The Liquid Biopsy in the Management of Colorectal Cancer: An Overview

Biomedicines - Tập 8 Số 9 - Trang 308
Marco Vacante1, Roberto Ciuni1, Francesco Basile1, Antonio Biondi1
1Department of General Surgery and Medical-Surgical Specialties, University of Catania, Via S. Sofia 78, 95123, Catania, Italy

Tóm tắt

Currently, there is a crucial need for novel diagnostic and prognostic biomarkers with high specificity and sensitivity in patients with colorectal cancer. A “liquid biopsy” is characterized by the isolation of cancer-derived components, such as circulating tumor cells, circulating tumor DNA, microRNAs, long non-coding RNAs, and proteins, from peripheral blood or other body fluids and their genomic or proteomic assessment. The liquid biopsy is a minimally invasive and repeatable technique that could play a significant role in screening and diagnosis, and predict relapse and metastasis, as well as monitoring minimal residual disease and chemotherapy resistance in colorectal cancer patients. However, there are still some practical issues that need to be addressed before liquid biopsy can be widely used in clinical practice. Potential challenges may include low amounts of circulating tumor cells and circulating tumor DNA in samples, lack of pre-analytical and analytical consensus, clinical validation, and regulatory endorsement. The aim of this review was to summarize the current knowledge of the role of liquid biopsy in the management of colorectal cancer.

Từ khóa


Tài liệu tham khảo

GBD 2017 Colorectal Cancer Collaborators (2019). The global, regional, and national burden of colorectal cancer and its attributable risk factors in 195 countries and territories, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol. Hepatol., 4, 913–933.

Bray, 2018, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries, CA Cancer J. Clin., 68, 394, 10.3322/caac.21492

Bishehsari, 2014, Epidemiological transition of colorectal cancer in developing countries: Environmental factors, molecular pathways, and opportunities for prevention, World J. Gastroenterol., 20, 6055, 10.3748/wjg.v20.i20.6055

Hui, L. (2018). Quantifying the effects of aging and urbanization on major gastrointestinal diseases to guide preventative strategies. BMC Gastroenterol., 18.

Valli, 2015, Hypoxia metabolism in ageing, Aging Albany N. Y., 7, 465, 10.18632/aging.100782

(2020, July 14). Cancer Facts & Figures 2020|American Cancer Society. Available online: https://www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/cancer-facts-figures-2020.html.

Stintzing, 2014, Management of colorectal cancer, F1000Prime Rep., 6, 108, 10.12703/P6-108

Nakayama, 2013, Current Options for the Diagnosis, Staging and Therapeutic Management of Colorectal Cancer, Gastrointest. Tumors, 1, 25, 10.1159/000354995

Bender, 2019, Adjuvant therapy for stages II and III colon cancer: Risk stratification, treatment duration, and future directions, Curr. Oncol., 26, S43, 10.3747/co.26.5605

Pfeiffer, 2019, The changing face of treatment for metastatic colorectal cancer, Expert Rev. Anticancer Ther., 19, 61, 10.1080/14737140.2019.1543593

Moriarity, 2016, Current targeted therapies in the treatment of advanced colorectal cancer: A review, Ther. Adv. Med. Oncol., 8, 276, 10.1177/1758834016646734

Norcic, G. (2018). Liquid biopsy in colorectal cancer-current status and potential clinical applications. Micromachines, 9.

Vacante, 2018, Biomarkers in colorectal cancer: Current clinical utility and future perspectives, World J. Clin. Cases, 6, 869, 10.12998/wjcc.v6.i15.869

Palmirotta, R., Lovero, D., Cafforio, P., Felici, C., Mannavola, F., Pellè, E., Quaresmini, D., Tucci, M., and Silvestris, F. (2018). Liquid biopsy of cancer: A multimodal diagnostic tool in clinical oncology. Ther. Adv. Med. Oncol., 10.

