Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

Circulating Tumor Cells and Cell-free Nucleic Acids as Biomarkers in Colorectal Cancer

Author(s): Ghazaleh Pourali, Ghazaleh Khalili-Tanha, Elham Nazari, Mina Maftooh, Mohammadreza Nassiri, Seyed Mahdi Hassanian, Majid Ghayour Mobarhan, Majid Khazaei*, Gordon Ferns and Amir Avan*

Volume 29, Issue 10, 2023

Published on: 04 April, 2023

Page: [748 - 765] Pages: 18

DOI: 10.2174/1381612829666230308102611

Price: $65

Abstract

Colorectal cancer (CRC) is currently the second most prevalent cancer diagnosed in women and the third most common kind of cancer in men. Despite tremendous efforts and advancements in diagnostic approaches and treatment options, the mortality rate of CRC accounts for around one million each year globally. The five-year survival rate of CRC is reported to be approximately 14 percent for patients diagnosed at an advanced stage. Due to its significant associated mortality and morbidity, diagnostic tools to identify the disease at its early stages are urgently required. Early diagnosis may lead to better outcomes. The gold standard approach for CRC diagnosis is colonoscopy with biopsy. However, it is an invasive process with a risk of complications and discomfort for the patient. Moreover, it is usually performed in symptomatic or high-risk individuals and therefore, asymptomatic patients might be missed. Thus, alternative non-invasive diagnostic techniques are required to improve CRC outcomes. The new era of personalized medicine is identifying novel biomarkers associated with overall survival and clinical outcomes. Recently, liquid biopsy, a minimally invasive analysis of body fluid biomarkers, has gained attention for diagnosis, evaluation of prognosis, and follow-up of patients with CRC. Several previous studies have demonstrated that this novel approach allows for better understanding of CRC tumor biology and leads to an improvement in clinical outcomes. Here, we explain the enrichment and detection methods of circulating biomarkers, including CTCs, ctDNA, miRNA, lncRNA, and circRNA. Furthermore, we provide an overview on their clinical potential as diagnostic, prognostic, and predictive biomarkers for CRC.

Keywords: Circulating biomarkers, colorectal cancer, early detection, clinical management, diagnostic, predictive, prognosis, epigenetic, CTC, ctDNA.

