Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2531 2022-11-16 10:48:34 |
2 update references and layout Meta information modification 2531 2022-11-17 02:25:05 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Savino, F.D.;  Rigali, F.;  Giustini, V.;  D’aliberti, D.;  Spinelli, S.;  Piazza, R.;  Sacco, A.;  Roccaro, A.M. Liquid Biopsy in Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/34910 (accessed on 06 July 2024).
Savino FD,  Rigali F,  Giustini V,  D’aliberti D,  Spinelli S,  Piazza R, et al. Liquid Biopsy in Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/34910. Accessed July 06, 2024.
Savino, Francesco D., Fabio Rigali, Viviana Giustini, Deborah D’aliberti, Silvia Spinelli, Rocco Piazza, Antonio Sacco, Aldo M. Roccaro. "Liquid Biopsy in Cancer" Encyclopedia, https://encyclopedia.pub/entry/34910 (accessed July 06, 2024).
Savino, F.D.,  Rigali, F.,  Giustini, V.,  D’aliberti, D.,  Spinelli, S.,  Piazza, R.,  Sacco, A., & Roccaro, A.M. (2022, November 16). Liquid Biopsy in Cancer. In Encyclopedia. https://encyclopedia.pub/entry/34910
Savino, Francesco D., et al. "Liquid Biopsy in Cancer." Encyclopedia. Web. 16 November, 2022.
Liquid Biopsy in Cancer
Edit

Liquid biopsy (LBx) is a novel and promising approach in precision medicine, suitable for patient management in a wide range of medical conditions. Its utility in oncology ranges from disease screening to early diagnosis and treatment. LBx has several strengths, such as safeness, quickness of execution, and repeatability, compared to old-fashioned solid biopsy techniques; indeed, LBx requires the collection of a small number of biospecimens. LBx has been proven to be accurate and reliable, as demonstrated in several clinical studies, and it could have a surprising impact on survival and quality of life for cancer patients in the near future.

liquid biopsy lymphoproliferative diseases onco-hematology

1. Introduction

Liquid Biopsy (LBx) represents an approach that allows the identification of disease-related biomarkers, through a simple and minimally or non-invasive procedure, such as a blood draw or collection of other body fluids, including saliva, urine, cerebrospinal fluid (CSF), and stool. Particular attention has been given to LBx within the oncology field, in relation to both solid tumors and hematologic malignancies. For instance, research studies have shown how LBx could be used to support the clinician in diagnosis, treatment selection, as well as disease monitoring [1][2][3][4][5][6].
By performing and interpreting an LBx researchers may get useful information related to the detection of circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), circulating RNAs, and extracellular vesicles (EVs) through a peripheral blood draw.
Technologies for processing, isolating, and collecting tumor material, together with analysis techniques and the discovery of new research targets, have rapidly evolved, especially in the last decade. The first PCR-based test assessed on humans goes back to 2016 with the approval of the FDA, followed by the first two NGS-based assays in 2020 [7][8][9]. The implementation of LBx in the clinical management of lung cancer patients has found its rationale in circumstances where low tumor tissue availability could have hindered the execution of a conventional solid biopsy [10][11][12]. LBx has been implemented in cancer screening [13], with CTCs and ctDNA analysis being used for supporting prognosis assessment, defining minimum residual disease (MRD) status, driving treatment selection, and supporting cancer patient follow-up [14][15]. An additional use for LBx refers to early cancer detection, as shown by the relevance of circulating miRNA and cf-DNA in supporting cancer diagnosis [13][16][17][18][19][20][21].
Compared to solid biopsy, LBx offers several advantages (Table 1). First of all, a blood draw is safer, faster, and cheaper than an image-guided invasive surgical procedure, which requires planning, coordination, and execution by dedicated professionals. Moreover, primary tumor sources usually do not involve metastatic foci, something that is, vice versa, detectable within the bloodstream; the traditional approach is also not easily re-accessible through time without exposing the patient to risks due to repeated biopsies. Moreover, procedure-related bleeding and pain may represent common concurrent complications of solid biopsy [16][22][23]. Besides the static information that can be acquired through an LBx (i.e., diagnostic biomarkers), the latter sounds appealing when it comes to monitoring dynamic parameters, such as the activity of the disease and the longitudinal treatment response. Somatic mutations are also variable parameters, given the changing nature of the tumor and metastasis genetic makeup, and are more likely to be investigated through circulating tumor-derived particles [24][25]. LBx could also reveal subclonal mutations responsible for both tumor progression and drug resistance [15].
Table 1. Pros and cons of LBx compared to solid tissue biopsy.
Finally, recent studies have highlighted the relevance of extracellular vesicles (EVs), for instance, exosomes, which could serve as a crucial source for developing novel application fields for LBx [26]. Circulating EVs present with unique surface markers and disease-specific content, thus leading to the development of biopanels related to specific diseases, functions, and other variables useful in several clinical settings. New technologies are continuously emerging to isolate, identify, and link distinct EV subpopulations to peculiar biological and clinical features, even at an individual and personalized level, as in the case of the innovative phage display approach recently described by Maisano et al. [26]. Once LBx becomes well integrated into the clinical practice, it could replace solid biopsy for those cases requiring multiple draws at close range over time, or for patients that are unfit for undergoing an invasive solid biopsy procedure. Patients unfit for a solid biopsy are, for example, immunosuppressed subjects, who may suffer more easily from procedure-related complications, thus contributing to lower diagnostic power and higher mortality rates [27][28].
The main issue with the application of LBx in clinical practice is the confidence level of the tests and pre-analytic phases. For instance, LBx would not represent the best approach to identifying genetic alterations at an early disease stage, because of the low ctDNA concentration, resulting from a low tumor burden. The same concerns would apply within the MRD setting [20][29][30].

2. Liquid Biopsy: New Techniques and Biomarkers

Performing an LBx allows physicians and scientists to investigate tumor cells at both the cellular and molecular level: for instance, ctDNA and CTCs are the most studied so far. Recent studies have also included miRNAs, methylation markers, and extracellular vesicles, as emerging and promising alternatives, widening the diagnostic and therapeutic possibilities of LBx (Figure 1).
Figure 1. Principal tumor-derived elements detectable in peripheral blood and the assessable information they may carry.

