Home Circulating cell-free DNA and its clinical utility in cancer
Article Open Access

Circulating cell-free DNA and its clinical utility in cancer

  • Amanda Salviano-Silva ORCID logo , Cecile L. Maire , Katrin Lamszus and Franz L. Ricklefs ORCID logo EMAIL logo
Published/Copyright: July 19, 2022
Become an author with De Gruyter Brill

Abstract

Liquid biopsies are a valuable non-invasive biomarker source for the diagnosis, prognosis and monitoring of cancer patients. The detection of circulating cell-free DNA (cfDNA) derived from tumor cells (ctDNA) has emerged as a promising clinical approach, as their levels are elevated in many cancers and contains tumor-related mutations and specific methylation patterns. ctDNA can be released from tumor cells into the bloodstream, either linked to extracellular vesicles (EV-DNA) or in an EV-free form when associated with nucleosomes and other proteins, or even as a component of macromolecular structures such as neutrophil extracellular traps (NET DNA). These different types of cfDNA can mirror cancer progression and predict patient outcome. This review presents the recent benefits of cfDNA in cancer, distinguishing between EV-DNA and EV-free DNA, and highlights their clinical utility.

Introduction

Finding minimally invasive techniques to detect, characterize and monitor cancer has long been a central goal in oncology research. In the past decade, there have been major breakthroughs in our ability to examine tumor-derived material in the circulation (blood/plasma) and other biofluids, including cerebrospinal fluid (CSF), saliva, and urine [1]. This has been possible due to the development of sensitive assays, capable of detecting the so-called needle in a haystack which refers to rare cancer-specific analytes hidden within the abundance of analytes of healthy cells. Current analytes used for liquid biopsy approaches include circulating tumor cells (CTCs), cell-free DNA (cfDNA), extracellular vesicles (EVs), tumor-educated platelets and other entities (i.e. proteins, mRNA, miRNAs, lncRNAs, metabolites, lipids). Each analyte has its advantages and disadvantages that must be considered when it is used to answer specific clinical questions. For example, cfDNA is an easily accessible and widely available analyte, yet its half-life is less than 1–2 h [2, 3], whereas EVs and CTCs can protect their cargo from degradation but require elaborated isolation techniques, resulting in relatively low quantities.

Most published studies used cfDNA as analyte for the detection of tumor-specific mutations via liquid biopsy [4], due to the relative easeness of DNA isolation and the availability of sequencing technologies to assess cancer-specific alterations. Circulating DNA is usually fragmented into ∼167 bp sequences, and samples from individuals with a high tumor burden show a trend toward even shorter sizes (<150 bp) [5]. It is assumed that the fragmented DNA suffices to detect tumor-specific mutations, however, the protocols that are used to isolate cfDNA from the blood of cancer patients usually do not involve an EV-removal step resulting in a mix of bona fide circulating DNA and EV-associated DNA [6]. While conflicting results regarding the proportion of EV-DNA within cfDNA have been reported (41–93%) [7, 8], EV-DNA has the advantage of being much larger (up to >10 kb) [9, 10]. It is thus unclear whether the circulating EV-DNA or fragmented cfDNA are the main source of the detected tumor-specific mutations and which analyte is better suited for identifying genome-wide mutations. Moreover, cfDNA sequencing can yield false positive results and detect mutations that are not present in the original tumor. Discordant results are sometimes due to clonal hematopoiesis, but also technical issues that compromise the specificity of mutation detection [4]. In this review, we summarize the main information contained in literature concerning circulating cfDNAs and their potential utility as tumor biomarkers in liquid biopsies.

Cell free DNA

Liquid biopsy from plasma, CSF or urine is a source of easily accessible cfDNA that due to advances in sequencing techniques is now used as biomarker in routine prenatal diagnostic screening [11], as well as in tumor classification, tumor heterogeneity assessment and therapy monitoring [12]. cfDNA combines genomic, mitochondrial and microbial DNA, double and single stranded degraded fragments from all kind of shredded or apoptotic cells. Despite the low amount and poor DNA quality, essential inputs can be inferred from bulk cfDNA and tumor-derived ctDNA to identify tumor specific genomic alterations and infer tumor subclasses, emergence of specific clones and treatment evolution. The confirmation that plasma ctDNA whole genome as well as whole exome sequencing is a valid method to identify unknown point mutations and copy number alterations from metastatic prostate and breast cancer [13] opened the door for ctDNA analysis as a diagnostic tool. Recently, WGS on ctDNA extracted from CSF of pediatric patients with medulloblastoma, was proven to be an essential asset to identify residual disease and for survival outcome prediction [14]. Targeted sequencing, an often more affordable approach, can also be used on plasma ctDNA to detect residual disease and patient outcome in gastric cancer [15]. In neurooncology, ctDNA analysis was until recently still extremely limited and ineffective due do the limiting amount of ctDNA from brain tumors that can be detected in the plasma and the low percentage of point mutations found in general in glioblastoma (GBM). However, the use of personalized sequencing panels designed from matched tumor combined with machine learning models was successful for glioma-ctDNA in plasma, urine, and CSF [16]. Whole genome bi-sulfite sequencing recently became a more promising technique to interrogate genome-wide methylation patterns as well as querying copy number variations and SNV indels. Such methylation profiling performed on ctDNA proved that methylation patterns are informative enough to detect over 50 different types of cancer in plasma [17]. Methylated DNA immunoprecipitation sequencing (MeDIP-seq), which provides a good coverage of the low CpG density genomic regions [18], has also been used successfully on ctDNA to identify differentially methylated regions which turn out to be sufficient to classify different types of brain tumors [19]. Genome wide DNA methylation profiling on ctDNA could also be achieved through CpG based arrays despite the plasma ctDNA degradation and can yield valuable information [20] as methylation profiling can be used to classify CNS tumors and assign them to specific subclasses with different susceptibility to treatment [21]. Additionally, specific mutations in the TERT promoter (C228T, C250T) have been recently identified to be present in more than 70% of GBM which opened the door for the use of digital droplet PCR to detect TERT mutations in GBM plasma ctDNA [22].

Whole genome sequencing and methylation profiling of cell free DNA led to new opportunities in early diagnosis as well as therapy monitoring and patient survival prediction, however, it is still unclear if the cfDNA used in most of these publications is linked with EVs or organized into DNA macromolecular complexes (Figure 1).

Figure 1: 
Main cfDNA types.
Figure 1:

Main cfDNA types.

Tumor cfDNA can reach the circulation being carried by extracellular vesicles (EV-DNA) (middle) or in an EV-free way, either by association with nucleosomes, as consequence of cell apoptosis (left), or as macromolecular complexes released from neutrophils (NET-DNA) (right).

