A, Gorzelanny, C, Schlesinger, M, et al.. Inhibition of tumor-host cell interactions using synthetic heparin mimetics. ACS Appl Mater Interfaces 2021;13:7080–93. https://doi.org/10.1021/acsami.0c20744.Search in Google Scholar PubMed
192. Bronkhorst, AJ, Wentzel, JF, Ungerer, V, Peters, DL, Aucamp, J, de Villiers, EP, et al.. Sequence analysis of cell-free DNA derived from cultured human bone osteosarcoma (143B) cells. Tumour Biol 2018;40:1010428318801190. https://doi.org/10.1177/1010428318801190.Search in Google Scholar PubMed
193. Olsson, I, Ronquist, G. Nucleic acid association to human prostasomes. Arch Androl 1990;24:1–10. https://doi.org/10.3109/01485019008986852.Search in Google Scholar PubMed
194. Ronquist, KG, Ronquist, G, Carlsson, L, Larsson, A. Human prostasomes contain chromosomal DNA. Prostate 2009;69:737–43. https://doi.org/10.1002/pros.20921.Search in Google Scholar PubMed
195. Guescini, M, Genedani, S, Stocchi, V, Agnati, LF. Astrocytes and glioblastoma cells release exosomes carrying mtDNA. J Neural Transm 2010;117:1–4. https://doi.org/10.1007/s00702-009-0288-8.Search in Google Scholar PubMed
196. Kalluri, R, LeBleu, VS, editors. Discovery of double-stranded genomic DNA in circulating exosomes. Cold Spring Harbor symposia on quantitative biology. New York: Cold Spring Harbor Laboratory Press; 2016:275–80 pp.10.1101/sqb.2016.81.030932Search in Google Scholar PubMed
197. Kahlert, C, Melo, SA, Protopopov, A, Tang, J, Seth, S, Koch, M, et al.. Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J Biol Chem 2014;289:3869–75. https://doi.org/10.1074/jbc.c113.532267.Search in Google Scholar PubMed PubMed Central
198. Sansone, P, Savini, C, Kurelac, I, Chang, Q, Amato, LB, Strillacci, A, et al.. Packaging and transfer of mitochondrial DNA via exosomes regulate escape from dormancy in hormonal therapy-resistant breast cancer. Proc Natl Acad Sci USA 2017;114:E9066–75. https://doi.org/10.1073/pnas.1704862114.Search in Google Scholar PubMed PubMed Central
199. Balaj, L, Lessard, R, Dai, L, Cho, YJ, Pomeroy, SL, Breakefield, XO, et al.. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat Commun 2011;2:180. https://doi.org/10.1038/ncomms1180.Search in Google Scholar PubMed PubMed Central
200. Waldenström, A, Gennebäck, N, Hellman, U, Ronquist, G. Cardiomyocyte microvesicles contain DNA/RNA and convey biological messages to target cells. PLoS One 2012;7:e34653. https://doi.org/10.1371/journal.pone.0034653.Search in Google Scholar PubMed PubMed Central
201. Yang, S, Che, SPY, Kurywchak, P, Tavormina, JL, LBa, G. Detection of mutant KRAS and TP53 DNA in circulating exosomes from healthy individuals and patients with pancreatic cancer. Cancer Biol Ther 2017;18:158–65. https://doi.org/10.1080/15384047.2017.1281499.Search in Google Scholar PubMed PubMed Central
202. Cai, J, Han, Y, Ren, H, Chen, C, He, D, Zhou, L, et al.. Extracellular vesicle-mediated transfer of donor genomic DNA to recipient cells is a novel mechanism for genetic influence between cells. J Mol Cell Biol 2013;5:227–38. https://doi.org/10.1093/jmcb/mjt011.Search in Google Scholar PubMed PubMed Central
203. Cai, J, Wu, G, Jose, PA, Zeng, C. Functional transferred DNA within extracellular vesicles. Exp Cell Res 2016;349:179–83. https://doi.org/10.1016/j.yexcr.2016.10.012.Search in Google Scholar PubMed