Crowley, 2013, Liquid biopsy: Monitoring cancer-genetics in the blood, Nat. Rev. Clin. Oncol., 10, 472, 10.1038/nrclinonc.2013.110

Fernández-Lázaro, D., García Hernández, J.L., García, A.C., Córdova Martínez, A., Mielgo-Ayuso, J., and Cruz-Hernández, J.J. (2020). Liquid biopsy as novel tool in precision medicine: Origins, properties, identification and clinical perspective of cancer’s biomarkers. Diagnostics, 10.

Otero, 2018, Exosomes: Definition, role in tumor development and clinical implications, Cancer Microenviron., 11, 13, 10.1007/s12307-018-0211-7

Wong, 2019, Clinical significance of exosomes as potential biomarkers in cancer, World J. Clin. Cases, 7, 171, 10.12998/wjcc.v7.i2.171

Lampis, A., Ghidini, M., Ratti, M., Mirchev, M.B., Okuducu, A.F., Valeri, N., and Hahne, J.C. (2020). Circulating tumour DNAs and Non-Coding RNAs as liquid biopsies for the management of colorectal cancer patients. Gastrointest. Disord., 2.

Osumi, 2019, Clinical utility of circulating tumor DNA for colorectal cancer, Cancer Sci., 110, 1148, 10.1111/cas.13972

Mathai, R.A., Vidya, R.V.S., Reddy, B.S., Thomas, L., Udupa, K., Kolesar, J., and Rao, M. (2019). Potential utility of liquid biopsy as a diagnostic and prognostic tool for the assessment of solid tumors: Implications in the precision oncology. J. Clin. Med., 8.

Rubis, 2019, Liquid biopsies in cancer diagnosis, monitoring, and prognosis, Trends Pharmacol. Sci., 40, 172, 10.1016/j.tips.2019.01.006

Gold, 2015, Do circulating tumor cells, exosomes, and circulating tumor nucleic acids have clinical utility? A report of the association for molecular pathology, J. Mol. Diagn., 17, 209, 10.1016/j.jmoldx.2015.02.001

Ding, Y., Li, W., Wang, K., Xu, C., Hao, M., and Ding, L. (2020, July 16). Perspectives of the Application of Liquid Biopsy in Colorectal Cancer. Available online: https://www.hindawi.com/journals/bmri/2020/6843180/.

Tsai, 2018, Prospective clinical study of circulating tumor cells for colorectal cancer screening, J. Clin. Oncol., 36, 556, 10.1200/JCO.2018.36.4_suppl.556

Bork, 2015, Circulating tumour cells and outcome in non-metastatic colorectal cancer: A prospective study, Br. J. Cancer, 112, 1306, 10.1038/bjc.2015.88

Gazzaniga, 2013, Circulating tumor cells in high-risk nonmetastatic colorectal cancer, Tumour Biol., 34, 2507, 10.1007/s13277-013-0752-9

Tsai, W.-S., Chen, J.-S., Shao, H.-J., Wu, J.-C., Lai, J.-M., Lu, S.-H., Hung, T.-F., Chiu, Y.-C., You, J.-F., and Hsieh, P.-S. (2016). Circulating tumor cell count correlates with colorectal neoplasm progression and is a prognostic marker for distant metastasis in non-metastatic patients. Sci. Rep., 6.

Musella, 2015, Circulating tumor cells as a longitudinal biomarker in patients with advanced chemorefractory, RAS-BRAF wild-type colorectal cancer receiving cetuximab or panitumumab, Int. J. Cancer, 137, 1467, 10.1002/ijc.29493

Krebs, 2015, Circulating Tumor Cell Enumeration in a Phase II Trial of a Four-Drug Regimen in Advanced Colorectal Cancer, Clin. Colorectal Cancer, 14, 115.e2, 10.1016/j.clcc.2014.12.006

Tie, 2016, Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer, Sci. Transl. Med., 8, 346ra92, 10.1126/scitranslmed.aaf6219