2.1. cfDNA, ctDNA, and Circulating RNA

Cell-free DNA (cfDNA) consists of total extracellular DNA detectable within the bloodstream, as well as in other biospecimens, such as cerebrospinal and serous fluids [31][32][33].
Cancer cells, as well as non-tumor cells, could shed their genetic material into the bloodstream, as circulating tumor DNA (ctDNA) [34], which has been proven to be useful in supporting diagnosis, prognosis, disease progression, MRD evaluation, and treatment response, within the context of non-small cell lung cancer (NSCLC) and other solid tumors [10][11][12][13][14][15][29][34][35].
cfDNA is mainly derived from hematopoietic cells circulating in the bloodstream [36], where its amount is typically limited and poor in terms of quality. It is often necessary to proceed with amplification processes, which may cause artifacts at both PCR and sequencing levels. Only a small fraction of cfDNA is represented by ctDNA, posing a challenge in terms of isolation, processing DNA, and analysis of the generated data. Moreover, discriminating relevant DNA variants from leucocyte hematopoietic clones, such as those found in clonal hematopoiesis of indeterminate potential, may represent another hurdle to overcome [37][38], along with the fact that cfDNA could be enriched from other pathological non-tumoral tissues [39][40], hence adding another confounding factor in comorbid patients. Furthermore, cfDNA evaluation can be distorted by acute trauma, infections, stroke, exercise, and transplantation [41][42][43][44][45], making it necessary to develop better techniques for identifying tumor-specific somatic DNA variants.
In its simplest form, ctDNA analysis can be assimilated into a particular type of genotyping technique, whose goal is to test the presence of a usually restricted number of cancer-related variants. Among the main challenges associated with the ctDNA “genotyping” researchers may consider: (i) low concentration of circulating DNA; (ii) the existence of fragmented ctDNA; (iii) ctDNA representing only a small fraction of the total cfDNA [46][47]. This is particularly true for those samples characterized by a low tumor burden, e.g., at MRD or early diagnosis levels. The latter point is crucial and explains why ctDNA analysis requires dedicated, and often relatively costly techniques, as opposed to conventional genotyping [48]. Schematically, researchers can distinguish two scenarios: in the first one, a standard NGS-based approach (e.g., whole-exome or targeted sequencing) is usually adopted using the primary tumor mass, thus identifying the mutational landscape of the tumor. This map can be subsequently used to search, a priori, at nucleotide resolution level, for all the variants that need to be monitored by ctDNA analysis: this is a typical scenario that could apply to the MRD monitoring setting. In the second one, the specific variants that are present in the sample are unknown, which may significantly increase the complexity of the analysis. This typically occurs in early detection screening protocols. In all cases, however, a classical approach based on PCR amplification of the target locus/loci followed by amplicon Sanger sequencing is simply unsuitable for ctDNA analysis, as the sensitivity of Sanger technology for variants occurring at low variant allele frequency (VAF) is too low to be of any practical use in this scenario.
ctDNA analysis typically relies on the following steps: (1) cfDNA extraction; (2) enrichment of the target regions; (3) NGS library preparation; (4) NGS sequencing. cfDNA extraction is usually performed using either spin column- or magnetic bead-based methods [49][50][51][52]. Despite the spin column-based kits being considered the first choice, when it comes to collecting cfDNA for molecular analyses, they are more expensive and have longer execution times, compared to magnetic bead-based ones [53].
The enrichment step usually involves the use of targeted gene panels, aiming to investigate disease-related highly recurrent mutated genes. Moreover, in cases when the specific target mutations are known, PCR-based enrichment can be considered as well. In both scenarios, precautions must be taken to avoid preparation artifacts such as amplification-based errors. In terms of the NGS library preparation step, since high sensitivity and specificity are major goals in virtually all ctDNA analyses, several NGS protocols have been recently developed to maximize the reliability of NGS-based ctDNA variant calling [54][55][56]; these include, for instance, CAPPSeq, iDES, and SAFE-seq techniques [57][58][59], whose detailed description goes beyond the purpose of this research. The final NGS sequencing step depends upon the amount of sequencing material, being roughly proportional to the number of profiled genes and the desired depth of the analysis [60].
In general, one of the significant advantages of NGS techniques is that they are particularly sensitive in spotting single nucleotide variants (SNVs), small insertions and deletions, and copy number variations (CNVs), while the gene fusion and exon skipping detection fall within the competence of PCR and RNA sequencing.
Currently available ctDNA assays use hybrid-capture-based or Amplicon/PCR-amplified-based methodologies and are limited to a strict selection of gene sets [61]. Ongoing studies are assessing whole genome sequencing as a cheaper, more rapid, and accessible methodology for obtaining significant information from the LBx [62].
In the context of the transcriptome evaluation, the dysregulation of microRNAs (miRNAs) plays a role in the pathogenesis of several diseases, including cancer, by taking part in post-transcriptional modification of genes related to apoptosis, stress response, mitosis, and cell differentiation [17][63][64][65][66]. MiRNAs also circulate in the bloodstream, configuring themselves as promising diagnostic biomarkers and, possibly, therapeutic targets [67][68][69].
The knowledge about the usefulness of the circulating tumor components and the appropriate techniques to evaluate the messages they carry is still scarce and requires further studies and insightful discoveries to be embedded within the clinical practice setting.

2.2. CTCs

CTCs derive from tumor foci, either primary or metastatic [70][71]. They are released from the primary source in form of clusters, but they can also be found as single cells, whose correlation with overall survival and prognosis has been established [72]. CTCs could provide early information mirroring the primary tumor, such as genomic alterations [73][74], and gene and protein expression [75][76][77][78] characterizing the tumor cells. The existence of sub-clonal CTC populations has been confirmed as related to the cancer metastatic spreading, [79] leading to possible new ways of diagnosing the disease progression beforehand. CTCs’ rarity, heterogeneity, and the difficulties involved in their analysis make their clinical involvement quite challenging [80][81][82]. Overall, given the ability of CTCs to mirror the tumor of origin, they have been shown to provide useful information about the tumor itself, its metastatic spreading, as well as resistance or sensitivity to therapies [83][84][85].
CTCs detection, unfortunately, requires complex enrichment techniques, on the basis of both biological (i.e., specific antibody affinity) and physical properties (i.e., selecting them through size or deformability) [86][87]. Similarly to cfDNA, the amount of CTCs compared to the number of normal nucleated blood cells is very low: this has represented a major challenge that could affect the reliability of the results [88].
For CTCs detection and counting, FDA has currently approved one platform (CellSearch® system, Menarini Silicon Biosystems, Firenze, Italy), based on EpCAM+ CTCs identification [89].