204. Sharma, A, Johnson, A. Exosome DNA: critical regulator of tumor immunity and a diagnostic biomarker. J Cell Physiol 2020;235:1921–32. https://doi.org/10.1002/jcp.29153.Search in Google Scholar PubMed
205. Fernando, MR, Jiang, C, Krzyzanowski, GD, Ryan, WL. New evidence that a large proportion of human blood plasma cell-free DNA is localized in exosomes. PLoS One 2017;12:e0183915. https://doi.org/10.1371/journal.pone.0183915.Search in Google Scholar PubMed PubMed Central
206. Holmgren, L, Szeles, A, Rajnavölgyi, E, Folkman, J, Klein, G, Ernberg, I, et al.. Horizontal transfer of DNA by the uptake of apoptotic bodies. Blood J Am Soc Hematol 1999;93:3956–63. https://doi.org/10.1182/blood.v93.11.3956.411k05_3956_3963.Search in Google Scholar
207. Bergsmedh, A, Szeles, A, Henriksson, M, Bratt, A, Folkman, MJ, Spetz, AL, et al.. Horizontal transfer of oncogenes by uptake of apoptotic bodies. Proc Natl Acad Sci USA 2001;98:6407–11. https://doi.org/10.1073/pnas.101129998.Search in Google Scholar PubMed PubMed Central
208. Dorward, DW, Garon, CF, Judd, RC. Export and intercellular transfer of DNA via membrane blebs of Neisseria gonorrhoeae. J Bacteriol 1989;171:2499–505. https://doi.org/10.1128/jb.171.5.2499-2505.1989.Search in Google Scholar PubMed PubMed Central
209. Garon, C, Dorward, D, Corwin, M. Structural features of borrelia burgdorferi—the Lyme disease spirochete: silver staining for nucleic acids. Scanning Microsc Suppl 1989;3:109–15.Search in Google Scholar
210. Soler, N, Marguet, E, Verbavatz, JM, Forterre, P. Virus-like vesicles and extracellular DNA produced by hyperthermophilic archaea of the order thermococcales. Res Microbiol 2008;159:390–9. https://doi.org/10.1016/j.resmic.2008.04.015.Search in Google Scholar PubMed
211. Bitto, NJ, Chapman, R, Pidot, S, Costin, A, Lo, C, Choi, J, et al.. Bacterial membrane vesicles transport their DNA cargo into host cells. Sci Rep 2017;7:7072. https://doi.org/10.1038/s41598-017-07288-4.Search in Google Scholar PubMed PubMed Central
212. Beiter, T, Fragasso, A, Hudemann, J, Schild, M, Steinacker, J, Mooren, FC, et al.. Neutrophils release extracellular DNA traps in response to exercise. J Appl Physiol 2014;117:325–33. https://doi.org/10.1152/japplphysiol.00173.2014.Search in Google Scholar PubMed
213. Sur Chowdhury, C, Hahn, S, Hasler, P, Hoesli, I, Lapaire, O, Giaglis, S. Elevated levels of total cell-free DNA in maternal serum samples arise from the generation of neutrophil extracellular traps. Fetal Diagn Ther 2016;40:263–7. https://doi.org/10.1159/000444853.Search in Google Scholar PubMed
214. Massberg, S, Grahl, L, von Bruehl, ML, Manukyan, D, Pfeiler, S, Goosmann, C, et al.. Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases. Nat Med 2010;16:887–96. https://doi.org/10.1038/nm.2184.Search in Google Scholar PubMed
215. Rykova, EY, Morozkin, ES, Ponomaryova, AA, Loseva, EM, Zaporozhchenko, IA, Cherdyntseva, NV, et al.. Cell-free and cell-bound circulating nucleic acid complexes: mechanisms of generation, concentration and content. Expet Opin Biol Ther 2012;12:S141–53. https://doi.org/10.1517/14712598.2012.673577.Search in Google Scholar PubMed