Sun, 2018, Monitoring colorectal cancer following surgery using plasma circulating tumor DNA, Oncol. Lett., 15, 4365

Tie, 2015, Circulating tumor DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer, Ann. Oncol., 26, 1715, 10.1093/annonc/mdv177

Tie, 2018, Serial circulating tumor DNA (ctDNA) analysis as a prognostic marker and a real-time indicator of adjuvant chemotherapy (CT) efficacy in stage III colon cancer (CC), J. Clin. Oncol., 36, 3516, 10.1200/JCO.2018.36.15_suppl.3516

Grasselli, 2017, Concordance of blood- and tumor-based detection of RAS mutations to guide anti-EGFR therapy in metastatic colorectal cancer, Ann. Oncol., 28, 1294, 10.1093/annonc/mdx112

Khan, 2018, Functional imaging and circulating biomarkers of response to regorafenib in treatment-refractory metastatic colorectal cancer patients in a prospective phase II study, Gut, 67, 1484, 10.1136/gutjnl-2017-314178

Flamini, 2006, Free DNA and carcinoembryonic antigen serum levels: An important combination for diagnosis of colorectal cancer, Clin. Cancer Res., 12, 6985, 10.1158/1078-0432.CCR-06-1931

Hao, 2014, Circulating cell-free DNA in serum as a biomarker for diagnosis and prognostic prediction of colorectal cancer, Br. J. Cancer, 111, 1482, 10.1038/bjc.2014.470

Sun, J., Fei, F., Zhang, M., Li, Y., Zhang, X., Zhu, S., and Zhang, S. (2019). The role of mSEPT9 in screening, diagnosis, and recurrence monitoring of colorectal cancer. BMC Cancer, 19.

Link, 2010, Fecal microRNAs as novel biomarkers for colon cancer screening, Cancer Epidemiol. Biomark. Prev., 19, 1766, 10.1158/1055-9965.EPI-10-0027

Ya, 2017, Serum miR-129 functions as a biomarker for colorectal cancer by targeting estrogen receptor (ER) β, Pharmazie, 72, 107

He, 2018, Low-level serum miR-24-2 is associated with the progression of colorectal cancer, Cancer Biomark., 21, 261, 10.3233/CBM-170321

Wang, Y., Chen, Z., and Chen, W. (2017). Novel circulating microRNAs expression profile in colon cancer: A pilot study. Eur. J. Med. Res., 22.

Toiyama, 2014, Serum miR-200c is a novel prognostic and metastasis-predictive biomarker in patients with colorectal cancer, Ann. Surg., 259, 735, 10.1097/SLA.0b013e3182a6909d

Tang, Y., Zhao, Y., Song, X., Song, X., Niu, L., and Xie, L. (2019). Tumor-derived exosomal miRNA-320d as a biomarker for metastatic colorectal cancer. J. Clin. Lab. Anal., 33.

Koga, 2013, Fecal miR-106a is a useful marker for colorectal cancer patients with false-negative results in immunochemical fecal occult blood test, Cancer Epidemiol. Biomark. Prev., 22, 1844, 10.1158/1055-9965.EPI-13-0512

Sazanov, 2017, Plasma and saliva miR-21 expression in colorectal cancer patients, J. Appl. Genet., 58, 231, 10.1007/s13353-016-0379-9

Fu, 2018, Circulating exosomal miR-17-5p and miR-92a-3p predict pathologic stage and grade of colorectal cancer, Transl. Oncol., 11, 221, 10.1016/j.tranon.2017.12.012

Tsukamoto, 2017, Circulating exosomal MicroRNA-21 as a biomarker in each tumor stage of colorectal cancer, Oncology, 92, 360, 10.1159/000463387

Liu, 2016, Serum exosomal miR-4772-3p is a predictor of tumor recurrence in stage II and III colon cancer, Oncotarget, 7, 76250, 10.18632/oncotarget.12841