2.3. Methylation Markers

DNA methylation has been reported to support the pathogenesis and disease progression of several cancer types. For instance, it can enhance the metastatic process, acting on promoter regions related to tumor suppressor genes [90][91][92]. It could also give information about the treatment response [93]. Since the methylation process could anticipate the manifestation of the full-blown disease [94], it is reasonable to think to integrate ctDNA methylation sequencing for early diagnosis, in a disease-related targeted way [95]. Based on this idea, Liu et al. conducted a study with a wide cohort of neoplastic patients, in which this method managed to diagnose more than 50 cancer types across all stages, with great specificity [96], but with low sensitivity level, particularly in early-stage cases [97]. The high specificity levels were justified by the rarity of DNA methylation findings in healthy samples’ CTCs and cfDNA [91][92][98][99]. In parallel, some studies have reported on the use of DNA methylation as a potential novel cancer biomarker, as shown for GSTPI, PITX2, and MGMT, within the context of prostate cancer, lymph node-negative breast cancer, and glioblastomas, respectively [100].

2.4. Extracellular Vesicles

Cells are capable of releasing vesicles across the extracellular space, as demonstrated within the context of both physiological and pathological conditions. These are heterogeneous particles, delimited by a phospholipid bilayer, responsible for several functions such as cell-to-cell communication [101], thus supporting the cross-talk between malignant cells and the microenvironment cells [102], as it has been shown within the specific context of multiple myeloma [103]. It is possible that, even when CTCs are below the threshold of detection, extracellular vesicles (EVs), for instance, nanovesicles, could be analyzed [104], and used to support the identification of cancer biomarkers. The content of these nanovesicles, together with lipids, proteins, and metabolites, often consists of small fragments of RNA and DNA [105]. They can generate from a tumor source, thus reflecting the primary tumor cell-specific mutational status, serving as a diagnostic biomarker [106][107]. In contrast to ctDNA, shed through cellular death mechanisms, EV-DNA is actively and selectively released by subclonal tumor cells and is wrapped in a membrane that protects the nucleic acids from a faster degradation, which makes it a more accurate depiction of the tumor and its diversified environment; nevertheless, the technological limitation in isolating EVs and analyzing them singularly prevents, at the moment, this promising opportunity from being adopted within clinical practice [108].
The dysregulation of microRNAs (miRNAs) plays a role in the pathogenesis of several diseases, including cancer, by taking part in post-transcriptional modification of apoptosis-, stress response-, mitosis-, and cell differentiation-related genes [17][63][64][65][66]. miRNAs circulate in the bloodstream both freely and as exosomal miRNAs, configuring themselves as promising diagnostic biomarkers and, possibly, therapeutic targets [67][68][69][109][110]. As reported in the literature, exosomal small RNAs (such as miRNAs) represent the main content of EVs [103][111][112].
EVs are usually characterized in terms of both morphology and content; this is achieved using several techniques, such as fluorescence-based platforms, electron microscopy, immunogold labeling, flow cytometry, and mass spectrometry [113][114], with great sensitivity and specificity degrees [103][115]. In the specific case of exosome characterization, for instance, CD63 and CD81 represent key markers to be investigated at the protein level [103][116][117].

