key: cord-0014225-nvrhejnz authors: Pös, Zuzana; Pös, Ondrej; Styk, Jakub; Mocova, Angelika; Strieskova, Lucia; Budis, Jaroslav; Kadasi, Ludevit; Radvanszky, Jan; Szemes, Tomas title: Technical and Methodological Aspects of Cell-Free Nucleic Acids Analyzes date: 2020-11-16 journal: Int J Mol Sci DOI: 10.3390/ijms21228634 sha: 7d1d93aff22fe858636327ad1fa4f1bd07d28545 doc_id: 14225 cord_uid: nvrhejnz Analyzes of cell-free nucleic acids (cfNAs) have shown huge potential in many biomedical applications, gradually entering several fields of research and everyday clinical care. Many biological properties of cfNAs can be informative to gain deeper insights into the function of the organism, such as their different types (DNA, RNAs) and subtypes (gDNA, mtDNA, bacterial DNA, miRNAs, etc.), forms (naked or vesicle bound NAs), fragmentation profiles, sequence composition, epigenetic modifications, and many others. On the other hand, the workflows of their analyzes comprise many important steps, from sample collection, storage and transportation, through extraction and laboratory analysis, up to bioinformatic analyzes and statistical evaluations, where each of these steps has the potential to affect the outcome and informational value of the performed analyzes. There are, however, no universal or standard protocols on how to exactly proceed when analyzing different cfNAs for different applications, at least according to our best knowledge. We decided therefore to prepare an overview of the available literature and products commercialized for cfNAs processing, in an attempt to summarize the benefits and limitations of the currently available approaches, devices, consumables, and protocols, together with various factors influencing the workflow, its processes, and outcomes. Cell-free nucleic acids (cfNAs), also called extracellular nucleic acids (ecNAs), represent fragments of DNA or RNA molecules that are actively or passively released from cells to different body fluids, including, but not limited to, blood, saliva, urine, or feces. Mechanisms of their release may be various, leaving typical molecular signatures for certain mechanisms and thus also for the physiological and pathophysiological process behind their release. Some cfNAs may be released passively following cell decomposition, for example by mechanisms such as apoptosis, necrosis, or physical damage, while others may be released actively to facilitate local or long-distance cell-cell communication, or even immune reactions, such in the case of NETosis or other similar processes. Their complex informational Processing and extraction of cfNAs should be performed as soon as possible after the sampling, therefore, the best practice may be to perform the whole processing at the same place at which the body fluids were sampled. These inter-step connections are highlighted in the scheme by red arrows and dashed lines. Although research of cell-free fetal nucleic acids (cffNAs) has primarily focused on maternal plasma, in line with the above delineated rule of body fluids becoming enriched with cfNAs derived from tissues and organs being in closest contact with the fluid, cffNAs might be found in much greater concentrations in the amniotic fluid, representing a pure fetal sample uncontaminated by maternal-and trophoblast-derived NAs. Technical progress has also made it possible to extract cffNAs, both cffDNA, and cffRNAs, of sufficient quality and quantity to perform functional genomic analyzes from amniotic fluid [9, 67] . Moreover, a large number of detectable miRNA species were reported to be present in amniotic fluid, having unique composition when compared to other body fluids, suggesting that the filtering process by the placenta reduces the exchange between amniotic and other body fluids [60] . cfDNA in the supernatant of pleural effusion (PE) is a valuable source that can be used to detect the driver as well as resistance mutations and can guide tyrosine kinase inhibitor treatment decisions [68] . High accordance in mutational spectra among matched cfDNA from plasma, malignant PE, and ascites collected from advanced gastric cancer patients have been discovered [69] . There is also evidence supporting the clinical utility of exosomal DNA from malignant PE of advanced lung adenocarcinoma patients. Targeted MPS analysis of PE-derived exosomal DNA and cfDNA exhibited highly concordant mutational profiles [70] . In addition to cfDNA also miRNA molecules have been extracted from the pleural fluid [60] . Likely because of the excess fluid accumulating inside the peritoneal cavity is a result of fluid leakage from blood vessels, as a consequence of various diseases, fragment sizes of ascites cfDNA were found to correlate with those seen in plasma. Analyzes of cfDNA from the ascites of individuals with metastatic cancer demonstrated the presence of tumorigenic CNVs in cancer-associated genes [71] , while mutation profiles of ascites derived cfDNA were also found to be highly concordant with cfDNA from plasma and even malignant pleural effusion in gastric cancer patients [69] . Liquid biopsy-based on bronchoalveolar lavage fluid (BALF) sampling has rarely been used, however, the molecular testing method for the detection of EGFR mutations in NSCLC patients using cfDNA from this source demonstrated high diagnostic value with 91.7% concordance with tumor biopsy results in the detection of EGFR mutations [72] . Intact RNA and miRNA were detected in cell-free supernatants of bronchial lavage fluid [60] . Moreover, similarly to other fluids, RNA concentrations were shown to be higher in lavage than in the serum samples of lung cancer patients [73] . Modern intravitreal chemotherapeutic techniques for retinoblastoma involve aspiration of aqueous humor, providing a novel sample source for analysis. Berry et al. demonstrated measurable concentrations of DNA, RNA, and miRNA in such samples [74] . Analysis of cfDNA from the aqueous humor fluid of retinoblastoma eyes was capable of detecting somatic variants and the results were comparable to current tests on enucleated tumor tissue [75] . Dysregulation of specific miRNAs in the vitreous humor might also be related to the alterations that characterize patients affected by the macular hole and epiretinal membrane [76] . There are also other body fluids in which cfNAs were assessed and evaluated, but studies for which are even scarcer, when compared to the abovementioned fluids. The presence of cfDNA in sweat, for example, was detected in 80% of tested healthy individuals, with an average concentration of 11.5 ng/mL [77] . Other body fluids and their cfNAs content are generally reported with respect to certain associated diseases. It was demonstrated, for example, that pancreatic juice is a useful source for genetic profiling of somatic alterations of pancreatic tumors and intraductal papillary mucinous neoplasm with a potential to assess also their malignant progression risk [78] . Since cervicovaginal secretion has close contact with certain types of tissues, it can be a useful specimen of genetic material for testing pathogens associated with sexually transmitted diseases or cervical cancer [10] . Somatic mutations, including single nucleotide variants (SNVs) and copy number variants (CNVs), were found to be detectable also within cfDNA obtained from the bile of biliary tract carcinoma patients. Interestingly, bile cfDNA was predominantly composed of long DNA fragments in contrast to plasma [79] . cfNAs content of synovial fluid was also assessed, while elevated cfDNA levels in the plasma and synovial fluid of rheumatoid arthritis patients were reported, with a median cfDNA concentration being~77-fold higher in the synovial fluid than that in the plasma of rheumatoid arthritis patients [80] . Potential role in diagnostics and prognosis of therapy-related peritoneal membrane degeneration was assigned to cfDNAs coming from the peritoneal effluent of peritoneal dialysis patients [81] . The quantification of peritoneal cfDNA could be an innovative method to determine acute damage and an inverse index of the repair process [82] . In peritoneal dialysis patients, also miRNA molecules were extracted from cell-free effluent samples, while specific miRNAs were present in a fraction of microvesicles and exosomes [83] . In addition to these body fluids, several types of cyst fluids were shown to be valuable sources of cfNAs and having potential clinical diagnostic utility, including cfDNA and miRNAs from pancreatic cyst fluid of pancreatic cancer patients [84] [85] [86] , or ovarian cancers with a potential to provide information about whether the cysts releasing them are benign or cancerous, but also about the presence of tumor-specific mutations of borderline tumors, type I cancers, and type II cancers [87] . Although there are highly specialized and mature analytical technologies able to characterize cfNAs, to ensure unbiased analyzes, preanalytical processes need to preserve the original state of cfNAs as much as possible. cfNAs could both be diluted by the post sampling release of cellular NAs, but also by their degradation. Compared to cfDNA, cfRNAs are, for example, very unstable due to RNase activity, unless they are protected by vesicles or macromolecular complexes, in which they were reported to be stable in several body fluids [88] . The first one of these preanalytical processes is sample collection, during which it is important therefore to secure at least the following points: (i) to prevent degradation of free cfNAs fragments; (ii) to stabilize cells to prevent the undesired release of NAs from them; and (iii) to stabilize EMVs and macromolecular complexes to protect their cfNAs cargos and contents. Many devices or solutions were developed for the stabilization and preservation of cfNAs in body fluids, however, the most evaluated are devices designed for the preservation of cfNAs in blood. Some of the available preservation solutions, dedicated for cfDNA, prevent cell lysis and reduce the releasing of non-vesicular miRNAs, however, they are not sufficient for the preservation of constant concentration of EMVs after blood collection [14] , since the time-dependent increase of cfRNA concentration was observed in the majority of these specialized collection tubes [89] . Among the wide range of body fluids, blood is the most commonly used and studied, for which a wide range of stabilization solutions are available, while recently they were developed also for saliva or urine (Table S3 ). With the increasing use of other body fluids for liquid biopsy, collection material and stabilization solutions dedicated for these should be developed. Until that time, collection of other body fluids need to rely on collection materials developed for the same body fluid but for the stabilization of total NAs, dedicated to other body fluids, or even on home-made solutions, all of which may, however, deteriorate the results unless their thorough evaluation and validation are performed. The volume of blood needed for the cfNAs analysis usually ranges between 6-10 mL, however, it depends upon the testing methodology. Commonly used collection tubes with anticoagulants such as di-or tripotassium ethylenediaminetetraacetic acid (K 2 EDTA, K 3 EDTA), lithium heparin, or sodium citrate can stabilize cfDNA only when plasma is processed within six hours, with EDTA performing slightly better than the other two at six hours and longer times [90] . For EDTA there were different time-frames reported for the preservation of cfDNA at room temperature, from a few hours up to 24 h [26, [91] [92] [93] or even up to 48 h [94] over time the lysis of blood cells was detected. Over that time, the storage of blood in EDTA at 4 • C seems to delay the lysis [91, 92, 95] up to three days [26] . Stability of cf-miRNA up to 12, or even 18 h, was also reported in EDTA tubes at room temperature with higher stability at 4 • C [96] . In general, however, it is recommended to process the blood samples, when collected either into EDTA, lithium heparin, or sodium citrate, immediately after collection, or at least as soon as possible [12, 94, 97, 98] . To prevent cell lysis when storing for a longer time, or during shipment to the laboratory, reagents such as formaldehyde or glutaraldehyde may be added, which, however, may cause damage of NAs due to cross-linking with proteins [99] [100] [101] . The best choice is to use specially designed collection tubes containing dedicated stabilization reagents, although the composition of which is generally not available [93, 102] . Storage times for these commercial products are claimed to range from three days up to 30 days at temperatures ranging from 4 • C up to 37 • C (Table S3 ). Although not claimed by the manufacturer, the LBgard Blood Tube (Biomatrica, San Diego, CA, USA) was reported to be effective in preserving cfDNA for up to 72 days. On the other hand, the majority of the collection tubes found by us are officially dedicated to the analysis of cfDNA or CTCs, while there are only two devices, the RNA Complete BCT tubes (Streck, La Vista, NE, USA) and the cf-DNA/cf-RNA Preservative Tubes (Norgen, Thorold, Ontario, Canada) reported to be suitable for the stabilization of cfRNAs in the blood (Table S3 ). However, apart from the claimed utility for cfDNA, both the PAXgene Blood ccfDNA Tubes (Qiagen, Hilden, Germany) and the Cell-Free DNA Collection Tubes (Roche, Basel, Switzerland) were found to be suitable for the preservation of cfRNA too, specifically of cf-miRNAs [96] . When compared to EDTA, and considering the effectivity of cell lysis prevention, claimed utilities of the available systems were proved in several studies, including the Streck Cell-Free DNA BCT tubes [13, 26, [91] [92] [93] 95, 103, 104] , the CellSave Preservative tubes (CellSearch, Huntington Valley, PA, USA) [97, 105] , the Qiagen PAXgene Blood ccfDNA Tube [92, 106] , the Roche Cell-Free DNA Collection Tubes [12, 98, 107] , the Norgen cf-DNA/cf-RNA Preservative Tubes [96] and Biomatrica LBgard Blood Tube [108] [109] [110] . Studies listing limitations are, however, also available. The suggested storage for up to 14 days was not confirmed, for example, for the Streck Cell-Free DNA BCT tubes in a study of Zhao et al. [12] . It should also be noted that temperatures outside the recommended range significantly affected the yield of cfDNA due to unprevented cell lysis, highlighting the importance to maintain temperatures above 10 • C and up to 37 • C to ensure plasma quality [13, 94, 111] . When using the Streck Cell-Free DNA BCT tubes, Qin et al. demonstrated their ability to stabilize proteins and mRNA for up to 4 days at room temperature [105] , however, other studies claimed that they are not suitable for the parallel analysis of other biomarkers, including cfRNA, and in some cases requiring modifications of the DNA extraction procedure [13, 92, 96] . The Streck Cell-Free DNA BCT tubes were reported to be inappropriate for the analysis of methylated sequences of cfDNA [106] , the Qiagen PAXgene Blood ccfDNA Tubes was reported to have no effect on methylation analyzes, in line with the manufacturer's claims [106] . On the other hand, another study claimed that neither the Streck Cell-Free DNA BCT tubes nor the Qiagen PAXgene Blood ccfDNA Tubes are suitable for the analysis of methylations [112] . When considering downstream applications, several studies compared common commercial