216. Tamkovich, S, Laktionov, P. Cell-surface-bound circulating DNA in the blood: biology and clinical application. IUBMB Life 2019;71:1201–10. https://doi.org/10.1002/iub.2070.Search in Google Scholar PubMed
217. Ungerer, V, Bronkhorst, AJ, Van den Ackerveken, P, Herzog, M, Holdenrieder, S. Serial profiling of cell-free DNA and nucleosome histone modifications in cell cultures. Sci Rep 2021;11:9460. https://doi.org/10.1038/s41598-021-88866-5.Search in Google Scholar PubMed PubMed Central
218. Bronkhorst, AJ, Wentzel, JF, Aucamp, J, van Dyk, E, du Plessis, L, Pretorius, PJ. Characterization of the cell-free DNA released by cultured cancer cells. Biochim Biophys Acta 2016;1863:157–65. https://doi.org/10.1016/j.bbamcr.2015.10.022.Search in Google Scholar PubMed
219. Aucamp, J, Bronkhorst, AJ, Peters, DL, Van Dyk, HC, Van der Westhuizen, FH, Pretorius, PJ. Kinetic analysis, size profiling, and bioenergetic association of DNA released by selected cell lines in vitro. Cell Mol Life Sci 2017;74:2689–707. https://doi.org/10.1007/s00018-017-2495-z.Search in Google Scholar PubMed
220. Mouliere, F, Chandrananda, D, Piskorz, AM, Moore, EK, Morris, J, Ahlborn, LB, et al.. Enhanced detection of circulating tumor DNA by fragment size analysis. Sci Transl Med 2018;10:eaat4921. https://doi.org/10.1126/scitranslmed.aat4921.Search in Google Scholar PubMed PubMed Central
221. Hudecova, I, Smith, CG, Hänsel-Hertsch, R, Chilamakuri, CS, Morris, JA, Vijayaraghavan, A, et al.. Characteristics, origin, and potential for cancer diagnostics of ultrashort plasma cell-free DNA. Genome Res 2022;32:215–27. https://doi.org/10.1101/gr.275691.121.Search in Google Scholar PubMed PubMed Central
222. Markus, H, Chandrananda, D, Moore, E, Mouliere, F, Morris, J, Brenton, JD, et al.. Refined characterization of circulating tumor DNA through biological feature integration. Sci Rep 2022;12:1928. https://doi.org/10.1038/s41598-022-05606-z.Search in Google Scholar PubMed PubMed Central
223. Sanchez, C, Snyder, MW, Tanos, R, Shendure, J, Thierry, AR. New insights into structural features and optimal detection of circulating tumor DNA determined by single-strand DNA analysis. NPJ Genom Med 2018;3:31. https://doi.org/10.1038/s41525-018-0069-0.Search in Google Scholar PubMed PubMed Central
224. Sanchez, C, Roch, B, Mazard, T, Blache, P, Dache, ZAA, Pastor, B, et al.. Circulating nuclear DNA structural features, origins, and complete size profile revealed by fragmentomics. JCI insight 2021;6:e144561. https://doi.org/10.1172/jci.insight.144561.Search in Google Scholar PubMed PubMed Central
225. Jiang, P, Sun, K, Tong, YK, Cheng, SH, Cheng, TH, Heung, MM, et al.. Preferred end coordinates and somatic variants as signatures of circulating tumor DNA associated with hepatocellular carcinoma. Proc Natl Acad Sci USA 2018;115:E10925–33. https://doi.org/10.1073/pnas.1814616115.Search in Google Scholar PubMed PubMed Central
226. Sun, K, Jiang, P, Wong, AIC, Cheng, YKY, Cheng, SH, Zhang, H, et al.. Size-tagged preferred ends in maternal plasma DNA shed light on the production mechanism and show utility in noninvasive prenatal testing. Proc Natl Acad Sci USA 2018;115:E5106–14. https://doi.org/10.1073/pnas.1804134115.Search in Google Scholar PubMed PubMed Central