DNA associated with extracellular vesicles (EV-DNA)

Extracellular vesicles (EVs) play an important role in intercellular communication between different tumor cells and also between tumor and normal somatic cells [23, 24]. EVs comprise a heterogenous population, with differences in biogenesis, content, and size. These parameters allow a differentiation of EVs in either small-EVs (S-EVs–including exosomes and small microvesicles) and large-EVs (L-EVs–including oncosomes and apoptotic bodies). S-EVs are ∼100 nm membrane vesicles, generally released into the extracellular space by multivesicular bodies that fuse with the plasma membrane. In contrast, L-EVs are usually bigger and detach from the plasma membrane, carrying unique surface markers. Both EV entities carry complex biological information, consisting of soluble and transmembrane proteins, RNAs and miRNAs, DNA, and lipids. EVs are taken up by recipient cells and release their bioactive contents, which can significantly alter the phenotype of the recipient cell [24]. Moreover, EVs are released from tumor cells into the bloodstream, and their cargo are protected from fragmentation and degradation by the membranous envelope, making circulating tumor EVs an optimal biomarker source for obtaining diagnostic information about the tumor and monitoring therapeutic progress by “liquid biopsy” [24].

Despite intensive research on EVs as potential sources of biomarkers and their role in physiological functions, the associated DNA remains largely unexplored. Most studies have so far focused almost exclusively on EV proteins, lipids and RNA cargo, and their composition. That EVs possess DNA is now widely accepted, but it remains uncertain where this DNA comes from, where it is located in relation to EVs, and what characteristics it possesses.

L-EVs detach from the plasma membrane and their seemingly non-specific loading mechanism can explain that their DNA content is reflective of the cell of origin seen at least evident for apoptotic bodies [25]. S-EVs, on the other hand, use more complex loading mechanisms using the ESCRT complex. However, it is not clear whether DNA can also be loaded into S-EVs via this loading mechanism. On the other hand, it has been shown that micronuclei, small constrictions of the nucleus as a result of DNA damage, can release DNA into the cytosol which then colocalizes with tetraspanin markers, which are established EV markers [26, 27]. These results support the early assumption that EVs are a waste removal system of cells to purify cytosolic DNA and maintain cellular homeostasis to avoid cellular senescence and apoptosis [28, 29].

Another topic that continues to be much debated is the localization of DNA in EVs. Most studies looking at L-EVs with or without comparison to S-EVs proved the presence of large gDNA (>2 million bp) in their vesicular lumen [30], [31], [32]. DNA associated with small EVs seems to be predominantly located to the outer membrane, yet a small proportion remains in the intraluminal space. Notably, although multiple groups provided strong evidence of DNA in EVs [9, 10, 28, 30, 32], [33], [34], [35], [36], a recent report questioned these findings and suggested that DNA is mainly associated with non-vesicular components [37].

The true localization of the DNA in EVs is clearly complex, and no consensus has yet been found. These differences are a result of the diversity of EV isolation and DNA characterization methods as well as the heterogeneity of EV DNA itself [38], [39], [40], [41]. Therefore, a consensus on EV DNA isolation and characterization methods must first be reached before an understanding of the true nature of EV-DNA localization can be obtained. In GBM patients’ tumor-specific DNA, especially mutant copies of IDH gene have been detected in EVs from glioma cell cultures or liquid biopsies [42], [43], [44], [45], [46], [47], [48]. While these results highlight the diagnostic potential of EVs, the significance of mutations in a few selected genes affected by recurrent hotspot mutations such as IDH1 or EGFR is limited to only a subset of patients with these alterations. More comprehensive profiling is needed to classify tumors with unknown genetic alterations and to monitor changes in genetic or epigenetic tumor makeup during the course of treatment and disease progression. Studies in other cancers have shown that high-molecular-weight double-stranded DNA from all chromosomes is present in EVs and can reflect the genome-wide mutational status of parental tumor cells [9, 10, 28, 30, 32]. Additionally, mutant KRAS alleles were found in late stages of pancreatic cancer progression to be associated not only to EVs, but also to histones in the circulation [49], suggesting both EV- and EV-free associated DNA as interesting sources for cancer biomarkers. The investigation of EV-DNA as a tumor biomarker and their role in therapeutic approaches has been extensively reviewed in the literature [50], [51], [52].

EV-free cfDNA (EF-DNA)

When not associated to EVs, cfDNA can be associated to many soluble proteins, including immunoglobulins [53], albumin [54], histones/nucleosomes [55] and lipoprotein-RNA complexes [56]. Such DNA/protein bindings are formed by electrostatic interactions and protect the extracellular DNA from degradation by nucleases.

Nucleosomes are stable complexes consisting of double stranded DNA arranged around a histone octamer. These nuclear structures can be found in the bloodstream as a result of cell apoptosis, carrying fragments of DNA with a characteristic size of approximately 150–200 bp, [5557]. The expression and structure of extracellular nucleosome DNA correlates with the cell of origin [57], is altered in various cancers [58], [59], [60] and carries tumor-derived mutations [49]. In addition, ctDNA is smaller and more fragmented than DNA from healthy cells [16, 57, 60], [61], [62].

CfDNA associated with nucleosomes can also exist in macromolecular structures called extracellular traps (ETs) [63]. The main types of ETs studied are neutrophil extracellular traps (NETs), which are released by activated neutrophils as a result of cell death (NETosis) or DNA extrusion without cell lysis. These structures are large fibers of nuclear or mitochondrial DNA, combined with histones and cytotoxic enzymes, such as myeloperoxidase (MPO) and neutrophil elastase (NE) [63]. NETs are mainly involved in pathogen recognition and inflammation initiation, and under pathological conditions they account for a significant proportion of EV-free cfDNA, as their increased production can be harmful to tissues [64], [65], [66]. Considering that inflammation is a hallmark of cancer and neutrophil infiltration is a common finding in tumor microenvironment, NETs have been increasingly investigated in the crosstalk between neutrophils and tumor cells [65, 67]. NETs are induced by several cancers, in response to reactive oxygen species (ROS) production, hypoxic conditions, proteases, interleukins, cytokines, and tumor-derived exosomes [68], supporting tumor growth and progression by different mechanisms. Citrullinated histone 3 (H3cit), MPO and NE, which are the main NET markers, contribute to platelets activation and cancer-associated thrombosis [69], [70], [71], [72], [73], [74], [75]. NETs are also involved in ECM disruption and cancer metastasis [68], as well as in exacerbated inflammation in advanced cancers [67].