[1]
Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mor-tality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021; 71(3): 209-49.
[http://dx.doi.org/10.3322/caac.21660] [PMID: 33538338]
[2]
Binefa G, Rodríguez-Moranta F, Teule A, Medina-Hayas M. Colorectal cancer: From prevention to personalized medicine. World J Gastroenterol 2014; 20(22): 6786-808.
[http://dx.doi.org/10.3748/wjg.v20.i22.6786] [PMID: 24944469]
[3]
Zaharie F, Muresan MS, Petrushev B, et al. Exosome-carried microRNA-375 inhibits cell progression and dissemination via Bcl-2 blocking in colon cancer. J Gastrointestin Liver Dis 2015; 24(4): 435-43.
[http://dx.doi.org/10.15403/jgld.2014.1121.244.375] [PMID: 26697569]
[4]
Świderska M, Choromańska B, Dąbrowska E, et al. Review the diagnostics of colorectal cancer. Contemp Oncol 2014; 1(1): 1-6.
[http://dx.doi.org/10.5114/wo.2013.39995] [PMID: 24876814]
[5]
Reumkens A, Rondagh EJ, Bakker MC, Winkens B, Masclee AA, Sanduleanu S. Post-colonoscopy complications: A systematic review, time trends, and meta-analysis of population-based studies. Am J Gastroenterol 2016; 111(8): 1092-101.
[6]
Vega P, Valentín F, Cubiella J. Colorectal cancer diagnosis: Pitfalls and opportunities. World J Gastrointest Oncol 2015; 7(12): 422-33.
[http://dx.doi.org/10.4251/wjgo.v7.i12.422] [PMID: 26690833]
[7]
Serrano MJ, Garrido-Navas MC. Diaz Mochon JJ, et al. Precision prevention and cancer intercep-tion: The new challenges of liquid biopsy. Cancer Discov 2020; 10(11): 1635-44.
[http://dx.doi.org/10.1158/2159-8290.CD-20-0466] [PMID: 33037026]
[8]
Wan JCM, Massie C, Garcia-Corbacho J, et al. Liquid biopsies come of age: Towards implementation of circulating tumour DNA. Nat Rev Cancer 2017; 17(4): 223-38.
[http://dx.doi.org/10.1038/nrc.2017.7] [PMID: 28233803]
[9]
Marrugo-Ramírez J, Mir M, Samitier J. Blood-based cancer biomarkers in liquid biopsy: A promising non-invasive alternative to tissue biopsy. Int J Mol Sci 2018; 19(10): 2877.
[http://dx.doi.org/10.3390/ijms19102877] [PMID: 30248975]
[10]
Yu M, Stott S, Toner M, Maheswaran S, Haber DA. Circulating tumor cells: Approaches to isola-tion and characterization. J Cell Biol 2011; 192(3): 373-82.
[http://dx.doi.org/10.1083/jcb.201010021] [PMID: 21300848]
[11]
Steinert G, Schölch S, Koch M, Weitz J. Biology and significance of circulating and disseminated tu-mour cells in colorectal cancer. Langenbecks Arch Surg 2012; 397(4): 535-42.
[http://dx.doi.org/10.1007/s00423-012-0917-9] [PMID: 22350614]
[12]
Hyun KA, Kim J, Gwak H, Jung HI. Isolation and enrichment of circulating biomarkers for cancer screen-ing, detection, and diagnostics. Analyst 2016; 141(2): 382-92.
[http://dx.doi.org/10.1039/C5AN01762A] [PMID: 26588824]
[13]
Pei H, Li L, Han Z, Wang Y, Tang B. Recent advances in microfluidic technologies for circulating tumor cells: Enrichment, single-cell analysis, and liquid biopsy for clinical applications. Lab Chip 2020; 20(21): 3854-75.
[http://dx.doi.org/10.1039/D0LC00577K] [PMID: 33107879]
[14]
Rushton AJ, Nteliopoulos G, Shaw JA, Coombes RC. A review of circulating tumour cell en-richment technologies. Cancers 2021; 13(5): 970.
[http://dx.doi.org/10.3390/cancers13050970] [PMID: 33652649]
[15]
Ligthart ST, Coumans FAW, Attard G. Mu-lick Cassidy A, de Bono JS, Terstappen LWMM. Unbiased and automated identification of a cir-culating tumour cell definition that associates with overall survival. PLoS One 2011; 6(11)e27419
[http://dx.doi.org/10.1371/journal.pone.0027419] [PMID: 22087312]