products. Sherwood et al., for example, demonstrated a significantly decreased detection rate of mutations after 72 h when stored in EDTA tubes compared to the Streck Cell-Free DNA BCT tubes and that different input of plasma volume can affect the DNA yield [91] . In another comparison, EDTA, the Streck Cell-Free DNA BCT tubes, and the CellSearch CellSave Preservative tubes were found to stabilize ctDNA at room temperature, or at 4 • C, for up to six hours before plasma processing, however, the Streck Cell-Free DNA BCT tubes and the CellSearch CellSave Preservative tubes demonstrated higher tumor mutation fraction than EDTA in which significant reduction of ctDNA occurred [94] . The Roche Cell-Free DNA Collection Tubes, the Qiagen PAXgene Blood ccfDNA Tubes, and the Streck Cell-Free DNA BCT tubes were found to be similarly effective in stabilizing ctDNA for a long time, however, in a case of low ctDNA concentration in blood only the Roche Cell-Free DNA Collection Tubes and the Qiagen PAXgene Blood ccfDNA Tubes were sufficient for the tumor mutation detection [113] . In the comparison study of Ward Gahlawat et al., the Norgen cf-DNA/cf-RNA Preservative Tubes and the Qiagen PAXgene Blood ccfDNA Tubes were found to be most suitable for the analysis of cfRNAs, while the Norgen cf-DNA/cf-RNA Preservative Tubes for combined analysis of both cfDNA and cfRNA [96] . In a study performed by Van Paemel et al., the Streck Cell-Free DNA BCT tubes, the Qiagen PAXgene Blood ccfDNA Tubes, the Roche Cell-Free DNA Collection Tubes, and Biomatrica LBgard Blood Tubes performed comparably in several downstream applications [108] . Since the introduction of the Streck Cell-Free DNA BCT tubes, various other collection tubes have been developed, while only a few of them were cross-evaluated for their effectiveness to stabilize and preserve cfNAs. There are, however, also newer collection systems, such as the CEE-Sure BCT (Biocept, San Diego, CA, USA), the Blood STASIS 21-cfDNA Blood Collection Tubes (MagBio Genomics, Gaithersburg, MD, USA) the Check cfDNA Tube (EONE-DIAGNOMICS NICE, Incheon, Korea) and the ImproGene Cell Free DNA Tube (Inresearch, Tsuen Wan, Hongkong) which were yet not studied as extensively as the abovementioned ones (for more detailed characteristics see Table S3 ). Saliva represents viable body fluid, which, unlike blood, does not coagulate, therefore it is easier to collect, transport, and store. On the other hand, saliva mainly consists of water, mucins, enzymes, and proteins, which all may affect downstream analyzes, while salivary proteins, cfNAs, and cfRNAs containing exosomes may very rapidly degrade when they are out of their environment [114] . Protocols for the whole saliva and also glandular secretion collection were established [115] , however, these were not focusing on the stabilization of cfNAs, neither optimized for all downstream molecular analytical methods [116] . Like blood, therefore, an optimized process for collection and processing is required for the stabilization of the salivary components including inhibitors for preserving the integrity of analyzed cfNAs or exosomes. Saliva can be collected under two conditions, -stimulated or unstimulated manner-by methods including spitting, suction, swabbing, and passive drooling. Preparation of salivary supernatant, encompassing collection on ice, centrifugation at 4 • C and addition of RNase and protein inhibitors, although labor-intensive, were found to be effective in improving cfRNAs and proteins stability [117] . Since saliva represents a valuable body fluid useful in early diagnostics, commercially available devices for the saliva collection were also developed. However, these are mainly designed for the analysis of genomic DNA, RNA, or proteins (Salivette from Sarstedt (Numbrecht, Germany), Saliva Collection System from Greiner Bio-One (Kremsmunster, Austria), SalivaBio Oral Swab from Salimetrics (Carlsbad, CA, USA), etc.), but some of them may also enable analyzes of cfNAs and exosomes [118] . On the other hand, when compared to blood, there is a significantly less heterogeneous collection of available commercial products for salivary cfNAs sampling (Table S3) , while the Pure•SAL and RNAPro•SAL (Oasis, Vancouver, WA, USA) were reported to be suitable for the analysis of salivary cfNAs [117] , the Saliva Exosome Collection and Preservation Kit (Norgen, Thorold, ON, Canada) was proved to be useful for cfRNAs isolation and downstream analyzes [119] . According to the manufacturer of this latter kit, salivary exosomes in preserved samples are stable for two years at room temperature (Table S3) , however, we yet did not find an independent evaluation of these claims. Saliva is viscous and may consist of food residue or other particles, therefore its pretreatment is necessary. However, not all methods are suitable when analyzing cfNAs or exosomes. For example, filtration methods aim to reduce viscosity by removing glycoproteins, but since exosomes are larger than proteins, they can be lost [30] . Several studies have been performed regarding the cfNAs analysis from saliva, however, only a few reported the exact collection tube or stabilizer agent used for the saliva collection [31, [120] [121] [122] as an example we present a study in which the Carlson Crittenden parotid collectors were used [123] . However, these are specifically developed collection devices for the type of the gland and not for the stabilization of the exosomes or cfNAs. Saliva represents a promising body fluid, therefore studies are focusing not only on the cfNAs analysis but also on the analysis of the salivary proteome. In these studies, saliva collection and processing were mainly performed either in accordance with previously published protocols (rinsed out the mouth with water and spit into 50 mL sterile Falcon tube) without adding any stabilizer [31, 121, 122, 124] or collected into Falcon tubes and adding a stabilizer [125] . In other studies, the collection and processing were performed according to Navazesh and Christensen [126] or Navazesh protocols [127] with adding RNase and protease inhibitors [120, 128, 129] or using dedicated collection devices and immediately after saliva collection protease inhibitors were added [130] . According to our knowledge, only one study evaluates different conditions of saliva samples storage. Its results have shown that salivary proteomes could be stabilized if the samples are kept at 4 • C with protease inhibitors or by adding absolute ethanol to the samples kept at room temperature [125] . Although saliva seems to be the easiest body fluid for the analysis of cfNAs as a liquid biopsy, several factors should be considered in saliva collection. Different glands produce different levels of salivary analytes [131] , therefore the location of sampling is very important. Saliva composition varies daily and is affected by many external factors, such as food, drinking, smoking, brushing the teeth, exercise, kissing, etc. [132, 133] , therefore the time and instruction for proper collection are also very important. Although urine represents a highly promising body fluid, yet little is known about the urinary cfNAs and the methods for their processing. Urinary cfDNA has a shorter half-life than blood derived cfDNA [134] because of high DNase I [135] and DNase II activity [37] . Therefore, inhibition of DNases might be crucial to prevent cfDNA degradation. One of the effective ways is to use treatment with EDTA and store urine samples at −70 • C or below for a long-time or to use sterile containers and store samples at 4 • C or on ice and process them as soon as possible [136] . Despite such conditions, the loss of cfDNA may occur [137] . Acquisition of high yields of cfNAs and their subsequent sensitive detection as biomarkers depend, therefore, on appropriate preanalytical conditions. When trying to evaluate these processes in the literature, we found that some of the studies do not describe the urine collection devices nor any preservative agent [138] [139] [140] [141] [142] , while others reported to use a sterile container without any stabilizer [121, 143] , used EDTA as a stabilizing agent [144, 145] , or used commercial preservation solution [146] . Only a few studies, to our knowledge, focused on the optimization and standardization of the preanalytical process for urine cfNAs studies. Various commercial stabilization and preservation solutions were developed and used for cfNAs applications (Table S3) , including the Streck Cell-Free DNA Urine Preserve [147] , Urine Conditioning Buffer [148] , Norgen Urine Preservation [149, 150] , Colli Pee (DNA Genotek, Ottawa, ON, Canada) [151] , or the Urine Collection Tube (Hunan UPSBio, Hunan, China) [136] . They are generally liquid reagents which can be added to the urine samples, or they may be available in a dried form in the collection tubes, to stabilize cfDNA and inhibit nuclease activity. Manufacturers claim storage times in a range from days up to years and storage temperatures mainly around room temperature (Table S3) . Although the suitable temperature ranges from 6 • C to 37 • C for the Streck Cell-Free DNA Urine Preserve, samples with this commercial preservative kept at 4 • C were found to be less stable, when mimicking transport condition, suggesting that storing or transporting samples at room temperature, or at higher temperatures, might be more suitable [147] . Unfortunately, studies evaluating the efficacy of these commercial products are largely missing. Recently, Augustus et al. compared the influence of the different preanalytical conditions on the concentration of cfDNA and gDNA. They found that it is inadequate to store samples without any preservatives for a week at room temperature resulting in depletion of a majority of DNA, however, keeping them at 4 • C can slow down the degradation and samples may remain stable for up to 96 h. Although their in-house developed preservative was shown to effectively preserve total DNA in urine, the Streck Cell-Free DNA Urine Preserve led to better results in the preservation of cfDNA [147] . Important differences in preanalytical conditions may exist, however, when evaluating DNA and RNA stability. When considering miRNA content encapsulated in EMVs, for example, the optimal handling condition includes sample storage at room temperature (around 20 • C) and further processing as soon as possible after collection. Alternately, urine held at 2 • C to 4 • C and processed within 24 h, as well as one-time freeze-thaw samples, may be used for miRNA profiling, however, a 5 min 37 • C sample equilibration to solubilize any precipitation for increased miRNA recovery is recommended. On the other hand, it is important to note that urine samples held at 37 • C for as little as four hours are significantly compromised and are not suitable for miRNA profiling [152] . Similarly to saliva, urine seems to be easy body fluid for liquid biopsy, however, amounts of cfNAs vary daily, may be higher in females than in males [153] and first morning urine seems to contain more degraded cfDNA [144] . It is essential to use freshly voided urine to limit the potential bacterial presence and also lifestyle or dietary changes should be considered [154] . All of the above points account to the numerous factors which could impact the downstream analyzes, so they should be kept in mind when designing analytical processes. Less conventional body fluids, for which standardized or commercial collection systems are yet missing, are generally collected into systems originally dedicated to other body fluids, or for the same body fluid but different analyte, such as conventional blood tubes or sterile containers [64] , supplied with EDTA or even without any stabilizer. In such cases, however, the performance of the system should be evaluated, or the samples should be processed as soon as possible. Since stool contains a really large portion of other components that could inhibit downstream analyzes, it is necessary to choose a collection method which allows the maximal recovery of human DNA, preserves human NAs, and inhibits undesirable components. There are some commercial collection devices or storage systems, containing specialized preservatives for the stabilization and protection of NAs in stool samples, available, including the DNA/RNA Shield Fecal Collection tubes (Zymo, Irvine, CA, USA) [155] , the Norgen Stool Nucleic Acid Collection and Preservation Tubes [156, 157] , or the OMNIgene-GUT buffer (DNA Genotek, Ottawa, ON, Canada) [158] , however, neither of them were found to be dedicated to cfNAs. They generally prevent the growth of many bacteria, fungi, and inactivate viruses, and also offer stabilization of NAs in stool samples from several days up to a few years, most commonly at room temperature, or in a frozen state (Table S3 ). However, it is not yet clear, whether these stabilization solutions are effective for the preservation of the wide range of different DNA length fragments [158] . For the collection of sputum, for example, there are various collection methods developed, likely due to the general use of the sputum in the detection of Mycobacterium tuberculosis. For cfDNA analyzes samples were reported to be collected into Fixcyt, comprising ethanol, polyethylene glycol, and ultrapure water [61] , or into a mixed solution of Saccomanno's fixative and dithiothreitol [62] . However, not each study describes the collection method in detail [159] . Breast milk collected into centrifuge tubes without any stabilizer was reported to contain relatively stable cfNAs even at room temperature for a week [63] . For milk samples, on the other hand, Norgen Milk DNA Preservation and Isolation Kit offers DNA preservation for up to one month at room temperature [160] , however, it is not specified whether this kit preserves also cfDNA. CSF was reported to be collected, following lumbar puncture, into the Streck Cell-Free DNA BCT tubes to prevent the release of gDNA [49] , but there are also studies which do not provide details regarding the collection procedure [48, 50] . Ejaculate was reported to be collected in sterile containers [56] , then was allowed to liquefy within 30 min at room temperature or at 37 • C, while in some studies a cryopreservation was added [55] , however, there are also studies which do not mention adding any stabilization nor describe the whole collection step [53, 54, 57] . Lampignano et al. suggested that downstream analytical processes, such as massively parallel sequencing (MPS) and droplet digital PCR, could be utilized with a variety of collection tubes [161] , however, there are some exceptions, usually when methylation analyzes should be performed. For example the Streck Cell-Free DNA BCT tubes do not seem to work for such analyzes, therefore the use of the Qiagen PAXgene Blood ccfDNA Tubes should be considered when designing methylation assays [106] , however, there are also reports of the use of the Streck Cell-Free DNA BCT tubes for epigenetic analysis of cfDNA [162] . Although K 2 EDTA showed worse effect then the Streck Cell-Free DNA BCT tubes on the cfDNA and ctDNA recovery, it seems that on the quality of MPS analyzes neither of these preservatives have noticeable effect [26] , even the Streck Cell-Free DNA BCT tubes can be used for the library preparation for aneuploidy detection in prenatal care without any unwanted changes [93] . These tubes