227. Cristiano, S, Leal, A, Phallen, J, Fiksel, J, Adleff, V, Bruhm, DC, et al.. Genome-wide cell-free DNA fragmentation in patients with cancer. Nature 2019;570:385–9. https://doi.org/10.1038/s41586-019-1272-6.Search in Google Scholar PubMed PubMed Central
228. Cheng, N, Skead, K, Ouellette, T, Bratman, S, De Carvalho, D, Soave, D, et al.. Early signatures of breast cancer up to seven years prior to clinical diagnosis in plasma cell-free DNA methylomes. 2022 [Preprint].10.21203/rs.3.rs-1203227/v1Search in Google Scholar
229. Chan, KC, Jiang, P, Chan, CW, Sun, K, Wong, J, Hui, EP, et al.. Noninvasive detection of cancer-associated genome-wide hypomethylation and copy number aberrations by plasma DNA bisulfite sequencing. Proc Natl Acad Sci USA 2013;110:18761–8. https://doi.org/10.1073/pnas.1313995110.Search in Google Scholar PubMed PubMed Central
230. Hulbert, A, Jusue-Torres, I, Stark, A, Chen, C, Rodgers, K, Lee, B, et al.. Early detection of lung cancer using DNA promoter hypermethylation in plasma and sputum. Clin Cancer Res 2017;23:1998–2005. https://doi.org/10.1158/1078-0432.ccr-16-1371.Search in Google Scholar
231. Zeng, H, He, B, Yi, C, Peng, J. Liquid biopsies: DNA methylation analyses in circulating cell-free DNA. J Genet Genom 2018;45:185–92. https://doi.org/10.1016/j.jgg.2018.02.007.Search in Google Scholar PubMed
232. Lubotzky, A, Zemmour, H, Neiman, D, Gotkine, M, Loyfer, N, Piyanzin, S, et al.. Liquid biopsy reveals collateral tissue damage in cancer. JCI Insight 2022;7:e153559. https://doi.org/10.1172/jci.insight.153559.Search in Google Scholar PubMed PubMed Central
233. Korshunova, Y, Maloney, RK, Lakey, N, Citek, RW, Bacher, B, Budiman, A, et al.. Massively parallel bisulphite pyrosequencing reveals the molecular complexity of breast cancer-associated cytosine-methylation patterns obtained from tissue and serum DNA. Genome Res 2008;18:19–29. https://doi.org/10.1101/gr.6883307.Search in Google Scholar PubMed PubMed Central
234. Shen, SY, Singhania, R, Fehringer, G, Chakravarthy, A, Roehrl, MHA, Chadwick, D, et al.. Sensitive tumour detection and classification using plasma cell-free DNA methylomes. Nature 2018;563:579–83. https://doi.org/10.1038/s41586-018-0703-0.Search in Google Scholar PubMed
235. Brock, MV, Hooker, CM, Ota-Machida, E, Han, Y, Guo, M, Ames, S, et al.. DNA methylation markers and early recurrence in stage I lung cancer. N Engl J Med 2008;358:1118–28. https://doi.org/10.1056/nejmoa0706550.Search in Google Scholar
236. Gezer, U, Üstek, D, Yörüker, EE, Cakiris, A, Abaci, N, Leszinski, G, et al.. Characterization of H3K9me3-and H4K20me3-associated circulating nucleosomal DNA by high-throughput sequencing in colorectal cancer. Tumor Biol 2013;34:329–36. https://doi.org/10.1007/s13277-012-0554-5.Search in Google Scholar PubMed
237. Gezer, U, Yörüker, EE, Keskin, M, Kulle, CB, Dharuman, Y, Holdenrieder, S. Histone methylation marks on circulating nucleosomes as novel blood-based biomarker in colorectal cancer. Int J Mol Sci 2015;16:29654–62. https://doi.org/10.3390/ijms161226180.Search in Google Scholar PubMed PubMed Central
238. Kneip, C, Schmidt, B, Seegebarth, A, Weickmann, S, Fleischhacker, M, Liebenberg, V, et al.. SHOX2 DNA methylation is a biomarker for the diagnosis of lung cancer in plasma. J Thorac Oncol 2011;6:1632–8. https://doi.org/10.1097/jto.0b013e318220ef9a.Search in Google Scholar PubMed