[16]
Swennenhuis JF, van Dalum G, Zeune LL, Terstappen LWMM. Improving the CellSearch® sys-tem. Expert Rev Mol Diagn 2016; 16(12): 1291-305.
[http://dx.doi.org/10.1080/14737159.2016.1255144] [PMID: 27797592]
[17]
Kölbl A, Jeschke U, Andergassen U. The sig-nificance of epithelial-to-mesenchymal transition for circulat-ing tumor cells. Int J Mol Sci 2016; 17(8): 1308.
[http://dx.doi.org/10.3390/ijms17081308] [PMID: 27529216]
[18]
Liu Z, Fusi A, Klopocki E, et al. Negative en-richment by immunomagnetic nanobeads for unbiased char-acterization of circulating tumor cells from peripheral blood of cancer patients. J Transl Med 2011; 9(1): 70.
[http://dx.doi.org/10.1186/1479-5876-9-70] [PMID: 21595914]
[19]
Dan Z, Daxiang C. Advances in isolation and detec-tion of circulating tumor cells based on microfluidics. Cancer Biol Med 2018; 15(4): 335-53.
[http://dx.doi.org/10.20892/j.issn.2095-3941.2018.0256] [PMID: 30766747]
[20]
Desitter I, Guerrouahen BS, Benali-Furet N, et al. A new device for rapid isolation by size and charac-terization of rare circulating tumor cells. Anticancer Res 2011; 31(2): 427-41.
[PMID: 21378321]
[21]
Rosenberg R, Gertler R, Friederichs J, et al. Comparison of two density gradient centrifugation systems for the enrichment of disseminated tumor cells in blood. Cytometry 2002; 49(4): 150-8.
[http://dx.doi.org/10.1002/cyto.10161] [PMID: 12454978]
[22]
Campton DE, Ramirez AB, Nordberg JJ, et al. High-recovery visual identification and single-cell re-trieval of circulating tumor cells for genomic analysis using a dual-technology platform integrated with automated immuno-fluorescence staining. BMC Cancer 2015; 15(1): 360.
[http://dx.doi.org/10.1186/s12885-015-1383-x] [PMID: 25944336]
[23]
Gupta V, Jafferji I, Garza M, et al. ApoStream ™, a new dielectrophoretic device for antibody independent isolation and recovery of viable cancer cells from blood. Biomicrofluidics 2012; 6(2)024133
[http://dx.doi.org/10.1063/1.4731647] [PMID: 23805171]
[24]
Srovnal J, Skalicky P, Rehulkova A, et al. Abstract 1949: Circulating tumor cells presence shorten the cancer specific survival in colorectal cancer: 10 years follow-up study. Cancer Res 2022; 82(12) (Supplement): 1949-9.
[http://dx.doi.org/10.1158/1538-7445.AM2022-1949]
[25]
Fehm TN, Meier-Stiegen F, Driemel C, et al. Diagnostic leukapheresis for CTC analysis in breast cancer patients: CTC frequency, clinical experiences and recom-mendations for standardized reporting. Cytometry A 2018; 93(12): 1213-9.
[http://dx.doi.org/10.1002/cyto.a.23669] [PMID: 30551262]
[26]
Scherag FD, Niestroj-Pahl R, Krusekopf S, Lücke K, Brandstetter T, Rühe J. Highly selec-tive capture surfaces on medical wires for fishing tumor cells in whole blood. Anal Chem 2017; 89(3): 1846-54.
[http://dx.doi.org/10.1021/acs.analchem.6b04219] [PMID: 28208267]
[27]
Donato C, Szczerba BM, Scheidmann MC, Castro-Giner F, Aceto N. Micromanipulation of circulating tumor cells for downstream molecular analysis and metastatic potential assessment. J Vis Exp 2019; (147): e59677
[PMID: 31157780]
[28]
Nelep C, Eberhardt J. Automated rare single cell picking with the ALS cellcelector™. Cytometry A 2018; 93(12): 1267-70.
[http://dx.doi.org/10.1002/cyto.a.23568] [PMID: 30184320]
[29]
Fontana F, Rapone C, Bregola G, et al. Isola-tion and genetic analysis of pure cells from forensic biologi-cal mixtures: The precision of a digital approach. Forensic Sci Int Genet 2017; 29: 225-41.
[http://dx.doi.org/10.1016/j.fsigen.2017.04.023] [PMID: 28511094]