proved their suitability for PCR and real time quantitative PCR [101] , but also for less conventional methods (e.g., BEAMing and Plasma Safe-Sequencing technologies) [13] . It was demonstrated that the Roche Cell-Free DNA Collection Tubes, similar to the Streck Cell-Free DNA BCT tubes, do not affect the quantity and complexity of MPS libraries. These two factors are very important in determining the quality of MPS, since they can reflect the bias of MPS-based results caused by cfDNA quality [12] . Using a microarray technology approximately 3000 different mRNAs were discovered in cell-free saliva. It points to that not only from blood, but cfNAs originating from all body fluids should be acquired in sufficient quality and quantity for plenty of downstream analysis regardless of whether they are performed in a diagnostic manner or in research [162, 163] . It is important to note that NAs extracted from body fluids are not automatically cell-free or extracellular, since the majority of body fluids contain also cells, either being physiologically present, or released into the fluid from the surrounding tissues, or even representing the microbiome. These need to be removed, most commonly by a series of centrifugation and/or filtration steps. Since these cells may release their cellular NAs content following lysis, the faster the processing is, the less contamination by cellular NAs may occur. Although the procedures may be different, there are also examples when the same processing procedures were used for several body fluids, like in the study of Weber et al., in which 12 different body fluids were studied and processed under the same conditions [60] . In addition, there are also procedures which allow to differentially isolate EMVs present in body fluids. Some of the extra steps used for exosome separation are described in the following text, however, there is also a variety of commercial kits dedicated to exosome isolation, which are suitable also for the isolation of other EMVs. These are summarized in Table S4 and are most commonly based on immunoassays, on size exclusion chromatography, or on precipitation, along with different other possibilities. Alternative approaches for isolation of exosomes may utilize microfluidics, which may employ methods such as immunoaffinity [164] [165] [166] , ciliated micropillars [167] , porous polymer monoliths (PPM)-based membrane filters [168] , functionalized surfaces [168, 169] , and droplet-based systems [170, 171] . Numerous centrifugation protocols were developed for blood plasma acquisition. Centrifugation of whole blood allows to separate liquid (serum and plasma) and cellular fraction, representing an important step to prevent the plasma contamination by cellular NAs. Usually, plasma preparation protocols are based on different centrifugal forces (lower or higher forces, or even ultracentrifugation), steps (one-step or two-step protocols) and temperatures (4 • C or at room temperature). Lower cellular DNA contamination was reported when the blood was processed by double centrifugation. It was found that double centrifugation at 2000× g for 10 min did not decrease DNA yield relative to single centrifugation of blood processed two hours post-collection, but the second step significantly reduced DNA yield in blood processed 72 h post-collection, suggesting the presence of contaminating cellular DNA at 72 h due to lysis of blood cells that was not efficiently pelleted by a single centrifugation step [13, 91] . Another described possibility is the use of an initial slow-speed centrifugation at 1600× g for 10 min followed by a second centrifugation of the collected plasma at high-speed 16,000× g for 10 min [91, 172] . The same two-step protocol with identical centrifugal forces but with different time and temperature, i.e., 15 min at 22 • C, was also reported [173] , while double centrifugation protocols were reported to be performed both at 4 • C [12, 91, 174, 175] as well as at room temperature [176] . In each case the samples should be removed carefully after the first centrifugation to avoid contamination by the buffy coat during the plasma separation [177] . It has been observed that combination of higher speed centrifugation at 14,000× g with the following filtration by 0.45 µm filter is suitable to isolate the purest plasma fraction when compared to low-speed centrifugation at 1600× g [178, 179] , however, no centrifugation time was specified in this study. A third centrifugation step at 16,000× g, on the other hand, did not result in any difference in the yield of plasmatic cfDNA [178] . It should be noted that there are also studies which did not observe statistically significant changes in plasma cfDNA yields using different centrifugal forces [95, 180] . The plasma storage conditions represent critical issues too, influencing, for example, cfNAs fragmentation. Once plasma is separated, it should be stored frozen at −20 • C [181] or even at −80 • C for a long time storage [182] , while due to cfDNA fragmentation no freeze-thaw cycles are recommended [183] . Plasma storage for two weeks at −80 • C and for four weeks at −20 • C was reported to have no effect on cfDNA extraction [184] . Longer periods of storage at −80 • C, for example 5-21 months, may, however, cause a decreased cfDNA yield [184, 185] . On the other hand, plasma samples stored at −80 • C from one to six years were reported to be still suitable for analyzes using MPS [186] . However, different types of collection tubes, together with external factors such as extreme temperature or long-time storage of the biological material in the collection tubes also affect processing. Under certain circumstances almost no visible clear plasma was present after the first centrifugation, for example in the Streck Cell-Free DNA BCT tubes [13] . Regardless of the collection tubes, volume of separable plasma was found to decrease over time by storage at room temperature [26] . Process of plasma preparation for cfRNAs extraction may be little different from that for cfDNA extraction. To separate plasma, for example, samples may be processed by two-step centrifugation too, but at 300× g for 20 min at room temperature at first, and after removal of the upper plasma layer and transferring to a new tube, the upper plasma may be centrifuged at 5000× g for 10 min, followed by storage at −80 • C [103] . The study of Sorber et al. provides a good comparison of different centrifugal forces for high-quality cfDNA and cfRNA yield in both EDTA and the Streck Cell-Free DNA BCT tubes [103, 180] , while the study of Zheng et al. compared one-step and two-step centrifugal methods that are routinely used in plasma miRNA analysis [187] . Since most miRNA molecules are bound to proteins [188] or encapsulated in vesicles, it has been shown that double centrifugation protocol decreases the quantity of plasma miRNAs, because exosomal or protein complexes containing miRNAs may be precipitated in a certain degree after two-step centrifugation [187] . In line with this, plasma preparation by the two-step centrifugation significantly resulted also in lower cf-miRNA levels, when compared to both plasma and serum samples processed by low-speed centrifugation [189] . In case of cf-lncRNAs analyzes, single low-speed centrifugation at 800× g for 10 min at room temperature can be used [190] . In addition to cfDNA and cfRNAs, cf-mtDNA can also be isolated from pure plasma samples. When comparing four different blood-processing treatments, including a standard double centrifugation protocol, filtration with 0.22 µm filter, ultracentrifugation at 99,960× g for two hours and no additional treatment, the two-step centrifugation followed by filtration or ultracentrifugation is more efficient for removing particle-associated mtDNA [191] . When combined with the recently described protocol for total RNA sequencing, which allows assembly of the complete mitochondrial genome, it could be an effective method for cell-free whole mitochondrial genome analysis [192] . After the cell-free plasma preparation and removal of cellular components, exosomes may be isolated using size exclusion chromatography [193] , using differential centrifugation coupled with ultracentrifugation [194] , epithelial cell adhesion molecule immunoaffinity pull-down, or using OptiPrep density gradient separation, while when comparing the latter three methods, OptiPrep density gradient was found to be superior in isolating pure exosomal populations from blood plasma [195] . The various potential biomarkers and the genetic material within saliva may require different conditions for optimal preservation. Furthermore, storage methods may differ based on the type of downstream application needed. Samples should be processed as soon as possible after collection, ideally within 24 h or stored immediately under ultralow temperatures at −80 • C. During transport, samples should be kept at 4 • C. A study by the UK Biobank showed that there was a little change in the quantity of many salivary fluid components, including genetic material such as mRNA, under the Biobank's normal handling conditions, which involves keeping saliva samples at 4 • C for 24 h prior to freezing them at −80 • C [196] . Moreover, the quantity and integrity of DNA and RNA within whole saliva samples have been shown to be relatively preserved for at least five years when samples were kept at −80 • C [197] . To remove cell debris, bacterial cells and contaminants, whole saliva samples require centrifugation. For cfDNA isolation, for a maximum of one hour after collection, the saliva samples are centrifuged at 300× g for 20 min and followed by at 10,000× g for 20 min [31] . In the study of El-Mogy et al. [198] the saliva samples were processed by single-step centrifugation at 200× g for 10 min to isolate small cfRNAs. Centrifugation at 3000× g for 15 min at 4 • C was used by Yu et al. to exosomal PD-L1 mRNA isolation [199] . As the gold standard method for salivary exosome isolation density gradient or sucrose cushion ultracentrifugation at 100,000× g was reported [200] . On the other hand, ultracentrifugation protocols at 160,000× g for one hour at 4 • C for exosomal isolation were used with previous double centrifugation at 1500× g for 10 min and 17,000× g for 15 min at 4 • C to remove cell debris [123] . In recent years, several exosome and EMVs extraction protocols have been proposed and some of them were summarized by Konoschenko et al. [201] . For example, to remove contaminants, the saliva samples could be centrifuged at 3000× g for 15 min. Then the supernatant is filtered with 0.2 µm filters and precipitation solution was added. Overnight incubation at 4 • C with following centrifugation at 3000× g for 30 min to vesicles precipitation and 1500× g for 5 min to remove residual supernatant were performed [122] . For exosomal precipitation the refrigeration at 4 • C overnight was also used, followed by exosomes collection using a two-step centrifugation at 1500× g for 30 min [199] . Centrifugation of the whole urine sample is required to separate the cell-free supernatant from residual cells. This could be performed as a single high-speed centrifugation, for example at 16,000× g for 10 min [202] , or a single low-speed centrifugation, for example at 850× g [148] , that was reported to be enough to well separate cells with no change in the yield and the quality of urinary cfDNA. Other single step protocols were also reported to be functional, such as single centrifugation at 4000× g for 10 min, or 3000× g for 15 min [147] , suggesting a wide functional range for centrifugation protocols. On the other hand, there are studies that prefer a double centrifugation protocol, first at 200× g followed by 1800× g for 10 min [153] , or first low-speed centrifugation at 1600× g for 10 min followed by second high-speed centrifugation at 16,000× g for 10 min at 4 • C [136] . Two-step centrifugation at 200× g for 10 min followed by a second centrifugation at 1000× g for 10 min was also reported to be used [152] . After removal of any residual contaminants, the cell-free supernatants are usually stored at −80 • C until further analysis [152, 203, 204] . Similarly to other body fluids, exosomes can be isolated from urine samples using a combination of several methods, including differential centrifugation coupled with ultracentrifugation, ultrafiltration, precipitation with polyethylene glycol, size exclusion chromatography and immunoisolation, which are summarized by Zhang et al. [205] . Although ultracentrifugation is a widely used method, combination of size exclusion chromatography and ultrafiltration may provide even higher urinary exosome concentration [206] . One of the alternative approaches to urine exosome isolation is filtration. There are many different methods based on filtration, including nanomembrane concentrators with a membrane pore size small enough to block exosomes and large enough for water, electrolytes, and soluble proteins to pass through. Centrifugation is used to enhance flow, but the forces required are lower, unlike ultracentrifugation [207] . Another option is to use dialysis membranes, although the purification time is typically longer [208] . Alternatively, size exclusion chromatography with exosomes collected in the initial flow-through fraction may be used [209] . Exosomes can be also precipitated by altering the properties of the surrounding fluid and further isolated by centrifugation at lower speed using standard laboratory centrifuges [210] . There are also commercial reagents available based on volume-excluding polymers [211] and several different polymers, while different molecular weights and concentrations of such polymers are suitable for certain body fluids. Polymers are sequestering water causing the precipitation of less soluble material. This method is simple, but the purity of the preparation is questionable [212, 213] . Exosomes can also be isolated based on their affinity to different substrates, such as heparin [214] and lectins [215] which have been used for their agglutination. Spin column-based kits [216] are now available utilizing an electrostatic interaction between the negatively charged exosomes and a capture surface functionalized with positively charged ions [217] . The greatest specificity offers the use of antibody-based affinity. This method potentially enables capture of exosomes from specific cell types or simultaneous capture and detection [218] . Exosomes can be further isolated using antibodies conjugated to beads [219] , magnetic beads [220] , porous silica substrates [221] , and directly in coated assay wells [218] . After urinary exosomal isolation the samples should be stored at −80 • C until further analysis [222] . When compared to conventionally used body fluids, standardized operational procedures for the processing of less commonly used biological fluids for cfNAs analyses is even scarcer. In case of CSF, the processing of collected samples was reported to involve either a one-step centrifugation at 1000× g for 10 min at 4 • C [223] , a double-step protocol at 1500× g for 10 min at 4 • C and 20,000× g for 10 min, with subsequent long-term storage at −80 • C [48] , or a single centrifugation at 2000× g for 10 min at 4 • C for cf-mtDNA isolation [224] . Bronchoalveolar lavage fluid samples were processed by one-step centrifugation at 2000× g for 30 min at 4 • C [225] , at 350× g for 