239. Li, M, Chen, WD, Papadopoulos, N, Goodman, SN, Bjerregaard, NC, Laurberg, S, et al.. Sensitive digital quantification of DNA methylation in clinical samples. Nat Biotechnol 2009;27:858. https://doi.org/10.1038/nbt.1559.Search in Google Scholar PubMed PubMed Central
240. Song, CX, Yin, S, Ma, L, Wheeler, A, Chen, Y, Zhang, Y, et al.. 5-hydroxymethylcytosine signatures in cell-free DNA provide information about tumor types and stages. Cell Res 2017;27:1231–42. https://doi.org/10.1038/cr.2017.106.Search in Google Scholar PubMed PubMed Central
241. Li, W, Zhang, X, Lu, X, You, L, Song, Y, Luo, Z, et al.. 5-hydroxymethylcytosine signatures in circulating cell-free DNA as diagnostic biomarkers for human cancers. Cell Res 2017;27:1243–57. https://doi.org/10.1038/cr.2017.121.Search in Google Scholar PubMed PubMed Central
242. McAnena, P, Brown, JA, Kerin, MJ. Circulating nucleosomes and nucleosome modifications as biomarkers in cancer. Cancers 2017;9:5. https://doi.org/10.3390/cancers9010005.Search in Google Scholar PubMed PubMed Central
243. Müller, HM, Widschwendter, A, Fiegl, H, Ivarsson, L, Goebel, G, Perkmann, E, et al.. DNA methylation in serum of breast cancer patients: an independent prognostic marker. Cancer Res 2003;63:7641–5.Search in Google Scholar
244. Nassiri, F, Chakravarthy, A, Feng, S, Shen, SY, Nejad, R, Zuccato, JA, et al.. Detection and discrimination of intracranial tumors using plasma cell-free DNA methylomes. Nat Med 2020;26:1044–7. https://doi.org/10.1038/s41591-020-0932-2.Search in Google Scholar PubMed PubMed Central
245. Schröck, A, Leisse, A, de Vos, L, Gevensleben, H, Dröge, F, Franzen, A, et al.. Free-circulating methylated DNA in blood for diagnosis, staging, prognosis, and monitoring of head and neck squamous cell carcinoma patients: an observational prospective cohort study. Clin Chem 2017;63:1288–96. https://doi.org/10.1373/clinchem.2016.270207.Search in Google Scholar PubMed
246. Thålin, C, Lundström, S, Seignez, C, Daleskog, M, Lundström, A, Henriksson, P, et al.. Citrullinated histone H3 as a novel prognostic blood marker in patients with advanced cancer. PLoS One 2018;13:e0191231. https://doi.org/10.1371/journal.pone.0191231.Search in Google Scholar PubMed PubMed Central
247. Xu, RH, Wei, W, Krawczyk, M, Wang, W, Luo, H, Flagg, K, et al.. Circulating tumour DNA methylation markers for diagnosis and prognosis of hepatocellular carcinoma. Nat Mater 2017;16:1155–61. https://doi.org/10.1038/nmat4997.Search in Google Scholar PubMed
248. de Castro Cuadrat, RR, Kratzer, A, Arnal, HG, Wreczycka, K, Blume, A, Ebenal, V, et al.. Cardiovascular disease biomarkers derived from circulating cell-free DNA methylation. medRxiv 2021 [Preprint].10.1101/2021.11.05.21265870Search in Google Scholar
249. Akirav, EM, Lebastchi, J, Galvan, EM, Henegariu, O, Akirav, M, Ablamunits, V, et al.. Detection of $\beta$ cell death in diabetes using differentially methylated circulating DNA. Proc Natl Acad Sci USA 2011;108:19018–23. https://doi.org/10.1073/pnas.1111008108.Search in Google Scholar PubMed PubMed Central
250. Zhang, K, Lin, G, Han, Y, Xie, J, Li, J. Circulating unmethylated insulin DNA as a potential non-invasive biomarker of beta cell death in type 1 diabetes: a review and future prospect. Clin Epigenet 2017;9:44. https://doi.org/10.1186/s13148-017-0343-5.Search in Google Scholar PubMed PubMed Central