References

  1. Siravegna, G.; Marsoni, S.; Siena, S.; Bardelli, A. Integrating liquid biopsies into the management of cancer. Nat. Rev. Clin. Oncol. 2017, 14, 531–548.
  2. Corcoran, R.B.; Chabner, B.A. Application of Cell-free DNA Analysis to Cancer Treatment. N. Engl. J. Med. 2018, 379, 1754–1765.
  3. Togneri, F.S.; Ward, D.G.; Foster, J.M.; Devall, A.J.; Wojtowicz, P.; Alyas, S.; Vasques, F.R.; Oumie, A.; James, N.D.; Cheng, K.K.; et al. Genomic complexity of urothelial bladder cancer revealed in urinary cfDNA. Eur. J. Hum. Genet. 2016, 24, 1167–1174.
  4. Kaczor-Urbanowicz, K.E.; Wei, F.; Rao, S.L.; Kim, J.; Shin, H.; Cheng, J.; Tu, M.; Wong, D.T.W.; Kim, Y. Clinical validity of saliva and novel technology for cancer detection. Biochim. Biophys. Acta Rev. Cancer 2019, 1872, 49–59.
  5. Imperiale, T.F.; Ransohoff, D.F.; Itzkowitz, S.H. Multitarget stool DNA testing for colorectal-cancer screening. N. Engl. J. Med. 2014, 371, 187–188.
  6. De Mattos-Arruda, L.; Mayor, R.; Ng, C.K.Y.; Weigelt, B.; Martinez-Ricarte, F.; Torrejon, D.; Oliveira, M.; Arias, A.; Raventos, C.; Tang, J.; et al. Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nat. Commun. 2015, 6, 8839.
  7. Malapelle, U.; Sirera, R.; Jantus-Lewintre, E.; Reclusa, P.; Calabuig-Farinas, S.; Blasco, A.; Pisapia, P.; Rolfo, C.; Camps, C. Profile of the Roche cobas(R) EGFR mutation test v2 for non-small cell lung cancer. Expert. Rev. Mol. Diagn. 2017, 17, 209–215.
  8. FDA. FDA Approves First Liquid Biopsy Next-Generation Sequencing Companion Diagnostic Test; FDA: Silver Spring, MD, USA, 2020.
  9. FDA. FDA Approves Liquid Biopsy NGS Companion Diagnostic Test for Multiple Cancers and Biomarkers; FDA: Silver Spring, MD, USA, 2020.
  10. Rolfo, C.; Mack, P.C.; Scagliotti, G.V.; Baas, P.; Barlesi, F.; Bivona, T.G.; Herbst, R.S.; Mok, T.S.; Peled, N.; Pirker, R.; et al. Liquid Biopsy for Advanced Non-Small Cell Lung Cancer (NSCLC): A Statement Paper from the IASLC. J. Thorac. Oncol. 2018, 13, 1248–1268.
  11. Leighl, N.B.; Page, R.D.; Raymond, V.M.; Daniel, D.B.; Divers, S.G.; Reckamp, K.L.; Villalona-Calero, M.A.; Dix, D.; Odegaard, J.I.; Lanman, R.B.; et al. Clinical Utility of Comprehensive Cell-free DNA Analysis to Identify Genomic Biomarkers in Patients with Newly Diagnosed Metastatic Non-small Cell Lung Cancer. Clin. Cancer Res. 2019, 25, 4691–4700.
  12. Aggarwal, C.; Thompson, J.C.; Black, T.A.; Katz, S.I.; Fan, R.; Yee, S.S.; Chien, A.L.; Evans, T.L.; Bauml, J.M.; Alley, E.W.; et al. Clinical Implications of Plasma-Based Genotyping With the Delivery of Personalized Therapy in Metastatic Non-Small Cell Lung Cancer. JAMA Oncol. 2019, 5, 173–180.
  13. Cescon, D.W.; Bratman, S.V.; Chan, S.M.; Siu, L.L. Circulating tumor DNA and liquid biopsy in oncology. Nat. Cancer 2020, 1, 276–290.
  14. Mosele, F.; Remon, J.; Mateo, J.; Westphalen, C.B.; Barlesi, F.; Lolkema, M.P.; Normanno, N.; Scarpa, A.; Robson, M.; Meric-Bernstam, F.; et al. Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: A report from the ESMO Precision Medicine Working Group. Ann. Oncol. 2020, 31, 1491–1505.
  15. Markou, A.; Tzanikou, E.; Lianidou, E. The potential of liquid biopsy in the management of cancer patients. Semin. Cancer Biol. 2022, 84, 69–79.
  16. Borghesi, M.; Ahmed, H.; Nam, R.; Schaeffer, E.; Schiavina, R.; Taneja, S.; Weidner, W.; Loeb, S. Complications After Systematic, Random, and Image-guided Prostate Biopsy. Eur. Urol. 2017, 71, 353–365.
  17. Lu, M.; Shi, B.; Wang, J.; Cao, Q.; Cui, Q. TAM: A method for enrichment and depletion analysis of a microRNA category in a list of microRNAs. BMC Bioinform. 2010, 11, 419.
  18. Roy, D.; Lucci, A.; Ignatiadis, M.; Jeffrey, S.S. Cell-free circulating tumor DNA profiling in cancer management. Trends Mol. Med. 2021, 27, 1014–1015.
  19. Roy, D.; Pascher, A.; Juratli, M.A.; Sporn, J.C. The Potential of Aptamer-Mediated Liquid Biopsy for Early Detection of Cancer. Int. J. Mol. Sci. 2021, 22, 5601.
  20. Sacher, A.G.; Paweletz, C.; Dahlberg, S.E.; Alden, R.S.; O’Connell, A.; Feeney, N.; Mach, S.L.; Janne, P.A.; Oxnard, G.R. Prospective Validation of Rapid Plasma Genotyping for the Detection of EGFR and KRAS Mutations in Advanced Lung Cancer. JAMA Oncol. 2016, 2, 1014–1022.
  21. Valihrach, L.; Androvic, P.; Kubista, M. Circulating miRNA analysis for cancer diagnostics and therapy. Mol. Aspects Med. 2020, 72, 100825.
  22. Tural-Kara, T.; Ozdemir, H.; Fitoz, S.; Ciftci, E.; Yalcinkaya, F. Bone marrow aspiration complications: Iliopsoas abscess and sacroiliac osteomyelitis. Turk J. Pediatr. 2016, 58, 562–565.
  23. Abhishek, K.; Khunger, N. Complications of skin biopsy. J. Cutan. Aesthet. Surg. 2015, 8, 239–241.
  24. Gerlinger, M.; Rowan, A.J.; Horswell, S.; Math, M.; Larkin, J.; Endesfelder, D.; Gronroos, E.; Martinez, P.; Matthews, N.; Stewart, A.; et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 2012, 366, 883–892.
  25. Osumi, H.; Shinozaki, E.; Yamaguchi, K.; Zembutsu, H. Clinical utility of circulating tumor DNA for colorectal cancer. Cancer Sci. 2019, 110, 1148–1155.
  26. Maisano, D.; Mimmi, S.; Dattilo, V.; Marino, F.; Gentile, M.; Vecchio, E.; Fiume, G.; Nistico, N.; Aloisio, A.; de Santo, M.P.; et al. A novel phage display based platform for exosome diversity characterization. Nanoscale 2022, 14, 2998–3003.