Lung cancer studies have shown that NET DNA can trap CTCs, promoting their adhesion in distant organs and leading to metastasis. This metastatic adhesion has been reversed by NET disruption in vitro and in vivo [76, 77], and is mediated in part by Integrin ß1 (78,79). Furthermore, integrin α3β1 signaling pathway can be activated by NET-induced proteolytic laminin remodeling during lung inflammation, leading to the awakening of dormant cancer cells and aggressive metastasis [78]. In sharp contrast, an antitumor effect of NETs has been observed in melanoma cells, where integrin-mediated NET adhesion can inhibit tumor cell migration [79]. Conflicting data have also been observed in ovarian cancer, where a higher proporation of NETs may be associated with a favorable outcome [80] or a deleterious effect promoting metastasis [81]. NETs production is stimulated by breast cancer cells and promotes cancer proliferation and dissemination [82], [83], [84]. NETs are also increased in different digestive malignancies [68], contributing to progression, hypercoagulation and metastasis in gastric [85, 86], pancreatic [87] and colorectal cancers [88], [89], [90].

High levels of free fatty acids stimulate NET formation in nonalcoholic steatohepatitis patients, increasing inflammation, leading to the progression to hepatocellular carcinoma (HCC) [91]. Increased NET levels are also observed in HCC patients to promote metastasis [92, 93], also by inducing inflammatory response [94], or even as a consequence of surgical stress [95]. Moreover, NETs are also elevated and associated with thrombosis in non-solid tumors, such as in myeloproliferative neoplasms (MPN) [96] and acute promyelocytic leukemia (APL) [97].

Furthermore, in a genome wide association study (GWAS) performed with chronic diseases patients, the plasma levels of NETs were associated with the presence of genetic variants located in genes predicted to be involved in cancer pathways, such as OR10H1 and KHDRBS1 [98]. This suggests the possibility of genetic markers for NET levels with prognostic value in cancer, although no studies were performed in this specific field until the moment.

New diagnostic approaches with circulating cell free DNA

The detection of ctDNA in liquid biopsies is a non-invasive method which allows a real-time monitoring of cancer patients, being attractive for diagnostic and prognostic purposes. Therefore, many efforts have been made in the last decades to integrate the use of cfDNA and other circulating biomolecules into clinical routine. In this context, numerous studies analyze cfDNA associated with EVs, as the nucleic acids are protected and less fragmented when carried by these nanoparticles, and therefore highly representative of their original cells. Therefore, it is not surprising that the number of studies with EV DNA is increased in different types of cancer [50], [51], [52], compared to NET DNA.

Although most studies using NETs have been conducted in inflammatory and autoimmune disorders, interest in these macromolecular DNA structures has increased in cancer studies due to their influence on the tumor microenvironment and their potential utility as clinical markers. As aforementioned, NET DNA is elevated in several cancers and has been associated with poor prognosis, whereas NET disruption with DNase I has been associated with better outcomes [67, 71]. Interestingly, the NET degradation with DNase I has been shown in vivo to decrease the anti-PD-1 blockade resistance and attenuate the tumor growth. In this context, DNase I administration is suggested to be combined with immune checkpoint inhibitors for colorectal cancer patients [99, 100]. Other proposed NET inhibitors include prostaglandin 2 [101, 102] and inhibitors of peptidylarginine deiminase [103].

Clinical trials are currently being conducted with cfDNA in cancer patients [104]. Among them, RB1 mutations carried by EVs are investigated in childhood cancer retinoblastoma (ClinicalTrials.gov identifier: NCT04164134), while EVs carrying T790M derived from non-small cell lung cancer cells are being used to evaluate the efficacy of a potential antitumor treatment (NCT03228277). EV-DNA is also currently researched in early diagnosis of lung (NCT04529915) and thyroid cancers (NCT04742608). NETs have also been investigated in clinical trials, such as for detection of occult cancers in patients with venous thromboembolism (NCT03781531), and also for the prognosis of patients with high risk for thromboembolism, as the case of hepatocellular carcinoma (NCT05040347), myeloproliferative neoplasms (NCT04177576), and pancreatic, gastric and colon cancers (NCT04294589). In breast cancer, clinical trials are currently being conducted to evaluate NETs side effects as response to Tamoxifen administration (NCT05056857). Further clinical trials have been conducted, although there is still a long way to go to incorporate EV- and NET-DNA samples into routine clinical practice.

Conclusions

Detection of ctDNA in liquid biopsies is a promising tool for monitoring patients and predicting tumor status, in real time and in a non-invasive way. EV-DNA, NET-DNA and other types of cfDNA have been increasingly studied and some of them are already presented in the literature as potential clinical biomarkers for tumor progression and prognosis. However, there are still many challenges regarding their low sensitivity in detection. In addition, some important questions regarding cfDNA remain to be addressed. For example, the proportion of ctDNA in total cfDNA is still unclear, and it is largely unknown how this proportion changes during treatment. There is therefore a great need to characterize different DNA analytes and directly compare their suitability for detecting tumor-specific mutations in cancer patients.


Corresponding author: Franz L. Ricklefs, Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany, Phone: +49-40-7410-0, fax: +49-40-7410-55590, E-mail:

  1. Research funding: None declared.

  2. Author contributions: All authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  3. Competing interests: Authors state no conflict of interest.

  4. Informed consent: Not applicable.

  5. Ethical approval: Not applicable.

References

1. Pantel, K, Alix-Panabières, C. Liquid biopsy and minimal residual disease — latest advances and implications for cure. Nat Rev Clin Oncol 2019;16:409–24. https://doi.org/10.1038/s41571-019-0187-3.Search in Google Scholar PubMed

2. Muhanna, N, di Grappa, MA, Chan, HHL, Khan, T, Jin, CS, Zheng, Y, et al.. Cell-free DNA kinetics in a pre-clinical model of head and neck cancer. Sci Rep 2017;7:16723. https://doi.org/10.1038/s41598-017-17079-6.Search in Google Scholar PubMed PubMed Central

3. Thierry, AR, Mouliere, F, Gongora, C, Ollier, J, Robert, B, Ychou, M, et al.. Origin and quantification of circulating DNA in mice with human colorectal cancer xenografts. Nucleic Acids Res 2010;38:6159–75. https://doi.org/10.1093/nar/gkq421.Search in Google Scholar PubMed PubMed Central

4. Keller, L, Belloum, Y, Wikman, H, Pantel, K. Clinical relevance of blood-based ctDNA analysis: mutation detection and beyond. Br J Cancer 2021;124:345–58. https://doi.org/10.1038/s41416-020-01047-5.Search in Google Scholar PubMed PubMed Central