251. Hardy, T, Zeybel, M, Day, CP, Dipper, C, Masson, S, McPherson, S, et al.. Plasma DNA methylation: a potential biomarker for stratification of liver fibrosis in non-alcoholic fatty liver disease. Gut 2017;66:1321–8. https://doi.org/10.1136/gutjnl-2016-311526.Search in Google Scholar PubMed PubMed Central
252. Lehmann-Werman, R, Magenheim, J, Moss, J, Neiman, D, Abraham, O, Piyanzin, S, et al.. Monitoring liver damage using hepatocyte-specific methylation markers in cell-free circulating DNA. JCI Insight 2018;3:e120687. https://doi.org/10.1172/jci.insight.120687.Search in Google Scholar PubMed PubMed Central
253. Dunaeva, M, Derksen, M, Pruijn, GJ. LINE-1 hypermethylation in serum cell-free DNA of relapsing remitting multiple sclerosis patients. Mol Neurobiol 2018;55:4681–8. https://doi.org/10.1007/s12035-017-0679-z.Search in Google Scholar PubMed PubMed Central
254. Lubotzky, A, Pelov, I, Teplitz, R, Neiman, D, Smadja, A, Zemmour, H, et al.. Elevated brain-derived cell-free DNA among patients with first psychotic episode-a proof-of-concept study. medRxiv 2022 [Preprint].10.7554/eLife.76391.sa2Search in Google Scholar
255. Laurent, D, Semple, F, Starkey Lewis, PJ, Rose, E, Black, HA, Coe, J, et al.. Absolute measurement of the tissue origins of cell-free DNA in the healthy state and following paracetamol overdose. BMC Med Genom 2020;13:60. https://doi.org/10.1186/s12920-020-0705-2.Search in Google Scholar PubMed PubMed Central
256. Guo, S, Diep, D, Plongthongkum, N, Fung, HL, Zhang, K, Zhang, K. Identification of methylation haplotype blocks aids in deconvolution of heterogeneous tissue samples and tumor tissue-of-origin mapping from plasma DNA. Nat Genet 2017;49:635–42. https://doi.org/10.1038/ng.3805.Search in Google Scholar PubMed PubMed Central
257. Lehmann-Werman, R, Neiman, D, Zemmour, H, Moss, J, Magenheim, J, Vaknin-Dembinsky, A, et al.. Identification of tissue-specific cell death using methylation patterns of circulating DNA. Proc Natl Acad Sci USA 2016;113:E1826–34.10.1073/pnas.1519286113Search in Google Scholar PubMed PubMed Central
258. Tang, W, Wan, S, Yang, Z, Teschendorff, AE, Zou, Q. Tumor origin detection with tissue-specific miRNA and DNA methylation markers. Bioinformatics 2017;34:398–406. https://doi.org/10.1093/bioinformatics/btx622.Search in Google Scholar PubMed
259. 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. https://doi.org/10.1016/j.cell.2015.11.050.Search in Google Scholar PubMed PubMed Central
260. Ulz, P, Perakis, S, Zhou, Q, Moser, T, Belic, J, Lazzeri, I, et al.. Inference of transcription factor binding from cell-free DNA enables tumor subtype prediction and early detection. Nat Commun 2019;10:4666. https://doi.org/10.1038/s41467-019-12714-4.Search in Google Scholar PubMed PubMed Central
261. Sadeh, R, Sharkia, I, Fialkoff, G, Rahat, A, Gutin, J, Chappleboim, A, et al.. ChIP-seq of plasma cell-free nucleosomes identifies gene expression programs of the cells of origin. Nat Biotechnol 2021;39:586–98. https://doi.org/10.1038/s41587-020-00775-6.Search in Google Scholar PubMed PubMed Central