  27. Plattner, B.W.; Chen, P.; Cross, R.; Leavitt, M.A.; Killen, P.D.; Heung, M. Complications and adequacy of transplant kidney biopsies: A comparison of techniques. J. Vasc. Access 2018, 19, 291–296.
  28. Cleveland, H.; Chau, A.; Jeng, Z.; Gardner, G.; Yoo, R.; Zhang, W.; Hernandez, J. Percutaneous Lung Biopsy in Immunocompromised Pediatric Patients. J. Vasc. Interv. Radiol. 2020, 31, 93–98.
  29. Siravegna, G.; Mussolin, B.; Venesio, T.; Marsoni, S.; Seoane, J.; Dive, C.; Papadopoulos, N.; Kopetz, S.; Corcoran, R.B.; Siu, L.L.; et al. How liquid biopsies can change clinical practice in oncology. Ann. Oncol. 2019, 30, 1580–1590.
  30. Rolfo, C.; Cardona, A.F.; Cristofanilli, M.; Paz-Ares, L.; Diaz Mochon, J.J.; Duran, I.; Raez, L.E.; Russo, A.; Lorente, J.A.; Malapelle, U.; et al. Challenges and opportunities of cfDNA analysis implementation in clinical practice: Perspective of the International Society of Liquid Biopsy (ISLB). Crit. Rev. Oncol. Hematol. 2020, 151, 102978.
  31. Reckamp, K.L.; Melnikova, V.O.; Karlovich, C.; Sequist, L.V.; Camidge, D.R.; Wakelee, H.; Perol, M.; Oxnard, G.R.; Kosco, K.; Croucher, P.; et al. A Highly Sensitive and Quantitative Test Platform for Detection of NSCLC EGFR Mutations in Urine and Plasma. J. Thorac. Oncol. 2016, 11, 1690–1700.
  32. Villatoro, S.; Mayo-de-Las-Casas, C.; Jordana-Ariza, N.; Viteri-Ramirez, S.; Garzon-Ibanez, M.; Moya-Horno, I.; Garcia-Pelaez, B.; Gonzalez-Cao, M.; Malapelle, U.; Balada-Bel, A.; et al. Prospective detection of mutations in cerebrospinal fluid, pleural effusion, and ascites of advanced cancer patients to guide treatment decisions. Mol. Oncol. 2019, 13, 2633–2645.
  33. Zheng, M.M.; Li, Y.S.; Tu, H.Y.; Jiang, B.Y.; Yang, J.J.; Zhou, Q.; Xu, C.R.; Yang, X.R.; Wu, Y.L. Genotyping of Cerebrospinal Fluid Associated With Osimertinib Response and Resistance for Leptomeningeal Metastases in EGFR-Mutated NSCLC. J. Thorac. Oncol. 2021, 16, 250–258.
  34. Diaz, L.A., Jr.; Bardelli, A. Liquid biopsies: Genotyping circulating tumor DNA. J. Clin. Oncol. 2014, 32, 579–586.
  35. Heitzer, E.; Ulz, P.; Geigl, J.B. Circulating tumor DNA as a liquid biopsy for cancer. Clin. Chem. 2015, 61, 112–123.
  36. Jiang, P.; Chan, K.C.A.; Lo, Y.M.D. Liver-derived cell-free nucleic acids in plasma: Biology and applications in liquid biopsies. J. Hepatol. 2019, 71, 409–421.
  37. Bauml, J.; Levy, B. Clonal Hematopoiesis: A New Layer in the Liquid Biopsy Story in Lung Cancer. Clin. Cancer Res. 2018, 24, 4352–4354.
  38. Dai, J.; Su, Y.; Zhong, S.; Cong, L.; Liu, B.; Yang, J.; Tao, Y.; He, Z.; Chen, C.; Jiang, Y. Exosomes: Key players in cancer and potential therapeutic strategy. Signal Transduct. Target. Ther. 2020, 5, 145.
  39. Moss, J.; Magenheim, J.; Neiman, D.; Zemmour, H.; Loyfer, N.; Korach, A.; Samet, Y.; Maoz, M.; Druid, H.; Arner, P.; et al. Comprehensive human cell-type methylation atlas reveals origins of circulating cell-free DNA in health and disease. Nat. Commun. 2018, 9, 5068.
  40. Liu, X.; Ren, J.; Luo, N.; Guo, H.; Zheng, Y.; Li, J.; Tang, F.; Wen, L.; Peng, J. Comprehensive DNA methylation analysis of tissue of origin of plasma cell-free DNA by methylated CpG tandem amplification and sequencing (MCTA-Seq). Clin. Epigenetics 2019, 11, 93.
  41. Rodrigues Filho, E.M.; Simon, D.; Ikuta, N.; Klovan, C.; Dannebrock, F.A.; Oliveira de Oliveira, C.; Regner, A. Elevated cell-free plasma DNA level as an independent predictor of mortality in patients with severe traumatic brain injury. J. Neurotrauma 2014, 31, 1639–1646.
  42. O’Connell, G.C.; Petrone, A.B.; Tennant, C.S.; Lucke-Wold, N.; Kabbani, Y.; Tarabishy, A.R.; Chantler, P.D.; Barr, T.L. Circulating extracellular DNA levels are acutely elevated in ischaemic stroke and associated with innate immune system activation. Brain Inj. 2017, 31, 1369–1375.
  43. Tug, S.; Helmig, S.; Deichmann, E.R.; Schmeier-Jurchott, A.; Wagner, E.; Zimmermann, T.; Radsak, M.; Giacca, M.; Simon, P. Exercise-induced increases in cell free DNA in human plasma originate predominantly from cells of the haematopoietic lineage. Exerc. Immunol. Rev. 2015, 21, 164–173.
  44. De Vlaminck, I.; Martin, L.; Kertesz, M.; Patel, K.; Kowarsky, M.; Strehl, C.; Cohen, G.; Luikart, H.; Neff, N.F.; Okamoto, J.; et al. Noninvasive monitoring of infection and rejection after lung transplantation. Proc. Natl. Acad. Sci. USA 2015, 112, 13336–13341.
  45. Phuong, N.T.N.; Manh, D.H.; Dumre, S.P.; Mizukami, S.; Weiss, L.N.; Van Thuong, N.; Ha, T.T.N.; Phuc, L.H.; Van An, T.; Tieu, T.M.; et al. Plasma cell-free DNA: A potential biomarker for early prediction of severe dengue. Ann. Clin. Microbiol. Antimicrob. 2019, 18, 10.
  46. Mouliere, F.; Chandrananda, D.; Piskorz, A.M.; Moore, E.K.; Morris, J.; Ahlborn, L.B.; Mair, R.; Goranova, T.; Marass, F.; Heider, K.; et al. Enhanced detection of circulating tumor DNA by fragment size analysis. Sci. Transl. Med. 2018, 10, eaat4921.
  47. Underhill, H.R.; Kitzman, J.O.; Hellwig, S.; Welker, N.C.; Daza, R.; Baker, D.N.; Gligorich, K.M.; Rostomily, R.C.; Bronner, M.P.; Shendure, J. Fragment Length of Circulating Tumor DNA. PLoS Genet. 2016, 12, e1006162.