5. Mouliere, F, Chandrananda, D, Piskorz, AM, Moore, EK, Morris, J, Ahlborn, LB, et al.. Enhanced detection of circulating tumor DNA by fragment size analysis. Sci Transl Med 2018;10:1–13. https://doi.org/10.1126/scitranslmed.aat4921.Search in Google Scholar PubMed PubMed Central

6. García-Silva, S, Gallardo, M, Peinado, H. DNA-loaded extracellular vesicles in liquid biopsy: tiny players with big potential? Front Cell Dev Biol 2020;8:622579.10.3389/fcell.2020.622579Search in Google Scholar PubMed PubMed Central

7. Fernando, MR, Jiang, C, Krzyzanowski, GD, Ryan, WL. New evidence that a large proportion of human blood plasma cell-free DNA is localized in exosomes. PLoS One 2017;12:e0183915. https://doi.org/10.1371/journal.pone.0183915.Search in Google Scholar PubMed PubMed Central

8. Wan, Y, Liu, B, Lei, H, Zhang, B, Wang, Y, Huang, H, et al.. Nanoscale extracellular vesicle-derived DNA is superior to circulating cell-free DNA for mutation detection in early-stage non-small-cell lung cancer. Ann Oncol 2018;29:2379–83. https://doi.org/10.1093/annonc/mdy458.Search in Google Scholar PubMed PubMed Central

9. Kahlert, C, Melo, SA, Protopopov, A, Tang, J, Seth, S, Koch, M, et al.. Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J Biol Chem 2014;289:3869–75. https://doi.org/10.1074/jbc.c113.532267.Search in Google Scholar PubMed PubMed Central

10. Thakur, BK, Zhang, H, Becker, A, Matei, I, Huang, Y, Costa-Silva, B, et al.. Double-stranded DNA in exosomes: a novel biomarker in cancer detection. Cell Res 2014;24:766–9. https://doi.org/10.1038/cr.2014.44.Search in Google Scholar PubMed PubMed Central

11. Pös, O, Budiš, J, Szemes, T. Recent trends in prenatal genetic screening and testing. F1000Res 2019;8:764.10.12688/f1000research.16837.1Search in Google Scholar PubMed PubMed Central

12. Szilágyi, M, Pös, O, Éva, M, Buglyó, G, Soltész, B, Keserű, J, et al.. Circulating cell-free nucleic acids: main characteristics and clinical application. Int J Mol Sci 2020;21:6827.10.3390/ijms21186827Search in Google Scholar PubMed PubMed Central

13. Adalsteinsson, VA, Ha, G, Freeman, SS, Choudhury, AD, Stover, DG, Parsons, HA, et al.. Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat Commun 2017;8:1324. https://doi.org/10.1038/s41467-017-00965-y.Search in Google Scholar PubMed PubMed Central

14. Liu, APY, Smith, KS, Kumar, R, Paul, L, Bihannic, L, Lin, T, et al.. Serial assessment of measurable residual disease in medulloblastoma liquid biopsies. Cancer Cell 2021;39:1519–30.e4. https://doi.org/10.1016/j.ccell.2021.09.012.Search in Google Scholar PubMed

15. Leal, A, van Grieken, NCT, Palsgrove, DN, Phallen, J, Medina, JE, Hruban, C, et al.. White blood cell and cell-free DNA analyses for detection of residual disease in gastric cancer. Nat Commun 2020;11:525. https://doi.org/10.1038/s41467-020-14310-3.Search in Google Scholar PubMed PubMed Central

16. Mouliere, F, Smith, CG, Heider, K, Su, J, van der Pol, Y, Thompson, M, et al.. Fragmentation patterns and personalized sequencing of cell-free DNA in urine and plasma of glioma patients. EMBO Mol Med 2021;13:e12881. https://doi.org/10.15252/emmm.202012881.Search in Google Scholar PubMed PubMed Central

17. Liu, MC, Oxnard, GR, Klein, EA, Swanton, C, Seiden, Mv, CCGA Consortium. Sensitive and specific multi-cancer detection and localization using methylation signatures in cell-free DNA. Ann Oncol 2020;31:745–59. https://doi.org/10.1016/j.annonc.2020.02.011.Search in Google Scholar PubMed PubMed Central

18. Beck, D, Ben Maamar, M, Skinner, MK. Genome-wide CpG density and DNA methylation analysis method (MeDIP, RRBS, and WGBS) comparisons. Epigenetics 2021;17:518–30. https://doi.org/10.1080/15592294.2021.1924970.Search in Google Scholar PubMed PubMed Central

19. Nassiri, F, Chakravarthy, A, Feng, S, Shen, SY, Nejad, R, Zuccato, JA, et al.. Detection and discrimination of intracranial tumors using plasma cell-free DNA methylomes. Nat Med 2020;26:1044–7. https://doi.org/10.1038/s41591-020-0932-2.Search in Google Scholar PubMed PubMed Central

20. Sabedot, TS, Malta, TM, Snyder, J, Nelson, K, Wells, M, deCarvalho, AC, et al.. A serum-based DNA methylation assay provides accurate detection of glioma. Neuro Oncol 2021;23:1494–508. https://doi.org/10.1093/neuonc/noab023.Search in Google Scholar PubMed PubMed Central

21. Capper, D, Jones, DTW, Sill, M, Hovestadt, V, Schrimpf, D, Sturm, D, et al.. DNA methylation-based classification of central nervous system tumours. Nature 2018;555:469–74. https://doi.org/10.1038/nature26000.Search in Google Scholar PubMed PubMed Central

22. Muralidharan, K, Yekula, A, Small, JL, Rosh, ZS, Kang, KM, Wang, L, et al.. TERT promoter mutation analysis for blood-based diagnosis and monitoring of gliomas. Clin Cancer Res 2021;27:169–78. https://doi.org/10.1158/1078-0432.ccr-20-3083.Search in Google Scholar

23. el Andaloussi, S, Mäger, I, Breakefield, XO, Wood, MJA. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov 2013;12:347–57. https://doi.org/10.1038/nrd3978.Search in Google Scholar PubMed

24. Westphal, M, Lamszus, K. Circulating biomarkers for gliomas. Nat Rev Neurol 2015;11:556–66. https://doi.org/10.1038/nrneurol.2015.171.Search in Google Scholar PubMed

25. Pisetsky, DS, Gauley, J, Ullal, AJ. Microparticles as a source of extracellular DNA. Immunol Res 2011;49:227–34. https://doi.org/10.1007/s12026-010-8184-8.Search in Google Scholar PubMed PubMed Central

26. Harding, SM, Benci, JL, Irianto, J, Discher, DE, Minn, AJ, Greenberg, RA. Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature 2017;548:466–70. https://doi.org/10.1038/nature23470.Search in Google Scholar PubMed PubMed Central