262. Cohen, JD, Javed, AA, Thoburn, C, Wong, F, Tie, J, Gibbs, P, et al.. Combined circulating tumor DNA and protein biomarker-based liquid biopsy for the earlier detection of pancreatic cancers. Proc Natl Acad Sci USA 2017;114:10202–7. https://doi.org/10.1073/pnas.1704961114.Search in Google Scholar PubMed PubMed Central
263. Cohen, JD, Li, L, Wang, Y, Thoburn, C, Afsari, B, Danilova, L, et al.. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science 2018;359:926–30. https://doi.org/10.1126/science.aar3247.Search in Google Scholar PubMed PubMed Central
264. Keup, C, Suryaprakash, V, Hauch, S, Storbeck, M, Hahn, P, Sprenger-Haussels, M, et al.. Integrative statistical analyses of multiple liquid biopsy analytes in metastatic breast cancer. Genome Med 2021;13:85. https://doi.org/10.1186/s13073-021-00902-1.Search in Google Scholar PubMed PubMed Central
265. Alix-Panabières, C, Schwarzenbach, H, Pantel, K. Circulating tumor cells and circulating tumor DNA. Annu Rev Med 2012;63:199–215. https://doi.org/10.1146/annurev-med-062310-094219.Search in Google Scholar PubMed
266. Alix-Panabières, C, Pantel, K. Circulating tumor cells: liquid biopsy of cancer. Clin Chem 2013;59:110–8. https://doi.org/10.1373/clinchem.2012.194258.Search in Google Scholar PubMed
267. Alix-Panabières, C, Pantel, K. Clinical applications of circulating tumor cells and circulating tumor DNA as liquid biopsy. Cancer Discov 2016;6:479–91. https://doi.org/10.1158/2159-8290.cd-15-1483.Search in Google Scholar PubMed
268. Keup, C, Storbeck, M, Hauch, S, Hahn, P, Sprenger-Haussels, M, Hoffmann, O, et al.. Multimodal targeted deep sequencing of circulating tumor cells and matched cell-free DNA provides a more comprehensive tool to identify therapeutic targets in metastatic breast cancer patients. Cancers 2020;12:1084. https://doi.org/10.3390/cancers12051084.Search in Google Scholar PubMed PubMed Central
269. Zhang, W, Xia, W, Lv, Z, Xin, Y, Ni, C, Yang, L. Liquid biopsy for cancer: circulating tumor cells, circulating free DNA or exosomes? Cell Physiol Biochem 2017;41:755–68. https://doi.org/10.1159/000458736.Search in Google Scholar PubMed
270. Keup, C, Suryaprakash, V, Storbeck, M, Hoffmann, O, Kimmig, R, Kasimir-Bauer, S. Longitudinal multi-parametric liquid biopsy approach identifies unique features of circulating tumor cell, extracellular vesicle, and cell-free DNA characterization for disease monitoring in metastatic breast cancer patients. Cells 2021;10:212. https://doi.org/10.3390/cells10020212.Search in Google Scholar PubMed PubMed Central
271. Mitchell, PS, Parkin, RK, Kroh, EM, Fritz, BR, Wyman, SK, Pogosova-Agadjanyan, EL, et al.. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA 2008;105:10513–8. https://doi.org/10.1073/pnas.0804549105.Search in Google Scholar PubMed PubMed Central
272. Kopreski, MS, Benko, FA, Kwak, LW, Gocke, CD. Detection of tumor messenger RNA in the serum of patients with malignant melanoma. Clin Cancer Res 1999;5:1961–5.Search in Google Scholar
273. Mayers, JR, Wu, C, Clish, CB, Kraft, P, Torrence, ME, Fiske, BP, et al.. Elevation of circulating branched-chain amino acids is an early event in human pancreatic adenocarcinoma development. Nat Med 2014;20:1193–8. https://doi.org/10.1038/nm.3686.Search in Google Scholar PubMed PubMed Central