Yan, 2018, Exosomal miR-6803-5p as potential diagnostic and prognostic marker in colorectal cancer, J. Cell. Biochem., 119, 4113, 10.1002/jcb.26609

Liu, 2018, Circulating exosomal miR-27a and miR-130a act as novel diagnostic and prognostic biomarkers of colorectal cancer, Cancer Epidemiol. Biomark. Prev., 27, 746, 10.1158/1055-9965.EPI-18-0067

Peng, 2018, Downregulation of exosome-encapsulated miR-548c-5p is associated with poor prognosis in colorectal cancer, J. Cell. Biochem., 120, 1457, 10.1002/jcb.27291

Jin, 2019, A panel of serum exosomal microRNAs as predictive markers for chemoresistance in advanced colorectal cancer, Cancer Chemother. Pharmacol., 84, 315, 10.1007/s00280-019-03867-6

Yagi, 2019, Plasma exosomal microRNA-125b as a monitoring biomarker of resistance to mFOLFOX6-based chemotherapy in advanced and recurrent colorectal cancer patients, Mol. Clin. Oncol., 11, 416

Wang, 2018, Long non-coding RNA H19 confers 5-Fu resistance in colorectal cancer by promoting SIRT1-mediated autophagy, Cell Death Dis., 9, 1, 10.1038/s41419-018-1187-4

Li, 2017, MEG3 is a prognostic factor for CRC and promotes chemosensitivity by enhancing oxaliplatin-induced cell apoptosis, Oncol. Rep., 38, 1383, 10.3892/or.2017.5828

Sun, 2019, The identification of CRNDE, H19, UCA1 and HOTAIR as the key lncRNAs involved in oxaliplatin or irinotecan resistance in the chemotherapy of colorectal cancer based on integrative bioinformatics analysis, Mol. Med. Rep., 20, 3583

Tang, 2019, LncRNA GLCC1 promotes colorectal carcinogenesis and glucose metabolism by stabilizing c-Myc, Nat. Commun., 10, 3499, 10.1038/s41467-019-11447-8

Liu, 2016, Exosomal long noncoding RNA CRNDE-h as a novel serum-based biomarker for diagnosis and prognosis of colorectal cancer, Oncotarget, 7, 85551, 10.18632/oncotarget.13465

Liang, 2019, LncRNA RPPH1 promotes colorectal cancer metastasis by interacting with TUBB3 and by promoting exosomes-mediated macrophage M2 polarization, Cell Death Dis., 10, 829, 10.1038/s41419-019-2077-0

Barkun, 2018, Systematic review of colorectal cancer screening guidelines for average-risk adults: Summarizing the current global recommendations, World J. Gastroenterol., 24, 124, 10.3748/wjg.v24.i1.124

Marcuello, 2019, Circulating biomarkers for early detection and clinical management of colorectal cancer, Mol. Asp. Med., 69, 107, 10.1016/j.mam.2019.06.002

Cima, 2016, Tumor-derived circulating endothelial cell clusters in colorectal cancer, Sci. Transl. Med., 8, 345ra89, 10.1126/scitranslmed.aad7369

Bi, 2020, Circulating tumor DNA in colorectal cancer: Opportunities and challenges, Am. J. Transl. Res., 12, 1044

Wang, 2018, Circulating cell free DNA as the diagnostic marker for colorectal cancer: A systematic review and meta-analysis, Oncotarget, 9, 24514, 10.18632/oncotarget.25314

Luo, H., Zhao, Q., Wei, W., Zheng, L., Yi, S., Li, G., Wang, W., Sheng, H., Pu, H., and Mo, H. (2020). Circulating tumor DNA methylation profiles enable early diagnosis, prognosis prediction, and screening for colorectal cancer. Sci. Transl. Med., 12.