27. Bakhoum, SF, Ngo, B, Laughney, AM, Cavallo, J-A, Murphy, CJ, Ly, P, et al.. Chromosomal instability drives metastasis through a cytosolic DNA response. Nature 2018;553:467–72. https://doi.org/10.1038/nature25432.Search in Google Scholar PubMed PubMed Central

28. Takahashi, A, Okada, R, Nagao, K, Kawamata, Y, Hanyu, A, Yoshimoto, S, et al.. Exosomes maintain cellular homeostasis by excreting harmful DNA from cells. Nat Commun 2017;8:15287. https://doi.org/10.1038/ncomms15287.Search in Google Scholar PubMed PubMed Central

29. Malkin, EZ, Bratman, Sv. Bioactive DNA from extracellular vesicles and particles. Cell Death Dis 2020;11:584. https://doi.org/10.1038/s41419-020-02803-4.Search in Google Scholar PubMed PubMed Central

30. Vagner, T, Spinelli, C, Minciacchi, VR, Balaj, L, Zandian, M, Conley, A, et al.. Large extracellular vesicles carry most of the tumour DNA circulating in prostate cancer patient plasma. J Extracell Vesicles 2018;7:1505403. https://doi.org/10.1080/20013078.2018.1505403.Search in Google Scholar PubMed PubMed Central

31. Lázaro‐Ibáñez, E, Sanz‐Garcia, A, Visakorpi, T, Escobedo‐Lucea, C, Siljander, P, Ayuso‐Sacido, Á, et al.. Different gDNA content in the subpopulations of prostate cancer extracellular vesicles: apoptotic bodies, microvesicles, and exosomes. Prostate 2014;74:1379–90.10.1002/pros.22853Search in Google Scholar PubMed PubMed Central

32. Balaj, L, Lessard, R, Dai, L, Cho, Y-J, Pomeroy, SL, Breakefield, XO, et al.. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat Commun 2011;2:180. https://doi.org/10.1038/ncomms1180.Search in Google Scholar PubMed PubMed Central

33. Lázaro-Ibáñez, E, Lässer, C, Shelke, GV, Crescitelli, R, Jang, SC, Cvjetkovic, A, et al.. DNA analysis of low- and high-density fractions defines heterogeneous subpopulations of small extracellular vesicles based on their DNA cargo and topology. J Extracell Vesicles 2019;8:1656993.10.1080/20013078.2019.1656993Search in Google Scholar PubMed PubMed Central

34. Fischer, S, Cornils, K, Speiseder, T, Badbaran, A, Reimer, R, Indenbirken, D, et al.. Indication of horizontal DNA gene transfer by extracellular vesicles. PLoS One 2016;11:e0163665. https://doi.org/10.1371/journal.pone.0163665.Search in Google Scholar PubMed PubMed Central

35. Lee, TH, Chennakrishnaiah, S, Audemard, E, Montermini, L, Meehan, B, Rak, J. Oncogenic ras-driven cancer cell vesiculation leads to emission of double-stranded DNA capable of interacting with target cells. Biochem Biophys Res Commun 2014;451:295–301. https://doi.org/10.1016/j.bbrc.2014.07.109.Search in Google Scholar PubMed

36. Yokoi, A, Villar-Prados, A, Oliphint, PA, Zhang, J, Song, X, de Hoff, P, et al.. Mechanisms of nuclear content loading to exosomes. Sci Adv 2019;5:1–16. https://doi.org/10.1126/sciadv.aax8849.Search in Google Scholar PubMed PubMed Central

37. Jeppesen, DK, Fenix, AM, Franklin, JL, Higginbotham, JN, Zhang, Q, Zimmerman, LJ, et al.. Reassessment of exosome composition. Cell 2019;177:428–45.e18. https://doi.org/10.1016/j.cell.2019.02.029.Search in Google Scholar PubMed PubMed Central

38. Willms, E, Cabañas, C, Mäger, I, Wood, MJA, Vader, P. Extracellular vesicle heterogeneity: subpopulations, isolation techniques, and diverse functions in cancer progression. Front Immunol 2018;30:9. https://doi.org/10.3389/fimmu.2018.00738.Search in Google Scholar PubMed PubMed Central

39. Théry, C, Witwer, KW, Aikawa, E, Alcaraz, MJ, Anderson, JD, Andriantsitohaina, R, et al.. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the international society for extracellular vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 2018;7:1535750. https://doi.org/10.1080/20013078.2018.1535750.Search in Google Scholar PubMed PubMed Central

40. Witwer, KW, Théry, C. Extracellular vesicles or exosomes? On primacy, precision, and popularity influencing a choice of nomenclature. J Extracell Vesicles 2019;8:1648167. https://doi.org/10.1080/20013078.2019.1648167.Search in Google Scholar PubMed PubMed Central

41. Lucchetti, D, Fattorossi, A, Sgambato, A. Extracellular vesicles in oncology: progress and pitfalls in the methods of isolation and analysis. Biotechnol J 2019;14:1700716. https://doi.org/10.1002/biot.201700716.Search in Google Scholar PubMed

42. Chen, WW, Balaj, L, Liau, LM, Samuels, ML, Kotsopoulos, SK, Maguire, CA, et al.. BEAMing and droplet digital PCR analysis of mutant IDH1 mRNA in glioma patient serum and cerebrospinal fluid extracellular vesicles. Mol Ther Nucleic Acids 2013;2:e109. https://doi.org/10.1038/mtna.2013.28.Search in Google Scholar PubMed PubMed Central

43. Figueroa, JM, Skog, J, Akers, J, Li, H, Komotar, R, Jensen, R, et al.. Detection of wild-type EGFR amplification and EGFRvIII mutation in CSF-derived extracellular vesicles of glioblastoma patients. Neuro Oncol 2017;19:1494–502. https://doi.org/10.1093/neuonc/nox085.Search in Google Scholar PubMed PubMed Central

44. García-Romero, N, Carrión-Navarro, J, Esteban-Rubio, S, Lázaro-Ibáñez, E, Peris-Celda, M, Alonso, MM, et al.. DNA sequences within glioma-derived extracellular vesicles can cross the intact blood-brain barrier and be detected in peripheral blood of patients. Oncotarget 2017;8:1416–28.10.18632/oncotarget.13635Search in Google Scholar PubMed PubMed Central

45. Graner, MW, Alzate, O, Dechkovskaia, AM, Keene, JD, Sampson, JH, Mitchell, DA, et al.. Proteomic and immunologic analyses of brain tumor exosomes. FASEB J 2009;23:1541–57. https://doi.org/10.1096/fj.08-122184.Search in Google Scholar PubMed PubMed Central