10 min at room temperature [226] , or at 1000× g for 10 min at 4 • C [72] . According to the WHO guidelines, to prevent sperm lysis, samples should be processed within two hours from collection at low-speed centrifugation 400× g for 10 min at room temperature, followed by high-speed centrifugation of the seminal plasma containing supernatant 16,000× g for 5 min at room temperature. Seminal plasma samples should be stored at −80 • C until cfDNA extraction [227, 228] . For isolation of exosomes from seminal plasma, after liquefication at 37 • C for 30 min, 1600× g for 10 min for first centrifugation and 16,000× g for 10 min at 4 • C for second centrifugation was used that was followed by one microfiltration step, with a pore size 0.22 µm, and ultracentrifugation at 100,000× g for two hours at 4 • C [229, 230] . Another study suggested, however, that seminal plasma filtered through the 0.22 µm filter resulted in similar cfDNA yield and size distribution than seminal plasma acquired by high-speed centrifugation (16000× g for 5 min) with or without initial low-speed centrifugation (initial spin at 400× g for 10 min followed by seminal plasma spin at 2000× g for 20 min) [54] . For the processing of whole breast milk a single-step centrifugation protocol was reported to be used, involving 1000× g for 10 min for RNA extraction [60] , or a two-step protocol, involving centrifugation at 1500× g for 10 min and at 10,000× g for 10 min at room temperature for cfDNA and cfRNA extraction [63] . Based on the few studies published, after removing the cells and debris by centrifugation, the pleural fluid supernatant should be used for cfNAs testing, while the described centrifugation protocols are mostly one-step, involving 1600× g for 15 min [231] , or 1000× g for 10 min at 4 • C [232] , with subsequent storage of supernatant either at −20 • C, or at −80 • C, respectively. In one study the same centrifugation protocol was used for the preparation of the cell-free ascites and PE supernatants. After the centrifugation at 1500 RPM (listed without specification of the rotor radius or centrifugal forces) for 5 min the samples were stored at −80 • C [71] . Like most other body fluids, pancreatic juice samples were reported to be processed for exosome isolation and cf-miRNA extraction by a two-step protocol combined with filtration and ultracentrifugation. Exosomes could be extracted by ultracentrifugation according to isolation protocol summarized in the study of Costa-Silva et al. [233] . Next, the pancreatic juice was spun at 300× g for 10 min at 4 • C followed by next centrifugation at 16,500× g for 20 min at the same temperature. Before ultracentrifugation at 140,000× g for 70 min at 4 • C, the supernatant was filtered through a 0.2 µm filter [234] . For tear fluid, after the sample collection a brief spin in a microcentrifuge is recommended, and then the tear samples should be kept on ice until cfDNA analyzes [59] . Compared to cfDNA, isolation of EMVs from tears includes initial centrifugation step at 20,000× g for 15 min at room temperature followed by dilution and filtration through 100 nm pore-size syringe filters and ultracentrifugation of filtrate at 100,000× g for 90 min at 4 • C to obtain a pellet containing EMVs [235] . For the processing of frozen amniotic fluid samples, thawing at 37 • C, vortexing for 15 s and centrifugation at 13,500× g was reported [67] . Although there are several studies describing cfNAs analyzes from less conventional body fluids, methodological sections of many of them contain incomplete information on analytical details, with the temperature at which samples should be processed missing relatively commonly. This pre-analytical factor, as well as the other steps, may influence the quantity and quality of isolated cfNAs. However, most studies agree that a second centrifugation step of suitable force reduces the cells and cell debris that can contaminate samples by cellular DNA to a minimum. Although laboratory developed assays, such as the conventional ethanol precipitation, phenol-chloroform or triton-heat-phenol-based assays, may be used for several cfNAs extraction procedures, the present day market is well equipped with commercial kits dedicated to the isolation of cfDNA and cfRNAs from the most commonly used body fluids, mostly using silica-based spin columns or magnetic beads [236] , or even the so called polymer mediated enrichment (PME) technology, which capture cfDNA by encapsulating it within the PME polymer (Table S5 ). In contrast to the previous processing steps, the "most commonly used" category in this section does not contain saliva, since we did not find commercial extraction assays dedicated to cfNAs for this body fluid. The most commonly used body fluid is blood, more specifically plasma, and since the prepared plasma is usually frozen at −80 • C, samples should be thawed prior to the extraction step and immediately processed using an appropriate cfNAs extraction kit [13] . However, these recommendations may be modified based on the recommendations of the manufacturer of the collection tube or preservation solution used for sample collection and stabilization. Despite dedicated efforts during sample processing, cfNAs in the blood may still face contamination from cellular NAs released after sampling, strongly suggesting the benefits of using extraction kits developed for cfNAs, however, the use of different kits may provide different yields of cfNAs. It should be mentioned, on the other hand, that extraction kits developed for other purposes, such as for isolation of viral NAs, were also widely used for cfNAs applications [237] . When concerning cfDNA extraction kits, in many applications the commercial spin column kits containing silica membranes are widely used [91, 238, 239] , in which NAs bind to the silica surface under high chaotropic salt conditions and are detached at low salt concentration [240] , providing relatively high yield and high purity of cfDNA [241] . From these, several comparative studies have identified either the Qiagen QIAamp Circulating Nucleic Acid Kit as one of the best-performing commercial options for plasma cfDNA extraction [238, [242] [243] [244] [245] , or the Norgen Plasma/Serum Cell-Free Circulating DNA Purification Kit [239] . With regard to this latter, although the kit has high overall yields from plasma, its recovery has been previously shown to decrease with the length of fragments [238, 244] . Raymond et al. described a two-step cfDNA purification method, relying on magnetic bead-based technology that entails extraction of total DNA followed by size-selective enrichment of the smaller fragments that are characteristic for DNA originating from fragmentation between nucleosomes. The first step of this method is extraction of total DNA from plasma with an emphasis on near-complete recovery of DNA present in the sample. The second step is size-based separation of nucleosomal-sized cfDNA fragments from high molecular weight gDNA, while the yields of cfDNA were reported to be directly comparable to the most popular QIAamp kits, but with lower overall costs [246] . The alternative approaches for the isolation of cfDNA are microfluidic and lab-on-a-chip technologies that can automate the process and reduce the time and labor cost. Compared to the common isolation kits, microfluidic technologies have several advantages, such as high throughput, sensitivity, purity and lower operating costs. There are various solid phase microfluidic extraction methods based on an extended surface area in microchannels [247] [248] [249] , miniaturized fluidic chips including silica membranes [250, 251] , or silica beads [252] [253] [254] . For cfDNA extraction and a cancer monitoring test a lab-on-a-disc system containing silica-coated beads [255] and surface modification using a non-chaotropic agent, dimethyl dithiobispropionimidate (DTBP) were used [256] . However, there are several aspects of the system, which need to be further developed for the use in clinical settings. These aspects include full automatization, large working capacity for simultaneous multiple extraction in a single operation, complete incorporation of separate experimental steps, and ease of use with low total cost. Lee et al. introduced a pressure and immiscibility-based extraction (PIBEX) method for the centrifugation-free extraction of cfDNA with a silica membrane under vacuum pressure using an immiscible liquid, such as mineral oil. However, microfluidic integration and automatic fluid control were not realized [257] . Principle of the PIBEX is based on the microfluidics associated with surface tension in silica membrane pores. Owing to these methods the entire extraction within a chip can be replaced with one continuously operating process. For the extraction of cfDNA from urine commercial kits based on silica gel membrane or magnetic beads can be used [137, 145, 147, 153] . Comparison studies are also available for several of them and in one of them the MagMAX Cell-Free DNA Isolation Kit (Thermo Fisher, Waltham, MA, USA) and the Qiagen QIAamp Circulating Nucleic Acid Kit provided the highest cfDNA yield in the 50-300 bp range, while the Thermo Fisher MagMAX Cell-Free DNA Isolation Kit and the Norgen Urine Cell-Free Circulating DNA Purification Midi Kit gave the highest cfDNA yield in the 50-100 bp range. In particular, the Urine Cell-Free Circulating DNA Purification Midi Kit was efficient for the isolation of more fragmented cfDNA in the range of 50-100 bp with the lowest cellular gDNA contamination. Another study reported that the Zymo Quick-DNA Urine Kit had the best cost-efficiency for isolating the same amount of urinary cfDNA [137] . However, it is possible to use other extraction methods too, such as poly-Lys-coated particles, which demonstrate strong affinity for negatively charged molecules such as DNA. The amounts of cfDNA (1.1-1.5 µg) recovered using this method was reported to be about three times higher than that previously obtained using the commercial Zymo Quick-DNA Urine Kit, with high purity [258] . However, fragment sizes of about 20 bp, while other studies reported size ranges of cfDNAs around 100-200 bp in urine [17, 138, [258] [259] [260] [261] , may suggest lower extraction efficiency for long DNA fragments as compared to that for short ones [258] . In a modified version using amine-modified silica particles, surface area and modification significantly improved DNA extraction [262] . The Wizard resin/guanidinium thiocyanate (Wizard/GuSCN) method uses high concentrations of chaotropic GuSCN to adsorb DNA to Wizard silica resin, but was reported to have low and variable recovery, with improved recovery of moderately short targets of about 40 bp, and inability to recover the shortest fragments of about 25 bp [145] . The Q Sepharose method uses a quaternary ammonium anion exchange resin to preconcentrate DNA before desalting on a silica spin column, while is characterized by a user friendly, ready-to-go protocol [145, 263] . When extreme sensitivity and retention of the shortest fragments are required, use of hybridization capture is recommended instead [145] . Laboratory developed extraction method based on hybridization capture for urine cfDNA using a biotinylated sequence specific probe and streptavidin-coated magnetic beads has perspective for urine cfDNA applications where maximum sensitivity is required, especially in the case of an ultrashort PCR target, (e.g., 25 bp). A key limitation of hybridization capture is that, unlike silica-based methods, it will only isolate specific targeted sequences [145] . The Norgen Urine Cell-Free Circulating DNA Purification Kit uses a hybrid silica/silicon carbide spin column, where addition of silicon carbide reportedly improves yield of short DNA compared with silica alone [264] . The biggest disadvantage of this method is consistent PCR inhibition, even when using only a small volume of eluate in PCR. Therefore, this kit is unreliable for quantification [265] . Qiagen QIAamp Circulating Nucleic Acid Kit uses a silica vacuum column and reportedly improves recovery of fragmented DNA compared with other Qiagen kits. It is one of the most widely used commercial kits for plasma cfDNA extraction [266] but is not commonly used for urine cfDNA. The results suggest that the QIAamp kit is inadequate for capturing short DNA fragments. The Thermo Fisher Scientific MagMAX Cell-Free DNA Isolation Kit uses Dynabeads MyOne Silane to maximize binding kinetics and capacity but is intended primarily for plasma cfDNA. The use of the MagMax kit in urine samples, where most cfDNA fragments are too short, is not recommended. This is because the kit is extremely dependent on fragment length, as expected for a silica-based method. Similarly to other processing steps, commercial kits for extraction of cfNAs from less commonly used body fluids are yet missing. However, since biological material coming from different body fluids tends to lose certain differences following the processing steps described in the previous section, commercial assays dedicated to other body fluids are commonly used in the literature, such as those listed for breast milk, CSF or amniotic fluid (Table S5 ). Weber et al. used, for example, the same commercial miRNA extraction kit, i.e., the miRNeasy kit manufactured by Qiagen, for 12 different body fluids [60] . In other cases, like in the case of saliva, kits developed for total DNA or RNA isolation from salivary samples are reported to be used in the literature (Table S5) . Based on a recent study, downstream analytical processes, such as MPS and droplet digital PCR, could be utilized with a variety of extraction kits or collection tubes, however, the amount of input DNA should be considered in the process of validation [161] . It was also reported that concentration of extracted cfDNA using vacuum concentration may enhance the detection of fetal fragments from maternal plasma. On the other hand, centrifugal filters and spin columns may also be used, for example if automation is considered, for which vacuum concentration is less suitable because of an uneven and unpredictable sample evaporation rate [267] . Since cfNAs may have a wide range of utilities, aims of the analyzes may also vary, posing thus specific challenges for analytical methods. In applications in which the amounts of cfNAs are of major interest, simple quantification may represent the final analysis of cfNAs, while it is generally sequence nonspecific. Like in other NAs, for simple quantification of cfNAs there are at least two commonly used approaches, based on spectrophotometry and on fluorimetry, both working on different principles and having different advantages and disadvantages. In cases where the extent of degradation, contamination, or the likely origin of the fragments are also of interest, quantification may be complemented by the estimation of the size distribution of cfNAs fragments. This exploit most typically capillary electrophoresis using fluorescently stained NAs, with the possibilities of determining also the concentration of fragments representing a certain choosable size range. To extend the informational value of cfNAs, more specialized sequence specific analyses may be performed, for example for detection of the presence/absence of certain sequences using PCR amplification followed by fragment analysis on genetic analyzers, or for absolute/relative