[30]
Snyder MW, Kircher M, Hill AJ, Daza RM, Shendure J. Cell-free DNA comprises an in vivo nucleo-some footprint that informs its tissues-of-origin. Cell 2016; 164(1-2): 57-68.
[http://dx.doi.org/10.1016/j.cell.2015.11.050] [PMID: 26771485]
[31]
Mayrhofer M, De Laere B, Whitington T, et al. Cell-free DNA profiling of metastatic prostate cancer re-veals microsatellite instability, structural rearrangements and clonal hematopoiesis. Genome Med 2018; 10(1): 85.
[http://dx.doi.org/10.1186/s13073-018-0595-5] [PMID: 30458854]
[32]
Elazezy M, Joosse SA. Techniques of using circulat-ing tumor DNA as a liquid biopsy component in cancer man-agement. Comput Struct Biotechnol J 2018; 16: 370-8.
[http://dx.doi.org/10.1016/j.csbj.2018.10.002]
[33]
Keller L, Belloum Y, Wikman H, Pantel K. Clinical relevance of blood-based ctDNA analysis: Mutation detection and beyond. Br J Cancer 2021; 124(2): 345-58.
[http://dx.doi.org/10.1038/s41416-020-01047-5] [PMID: 32968207]
[34]
Chan HT, Chin YM, Nakamura Y, Low SK. Clonal hematopoiesis in liquid biopsy: From biological noise to valuable clinical implications. Cancers 2020; 12(8): 2277.
[http://dx.doi.org/10.3390/cancers12082277] [PMID: 32823942]
[35]
Wang H, Jiang J, Mostert B, et al. Allele-specific, non-extendable primer blocker PCR (AS-NEPB-PCR) for DNA mutation detection in cancer. J Mol Diagn 2013; 15(1): 62-9.
[http://dx.doi.org/10.1016/j.jmoldx.2012.08.007] [PMID: 23159590]
[36]
Burch JA, Soares-Weiser K, St John DJB, et al. Diagnostic accuracy of faecal occult blood tests used in screening for colorectal cancer: A systematic review. J Med Screen 2007; 14(3): 132-7.
[http://dx.doi.org/10.1258/096914107782066220] [PMID: 17925085]
[37]
Glenn TC. Field guide to next-generation DNA sequenc-ers. Mol Ecol Resour 2011; 11(5): 759-69.
[http://dx.doi.org/10.1111/j.1755-0998.2011.03024.x] [PMID: 21592312]
[38]
Veldore V, Choughule A, Routhu T, et al. Validation of liquid biopsy: Plasma cell-free DNA testing in clinical management of advanced non-small cell lung cancer. Lung Cancer 2018; 9: 1-11.
[http://dx.doi.org/10.2147/LCTT.S147841] [PMID: 29379323]
[39]
Kim HR, Lee SY, Hyun DS, et al. Detection of EGFR mutations in circulating free DNA by PNA-mediated PCR clamping. J Exp Clin Cancer Res 2013; 32(1): 50.
[http://dx.doi.org/10.1186/1756-9966-32-50] [PMID: 23927790]
[40]
Freidin MB, Freydina DV, Leung M. Mon-tero Fernandez A, Nicholson AG, Lim E. Circulating tumor DNA outperforms circulating tumor cells for KRAS mutation detection in thoracic malignancies. Clin Chem 2015; 61(10): 1299-304.
[http://dx.doi.org/10.1373/clinchem.2015.242453] [PMID: 26272233]
[41]
Sefrioui D, Sarafan-Vasseur N, Beaussire L, et al. Clinical value of chip-based digital-PCR platform for the detection of circulating DNA in metastatic colorectal can-cer. Dig Liver Dis 2015; 47(10): 884-90.
[http://dx.doi.org/10.1016/j.dld.2015.05.023] [PMID: 26160500]
[42]
Li M, Diehl F, Dressman D, Vogelstein B, Kinzler KW. BEAMing up for detection and quantifica-tion of rare sequence variants. Nat Methods 2006; 3(2): 95-7.
[http://dx.doi.org/10.1038/nmeth850] [PMID: 16432518]
[43]
Wee EJH, Wang Y, Tsao SCH, Trau M. Sim-ple, sensitive and accurate multiplex detection of clinically important melanoma DNA mutations in circulating tumour DNA with SERS nanotags. Theranostics 2016; 6(10): 1506-13.
[http://dx.doi.org/10.7150/thno.15871] [PMID: 27446486]
[44]