46. Lee, K, Fraser, K, Ghaddar, B, Yang, K, Kim, E, Balaj, L, et al.. Multiplexed profiling of single extracellular vesicles. ACS Nano 2018;12:494–503. https://doi.org/10.1021/acsnano.7b07060.Search in Google Scholar PubMed PubMed Central

47. Skog, J, Würdinger, T, van Rijn, S, Meijer, DH, Gainche, L, Curry, WT, et al.. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol 2008;10:1470–6. https://doi.org/10.1038/ncb1800.Search in Google Scholar PubMed PubMed Central

48. Maire, CL, Fuh, MM, Kaulich, K, Fita, KD, Stevic, I, Heiland, DH, et al.. Genome-wide methylation profiling of glioblastoma cell-derived extracellular vesicle DNA allows tumor classification. Neuro Oncol 2021;23:1087–99. https://doi.org/10.1093/neuonc/noab012.Search in Google Scholar PubMed PubMed Central

49. Hagey, DW, Kordes, M, Görgens, A, Mowoe, MO, Nordin, JZ, Moro, CF, et al.. Extracellular vesicles are the primary source of blood-borne tumour-derived mutant KRAS DNA early in pancreatic cancer. J Extracell Vesicles 2021;10:1–10. https://doi.org/10.1002/jev2.12142.Search in Google Scholar PubMed PubMed Central

50. Elzanowska, J, Semira, C, Costa‐Silva, B. DNA in extracellular vesicles: biological and clinical aspects. Mol Oncol 2021;15:1701–14. https://doi.org/10.1002/1878-0261.12777.Search in Google Scholar PubMed PubMed Central

51. Hur, JY, Lee, KY. Characteristics and clinical application of extracellular vesicle-derived DNA. Cancers (Basel) 2021;13:3827. https://doi.org/10.3390/cancers13153827.Search in Google Scholar

52. Abhange, K, Makler, A, Wen, Y, Ramnauth, N, Mao, W, Asghar, W, et al.. Small extracellular vesicles in cancer. Bioact Mater 2021;6:3705–43. https://doi.org/10.1016/j.bioactmat.2021.03.015.Search in Google Scholar

53. EYu, R, Pautova, LV, Yakubov, LA, Karamyshev, VN, Vlassov, VV. Serum immunoglobulins interact with oligonucleotides. FEBS (Fed Eur Biochem Soc) Lett 1994;344:96–8. https://doi.org/10.1016/0014-5793(94)00360-2.Search in Google Scholar

54. Chelobanov, BP, Laktionov, PP, Vlasov, Vv. Proteins involved in binding and cellular uptake of nucleic acids. Biochemistry (Moscow) 2006;71:583–96. https://doi.org/10.1134/s0006297906060010.Search in Google Scholar PubMed

55. Rumore, PM, Steinman, CR. Endogenous circulating DNA in systemic lupus erythematosus. Occurrence as multimeric complexes bound to histone. J Clin Invest 1990;86:69–74. https://doi.org/10.1172/jci114716.Search in Google Scholar

56. Gahan, PB, Stroun, M. The virtosome-a novel cytosolic informative entity and intercellular messenger. Cell Biochem Funct 2010;28:529–38. https://doi.org/10.1002/cbf.1690.Search in Google Scholar PubMed

57. Snyder, MW, Kircher, M, Hill, AJ, Daza, RM, Shendure, J. Cell-free DNA comprises an in vivo nucleosome footprint that informs its tissues-of-origin. Cell 2016;164:57–68. https://doi.org/10.1016/j.cell.2015.11.050.Search in Google Scholar PubMed PubMed Central

58. McAnena, P, Brown, J, Kerin, M. Circulating nucleosomes and nucleosome modifications as biomarkers in cancer. Cancers (Basel) 2017;9:5. https://doi.org/10.3390/cancers9010005.Search in Google Scholar PubMed PubMed Central

59. Ulz, P, Thallinger, GG, Auer, M, Graf, R, Kashofer, K, Jahn, SW, et al.. Inferring expressed genes by whole-genome sequencing of plasma DNA. Nat Genet 2016;48:1273–8. https://doi.org/10.1038/ng.3648.Search in Google Scholar PubMed

60. Cristiano, S, Leal, A, Phallen, J, Fiksel, J, Adleff, V, Bruhm, DC, et al.. Genome-wide cell-free DNA fragmentation in patients with cancer. Nature 2019;570:385–9. https://doi.org/10.1038/s41586-019-1272-6.Search in Google Scholar PubMed PubMed Central

61. Mouliere, F, Robert, B, Arnau Peyrotte, E, del Rio, M, Ychou, M, Molina, F, et al.. High fragmentation characterizes tumour-derived circulating DNA. PLoS One 2011;6:e23418. https://doi.org/10.1371/journal.pone.0023418.Search in Google Scholar PubMed PubMed Central

62. Mathios, D, Johansen, JS, Cristiano, S, Medina, JE, Phallen, J, Larsen, KR, et al.. Detection and characterization of lung cancer using cell-free DNA fragmentomes. Nat Commun 2021;12:5060. https://doi.org/10.1038/s41467-021-24994-w.Search in Google Scholar PubMed PubMed Central

63. Conceição-Silva, F, Reis, CSM, de Luca, PM, Leite-Silva, J, Santiago, MA, Morrot, A, et al.. The immune system throws its traps: cells and their extracellular traps in disease and protection. Cells 2021;10:1891. https://doi.org/10.3390/cells10081891.Search in Google Scholar PubMed PubMed Central

64. Jorch, SK, Kubes, P. An emerging role for neutrophil extracellular traps in noninfectious disease. Nat Med 2017;23:279–87. https://doi.org/10.1038/nm.4294.Search in Google Scholar PubMed

65. Liu, Y, Liu, L. The pro-tumor effect and the anti-tumor effect of neutrophils extracellular traps. Biosci Trends 2020;13:469–75. https://doi.org/10.1201/9781351187435-57.Search in Google Scholar

66. Tsourouktsoglou, T-D, Warnatsch, A, Ioannou, M, Hoving, D, Wang, Q, Papayannopoulos, V. Histones, DNA, and citrullination promote neutrophil extracellular trap inflammation by regulating the localization and activation of TLR4. Cell Rep 2020;31:107602. https://doi.org/10.1016/j.celrep.2020.107602.Search in Google Scholar PubMed

67. Thålin, C, Lundström, S, Seignez, C, Daleskog, M, Lundström, A, Henriksson, P, et al.. Citrullinated histone H3 as a novel prognostic blood marker in patients with advanced cancer. PLoS One 2018;13:e0191231.10.1371/journal.pone.0191231Search in Google Scholar PubMed PubMed Central