Nian, 2017, Diagnostic accuracy of methylated SEPT9 for blood-based colorectal cancer detection: A systematic review and meta-analysis, Clin. Transl. Gastroenterol., 8, e216, 10.1038/ctg.2016.66

Cohen, 2018, Detection and localization of surgically resectable cancers with a multi-analyte blood test, Science, 359, 926, 10.1126/science.aar3247

Hayder, 2018, Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation, Front. Endocrinol. Lausanne, 9, 402, 10.3389/fendo.2018.00402

Ragusa, 2012, Specific alterations of the microRNA transcriptome and global network structure in colorectal cancer after treatment with MAPK/ERK inhibitors, J. Mol. Med., 90, 1421, 10.1007/s00109-012-0918-8

Chen, 2019, Emerging microRNA biomarkers for colorectal cancer diagnosis and prognosis, Open Biol., 9, 180212, 10.1098/rsob.180212

Majem, 2018, Human salivary microRNAs in Cancer, J. Cancer, 9, 638, 10.7150/jca.21180

Heymann, 2019, Circulating tumor cells as a tool for assessing tumor heterogeneity, Theranostics, 9, 4580, 10.7150/thno.34337

Huang, X., Gao, P., Song, Y., Sun, J., Chen, X., Zhao, J., Liu, J., Xu, H., and Wang, Z. (2014). Relationship between circulating tumor cells and tumor response in colorectal cancer patients treated with chemotherapy: A meta-analysis. BMC Cancer, 14.

Bettegowda, 2014, Detection of Circulating Tumor DNA in Early- and Late-Stage Human Malignancies, Sci. Transl. Med., 6, 224ra24, 10.1126/scitranslmed.3007094

Dasari, A., Morris, V.K., Allegra, C.J., Atreya, C., Benson, A.B., Boland, P., Chung, K., Copur, M.S., Corcoran, R.B., and Deming, D.A. (2020). ctDNA applications and integration in colorectal cancer: An NCI Colon and Rectal–Anal Task Forces whitepaper. Nat. Rev. Clin. Oncol., 1–14.

Osumi, H., Shinozaki, E., and Yamaguchi, K. (2020). Circulating tumor DNA as a novel biomarker optimizing chemotherapy for colorectal cancer. Cancers, 12.

Siravegna, 2015, Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients, Nat. Med., 21, 795, 10.1038/nm.3870

Khan, 2018, longitudinal liquid biopsy and mathematical modeling of clonal evolution forecast time to treatment failure in the PROSPECT-C Phase II colorectal cancer clinical trial, Cancer Discov., 8, 1270, 10.1158/2159-8290.CD-17-0891

Siravegna, 2019, Plasma HER2 (ERBB2) Copy Number Predicts Response to HER2-targeted Therapy in Metastatic Colorectal Cancer, Clin. Cancer Res., 25, 3046, 10.1158/1078-0432.CCR-18-3389

To, 2018, MicroRNAs in the prognosis and therapy of colorectal cancer: From bench to bedside, World J. Gastroenterol., 24, 2949, 10.3748/wjg.v24.i27.2949

Zhang, 2017, Prognostic value of high-expression of miR-17-92 cluster in various tumors: Evidence from a meta-analysis, Sci. Rep., 7, 8375, 10.1038/s41598-017-08349-4

Ma, 2018, miR-1290 Contributes to Colorectal Cancer Cell Proliferation by Targeting INPP4B, Oncol. Res., 26, 1167, 10.3727/096504017X15051741798389

Falzone, 2018, Integrated analysis of colorectal cancer microRNA datasets: Identification of microRNAs associated with tumor development, Aging Albany N. Y, 10, 1000, 10.18632/aging.101444

Ragusa, 2010, Specific alterations of microRNA transcriptome and global network structure in colorectal carcinoma after cetuximab treatment, Mol. Cancer Ther., 9, 3396, 10.1158/1535-7163.MCT-10-0137

Nagano, 2011, No-nonsense functions for long noncoding RNAs, Cell, 145, 178, 10.1016/j.cell.2011.03.014

Zhang, X., Wang, W., Zhu, W., Dong, J., Cheng, Y., Yin, Z., and Shen, F. (2019). Mechanisms and Functions of Long Non-Coding RNAs at Multiple Regulatory Levels. Int. J. Mol. Sci., 20.