quantification using methods such as droplet digital PCR or real-time PCR, both representing quantitative PCR methods, however, working on completely different principles. From the actually available methods, however, MPS offers the most sophisticated sequence specific analytical methods for cfNAs applications, allowing, for example, relative quantification, size distribution estimations, as well as sequence and epigenetic characterization of the analyzed fragments. Importantly, sequence specific techniques allow for the detection and characterization of sequence variability, having immense biomedical significance. When considering hierarchy between these methods, it may be important to note that accurate quantification and size range estimation of cfNAs, for example using simple fluorimetry, capillary electrophoresis, real-time PCR, or even droplet digital PCR, may represent quality control steps for more sophisticated analyzes, such as MPS. Spectrophotometry, historically the first NAs quantification method, measures how much a chemical substance absorbs light by measuring the intensity of different wavelengths of the light passing through a sample solution. Measurement of absorbed or transmitted light can be used to measure the amount of a known chemical substance considering each chemical compound absorbs or transmits light over a particular wavelength range, while not only NAs have specific absorbance ranges, but also possible contaminants of the sample, such as chemicals or proteins. This can be considered for a drawback, since different substances may have the same or similar absorbance spectra, possibly leading to signal overlap and concentration overestimation [268] , but also for an advantage, with an ability to provide purity measurements of the analyzed samples. Contaminants may represent spectrophotometry-associated quantification problems in a concentration dependent manner, posing thus certain complications in cfNAs applications. Therefore, despite the generally wide dynamic range of measurable concentrations (usable concentration range, for example in NanoDrop is 0.4-15,000 ng/µL, according to the manufacturer [269]), low cfNAs concentrations, together with residual organic solvents left behind after extraction, may create certain lower limits of quantification [270] . With regard to sample volumes required for analysis, spectrophotometric applications offer as small as 0.5 µL as an analyzable sample volume, when using, for example, the pedestal forms of the NanoDrop series microvolume spectrophotometers. From the viewpoint of user friendliness, there are also possibilities of analytical parallelization both in smaller scale, up to eight samples with NanoDrop, but also in larger scale, such as when using microplate readers accommodating 96-or 384-well microplates, like the SpectraMax (Molecular Devices) or Safire (Tecan) series readers. When considering simplicity, together with the time and the cost of the analyses, spectrophotometric measurements are cheaper and faster compared to other quantification methods [271] . Methods based on fluorometry quantification rely on the detection of an excitation driven fluorescence emission of NAs binding molecules, fluorescent dyes, which may be specific to DNA (ssDNA or dsDNA) or RNA [272] . Although they generally emit light even in an unbound state, their fluorescence emission tends to increase several folds when bound directly to their target molecules [273] , so detection and quantification of NAs, including cfNAs, is highly precise and sensitive. If relatively simple fluorescence intensity measurements of a solution needs to be performed, we will refer to these methods as simple fluorometric quantification, there are several technical possibilities, from easy to use single sample measurements using fluorometers like Qubit (Invitrogen) or Quantus (Promega), offering single-or dual-channel measurements, respectively, up to higher throughput microplate readers, like the Safire series (Tecan) which allows measurements in a continuous spectrum. When compared to spectrophotometry, fluorometry is not so convenient in the estimation of the presence of different contaminants, however, it is also less susceptible to concentration overestimations occurring due to the presence of such contaminations. In addition, it has a sensitivity to accurately measure even lower concentrations of cfNAs [271] , while there are several assays for different concentration distributions (from tens of picograms up to hundreds of nanograms per microliter), as well as for different types of NAs (dsDNA, ssDNA, RNA, miRNA) [274] . Fluorometry based methods provide accurate quantification, but do not allow quantification of selective size range [275] . Partly based on the same physical principles as simple fluorometry, i.e., staining of NAs through sequence nonspecific DNA binding fluorescent dyes, capillary electrophoresis is performed in a time-resolved manner during an electrophoretic separation of NAs fragments. Accurate quantification of cfNAs on automated capillary electrophoresis provides, therefore, highly precise analytical evaluation of various NAs, even with regard to the size of the fragments. Although capillary electrophoresis of sequence specific PCR amplicons, labeled with covalently bound fluorophores, on genetic analyzers may also be used as a main analytical method [267] , sequence nonspecific quantification and sizing of cfNAs performed by capillary electrophoresis was shown to be sensitive and demonstrated good quantitation accuracy and reproducibility [270] moreover it is able to detect post-sampling cellular DNA contamination that may often be observed after cfNAs isolation [270] . Instruments like the Bioanalyzer [276] and TapeStation (Agilent, Santa Clara, CA, USA) [270, 276] , or Experion (Bio-Rad, Hercules, CA, USA) [277, 278] , for example, enable highly precise characterization of NAs concentrations and distribution of fragment sizes, however, only in certain detection ranges, which depends on the used consumables and kits. Since for DNA it is around from few picograms up to a thousand of picograms per microliter, while for RNA from tens of picograms up to thousands of picograms per microliter [279] , it may happen that the concentration of cfDNA/cfRNA will not fall within this range, whereupon additional quantification by another method may be necessary [270] . Despite certain limitations, the spread of MPS put electrophoresis, together with simple fluorimetry-based quantification, to the center of sequencing library preparation procedures, and are therefore considered prerequisites for more complex analyses of cfNAs applications. Conventional polymerase chain reaction (PCR), performed using fluorescently labelled oligonucleotide primers, coupled with an end-point detection of amplified products using capillary electrophoresis on genetic analyzers may be used for analyzes of cfNAs. It was reported to allow, for example, detection and genotyping of paternally inherited Y chromosome specific microsatellite alleles in cffDNA fragments of male fetuses present in maternal plasma [267] . Different modifications of PCR are, however, widely used among cfNAs analytical methods. Real-time quantitative PCR (qPCR) is a widely used detection and quantification method, sensitive enough to work also with low amounts of nucleic acids, including cfDNA [238] . Employing one of the several detection techniques, from DNA binding fluorescent dyes up to sequence-specific fluorescently labelled probes, it allows continuous monitoring of the amplification progression [280] . Since the increase of the amplification product depends on the initial template copy number and on the amplification efficiency, qPCR is generally used for the detection of cfNAs fragments of interest, estimation of PCR inhibition [281] , as well as for absolute or relative quantification using carefully selected reference genes [282, 283] . With regard to quantification, however, it was reported that cfDNA quantities based on measurements of some target genes were, on average, more than twofold higher than those of other assays, suggesting that analysis and averaging of multiple reference may lead to more reliable estimates of total cfDNA quantity [238] . Since the used oligonucleotides are sequence specific, qPCR allows also to determine the origin of cfNAs, discriminating, for example, between fetal and maternal fragments [284] , or between genomic and mitochondrial cfNAs [283] . In droplet digital PCR (ddPCR), sometimes regarded as a third generation of PCR technologies, target samples are partitioned, for example using oil droplets, allowing highly parallel but isolated amplification of single molecules of the same target DNA sequence, while the amplification itself, i.e., the presence or absence of the target molecule in the droplet is detected through fluorescent probes, but in and end-point manner [7] . Since it allows absolute quantification achieving orders of magnitude more precision and sensitivity than qPCR, ddPCR may be considered if quantification, but not complex size distribution estimation of NAs is required. In addition to quantification, for example of circulating fetal and maternal DNA from cell-free plasma [7] , however, ddPCR was also used for determination of sample quality and the degree of cellular DNA contamination using differently sized amplicons of a single target region [285] , or even of multiple target regions to ensure higher precision from limited quantities of cfDNA [286] . It should be mentioned here that significant differences were detected between archived and immediately processed samples, in the term of quality, when using ddPCR and comparing various extraction kits. On the other hand, comparing different collection protocols near no differences were detected in the term of quality and quantity [286] . Although conventional dideoxynucleotide-termination-based Sanger sequencing, i.e., one of the most commonly used first generation sequencing methods, may have a relatively common role in everyday clinical practice utilizing cfNAs, this method is not conventionally listed among the main cfNAs analytical technologies. It is used most commonly if the detection of certain sequences, or specific mutations, is of interest in cfNAs derived from specific body fluids, most typically in the field of oncogenetics [287] . Even in the field of cfNAs applications, MPS based second and third generation sequencing methods are gaining more and more attention [288] , especially since the housing of low-coverage whole genome sequencing with the recognition of the presence of cffDNA in maternal plasma. Following this they quickly became a popular and globally used testing strategy with rapidly increasing numbers of analyzes carried out annually [289] [290] [291] . Moreover, beyond NIPT, the use of this technique started to show not only secondary possibilities of NIPT data re-use for large-scale but low-cost population studies [5, 6] , but also an outbreak into unrelated clinical fields, such as oncology in a form of liquid biopsy-based non-invasive cancer diagnostics (NICD) [292] . Although both second and the third-generation sequencing is represented by many methods using different technological procedures, they generally work on the principles of miniaturized parallel sequencing of millions of DNA fragments. In second generation methods, fragments undergo clonal amplification during sequencing library preparation, while the sequencing length typically ranges from tens to hundreds of bases [293] . In contrast, third generation methods rely on single-molecule sequencing, i.e., they do not need clonal amplification, while they tend to have read lengths in several kilobases [294] . With regard to cfNAs applications, even in cases of fragments having different origins, such as fetal NAs in maternal plasma or cancer genomes in patient plasma, entire genomes can be characterized using MPS. Although using highly different read depths, genome-scale high-throughput screening and genotyping of the majority of DNA variant types was reported, from characterization of epigenetic markers of NAs [295, 296] , through SNVs and a wide range of CNVs [297] [298] [299] , including even microsatellites [300] and chromosomal aneuploidies [301] . Moreover, highly sensitive MPS approaches may be further modified to achieve even higher sensitivity of detection and quantification of low frequency mutations in plasma, for example using modifications such as the Safe-Sequencing System (Safe-SeqS). This utilizes assignment of unique molecular identifiers to each template molecule, clonal amplification of each uniquely tagged template molecule, followed by redundant sequencing of the amplification products [13, 302] . In addition, apart from the ability to detect a wide range of variations and modifications, the impact of MPS on cfNAs applications is further enhanced by its high sensitivity, but also by its ability to characterize both the ratios of cfNAs of different origins and the size distributions of the fragments. Determination of the ratios is based on the sequence composition itself and on the epigenetic landscape of the fragments, while their size distribution characterization may be based on insert sizes when using paired-end sequencing [297] . When estimating size ranges, however, specific steps of library preparation should be considered and possibly modified to prevent the loss of certain fragments during this highly selective process. Alongside with the above mentioned methods, although less commonly reported, there are also other analytical techniques for characterization of different aspects of cfNAs. For example for detection of low frequency mutations in tumor associated cfDNAs, high specificity and sensitivity was reported using the combination of common methods like high-resolution melting (HRM) analysis of PCR amplicons, restriction fragment length polymorphism (RFLP) assays and Sanger sequencing [287] . BEAMing, called on the basis of four principal components of the method (i.e., beads, emulsion, amplification, and magnetics) combines oligonucleotide-coupled-bead-based emulsion PCR amplification, single base extension and counting of fluorescently labeled particles via flow cytometry [303] , allowing for highly sensitive detection and quantification of somatic mutations in the plasma of patients, for example suffering from colorectal tumors [304] . From the highly parallel genome-wide techniques, although high-throughput microarray-based assays (like array-based comparative genomic hybridization) are not so frequently used in cfNAs applications, examples of their use do also exist [129, 305] . Fluorometric quantification using fluorescent dyes is an approach that compensates for some disadvantages of spectrophotometry-based quantification. Nevertheless, these two quantification methods allow the most effective, fast, very reliable and not much expensive cfNAs quantification that can be repeated many times from low amounts of input material. It is highly recommended to use the electrophoresis based method in parallel with fluorometric methods when quantification and sizing is required for the final evaluation of cfNAs profiles, or for downstream analyzes [177, 275, 306] . However, the detection range of some electrophoresis-based methods is very narrow and it is unlikely that the majority of samples will have