  48. Abbosh, C.; Birkbak, N.J.; Wilson, G.A.; Jamal-Hanjani, M.; Constantin, T.; Salari, R.; Le Quesne, J.; Moore, D.A.; Veeriah, S.; Rosenthal, R.; et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 2017, 545, 446–451.
  49. Kloten, V.; Ruchel, N.; Bruchle, N.O.; Gasthaus, J.; Freudenmacher, N.; Steib, F.; Mijnes, J.; Eschenbruch, J.; Binnebosel, M.; Knuchel, R.; et al. Liquid biopsy in colon cancer: Comparison of different circulating DNA extraction systems following absolute quantification of KRAS mutations using Intplex allele-specific PCR. Oncotarget 2017, 8, 86253–86263.
  50. Perez-Barrios, C.; Nieto-Alcolado, I.; Torrente, M.; Jimenez-Sanchez, C.; Calvo, V.; Gutierrez-Sanz, L.; Palka, M.; Donoso-Navarro, E.; Provencio, M.; Romero, A. Comparison of methods for circulating cell-free DNA isolation using blood from cancer patients: Impact on biomarker testing. Transl. Lung Cancer Res. 2016, 5, 665–672.
  51. Sorber, L.; Zwaenepoel, K.; Deschoolmeester, V.; Roeyen, G.; Lardon, F.; Rolfo, C.; Pauwels, P. A Comparison of Cell-Free DNA Isolation Kits: Isolation and Quantification of Cell-Free DNA in Plasma. J. Mol. Diagn. 2017, 19, 162–168.
  52. Warton, K.; Graham, L.J.; Yuwono, N.; Samimi, G. Comparison of 4 commercial kits for the extraction of circulating DNA from plasma. Cancer Genet. 2018, 228–229, 143–150.
  53. Diefenbach, R.J.; Lee, J.H.; Kefford, R.F.; Rizos, H. Evaluation of commercial kits for purification of circulating free DNA. Cancer Genet. 2018, 228–229, 21–27.
  54. Saelee, S.L.; Lovejoy, A.F.; Hinzmann, B.; Mayol, K.; Huynh, S.; Harrell, A.; Lefkowitz, J.; Deodhar, N.; Garcia-Montoya, G.; Yaung, S.J.; et al. Quantitative PCR-Based Method to Assess Cell-Free DNA Quality, Adjust Input Mass, and Improve Next-Generation Sequencing Assay Performance. J. Mol. Diagn. 2022, 24, 566–575.
  55. Gong, B.; Deveson, I.W.; Mercer, T.; Johann, D.J., Jr.; Jones, W.; Tong, W.; Xu, J. Ultra-deep sequencing data from a liquid biopsy proficiency study demonstrating analytic validity. Sci. Data 2022, 9, 170.
  56. Lander, E.S.; Linton, L.M.; Birren, B.; Nusbaum, C.; Zody, M.C.; Baldwin, J.; Devon, K.; Dewar, K.; Doyle, M.; FitzHugh, W.; et al. Initial sequencing and analysis of the human genome. Nature 2001, 409, 860–921.
  57. Newman, A.M.; Bratman, S.V.; To, J.; Wynne, J.F.; Eclov, N.C.; Modlin, L.A.; Liu, C.L.; Neal, J.W.; Wakelee, H.A.; Merritt, R.E.; et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat. Med. 2014, 20, 548–554.
  58. Newman, A.M.; Lovejoy, A.F.; Klass, D.M.; Kurtz, D.M.; Chabon, J.J.; Scherer, F.; Stehr, H.; Liu, C.L.; Bratman, S.V.; Say, C.; et al. Integrated digital error suppression for improved detection of circulating tumor DNA. Nat. Biotechnol. 2016, 34, 547–555.
  59. Tie, J.; Kinde, I.; Wang, Y.; Wong, H.L.; Roebert, J.; Christie, M.; Tacey, M.; Wong, R.; Singh, M.; Karapetis, C.S.; et al. Circulating tumor DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer. Ann. Oncol. 2015, 26, 1715–1722.
  60. Mellert, H.; Reese, J.; Jackson, L.; Maxwell, V.; Tschida, C.; Pestano, G.A. Targeted Next-Generation Sequencing of Liquid Biopsy Samples from Patients with NSCLC. Diagnostics 2021, 11, 155.
  61. Garcia-Pardo, M.; Makarem, M.; Li, J.J.N.; Kelly, D.; Leighl, N.B. Integrating circulating-free DNA (cfDNA) analysis into clinical practice: Opportunities and challenges. Br. J. Cancer 2022, 127, 592–602.
  62. Raman, L.; Van der Linden, M.; Van der Eecken, K.; Vermaelen, K.; Demedts, I.; Surmont, V.; Himpe, U.; Dedeurwaerdere, F.; Ferdinande, L.; Lievens, Y.; et al. Shallow whole-genome sequencing of plasma cell-free DNA accurately differentiates small from non-small cell lung carcinoma. Genome Med. 2020, 12, 35.
  63. Wang, Y.; Lee, C.G. MicroRNA and cancer--focus on apoptosis. J. Cell. Mol. Med. 2009, 13, 12–23.
  64. Jiang, Q.; Wang, Y.; Hao, Y.; Juan, L.; Teng, M.; Zhang, X.; Li, M.; Wang, G.; Liu, Y. miR2 Disease: A manually curated database for microRNA deregulation in human disease. Nucleic Acids Res. 2009, 37, D98–D104.
  65. Chen, X.; Liu, M.X.; Yan, G.Y. RWRMDA: Predicting novel human microRNA-disease associations. Mol. Biosyst. 2012, 8, 2792–2798.
  66. Li, Y.; Qiu, C.; Tu, J.; Geng, B.; Yang, J.; Jiang, T.; Cui, Q. HMDD v2.0: A database for experimentally supported human microRNA and disease associations. Nucleic Acids Res. 2014, 42, D1070–D1074.
  67. Chen, G.; Wang, J.; Cui, Q. Could circulating miRNAs contribute to cancer therapy? Trends Mol. Med. 2013, 19, 71–73.
  68. Keller, A.; Leidinger, P.; Bauer, A.; Elsharawy, A.; Haas, J.; Backes, C.; Wendschlag, A.; Giese, N.; Tjaden, C.; Ott, K.; et al. Toward the blood-borne miRNome of human diseases. Nat. Methods 2011, 8, 841–843.
  69. Mo, M.H.; Chen, L.; Fu, Y.; Wang, W.; Fu, S.W. Cell-free Circulating miRNA Biomarkers in Cancer. J. Cancer 2012, 3, 432–448.
  70. Chaffer, C.L.; Weinberg, R.A. A perspective on cancer cell metastasis. Science 2011, 331, 1559–1564.
  71. Alix-Panabieres, C.; Pantel, K. Circulating tumor cells: Liquid biopsy of cancer. Clin. Chem. 2013, 59, 110–118.