Mosko MJ, Nakorchevsky AA, Flores E, et al. Ultrasensitive detection of multiplexed somatic muta-tions using MALDI-TOF mass spectrometry. J Mol Diagn 2016; 18(1): 23-31.
[http://dx.doi.org/10.1016/j.jmoldx.2015.08.001] [PMID: 26596526]
[45]
Narayan A, Carriero NJ, Gettinger SN, et al. Ultrasensitive measurement of hotspot mutations in tumor DNA in blood using error-suppressed multiplexed deep se-quencing. Cancer Res 2012; 72(14): 3492-8.
[http://dx.doi.org/10.1158/0008-5472.CAN-11-4037] [PMID: 22581825]
[46]
Gale D, Lawson ARJ, Howarth K, et al. De-velopment of a highly sensitive liquid biopsy platform to de-tect clinically-relevant cancer mutations at low allele fractions in cell-free DNA. PLoS One 2018; 13(3)e0194630
[http://dx.doi.org/10.1371/journal.pone.0194630] [PMID: 29547634]
[47]
Danese E, Montagnana M, Minicozzi AM, et al. Real-time polymerase chain reaction quantification of free DNA in serum of patients with polyps and colorectal cancers. Clin Chem Lab Med 2010; 48(11): 1665-8.
[http://dx.doi.org/10.1515/CCLM.2010.301] [PMID: 20704532]
[48]
Kinde I, Wu J, Papadopoulos N, Kinzler KW, Vogelstein B. Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci USA 2011; 108(23): 9530-5.
[http://dx.doi.org/10.1073/pnas.1105422108] [PMID: 21586637]
[49]
Belic J. mFast-SeqS as a monitoring and pre-screening tool for tumor-specific aneuploidy in plasma DNA. In: Circulating nucleic acids in serum and plasma - CNAPS IX advances in experimental medicine and biology. Cham: Springer 2016; 924: pp. 147-55.
[http://dx.doi.org/10.1007/978-3-319-42044-8_28]
[50]
Newman AM, Bratman SV, To J, et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med 2014; 20(5): 548-54.
[http://dx.doi.org/10.1038/nm.3519] [PMID: 24705333]
[51]
Nishida N, Yamashita S, Mimori K, et al. MicroRNA-10b is a prognostic indicator in colorectal cancer and confers resistance to the chemotherapeutic agent 5-fluorouracil in colorectal cancer cells. Ann Surg Oncol 2012; 19(9): 3065-71.
[http://dx.doi.org/10.1245/s10434-012-2246-1] [PMID: 22322955]
[52]
Newman AM, Lovejoy AF, Klass DM, et al. Integrated digital error suppression for improved detection of circulating tumor DNA. Nat Biotechnol 2016; 34(5): 547-55.
[http://dx.doi.org/10.1038/nbt.3520] [PMID: 27018799]
[53]
Abbosh C, Birkbak NJ, Wilson GA, et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 2017; 545(7655): 446-51.
[http://dx.doi.org/10.1038/nature22364] [PMID: 28445469]
[54]
Paweletz CP, Sacher AG, Raymond CK, et al. Bias-corrected targeted next-generation sequencing for rapid, multiplexed detection of actionable alterations in cell-free DNA from advanced lung cancer patients. Clin Cancer Res 2016; 22(4): 915-22.
[http://dx.doi.org/10.1158/1078-0432.CCR-15-1627-T] [PMID: 26459174]
[55]
Wen L, Li J, Guo H, et al. Genome-scale detection of hypermethylated CpG islands in circulating cell-free DNA of hepatocellular carcinoma patients. Cell Res 2015; 25(11): 1250-64.
[http://dx.doi.org/10.1038/cr.2015.126] [PMID: 26516143]
[56]
Köhn L, Johansson M, Grankvist K, Nilsson J. Liquid biopsies in lung cancer-time to implement research technologies in routine care? Ann Transl Med 2017; 5(13): 278.
[http://dx.doi.org/10.21037/atm.2017.04.12] [PMID: 28758104]
[57]
Wei L, Wang X, Lv L, Zheng Y, Zhang N, Yang M. The emerging role of noncoding RNAs in colo-rectal cancer chemoresistance. Cell Oncol 2019; 42(6): 757-68.
[http://dx.doi.org/10.1007/s13402-019-00466-8] [PMID: 31359293]
[58]
Wang L, Duan W, Yan S, Xie Y, Wang C. Circulating long non-coding RNA colon cancer-associated transcript 2 protected by exosome as a potential biomarker for colorectal cancer. Biomed Pharmacother 2019; 113108758