274. Ulz, P, Thallinger, GG, Auer, M, Graf, R, Kashofer, K, Jahn, SW, et al.. Inferring expressed genes by whole-genome sequencing of plasma DNA. Nat Genet 2016;48:1273–8. https://doi.org/10.1038/ng.3648.Search in Google Scholar PubMed
275. Sina, AA, Carrascosa, LG, Liang, Z, Grewal, YS, Wardiana, A, Shiddiky, MJA, et al.. Epigenetically reprogrammed methylation landscape drives the DNA self-assembly and serves as a universal cancer biomarker. Nat Commun 2018;9:4915. https://doi.org/10.1038/s41467-018-07214-w.Search in Google Scholar PubMed PubMed Central
© 2022 the author(s), published by De Gruyter, Berlin/Boston
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Frontmatter
- Editorial
- Liquid profiling – circulating nucleic acid diagnostics gains momentum
- Articles
- The rising tide of cell-free DNA profiling: from snapshot to temporal genome analysis
- Liquid profiling for cancer patient stratification in precision medicine – current status and challenges for successful implementation in standard care
- Status of liquid profiling in precision oncology – the need for integrative diagnostics for successful implementation into standard care
- Pan-cancer screening by circulating tumor DNA (ctDNA) – recent breakthroughs and chronic pitfalls
- Multimodality in liquid biopsy: does a combination uncover insights undetectable in individual blood analytes?
- Circulating cell-free DNA and its clinical utility in cancer
- Are extracellular vesicles ready for the clinical laboratory?
- Profiling disease and tissue-specific epigenetic signatures in cell-free DNA
- Cell-free DNA in sports medicine: implications for clinical laboratory medicine
- Clonal hematopoiesis of indeterminate potential: clinical relevance of an incidental finding in liquid profiling
- Non-invasive prenatal screening tests – update 2022
- The DKTK EXLIQUID consortium – exploiting liquid biopsies to advance cancer precision medicine for molecular tumor board patients
Articles in the same Issue
- Frontmatter
- Editorial
- Liquid profiling – circulating nucleic acid diagnostics gains momentum
- Articles
- The rising tide of cell-free DNA profiling: from snapshot to temporal genome analysis
- Liquid profiling for cancer patient stratification in precision medicine – current status and challenges for successful implementation in standard care
- Status of liquid profiling in precision oncology – the need for integrative diagnostics for successful implementation into standard care
- Pan-cancer screening by circulating tumor DNA (ctDNA) – recent breakthroughs and chronic pitfalls
- Multimodality in liquid biopsy: does a combination uncover insights undetectable in individual blood analytes?
- Circulating cell-free DNA and its clinical utility in cancer
- Are extracellular vesicles ready for the clinical laboratory?
- Profiling disease and tissue-specific epigenetic signatures in cell-free DNA
- Cell-free DNA in sports medicine: implications for clinical laboratory medicine
- Clonal hematopoiesis of indeterminate potential: clinical relevance of an incidental finding in liquid profiling
- Non-invasive prenatal screening tests – update 2022
- The DKTK EXLIQUID consortium – exploiting liquid biopsies to advance cancer precision medicine for molecular tumor board patients