  72. Bidard, F.C.; Peeters, D.J.; Fehm, T.; Nole, F.; Gisbert-Criado, R.; Mavroudis, D.; Grisanti, S.; Generali, D.; Garcia-Saenz, J.A.; Stebbing, J.; et al. Clinical validity of circulating tumour cells in patients with metastatic breast cancer: A pooled analysis of individual patient data. Lancet Oncol. 2014, 15, 406–414.
  73. Ni, X.; Zhuo, M.; Su, Z.; Duan, J.; Gao, Y.; Wang, Z.; Zong, C.; Bai, H.; Chapman, A.R.; Zhao, J.; et al. Reproducible copy number variation patterns among single circulating tumor cells of lung cancer patients. Proc. Natl. Acad. Sci. USA 2013, 110, 21083–21088.
  74. Lohr, J.G.; Adalsteinsson, V.A.; Cibulskis, K.; Choudhury, A.D.; Rosenberg, M.; Cruz-Gordillo, P.; Francis, J.M.; Zhang, C.Z.; Shalek, A.K.; Satija, R.; et al. Whole-exome sequencing of circulating tumor cells provides a window into metastatic prostate cancer. Nat. Biotechnol. 2014, 32, 479–484.
  75. Kalinsky, K.; Mayer, J.A.; Xu, X.; Pham, T.; Wong, K.L.; Villarin, E.; Pircher, T.J.; Brown, M.; Maurer, M.A.; Bischoff, F.Z. Correlation of hormone receptor status between circulating tumor cells, primary tumor, and metastasis in breast cancer patients. Clin. Transl. Oncol. 2015, 17, 539–546.
  76. Yu, M.; Ting, D.T.; Stott, S.L.; Wittner, B.S.; Ozsolak, F.; Paul, S.; Ciciliano, J.C.; Smas, M.E.; Winokur, D.; Gilman, A.J.; et al. RNA sequencing of pancreatic circulating tumour cells implicates WNT signalling in metastasis. Nature 2012, 487, 510–513.
  77. Wallwiener, M.; Hartkopf, A.D.; Riethdorf, S.; Nees, J.; Sprick, M.R.; Schonfisch, B.; Taran, F.A.; Heil, J.; Sohn, C.; Pantel, K.; et al. The impact of HER2 phenotype of circulating tumor cells in metastatic breast cancer: A retrospective study in 107 patients. BMC Cancer 2015, 15, 403.
  78. Kalinich, M.; Bhan, I.; Kwan, T.T.; Miyamoto, D.T.; Javaid, S.; LiCausi, J.A.; Milner, J.D.; Hong, X.; Goyal, L.; Sil, S.; et al. An RNA-based signature enables high specificity detection of circulating tumor cells in hepatocellular carcinoma. Proc. Natl. Acad. Sci. USA 2017, 114, 1123–1128.
  79. Keller, L.; Pantel, K. Unravelling tumour heterogeneity by single-cell profiling of circulating tumour cells. Nat. Rev. Cancer 2019, 19, 553–567.
  80. Bardelli, A.; Pantel, K. Liquid Biopsies, What We Do Not Know (Yet). Cancer Cell 2017, 31, 172–179.
  81. Lianidou, E.S. Gene expression profiling and DNA methylation analyses of CTCs. Mol. Oncol. 2016, 10, 431–442.
  82. Boral, D.; Vishnoi, M.; Liu, H.N.; Yin, W.; Sprouse, M.L.; Scamardo, A.; Hong, D.S.; Tan, T.Z.; Thiery, J.P.; Chang, J.C.; et al. Molecular characterization of breast cancer CTCs associated with brain metastasis. Nat. Commun. 2017, 8, 196.
  83. Jakabova, A.; Bielcikova, Z.; Pospisilova, E.; Matkowski, R.; Szynglarewicz, B.; Staszek-Szewczyk, U.; Zemanova, M.; Petruzelka, L.; Eliasova, P.; Kolostova, K.; et al. Molecular characterization and heterogeneity of circulating tumor cells in breast cancer. Breast Cancer Res. Treat. 2017, 166, 695–700.
  84. Markou, A.; Strati, A.; Malamos, N.; Georgoulias, V.; Lianidou, E.S. Molecular characterization of circulating tumor cells in breast cancer by a liquid bead array hybridization assay. Clin. Chem. 2011, 57, 421–430.
  85. Strati, A.; Markou, A.; Parisi, C.; Politaki, E.; Mavroudis, D.; Georgoulias, V.; Lianidou, E. Gene expression profile of circulating tumor cells in breast cancer by RT-qPCR. BMC Cancer 2011, 11, 422.
  86. Alix-Panabieres, C.; Pantel, K. Challenges in circulating tumour cell research. Nat. Rev. Cancer 2014, 14, 623–631.
  87. Desitter, I.; Guerrouahen, B.S.; Benali-Furet, N.; Wechsler, J.; Janne, P.A.; Kuang, Y.; Yanagita, M.; Wang, L.; Berkowitz, J.A.; Distel, R.J.; et al. A new device for rapid isolation by size and characterization of rare circulating tumor cells. Anticancer Res. 2011, 31, 427–441.
  88. Liu, Y.; Li, R.; Zhang, L.; Guo, S. Nanomaterial-Based Immunocapture Platforms for the Recognition, Isolation, and Detection of Circulating Tumor Cells. Front. Bioeng. Biotechnol. 2022, 10, 850241.
  89. Riethdorf, S.; O’Flaherty, L.; Hille, C.; Pantel, K. Clinical applications of the CellSearch platform in cancer patients. Adv. Drug Deliv. Rev. 2018, 125, 102–121.
  90. Esteller, M. Epigenetics in cancer. N. Engl. J. Med. 2008, 358, 1148–1159.
  91. Pixberg, C.F.; Schulz, W.A.; Stoecklein, N.H.; Neves, R.P. Characterization of DNA Methylation in Circulating Tumor Cells. Genes 2015, 6, 1053–1075.
  92. Chimonidou, M.; Strati, A.; Tzitzira, A.; Sotiropoulou, G.; Malamos, N.; Georgoulias, V.; Lianidou, E.S. DNA methylation of tumor suppressor and metastasis suppressor genes in circulating tumor cells. Clin. Chem. 2011, 57, 1169–1177.
  93. Sigin, V.O.; Kalinkin, A.I.; Kuznetsova, E.B.; Simonova, O.A.; Chesnokova, G.G.; Litviakov, N.V.; Slonimskaya, E.M.; Tsyganov, M.M.; Ibragimova, M.K.; Volodin, I.V.; et al. DNA methylation markers panel can improve prediction of response to neoadjuvant chemotherapy in luminal B breast cancer. Sci. Rep. 2020, 10, 9239.
  94. Kanwal, R.; Gupta, K.; Gupta, S. Cancer epigenetics: An introduction. Methods Mol. Biol. 2015, 1238, 3–25.
  95. Liu, L.; Toung, J.M.; Jassowicz, A.F.; Vijayaraghavan, R.; Kang, H.; Zhang, R.; Kruglyak, K.M.; Huang, H.J.; Hinoue, T.; Shen, H.; et al. Targeted methylation sequencing of plasma cell-free DNA for cancer detection and classification. Ann. Oncol. 2018, 29, 1445–1453.