68. Chen, Y, Han, L, Qiu, X, Wang, G, Zheng, J. Neutrophil extracellular traps in digestive cancers: warrior or accomplice. Front Oncol 2021;11:1–17. https://doi.org/10.3389/fonc.2021.766636.Search in Google Scholar PubMed PubMed Central

69. Demers, M, Krause, DS, Schatzberg, D, Martinod, K, Voorhees, JR, Fuchs, TA, et al.. Cancers predispose neutrophils to release extracellular DNA traps that contribute to cancer-associated thrombosis. Proc Natl Acad Sci USA 2012;109:13076–81. https://doi.org/10.1073/pnas.1200419109.Search in Google Scholar PubMed PubMed Central

70. Cedervall, J, Hamidi, A, Platelets, OA-K. NETs and cancer. Thromb Res 2018;164:S148–52. https://doi.org/10.1016/j.thromres.2018.01.049.Search in Google Scholar PubMed

71. Grilz, E, Mauracher, L, Posch, F, Königsbrügge, O, Zöchbauer‐Müller, S, Marosi, C, et al.. Citrullinated histone H3, a biomarker for neutrophil extracellular trap formation, predicts the risk of mortality in patients with cancer. Br J Haematol 2019;186:311–20. https://doi.org/10.1111/bjh.15906.Search in Google Scholar PubMed PubMed Central

72. Jung, HS, Gu, J, Kim, J-E, Nam, Y, Song, JW, Kim, HK. Cancer cell-induced neutrophil extracellular traps promote both hypercoagulability and cancer progression. PLoS One 2019;14:e0216055. https://doi.org/10.1371/journal.pone.0216055.Search in Google Scholar PubMed PubMed Central

73. Hisada, Y, Grover, SP, Maqsood, A, Houston, R, Ay, C, Noubouossie, DF, et al.. Neutrophils and neutrophil extracellular traps enhance venous thrombosis in mice bearing human pancreatic tumors. Haematologica 2020;105:218–25. https://doi.org/10.3324/haematol.2019.217083.Search in Google Scholar PubMed PubMed Central

74. Efrimescu, CI, Buggy, PM, Buggy, DJ. Neutrophil extracellular trapping role in cancer, metastases, and cancer-related thrombosis: a narrative review of the current evidence base. Curr Oncol Rep 2021;23:118. https://doi.org/10.1007/s11912-021-01103-0.Search in Google Scholar PubMed PubMed Central

75. Zhang, S, Guo, M, Liu, Q, Liu, J, Cui, Y. Neutrophil extracellular traps induce a hypercoagulable state in glioma. Immun Inflamm Dis 2021;9:1383–93. https://doi.org/10.1002/iid3.488.Search in Google Scholar PubMed PubMed Central

76. Cools-Lartigue, J, Spicer, J, McDonald, B, Gowing, S, Chow, S, Giannias, B, et al.. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J Clin Invest 2013;123:3446–58. https://doi.org/10.1172/jci67484.Search in Google Scholar

77. Najmeh, S, Cools-Lartigue, J, Rayes, RF, Gowing, S, Vourtzoumis, P, Bourdeau, F, et al.. Neutrophil extracellular traps sequester circulating tumor cells via β1-integrin mediated interactions. Int J Cancer 2017;140:2321–30. https://doi.org/10.1002/ijc.30635.Search in Google Scholar PubMed

78. Albrengues, J, Shields, MA, Ng, D, Park, CG, Ambrico, A, Poindexter, ME, et al.. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science 2018;361:1–14. https://doi.org/10.1126/science.aao4227.Search in Google Scholar PubMed PubMed Central

79. Schedel, F, Mayer-Hain, S, Pappelbaum, KI, Metze, D, Stock, M, Goerge, T, et al.. Evidence and impact of neutrophil extracellular traps in malignant melanoma. Pigment Cell Melanoma Res 2020;33:63–73. https://doi.org/10.1111/pcmr.12818.Search in Google Scholar PubMed

80. Muqaku, B, Pils, D, Mader, JC, Aust, S, Mangold, A, Muqaku, L, et al.. Neutrophil extracellular trap formation correlates with favorable overall survival in high grade ovarian cancer. Cancers (Basel) 2020;12:505. https://doi.org/10.3390/cancers12020505.Search in Google Scholar PubMed PubMed Central

81. Lee, W, Ko, SY, Mohamed, MS, Kenny, HA, Lengyel, E, Naora, H. Neutrophils facilitate ovarian cancer premetastatic niche formation in the omentum. J Exp Med 2019;216:176–94. https://doi.org/10.1084/jem.20181170.Search in Google Scholar PubMed PubMed Central

82. Park, J, Wysocki, RW, Amoozgar, Z, Maiorino, L, Fein, MR, Jorns, J, et al.. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci Transl Med 2016;8:1–12. https://doi.org/10.1126/scitranslmed.aag1711.Search in Google Scholar PubMed PubMed Central

83. Guo, L, Chen, G, Zhang, W, Zhou, L, Xiao, T, Di, X, et al.. A high-risk luminal a dominant breast cancer subtype with increased mobility. Breast Cancer Res Treat 2019;175:459–72. https://doi.org/10.1007/s10549-019-05135-w.Search in Google Scholar PubMed PubMed Central

84. Snoderly, HT, Boone, BA, Bennewitz, MF. Neutrophil extracellular traps in breast cancer and beyond: current perspectives on NET stimuli, thrombosis and metastasis, and clinical utility for diagnosis and treatment. Breast Cancer Res 2019;21:145. https://doi.org/10.1186/s13058-019-1237-6.Search in Google Scholar PubMed PubMed Central

85. Yang, C, Sun, W, Cui, W, Li, X, Yao, J, Jia, X, et al.. Procoagulant role of neutrophil extracellular traps in patients with gastric cancer. Int J Clin Exp Pathol 2015;8:14075–86.Search in Google Scholar

86. Zhu, T, Zou, X, Yang, C, Li, L, Wang, B, Li, R, et al.. Neutrophil extracellular traps promote gastric cancer metastasis by inducing epithelial-mesenchymal transition. Int J Mol Med 2021;48:127. https://doi.org/10.3892/ijmm.2021.4960.Search in Google Scholar PubMed PubMed Central

87. Pratt, HG, Steinberger, KJ, Mihalik, NE, Ott, S, Whalley, T, Szomolay, B, et al.. Macrophage and neutrophil interactions in the pancreatic tumor microenvironment drive the pathogenesis of pancreatic cancer. Cancers (Basel) 2021;14:194. https://doi.org/10.3390/cancers14010194.Search in Google Scholar PubMed PubMed Central