similar concentration which will fall within its upper limit of quantification. The overestimated concentration may lead to chip overloading, which in turn provides inaccurate results, so this quantification is generally not suitable for first-line quantification of cfDNA [270] . Majority of laboratories prefer simple fluorometric quantification, which provides an easy workflow and saves time as compared to qPCR. qPCR is a sensitive, accurate and reproducible quantification method, however, it requires broad optimization of the PCR reaction conditions [177] or to obtain pre-optimized commercial qPCR assays which are generally much more expensive. ddPCR quantification is highly sensitive, reproducible and modern technology, but comparing the cost and time required for measurement is very convenient [307] . Despite several advantages, neither qPCR nor ddPCR is able to determine the size range of fragments, however, with certain modifications they may be used to estimate degradation status of the sample [285, 286] . On the other hand, it is absolutely important to realize when designing and performing cfNAs analyzes that concentration estimates generally differ when using different methods, even for the same sample, mainly because of different principles and limitations of the methods, as well as because several factors may deteriorate results [268] . When comparing spectrophotometric, simple fluorometric (using PicoGreen) and qPCR-based (using SYBR Green I assay) quantification methods in the light of DNA fragmentation, although the overall sensitivity of the spectrophotometric measurement was found to be the lowest, this was the only method remaining unaffected by fragmentation of the measured fragments. Both the fluorometric and the qPCR-based assays were significantly influenced, with a decrease in measured concentration connected to more intensive DNA fragmentation, suggesting that DNA quantification should be performed using equally fragmented control DNAs [308] . When considering the sequencing methods, it is important to keep in mind that different sequencing platforms suffer from contrasting limitations and laboratory biases, making them suitable for different classes of problems. As we mentioned above, second generation sequencers generate high yields of relatively high-quality sequenced fragments that have established their dominant position in the direct detection of small genomic variations in human genomes, especially SNVs and small CNVs, or methylation patterns [309] . However, due to their short size, they are impractical for direct detection of large genomic variations, such as larger CNVs or other structural rearrangements [310] . When these types of variants need to be assessed from short read sequencing data, indirect statistical evaluation of target region coverage is required (for details see the next chapter and citations there), while this is impractical for the detection of balanced rearrangements. Third generation sequencers promise to alleviate this problem by sequencing of much longer fragments, albeit yet at the cost of much lower base quality. The appropriate platform should be, therefore, carefully selected according to the goals of the testing, while combination of several platforms may also offer the potential to employ their complementary advantages to improve overall results [311] . Many of the abovementioned analytical methods rely on computational data collection, processing and evaluation, while in the majority of cases the relevant instruments are accompanied with dedicated commercial software. This is yet highly different in case of MPS. Although there are dedicated platform-associated bioinformatic tools too, extensive genome-scale data sets, having wide potential for further reanalysis and reuse also for other purposes than for which they were originally generated, promote the development of newer and newer bioinformatic tools and pipelines. As a result, bioinformatic tools for processing MPS data are still under intensive development and diversification [312] . On the other hand, an unprecedented amount of genomic data is generated every day, even for cfNAs, which poses a major challenge for computational and statistical methods to reveal usable patterns and answers to the diagnostic question, or to the experimental hypotheses. However, virtually all applications are based on sampling, laboratory processing, and measurements, which inevitably bias the generated data from the actual genomic content of the sample. Improperly prepared cfNAs may lead to unsuccessful method performance and consequently to the results misinterpretation. Therefore, the generated data should always be checked for known forms of distortions [313] , and state-of-the-art methods should be applied to mitigate their effects and remove sequencing artifacts. Although some bias can be reduced without losing sequenced data [314, 315] , it is more common to remove sequencing artifacts such as low-quality base calls [316] , remnants of sequencing adapters [316, 317] , contamination induced by laboratory processing [318] , or artificial duplications induced by PCR amplification [319] . Since each such preprocessing step lengthens the computational analysis, makes it more complex, and therefore more prone to failure, and most importantly reduces the data set, refinements in the laboratory methods should be preferred to increase the yield of high-quality genomic data for subsequent analyzes. In general, if germline variants of the analyzed individual are to be detected from his own cfNAs, they are present in a large portion of the sequenced fragments making them easier to detect. Conventional tools may be utilized for data processing, such as Bowtie 2 for alignment with subsequent specific normalization steps and, for example CNV calling on normalized bin counts using circular binary segmentation algorithm provided by the R package DNAcopy [6, 299] . On the other hand, the great challenge is to uncover the indicative patterns in fragments that are present only in the small fraction of the DNA mixture. Since oncological diseases are typical with frequent somatic variations due to defects in genome maintenance [320] , ranging from small variations [321] to even large structural variations [322] and methylation patterns [323] , detection of unusually high, but low frequency variation among the sequenced fragments may be indicative [324, 325] . Increasing sequencing depth is a widely used approach to improve mutation calling performance, which can be complemented by fine tuning of pipelines to ensure high specificity in identifying somatic variants supported by only a few reads, since variant caller tools such as Mutect [326] , Varscan [327] , Vardict [328] , Strelka2 [329] , may perform differently in different sequencing depth and variant frequency [330] . The problem of low frequency variants is similar also in non-invasive prenatal care, specifically in cases when germline sequence variants of a fetus are to be detected among the cfDNA fragments of the mother. When identifying larger genomic variants, such as CNVs, derived for example from the fetal genome but present in the maternal plasma, detection methods generally does not rely on the direct identification of the variant in the reads, due to length limitations, rather on indirect assessment through the relative number of mapped reads to different genomic regions. These measurements are quantitative, while the aberrant number of DNA material from the genomic region indicates the duplication or deletion of the affected chromosomal segment. Similarly to maternal CNVs described above, sample normalization, read counting for individual bins and CNV calling was described to be efficient for the identification of fetal CNVs in cfDNA isolated from maternal plasma [299, 331] . The main problem of this field is, however, still the relatively small proportion of fetal DNA present in the maternal blood, typically from five to 15% of the cfDNA mixture, that is most commonly alleviated by large amounts of sequenced DNA fragments that allow to more precisely distinguish the aberrant number of fragments from the affected fetal chromosome from the inaccuracies of measurements. A lot of effort has been put into developing more robust biostatistical methods to reduce the number of required sequenced fragments, and thus increase the viability of tests by statistical highlighting of fragments that are more likely to originate from the tissue of interest. So far, several characteristics with different separation capabilities have been described. The combination of these complementing characteristics into a single meta-predictor has great promise for improving overall accuracy and detecting new useful patterns in the sequenced fragments [332, 333] . Fragment lengths are a well-studied aspect of the sequenced cfNA fragments in several clinical domains [334] , since they tend to differ in different body fluids (Table S2 ), but also with regard to the cells of origin and mechanisms of release [335] . Although length alone cannot be unambiguously used to distinguish fragments, it can be used to estimate the proportions of a DNA mixture [333] or to focus analysis on fragments belonging to selected size ranges using robust statistical modeling [331, 332] . Length of cfDNA fragments may be determined as the difference of the leftmost and the rightmost mapped base of the corresponding read pair, following, for example, mapping using the Bowtie 2 algorithm [333, 336] . Mapping location is also a powerful metric for finding relevant patterns. Although analysis focused only on a specific region is too unreliable, the combination of a signal along the entire genome can reveal useful patterns [337, 338] . In addition, profiling end motifs and centering around nucleosomes may also indicate the tissue of origin, albeit typically with less accuracy [339] . Moreover, in addition to the high importance of germline and somatic genomic variation of the analyzed individual, a significant amount of genomic material sequenced from the sampled material cannot be directly attributed to the known human genome, i.e., remains unmapped following alignment to the human reference genome. This genomic "dark matter" typically represents the organisms present in the host, and so their sequencing represents a great opportunity to monitor the balance of the microbiome or to detect potentially harmful pathogens [340] . With the improvements of the sequencing technologies, algorithms, and statistical methods, along with ongoing efforts to capture genomic diversity of all living organisms, we can expect massive improvements in the detection of pathogens or harmful disbalances of the microbiome. cfNAs are present in various body fluids, including those commonly assessed, such as blood plasma, saliva, or urine, but also in less commonly considered body fluids, which may be, on the other hand, in direct contact with hardly accessible tissues or organs. They may reflect genomic alterations, ongoing physiological processes as well as changes in the organism in response to various stimuli. Therefore, main biomedical applications consider and use them as biomarkers of several pathological conditions, but their role as potential therapeutic targets is also emerging. cfNAs content of many body fluids in certain diseases is still under intensive research, so they had limited or no use in clinical practice, but certainly not to the same extent as blood plasma. Although cfNAs represent promising material, their analyzes is not so easy with many different approaches existing, since standardized protocols for the processing of cfNAs are still missing. Moreover, currently available methods for analyzes require cfNAs input of sufficient quality and quantity to obtain reliable results, therefore individual steps of the whole workflow should be carefully considered and designed. As we described above, this workflow consists of several steps, including (i) sample collection, storage, and transport, (ii) processing of biological material, (iii) extraction of cfNAs, (iv) analytical process, and (v) data analysis and interpretation of the results. Each level of this workflow may be affected by environmental and technical factors, which should be considered, especially in the case of those applications where the collection site of the biological material is usually elsewhere than the processing laboratory. Since cfNAs present in plasma are the most studied, the majority of sampling devices, buffers, protocols for processing, or extraction kits are designed for the analysis of cfNAs originated from this source. For those less commonly analyzed body fluids, specialized collection devices and/or preservation solutions have not yet been developed. Because of these limitations, commercial assays developed for total DNA or RNA extraction from the matching body fluid, or assays dedicated to extraction of cfNAs but from other body fluids, are commonly reported in the literature. Taken together, since each of the delineated steps and procedures may contain several more or less appropriate modules, the best choice to design the most appropriate combination will always depend on several factors, including the aims of the analyzes, application requirements, available budget, but also the technical and personal infrastructure of the workplaces involved in the analyzes. And last but not least, thorough validation of the whole complex process should always be performed before its implementation takes place. Supplementary Materials: Supplementary material contains five tables and can be found at http://www.mdpi. com/1422-0067/21/22/8634/s1. Table S1 Concentrations of cfNAs in different human body fluids., Table S2 The size range of cfNAs in different human body fluids., Table S3 The list of collection tubes or stabilization solutions developed for the preservation of cfNAs from different body fluids., Table S4 The list of methods developed for the isolation of exosomes from different body fluids., Table S5 The list of methods developed for the isolation of cfNAs from different body fluids. Circulating cell-free nucleic acids: Characteristics and applications Recent trends in prenatal genetic screening and testing Quantification of peripheral whole blood, cell-free plasma and exosome encapsulated mitochondrial DNA copy numbers in patients with atrial fibrillation Cell-Free Nucleic Acids and their Emerging Role in the Pathogenesis and Clinical Management of Inflammatory Bowel Disease Non-invasive prenatal testing as a valuable source of population specific allelic frequencies Identification of Structural Variation from NGS-Based Non-Invasive Prenatal Testing High-throughput droplet digital PCR system for absolute quantitation of DNA copy number Does maternal saliva contain fetal DNA usable for prenatal diagnostics? Cell-free fetal nucleic acids in amniotic fluid Application of cell-free DNA sequencing in characterization of bloodborne microbes and the study of microbe-disease interactions Cell-Free DNA: An Overview of Sample Types and Isolation Procedures Performance comparison of blood collection tubes as liquid biopsy storage system for minimizing cfDNA contamination from genomic DNA Performance of Streck cfDNA Blood Collection Tubes for Liquid Biopsy Testing Liquid Biopsy Preservation Solutions for Standardized Pre-Analytical Workflows-Venous Whole Blood and Plasma