  96. Liu, M.C.; Oxnard, G.R.; Klein, E.A.; Swanton, C.; Seiden, M.V.; Consortium, C. Sensitive and specific multi-cancer detection and localization using methylation signatures in cell-free DNA. Ann. Oncol. 2020, 31, 745–759.
  97. Klein, E.A.; Richards, D.; Cohn, A.; Tummala, M.; Lapham, R.; Cosgrove, D.; Chung, G.; Clement, J.; Gao, J.; Hunkapiller, N.; et al. Clinical validation of a targeted methylation-based multi-cancer early detection test using an independent validation set. Ann. Oncol. 2021, 32, 1167–1177.
  98. Chimonidou, M.; Tzitzira, A.; Strati, A.; Sotiropoulou, G.; Sfikas, C.; Malamos, N.; Georgoulias, V.; Lianidou, E. CST6 promoter methylation in circulating cell-free DNA of breast cancer patients. Clin. Biochem. 2013, 46, 235–240.
  99. Chimonidou, M.; Strati, A.; Malamos, N.; Georgoulias, V.; Lianidou, E.S. SOX17 promoter methylation in circulating tumor cells and matched cell-free DNA isolated from plasma of patients with breast cancer. Clin. Chem. 2013, 59, 270–279.
  100. Duffy, M.J.; Napieralski, R.; Martens, J.W.; Span, P.N.; Spyratos, F.; Sweep, F.C.; Brunner, N.; Foekens, J.A.; Schmitt, M.; Group, E.P. Methylated genes as new cancer biomarkers. Eur. J. Cancer 2009, 45, 335–346.
  101. Maas, S.L.N.; Breakefield, X.O.; Weaver, A.M. Extracellular Vesicles: Unique Intercellular Delivery Vehicles. Trends Cell Biol. 2017, 27, 172–188.
  102. Muralidharan-Chari, V.; Clancy, J.W.; Sedgwick, A.; D’Souza-Schorey, C. Microvesicles: Mediators of extracellular communication during cancer progression. J. Cell Sci. 2010, 123, 1603–1611.
  103. Roccaro, A.M.; Sacco, A.; Maiso, P.; Azab, A.K.; Tai, Y.T.; Reagan, M.; Azab, F.; Flores, L.M.; Campigotto, F.; Weller, E.; et al. BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression. J. Clin. Investig. 2013, 123, 1542–1555.
  104. Longjohn, M.N.; Hudson, J.B.J.; Smith, N.C.; Rise, M.L.; Moorehead, P.C.; Christian, S.L. Deciphering the messages carried by extracellular vesicles in hematological malignancies. Blood Rev. 2021, 46, 100734.
  105. Jurj, A.; Zanoaga, O.; Braicu, C.; Lazar, V.; Tomuleasa, C.; Irimie, A.; Berindan-Neagoe, I. A Comprehensive Picture of Extracellular Vesicles and Their Contents. Molecular Transfer to Cancer Cells. Cancers 2020, 12, 298.
  106. Kahlert, C.; Melo, S.A.; Protopopov, A.; Tang, J.; Seth, S.; Koch, M.; Zhang, J.; Weitz, J.; Chin, L.; Futreal, A.; 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, 3869–3875.
  107. Thakur, B.K.; Zhang, H.; Becker, A.; Matei, I.; Huang, Y.; Costa-Silva, B.; Zheng, Y.; Hoshino, A.; Brazier, H.; Xiang, J.; et al. Double-stranded DNA in exosomes: A novel biomarker in cancer detection. Cell Res. 2014, 24, 766–769.
  108. Ghanam, J.; Chetty, V.K.; Barthel, L.; Reinhardt, D.; Hoyer, P.F.; Thakur, B.K. DNA in extracellular vesicles: From evolution to its current application in health and disease. Cell Biosci. 2022, 12, 37.
  109. Azmi, A.S.; Bao, B.; Sarkar, F.H. Exosomes in cancer development, metastasis, and drug resistance: A comprehensive review. Cancer Metastasis. Rev. 2013, 32, 623–642.
  110. Yeh, Y.Y.; Ozer, H.G.; Lehman, A.M.; Maddocks, K.; Yu, L.; Johnson, A.J.; Byrd, J.C. Characterization of CLL exosomes reveals a distinct microRNA signature and enhanced secretion by activation of BCR signaling. Blood 2015, 125, 3297–3305.
  111. Berardocco, M.; Radeghieri, A.; Busatto, S.; Gallorini, M.; Raggi, C.; Gissi, C.; D’Agnano, I.; Bergese, P.; Felsani, A.; Berardi, A.C. RNA-seq reveals distinctive RNA profiles of small extracellular vesicles from different human liver cancer cell lines. Oncotarget 2017, 8, 82920–82939.
  112. Lunavat, T.R.; Cheng, L.; Kim, D.K.; Bhadury, J.; Jang, S.C.; Lasser, C.; Sharples, R.A.; Lopez, M.D.; Nilsson, J.; Gho, Y.S.; et al. Small RNA deep sequencing discriminates subsets of extracellular vesicles released by melanoma cells--Evidence of unique microRNA cargos. RNA Biol. 2015, 12, 810–823.
  113. Thery, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; 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.
  114. Szatanek, R.; Baj-Krzyworzeka, M.; Zimoch, J.; Lekka, M.; Siedlar, M.; Baran, J. The Methods of Choice for Extracellular Vesicles (EVs) Characterization. Int. J. Mol. Sci. 2017, 18, 1153.
  115. Hoshino, A.; Kim, H.S.; Bojmar, L.; Gyan, K.E.; Cioffi, M.; Hernandez, J.; Zambirinis, C.P.; Rodrigues, G.; Molina, H.; Heissel, S.; et al. Extracellular Vesicle and Particle Biomarkers Define Multiple Human Cancers. Cell 2020, 182, 1044–1061.e18.
  116. Paolino, G.; Huber, V.; Camerini, S.; Casella, M.; Macone, A.; Bertuccini, L.; Iosi, F.; Moliterni, E.; Cecchetti, S.; Ruspantini, I.; et al. The Fatty Acid and Protein Profiles of Circulating CD81-Positive Small Extracellular Vesicles Are Associated with Disease Stage in Melanoma Patients. Cancers 2021, 13, 4157.
  117. Schey, K.L.; Luther, J.M.; Rose, K.L. Proteomics characterization of exosome cargo. Methods 2015, 87, 75–82.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , ,
View Times: 484
Revisions: 2 times (View History)
Update Date: 17 Nov 2022
1000/1000
Video Production Service