88. Arelaki, S, Arampatzioglou, A, Kambas, K, Papagoras, C, Miltiades, P, Angelidou, I, et al.. Gradient infiltration of neutrophil extracellular traps in colon cancer and evidence for their involvement in tumour growth. PLoS One 2016;11:e0154484. https://doi.org/10.1371/journal.pone.0154484.Search in Google Scholar PubMed PubMed Central

89. Yazdani, HO, Roy, E, Comerci, AJ, van der Windt, DJ, Zhang, H, Huang, H, et al.. Neutrophil extracellular traps drive mitochondrial homeostasis in tumors to augment growth. Cancer Res 2019;79:5626–39. https://doi.org/10.1158/0008-5472.can-19-0800.Search in Google Scholar

90. Yang, L, Liu, L, Zhang, R, Hong, J, Wang, Y, Wang, J, et al.. IL-8 mediates a positive loop connecting increased neutrophil extracellular traps (NETs) and colorectal cancer liver metastasis. J Cancer 2020;11:4384–96. https://doi.org/10.7150/jca.44215.Search in Google Scholar PubMed PubMed Central

91. van der Windt, DJ, Sud, V, Zhang, H, Varley, PR, Goswami, J, Yazdani, HO, et al.. Neutrophil extracellular traps promote inflammation and development of hepatocellular carcinoma in nonalcoholic steatohepatitis. Hepatology 2018;68:1347–60. https://doi.org/10.1002/hep.29914.Search in Google Scholar PubMed PubMed Central

92. Guan, X, Lu, Y, Zhu, H, Yu, S, Zhao, W, Chi, X, et al.. The crosstalk between cancer cells and neutrophils enhances hepatocellular carcinoma metastasis via neutrophil extracellular traps-associated cathepsin G component: a potential therapeutic target. J Hepatocell Carcinoma 2021;8:451–65. https://doi.org/10.2147/jhc.s303588.Search in Google Scholar PubMed PubMed Central

93. Yang, L, Liu, Q, Zhang, X, Liu, X, Zhou, B, Chen, J, et al.. DNA of neutrophil extracellular traps promotes cancer metastasis via CCDC25. Nature 2020;583:133–8. https://doi.org/10.1038/s41586-020-2394-6.Search in Google Scholar PubMed

94. Yang, L-Y, Luo, Q, Lu, L, Zhu, W-W, Sun, H-T, Wei, R, et al.. Increased neutrophil extracellular traps promote metastasis potential of hepatocellular carcinoma via provoking tumorous inflammatory response. J Hematol Oncol 2020;13:3. https://doi.org/10.1186/s13045-019-0836-0.Search in Google Scholar PubMed PubMed Central

95. Tohme, S, Yazdani, HO, Al-Khafaji, AB, Chidi, AP, Loughran, P, Mowen, K, et al.. Neutrophil extracellular traps promote the development and progression of liver metastases after surgical stress. Cancer Res 2016;76:1367–80. https://doi.org/10.1158/0008-5472.can-15-1591.Search in Google Scholar

96. Ferrer-Marín, F, Cuenca-Zamora, EJ, Guijarro-Carrillo, PJ, Teruel-Montoya, R. Emerging role of neutrophils in the thrombosis of chronic myeloproliferative neoplasms. Int J Mol Sci 2021;22:1143. https://doi.org/10.3390/ijms22031143.Search in Google Scholar PubMed PubMed Central

97. Li, T, Ma, R, Zhang, Y, Mo, H, Yang, X, Hu, S, et al.. Arsenic trioxide promoting ETosis in acute promyelocytic leukemia through mTOR-regulated autophagy. Cell Death Dis 2018;9:75. https://doi.org/10.1038/s41419-017-0018-3.Search in Google Scholar PubMed PubMed Central

98. Donkel, SJ, Portilla Fernández, E, Ahmad, S, Rivadeneira, F, van Rooij, FJA, Ikram, MA, et al.. Common and rare variants genetic association analysis of circulating neutrophil extracellular traps. Front Immunol 2021;12:1–10. https://doi.org/10.3389/fimmu.2021.615527.Search in Google Scholar PubMed PubMed Central

99. Zhang, H, Wang, Y, Onuma, A, He, J, Wang, H, Xia, Y, et al.. Neutrophils extracellular traps inhibition improves PD-1 blockade immunotherapy in colorectal cancer. Cancers (Basel) 2021;13:5333. https://doi.org/10.3390/cancers13215333.Search in Google Scholar PubMed PubMed Central

100. Kaltenmeier, C, Yazdani, HO, Morder, K, Geller, DA, Simmons, RL, Tohme, S. Neutrophil extracellular traps promote T cell exhaustion in the tumor microenvironment. Front Immunol 2021;12:1–14. https://doi.org/10.3389/fimmu.2021.785222.Search in Google Scholar PubMed PubMed Central

101. Domingo-Gonzalez, R, Martínez-Colón, GJ, Smith, AJ, Smith, CK, Ballinger, MN, Xia, M, et al.. Inhibition of neutrophil extracellular trap formation after stem cell transplant by prostaglandin E2. Am J Respir Crit Care Med 2016;193:186–97. https://doi.org/10.1164/rccm.201501-0161oc.Search in Google Scholar

102. Shishikura, K, Horiuchi, T, Sakata, N, Trinh, D-A, Shirakawa, R, Kimura, T, et al.. Prostaglandin E 2 inhibits neutrophil extracellular trap formation through production of cyclic AMP. Br J Pharmacol 2016;173:319–31. https://doi.org/10.1111/bph.13373.Search in Google Scholar PubMed PubMed Central

103. Knight, JS, Subramanian, V, O’Dell, AA, Yalavarthi, S, Zhao, W, Smith, CK, et al.. Peptidylarginine deiminase inhibition disrupts NET formation and protects against kidney, skin and vascular disease in lupus-prone MRL/lpr mice. Ann Rheum Dis 2015;74:2199–206. https://doi.org/10.1136/annrheumdis-2014-205365.Search in Google Scholar PubMed PubMed Central

104. U.S. National. Library of Medicine. Available from: clinicaltrials.gov [Accessed Apr 2022].Search in Google Scholar

Received: 2022-04-06
Accepted: 2022-06-09
Published Online: 2022-07-19
Published in Print: 2022-08-26

© 2022 the author(s), published by De Gruyter, Berlin/Boston

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 21.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/labmed-2022-0047/html?lang=en&srsltid=AfmBOoq17r4s0dOoZp3HfhhaIySlMv8dFcDl1FQ_UvaBJ1R79Q2vffBb
Scroll to top button