Xie, 2015, Long non-coding RNAs in colorectal cancer, Oncotarget, 7, 5226, 10.18632/oncotarget.6446

Siddiqui, 2019, Roles of long non-coding RNAs in colorectal cancer tumorigenesis: A Review, Mol. Clin. Oncol., 11, 167

He, Q., Long, J., Yin, Y., Li, Y., Lei, X., Li, Z., and Zhu, W. (2020). Emerging Roles of lncRNAs in the Formation and Progression of Colorectal Cancer. Front. Oncol., 9.

Han, 2015, Long noncoding RNAs: Novel players in colorectal cancer, Cancer Lett., 361, 13, 10.1016/j.canlet.2015.03.002

Luo, 2017, Long non-coding RNAs: A rising biotarget in colorectal cancer, Oncotarget, 8, 22187, 10.18632/oncotarget.14728

Valli, 2019, Adaptation to HIF1α Deletion in Hypoxic Cancer Cells by Upregulation of GLUT14 and Creatine Metabolism, Mol. Cancer Res., 17, 1531, 10.1158/1541-7786.MCR-18-0315

Barbagallo, 2018, LncRNA UCA1, Upregulated in CRC Biopsies and Downregulated in Serum Exosomes, Controls mRNA Expression by RNA-RNA Interactions, Mol. Ther. Nucleic Acids, 12, 229, 10.1016/j.omtn.2018.05.009

Beije, 2019, Incorporating liquid biopsies into treatment decision-making: Obstacles and possibilities, Drug Discov. Today, 24, 1715, 10.1016/j.drudis.2019.05.028

Kolenčík, D., Shishido, S.N., Pitule, P., Mason, J., Hicks, J., and Kuhn, P. (2020). Liquid Biopsy in Colorectal Carcinoma: Clinical Applications and Challenges. Cancers, 12.

Leers, 2020, Circulating tumor DNA and their added value in molecular oncology, Clin. Chem. Lab. Med. CCLM, 58, 152, 10.1515/cclm-2019-0436

Hong, 2013, Detecting circulating tumor cells: Current challenges and new trends, Theranostics, 3, 377, 10.7150/thno.5195

Mamdouhi, 2019, Fugitives on the run: Circulating tumor cells (CTCs) in metastatic diseases, Cancer Metastasis Rev., 38, 297, 10.1007/s10555-019-09795-4

Millner, 2013, Circulating tumor cells: A review of present methods and the need to identify heterogeneous phenotypes, Ann. Clin. Lab. Sci., 43, 295

Barrière, G., Tartary, M., and Rigaud, M. (2012). Epithelial mesenchymal transition: A new insight into the detection of circulating tumor cells. ISRN Oncol., 2012.

Castro-Giner, F., Gkountela, S., Donato, C., Alborelli, I., Quagliata, L., Ng, C.K.Y., Piscuoglio, S., and Aceto, N. (2018). Cancer diagnosis using a liquid biopsy: Challenges and expectations. Diagnostics, 8.