Sources and Functions of Extracellular Small RNAs in Human Circulation Non-invasive prenatal diagnosis from the perspective of a low-resource country Genetic analysis of DNA excreted in urine: A new approach for detecting specific genomic DNA sequences from cells dying in an organism DNA fragments in the blood plasma of cancer patients: Quantitations and evidence for their origin from apoptotic and necrotic cells Predominant hematopoietic origin of cell-free DNA in plasma and serum after sex-mismatched bone marrow transplantation Circulating DNA and myeloperoxidase indicate disease activity in patients with thrombotic microangiopathies The importance of examining the proportion of circulating DNA originating from tumor, microenvironment and normal cells in colorectal cancer patients Circulating mRNAs and miRNAs as candidate markers for the diagnosis and prognosis of prostate cancer Recovering circulating extracellular or cell-free RNA from bodily fluids Extracellular vesicles: Biology and emerging therapeutic opportunities Cell-free nucleic acids as noninvasive biomarkers for colorectal cancer detection The Effect of Preservative and Temperature on the Analysis of Circulating Tumor DNA Cell-free DNA in maternal plasma and serum: A comparison of quantity, quality and tissue origin using genomic and epigenomic approaches Analytical and clinical validation of a microbial cell-free DNA sequencing test for infectious disease Free DNA, a reason for severe COVID-19 infection? Saliva-derived cfDNA is applicable for EGFR mutation detection but not for quantitation analysis in non-small cell lung cancer Characterization of RNA in saliva RNA Sequencing Analysis of Salivary Extracellular RNA Removal of high-molecular-weight DNA by carboxylated magnetic beads enhances the detection of mutated K-ras DNA in urine Cell-free nucleic acids in urine as potential biomarkers of kidney disease High resolution size analysis of fetal DNA in the urine of pregnant women by paired-end massively parallel sequencing Extracellular Nucleic Acids in Urine: Sources, Structure, Diagnostic Potential Clinical applications of urinary cell-free DNA in cancer: Current insights and promising future DNA alterations in body fluids as molecular tumor markers for urological malignancies Liquid biopsy: A step forward towards precision medicine in urologic malignancies Cell-free fetal DNA in the cerebrospinal fluid of women during the peripartum period Multitarget stool DNA testing for colorectal-cancer screening Cell-free DNA testing: Future applications in gastroenterology and hepatology The Host Shapes the Gut Microbiota via Fecal MicroRNA Regulation of cerebrospinal fluid (CSF) flow in neurodegenerative, neurovascular and neuroinflammatory disease Preanalytical variables that affect the outcome of cell-free DNA measurements Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma Detection of cell-free DNA fragmentation and copy number alterations in cerebrospinal fluid from glioma patients Therapeutic management of neuro-oncologic patients-Potential relevance of CSF liquid biopsy Clinical Utility of Cerebrospinal Fluid Cell-Free DNA as Liquid Biopsy for Leptomeningeal Metastases in ALK-Rearranged NSCLC The biomarker potential of cell-free microRNA from cerebrospinal fluid in Parkinsonian Syndromes More than cell dust: Microparticles isolated from cerebrospinal fluid of brain injured patients are messengers carrying mRNAs, miRNAs, and proteins Quick recovery and characterization of cell-free DNA in seminal plasma of normozoospermia and azoospermia: Implications for non-invasive genetic utilities Seminal cell-free DNA levels measured by PicoGreen fluorochrome are associated with sperm fertility criteria Double Stranded RNA in Human Seminal Plasma Cell-free seminal mRNA and microRNA exist in different forms Enríquez-de-Salamanca, A. Tear fluid biomarkers in ocular and systemic disease: Potential use for predictive, preventive and personalised medicine Tear fluid extracellular DNA: Diagnostic and therapeutic implications in dry eye disease The microRNA spectrum in 12 body fluids Can free DNA be detected in sputum of lung cancer patients? Lung Cancer Differences in the genomic profiles of cell-free DNA between plasma, sputum, urine, and tumor tissue in advanced NSCLC Potential of Using Cell-Free DNA and miRNA in Breast Milk to Screen Early Breast Cancer Exosomes with immune modulatory features are present in human breast milk Human saliva, plasma and breast milk exosomes contain RNA: Uptake by macrophages Transcriptome profiling of microRNA by Next-Gen deep sequencing reveals known and novel miRNA species in the lipid fraction of human breast milk Large amounts of cell-free fetal DNA are present in amniotic fluid Cell-free DNA in the supernatant of pleural effusion can be used to detect driver and resistance mutations, and can guide tyrosine kinase inhibitor treatment decisions Next-generation sequencing reveals mutational accordance between cell-free DNA from plasma, malignant pleural effusion and ascites and directs targeted therapy in a gastric cancer patient Liquid biopsies using pleural effusion-derived exosomal DNA in advanced lung adenocarcinoma Cell-Free DNA from Ascites and Pleural Effusions: Molecular Insights into Genomic Aberrations and Disease Biology Assessment of EGFR mutation status using cell-free DNA from bronchoalveolar lavage fluid Quantification of free RNA in serum and bronchial lavage: A new diagnostic tool in lung cancer detection? Lung Cancer Potential of Aqueous Humor as a Surrogate Tumor Biopsy for Retinoblastoma Non-invasive diagnosis of retinoblastoma using cell-free DNA from aqueous humour the vitreous humor of patients affected by idiopathic epiretinal membrane and macular hole Cell free DNA as a component of forensic evidence recovered from touched surfaces Genomic analysis of pancreatic juice DNA assesses malignant risk of intraductal papillary mucinous neoplasm of pancreas Bile cell-free DNA as a novel and powerful liquid biopsy for detecting somatic variants in biliary tract cancer Identification of Specific Joint-Inflammatogenic Cell-Free DNA Molecules From Synovial Fluids of Patients With Rheumatoid Arthritis. Front Cell-free DNA in the peritoneal effluent of peritoneal dialysis solutions Peritoneal Cell-free DNA: An innovative method for determining acute cell damage in peritoneal membrane and for monitoring the recovery process after peritonitis Neutrophil-derived miR-223 as local biomarker of bacterial peritonitis Pancreatic cyst fluid DNA analysis in evaluating pancreatic cysts: A report of the PANDA study Mutational Mosaics of Cell-Free DNA from Pancreatic Cyst Fluids Next generation sequencing of pancreatic cyst fluid microRNAs from low grade-benign and high grade-invasive lesions Diagnostic potential of tumor DNA from ovarian cyst fluid Circulating microRNAs as stable blood-based markers for cancer detection Abstract 4586: Evaluation of available blood collection tubes for use in stabilizing concentrations of extracellular vesicles / exosomes and associated cell-free RNA EDTA is a better anticoagulant than heparin or citrate for delayed blood processing for plasma DNA analysis Optimised Pre-Analytical Methods Improve KRAS Mutation Detection in Circulating Tumour DNA (ctDNA) from Patients with Non-Small Cell Lung Cancer (NSCLC) Evaluation of Streck BCT and PAXgene Stabilised Blood Collection Tubes for Cell-Free Circulating DNA Studies in Plasma Optimizing blood collection, transport and storage conditions for cell free DNA increases access to prenatal testing Comparative analysis of circulating tumor DNA stability In K3EDTA, Streck, and CellSave blood collection tubes Effects of Collection and Processing Procedures on Plasma Circulating Cell-Free DNA from Cancer Patients Evaluation of Storage Tubes for Combined Analysis of Circulating Nucleic Acids in Liquid Biopsies Genetic profiling of tumours using both circulating free DNA and circulating tumour cells isolated from the same preserved whole blood sample Plasma free DNA: Evaluation of temperature-associated storage effects observed for Roche Cell-Free DNA collection tubes Carbon-13 nuclear magnetic resonance analysis of formaldehyde free preservatives Modifications of human and viral deoxyribonucleic acid by formaldehyde fixation Effects of a novel cell stabilizing reagent on DNA amplification by PCR as compared to traditional stabilizing reagents Specialized Blood Collection Tubes for Liquid Biopsy: Improving the Pre-analytical Conditions Stabilization of cell-free RNA in blood samples using a new collection device A novel blood collection device stabilizes cell-free RNA in blood during sample shipping and storage Stabilization of circulating tumor cells in blood using a collection device with a preservative reagent Liquid biopsy-Performance of the PAXgene®Blood ccfDNA Tubes for the isolation and characterization of cell-free plasma DNA from tumor patients Comparison of Roche Cell-Free DNA collection Tubes to Streck Cell-Free DNA BCT s for sample stability using healthy volunteers Genome-wide study of the effect of blood collection tubes on the cell-free DNA methylome Abstract 2758: Comparison of cell-free DNA blood collection tubes Comparison of Cell-Free DNA Blood Collection Tubes for Low-Frequency Variant Detection Influence of temperature during transportation on cell-free DNA analysis Evaluation of Different Blood Collection Tubes and Blood Storage Conditions for the Preservation and Stability of Cell-Free Circulating DNA for the Analysis of the Methylated mSEPT9 Colorectal Cancer Screening Marker Comparison of Blood Collection Tubes from Three Different Manufacturers for the Collection of Cell-Free DNA for Liquid Biopsy Mutation Testing Saliva: A dynamic proteome Collection, storage, and processing of saliva samples for downstream molecular applications Collection and extraction of saliva DNA for next generation sequencing RNAPro•SAL: A device for rapid and standardized collection of saliva RNA and proteins Human salivary protein extraction from RNAPro·SAL TM , Pure·SAL TM , and passive drooling method Comparability of the small RNA secretome across human biofluids concomitantly collected from healthy adults Characterization of cellular and extracellular DNA in saliva Total Extracellular Small RNA Profiles from Plasma, Saliva, and Urine of Healthy Subjects. Sci. Rep. 2017 Salivary extracellular vesicle-associated miRNAs as potential biomarkers in oral squamous cell carcinoma Exosomes from human saliva as a source of microRNA biomarkers Possible role for interleukins as biomarkers for mortality and recurrence in oral cancer Method development for proteome stabilization in human saliva A comparison of whole mouth resting and stimulated salivary measurement procedures Methods for collecting saliva Interleukin 6 and interleukin 8 as potential biomarkers for oral cavity and oropharyngeal squamous cell carcinoma RNA profiling of cell-free saliva using microarray technology Salivary proteomic and genomic biomarkers for primary Sjögren's syndrome. Arthritis Rheum Biochemistry of human alpha amylase isoenzymes Saliva as a matrix for human biomonitoring in occupational and environmental medicine Prevalence and persistence of male DNA identified in mixed saliva samples after intense kissing Evaluation and comparison of in vitro degradation kinetics of DNA in serum, urine and saliva: A qualitative study Measurement of deoxyribonuclease I activity in human tissues and body fluids by a single radial enzyme-diffusion method New method to preserve the original proportion and integrity of urinary cell-free DNA Comparison of Four Commercial Kits for Isolation of Urinary Cell-Free DNA and Sample Storage Conditions Isolation and comparative study of cell-free nucleic acids from human urine Cell-Free DNA Integrity Analysis in Urine Samples Evaluation of cell-free DNA in urine as a marker for bladder cancer diagnosis Urinary cell-free DNA is a versatile analyte for monitoring infections of the urinary tract Urine Cell-Free DNA Integrity Analysis for Early Detection of Prostate Cancer Patients Analysis of extracellular mRNA in human urine reveals splice variant biomarkers of muscular dystrophies Urinary Cell-Free DNA Quantification as Non-Invasive Biomarker in Patients with Bladder Cancer Analytical Comparison of Methods for Extraction of Short Cell-Free DNA from Urine RNA Detection in Urine: From RNA Extraction to Good Normalizer Molecules The art of obtaining a high yield of cell-free DNA from urine Urinary Cell-Free DNA: Isolation, Quantification, and Quality Assessment Evaluation of Cell-Free Urine microRNAs Expression for the Use in Diagnosis of Ovarian and Endometrial Cancers Cell-Free Urinary MicroRNAs Expression in Small-Scale Experiments Evaluation of the "Colli-Pee", a first-void urine collection device for self-sampling at home for the detection of sexually transmitted infections, versus a routine clinic-based urine collection in a one-to-one comparison study design: Efficacy and acceptability among MSM in Belgium Pre-Analytical Handling Conditions and Small RNA Recovery from Urine for miRNA Profiling Quantifying cell free DNA in urine: Comparison between commercial kits, impact of gender and inter-individual variation Standardization of sample collection, isolation and analysis methods in extracellular vesicle research A comprehensive automated pipeline for human microbiome sampling, 16S rRNA gene sequencing and bioinformatics processing Altered Fecal Small RNA Profiles in Colorectal Cancer Reflect Gut Microbiome Composition in Stool Samples Human faecal collection methods demonstrate a bias in microbiome composition by cell wall structure A Pipeline for Faecal Host DNA Analysis by Absolute Quantification of LINE-1 and Mitochondrial Genomic Elements Using ddPCR Sputum Cell-Free DNA: Valued Surrogate Sample for Detection of EGFR Mutation in Patients with Advanced Lung Adenocarcinoma Milk DNA Preservation and Isolation Kit (Cat. 44800) Multicenter Evaluation of Circulating Cell-Free DNA Extraction and Downstream Analyses for the Development of Standardized (Pre)analytical Work Flows cfNOMe-A single assay for comprehensive epigenetic analyses of cell-free DNA Salivary Exosomes as Nanocarriers for Cancer Biomarker Delivery Integrated immunoisolation and protein analysis of circulating exosomes using microfluidic technology Chip-based analysis of exosomal mRNA mediating drug resistance in glioblastoma A microfluidic ExoSearch chip for multiplexed exosome detection towards blood-based ovarian cancer diagnosis Ciliated micropillars for the microfluidic-based isolation of nanoscale lipid vesicles Microfluidic filtration system to isolate extracellular vesicles from blood Microfluidic device (ExoChip) for on-chip isolation, quantification and characterization of circulating exosomes Quantitative and sensitive detection of rare mutations using droplet-based microfluidics Single-molecule detection of epidermal growth factor receptor mutations in plasma by microfluidics digital PCR in non-small cell lung cancer patients Cell-free DNA release under psychosocial and physical stress conditions Multisociety Consensus Quality Improvement Revised Consensus Statement for Endovascular Therapy of Acute Ischemic Stroke Circulating nucleic acids in cancer and pregnancy Application and optimization of minimally invasive cell-free DNA techniques in oncogenomics Plasma cell-free DNA level and its integrity as