Elazezy, 2018, Techniques of using circulating tumor DNA as a liquid biopsy component in cancer management, Comput. Struct. Biotechnol. J., 16, 370, 10.1016/j.csbj.2018.10.002

Fontanges, 2018, Clinical application of targeted next-generation sequencing for colorectal cancer patients: A multicentric Belgian experience, Oncotarget, 9, 20761, 10.18632/oncotarget.25099

Qin, 2019, Next-generation sequencing and its clinical application, Cancer Biol. Med., 16, 4, 10.20892/j.issn.2095-3941.2018.0055

Narayan, 2012, Ultrasensitive measurement of hotspot mutations in tumor DNA in blood using error-suppressed multiplexed deep sequencing, Cancer Res., 72, 3492, 10.1158/0008-5472.CAN-11-4037

Couraud, 2014, Noninvasive diagnosis of actionable mutations by deep sequencing of circulating free DNA in lung cancer from never-smokers: A proof-of-concept study from BioCAST/IFCT-1002, Clin. Cancer Res., 20, 4613, 10.1158/1078-0432.CCR-13-3063

Fontanges, Q., De Mendonca, R., Salmon, I., Le Mercier, M., and D’Haene, N. (2016). Clinical application of targeted next generation sequencing for colorectal cancers. Int. J. Mol. Sci., 17.

Grada, 2013, Next-generation sequencing: Methodology and application, J. Investig. Dermatol., 133, e11, 10.1038/jid.2013.248

Ng, 2019, Genomics and metagenomics of colorectal cancer, J. Gastrointest. Oncol., 10, 1164, 10.21037/jgo.2019.06.04

Kim, 2011, Genetic-based biomarkers and next-generation sequencing: The future of personalized care in colorectal cancer, Pers. Med., 8, 331, 10.2217/pme.11.16

Wadapurkar, 2018, Computational analysis of next generation sequencing data and its applications in clinical oncology, Inform. Med. Unlocked, 11, 75, 10.1016/j.imu.2018.05.003

Rachiglio, 2016, Limits and potential of targeted sequencing analysis of liquid biopsy in patients with lung and colon carcinoma, Oncotarget, 7, 66595, 10.18632/oncotarget.10704

Mastroiaco, 2017, Next-generation sequencing: Recent applications to the analysis of colorectal cancer, J. Transl. Med., 15, 246, 10.1186/s12967-017-1353-y

Soda, 2019, Advanced liquid biopsy technologies for circulating biomarker detection, J. Mater. Chem. B, 7, 6670, 10.1039/C9TB01490J

Risberg, 2018, Effects of Collection and Processing Procedures on Plasma Circulating Cell-Free DNA from Cancer Patients, J. Mol. Diagn., 20, 883, 10.1016/j.jmoldx.2018.07.005

Hauch, 2018, Liquid Biopsy Preservation Solutions for Standardized Pre-Analytical Workflows—Venous Whole Blood and Plasma, Curr. Pathobiol. Rep., 6, 275, 10.1007/s40139-018-0180-z

Harouaka, 2014, Circulating tumor cells: Advances in isolation and analysis, and challenges for clinical applications, Pharmacol. Ther., 141, 209, 10.1016/j.pharmthera.2013.10.004

Tamkovich, 2006, Circulating DNA and DNase activity in human blood, Ann. N. Y. Acad. Sci., 1075, 191, 10.1196/annals.1368.026

Siravegna, 2019, How liquid biopsies can change clinical practice in oncology, Ann. Oncol., 30, 1580, 10.1093/annonc/mdz227

Heitzer, 2019, Current and future perspectives of liquid biopsies in genomics-driven oncology, Nat. Rev. Genet., 20, 71, 10.1038/s41576-018-0071-5

Haselmann, 2018, Results of the first external quality assessment scheme (EQA) for isolation and analysis of circulating tumour DNA (ctDNA), Clin. Chem. Lab. Med., 56, 220, 10.1515/cclm-2017-0283

Vivancos, A., Aranda, E., Benavides, M., Élez, E., Gómez-España, M.A., Toledano, M., Alvarez, M., Parrado, M.R.C., García-Barberán, V., and Diaz-Rubio, E. (2019). Comparison of the Clinical Sensitivity of the Idylla Platform and the OncoBEAM RAS CRC Assay for KRAS Mutation Detection in Liquid Biopsy Samples. Sci. Rep., 9.