biomarkers to distinguish non-small cell lung cancer from tuberculosis Guidelines for the Preanalytical Conditions for Analyzing Circulating Cell-Free DNA Circulating cell free DNA: Preanalytical considerations Canadian Institutes of Health Research Team on Cellular Aging and HIV Comorbidities in Women and Children (CARMA) Circulating Cell-Free DNA and RNA Analysis as Liquid Biopsy: Optimal Centrifugation Protocol Preanalytical blood sample workup for cell-free DNA analysis using Droplet Digital PCR for future molecular cancer diagnostics A Collection of Tissue, Fluid Samples and Associated Data of Oncology Patients for the Use in Translational Research Quantifying circulating cell-free DNA in humans Cell-free DNA: Preanalytical variables Circulating cell-free DNA in cancer Feasibility Analysis of Cell-Free DNA Derived from Plasma of Lung Cancer Patients for Next-Generation Sequencing Centrifugation: An important pre-analytic procedure that influences plasma microRNA quantification during blood processing Argonaute2 expression is post-transcriptionally coupled to microRNA abundance Future-Proofing" Blood Processing for Measurement of Circulating miRNAs in Samples from Biobanks and Prospective Clinical Trials Circulating lncRNAs associated with occurrence of colorectal cancer progression Quantitative analysis of circulating mitochondrial DNA in plasma Ultracentrifugation enrichment protocol followed by total RNA sequencing allows assembly of the complete mitochondrial genome Specialized Cell-Free DNA Blood Collection Tubes Can Be Repurposed for Extracellular Vesicle Isolation: A Pilot Study Exosomes provide a protective and enriched source of miRNA for biomarker profiling compared to intracellular and cell-free blood Comparative proteomics evaluation of plasma exosome isolation techniques and assessment of the stability of exosomes in normal human blood plasma Effects of the UK Biobank collection protocol on potential biomarkers in saliva Quality assessment of saliva bank samples Diversity and signature of small RNA in different bodily fluids using next generation sequencing Detection of Exosomal PD-L1 RNA in Saliva of Patients With Periodontitis Isolation and characterization of exosomes from cell culture supernatants and biological fluids Isolation of Extracellular Vesicles: General Methodologies and Latest Trends Urinary Biomarkers Indicative of Apoptosis and Acute Kidney Injury in the Critically Ill Detection of bladder cancer using urinary cell-free DNA and cellular DNA Clinical Significance of Hotspot Mutation Analysis of Urinary Cell-Free DNA in Urothelial Bladder Cancer Methods and Technologies for Exosome Isolation and Characterization Development and comparative analysis of human urine exosome isolation strategies Rapid isolation of urinary exosomal biomarkers using a nanomembrane ultrafiltration concentrator A simplified method to recover urinary vesicles for clinical applications, and sample banking Ready-made chromatography columns for extracellular vesicle isolation from plasma Isolation of urinary exosomes for RNA biomarker discovery using a simple, fast, and highly scalable method Methods and Compositions for Exosome Isolation Optimized exosome isolation protocol for cell culture supernatant and human plasma The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling Heparin affinity purification of extracellular vesicles Lectin-induced agglutination method of urinary exosomes isolation followed by mi-RNA analysis: Application for prostate cancer diagnostic Characterization of RNA from Exosomes and Other Extracellular Vesicles Isolated by a Novel Spin Column-Based Method Procédés d'isolement de Microvésicules. World Patent WO2014107571A1 Immuno-based detection of extracellular vesicles in urine as diagnostic marker for prostate cancer Cancer cell-derived microvesicles induce transformation by transferring tissue transglutaminase and fibronectin to recipient cells Magnetic bead-based isolation of exosomes Antibody-coupled monolithic silica microtips for highthroughput molecular profiling of circulating exosomes Droplet digital PCR improves urinary exosomal miRNA detection compared to real-time PCR Tumor DNA in cerebral spinal fluid reflects clinical course in a patient with melanoma leptomeningeal brain metastases Biomarkers in Cerebrospinal Fluid: Analysis of Cell-Free Circulating Mitochondrial DNA by Digital PCR Cell-Free DNA and CXCL10 Derived from Bronchoalveolar Lavage Predict Lung Transplant Survival Detection of cell-free DNA in bronchial lavage fluid supernatants of patients with lung cancer Quick assessment of cell-free DNA in seminal fluid and fragment size for early non-invasive prostate cancer diagnosis WHO Laboratory Manual for the Examination and Processing of Human Semen Semen miRNAs Contained in Exosomes as Non-Invasive Biomarkers for Prostate Cancer Diagnosis Exosomal microRNAs in seminal plasma are markers of the origin of azoospermia and can predict the presence of sperm in testicular tissue Cell-free DNA concentration in pleural fluid and serum: Quantitative approach and potential prognostic factor in patients with cancer and pleural effusions Liquid biopsy using the supernatant of a pleural effusion for EGFR genotyping in pulmonary adenocarcinoma patients: A comparison between cell-free DNA and extracellular vesicle-derived DNA Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver Pancreatic Juice Exosomal MicroRNAs as Biomarkers for Detection of Pancreatic Ductal Adenocarcinoma What information can be obtained from the tears of a patient with primary open angle glaucoma? Liquid biopsies come of age: Towards implementation of circulating tumour DNA Selection of the optimal manual method of cell free fetal DNA isolation from maternal plasma Towards standardisation of cell-free DNA measurement in plasma: Controls for extraction efficiency, fragment size bias and quantification Comprehensive evaluation of methods to isolate, quantify, and characterize circulating cell-free DNA from small volumes of plasma Driving Forces for DNA Adsorption to Silica in Perchlorate Solutions DNA, RNA, and protein extraction: The past and the present Influence of plasma processing on recovery and analysis of circulating nucleic acids A Comparison of Cell-Free DNA Isolation Kits: Isolation and Quantification of Cell-Free DNA in Plasma Evaluation of commercial kits for purification of circulating free DNA Comparison of Circulating Cell-Free DNA Extraction Methods for Downstream Analysis in Cancer Patients UltraPrep is a scalable, cost-effective, bead-based method for purifying cell-free DNA A novel miniaturized biofilter based on silicon micropillars for nucleic acid extraction Rapid microfluidic solid-phase extraction system for hyper-methylated DNA enrichment and epigenetic analysis Functional integration of DNA purification and concentration into a real time micro-PCR chip An integrated, self-contained microfluidic cassette for isolation, amplification, and detection of nucleic acids A portable, integrated analyzer for microfluidic-Based molecular analysis Continuous microfluidic DNA extraction using phase-transfer magnetophoresis Chitosan-coated silica as a solid phase for RNA purification in a microfluidic device Microchip-based solid-phase purification of RNA from biological samples Fully automated, on-site isolation of cfDNA from whole blood for cancer therapy monitoring Simple and Low-Cost Sampling of Cell-Free Nucleic Acids from Blood Plasma for Rapid and Sensitive Detection of Circulating Tumor DNA Centrifugation-free extraction of circulating nucleic acids using immiscible liquid under vacuum pressure Extraction of cell-free DNA from urine, using polylysine-coated silica particles Human urine contains small, 150 to 250 nucleotide-sized, soluble DNA derived from the circulation and may be useful in the detection of colorectal cancer Transrenal DNA as a diagnostic tool: Important technical notes Evaluation of six methods for extraction and purification of viral DNA from urine and serum samples Extraction of urinary cell-free DNA by using triamine-modified silica particles for liquid biopsy Single-stranded DNA library preparation uncovers the origin and diversity of ultrashort cell-free DNA in plasma Methods and Columns for Nucleic Acid Purification Differential susceptibility of PCR reactions to inhibitors: An important and unrecognised phenomenon Factors that influence quality and yield of circulating-free DNA: A systematic review of the methodology literature Effect of different DNA concentration methods on performance of non-invasive fetal y-chromosomal short tandem repeat profiling from maternal plasma Quantification of Cell-Free DNA: A Comparative Study of Three Different Methods Comparison of methods for the quantification of cell-free DNA isolated from cell culture supernatant The value of fluorimetry (Qubit) and spectrophotometry (NanoDrop) in the quantification of cell-free DNA (cfDNA) in malignant melanoma and prostate cancer patients Pitfalls of DNA Quantification Using DNA-Binding Fluorescent Dyes and Suggested Solutions Comparison of different DNA binding fluorescent dyes for applications of high-resolution melting analysis Quantification of massively parallel sequencing libraries-A comparative study of eight methods Clinical relevance of size selection of circulating DNA station?ID=e88b061b-37dc-4610-b4b7-0ef925f622f9#fragment-doc PCR experion automated electrophoresis system to detect Listeria monocytogenes in foods Automated Electrophoresis An overview of real-time quantitative PCR: Applications to quantify cytokine gene expression Considerations and quality controls when analyzing cell-free tumor DNA Plasma cell-free DNA in ovarian cancer: An independent prognostic biomarker Extracellular DNA as a Prognostic and Therapeutic Target in Mouse Colitis under DNase I Treatment Vanishing twin as a potential source of bias in non-invasive fetal sex determination: A case report A stabilizing reagent prevents cell-free DNA contamination by cellular DNA in plasma during blood sample storage and shipping as determined by digital PCR Evaluation of pre-analytical factors affecting plasma DNA analysis Evaluation of PCR-HRM, RFLP, and direct sequencing as simple and cost-effective methods to detect common EGFR mutations in plasma cell-free DNA of non-small cell lung cancer patients The future of DNA sequencing Noninvasive prenatal screening for fetal aneuploidy, 2016 update: A position statement of the American College of Medical Genetics and Genomics Global perspectives on clinical adoption of NIPT Noninvasive Prenatal Genetic Testing: Current and Emerging Ethical, Legal, and Social Issues An Overview of Liquid Biopsy for Screening and Early Detection of Cancer. In CADTH Issues in Emerging Health Technologies; Canadian Agency for Drugs and Technologies in Health Next-generation DNA sequencing methods A window into third-generation sequencing Plasma DNA tissue mapping by genome-wide methylation sequencing for noninvasive prenatal, cancer, and transplantation assessments Nanopore native RNA sequencing of a human poly(A) transcriptome Maternal plasma DNA sequencing reveals the genome-wide genetic and mutational profile of the fetus Non-invasive prenatal measurement of the fetal genome Non-invasive prenatal testing (NIPT) by low coverage genomic sequencing: Detection limits of screened chromosomal microdeletions Noninvasive Detection of Microsatellite Instability and High Tumor Mutation Burden in Cancer Patients Treated with PD-1 Blockade Result of Prospective Validation of the Trisomy Test for the Detection of Chromosomal Trisomies. Diagnostics Detection and quantification of rare mutations with massively parallel sequencing Transforming single DNA molecules into fluorescent magnetic particles for detection and enumeration of genetic variations Detection and quantification of mutations in the plasma of patients with colorectal tumors Genomic Copy Number Profiling Using Circulating Free Tumor DNA Highlights Heterogeneity in Neuroblastoma Next-generation sequencing of circulating tumor DNA for detection of gene mutations in lung cancer: Implications for precision treatment Comparison of DNA Quantification Methods for Next Generation Sequencing Fragmentation of DNA affects the accuracy of the DNA quantitation by the commonly used methods Systematic comparison of germline variant calling pipelines cross multiple next-generation sequencers Structural variant calling: The long and the short of it Hybrid assembly of ultra-long Nanopore reads augmented with 10x-Genomics contigs: Demonstrated with a human genome Evaluating Variant Calling Tools for Non-Matched Next-Generation Sequencing Data Summarize analysis results for multiple tools and samples in a single report Summarizing and correcting the GC content bias in high-throughput sequencing A Novel Statistical Method to Diagnose, Quantify and Correct Batch Effects in Genomic Studies Trimmomatic: A flexible trimmer for Illumina sequence data Cutadapt removes adapter sequences from high-throughput sequencing reads From trash to treasure: Detecting unexpected contamination in unmapped NGS data A Fast De Novo Duplicates Removal Tool for Paired Short Reads Role of DNA mismatch repair genes in lung and head and neck cancer (Review) COSMIC: The Catalogue Of Somatic Mutations In Cancer Genome-Wide Sequencing of Cell-Free DNA Identifies Copy-Number Alterations That Can Be Used for Monitoring Response to Immunotherapy in Cancer Patients Aberrantly Methylated cfDNA in Body Fluids as a Promising Diagnostic Tool for Early Detection of Breast Cancer Circulating tumor DNA as an early cancer detection tool From Somatic Variants Toward Precision Oncology: An Investigation of Reporting Practice for Next-Generation Sequencing-Based Circulating Tumor DNA Analysis Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples VarScan 2: Somatic mutation and copy number alteration discovery in cancer by exome sequencing VarDict: A novel and versatile variant caller for next-generation sequencing in cancer research Strelka2: Fast and accurate calling of germline and somatic variants Systematic comparison of somatic variant calling performance among different sequencing depth and mutation frequency Utilization of Benchtop Next Generation Sequencing Platforms Ion Torrent PGM and MiSeq in Noninvasive Prenatal Testing for Chromosome 21 Trisomy and Testing of Impact of In Silico and Physical Size Selection on Its Analytical Performance Combining count-and length-based z-scores leads to improved predictions in non-invasive prenatal testing Combination of Fetal Fraction Estimators Based on Fragment Lengths and Fragment Counts in Non-Invasive Prenatal Testing Size profile of cell-free DNA: A beacon guiding the practice and innovation of clinical testing High fragmentation characterizes tumour-derived circulating DNA Fast gapped-read alignment with Bowtie 2 Determination of fetal DNA fraction from the plasma of pregnant women using sequence read counts PREFACE: In silico pipeline for accurate cell-free fetal DNA fraction prediction Calculating the fetal fraction for noninvasive prenatal testing based on genome-wide nucleosome profiles Liquid biopsy for infectious diseases: Sequencing of cell-free plasma to detect pathogen DNA in patients with invasive fungal disease The authors declare no conflict of interest. The sponsors had no role in the design, execution, interpretation, or writing of the study.