[http://dx.doi.org/10.1016/j.biopha.2019.108758] [PMID: 30877883]
[59]
Lin S, Gregory RI. MicroRNA biogenesis pathways in cancer. Nat Rev Cancer 2015; 15(6): 321-33.
[http://dx.doi.org/10.1038/nrc3932] [PMID: 25998712]
[60]
Salmaninejad A, Pourali G, Shahini A, Da-rabi H, Azhdari S. MicroRNA and exosome in retinal-related diseases: Their roles in the pathogenesis and diagno-sis. Comb Chem High Throughput Screen 2022; 25(2): 211-28.
[http://dx.doi.org/10.2174/1386207323999201230205435] [PMID: 33390101]
[61]
Suzuki HI, Katsura A, Matsuyama H, Miya-zono K. MicroRNA regulons in tumor microenvironment. Oncogene 2015; 34(24): 3085-94.
[http://dx.doi.org/10.1038/onc.2014.254] [PMID: 25132266]
[62]
Samami E. The potential diagnostic and prognostic value of circulating MicroRNAs in the assessment of patients with prostate cancer: Rational and progress. 2021; 11: 5977.
[63]
Slattery ML, Herrick JS, Mullany LE, et al. The co-regulatory networks of tumor suppressor genes, on-cogenes, and miRNAs in colorectal cancer. Genes Chromosomes Cancer 2017; 56(11): 769-87.
[http://dx.doi.org/10.1002/gcc.22481] [PMID: 28675510]
[64]
Kosaka N, Iguchi H, Ochiya T. Circulating mi-croRNA in body fluid: A new potential biomarker for cancer diagnosis and prognosis. Cancer Sci 2010; 101(10): 2087-92.
[http://dx.doi.org/10.1111/j.1349-7006.2010.01650.x] [PMID: 20624164]
[65]
Redova M, Sana J, Slaby O. Circulating miRNAs as new blood-based biomarkers for solid cancers. Future Oncol 2013; 9(3): 387-402.
[http://dx.doi.org/10.2217/fon.12.192] [PMID: 23469974]
[66]
Yang G, Lu X, Yuan L. LncRNA: A link between RNA and cancer. Biochim Biophys Acta Gene Regul Mech 2014; 1839(11): 1097-109.
[67]
Jiang M-C, Ni J-J, Cui W-Y, Wang B-Y, Zhuo W. Emerging roles of lncRNA in cancer and thera-peutic opportunities. Am J Cancer Res 2019; 9(7): 1354-66.
[PMID: 31392074]
[68]
Hamidi AA, Khalili-Tanha G. Nasrpour Na-vaei Z, Moghbeli M. Long non-coding RNAs as the critical regulators of epithelial mesenchymal transition in colorectal tumor cells: An overview. Cancer Cell Int 2022; 22(1): 71.
[http://dx.doi.org/10.1186/s12935-022-02501-5] [PMID: 35144601]
[69]
Yu C-Y, Kuo H-C. The emerging roles and functions of circular RNAs and their generation. J Biomed Sci 2019; 26(1): 29.
[http://dx.doi.org/10.1186/s12929-019-0523-z]
[70]
Memczak S, Papavasileiou P, Peters O, Ra-jewsky N. Identification and characterization of circular RNAs as a new class of putative biomarkers in human blood. PLoS One 2015; 10(10)e0141214
[http://dx.doi.org/10.1371/journal.pone.0141214] [PMID: 26485708]
[71]
Wang Y, Liu J, Ma J, et al. Exosomal circRNAs: Biogenesis, effect and application in human diseases. Mol Cancer 2019; 18(1): 116.
[http://dx.doi.org/10.1186/s12943-019-1041-z] [PMID: 31277663]
[72]
Bankó P, Lee SY, Nagygyörgy V, et al. Technologies for circulating tumor cell separation from whole blood. J Hematol Oncol 2019; 12(1): 48.
[http://dx.doi.org/10.1186/s13045-019-0735-4] [PMID: 31088479]
[73]
Kamel F, Eltarhoni K, Nisar P, Soloviev M. Colorectal cancer diagnosis: The obstacles we face in deter-mining a non-invasive test and current advances in bi-omarker detection. Cancers (Basel) 2022; 14(8): 1889.
[http://dx.doi.org/10.3390/cancers14081889] [PMID: 35454792]
[74]
Engell HC. Cancer cells in the circulating blood; a clini-cal study on the occurrence of cancer cells in the peripheral blood and in venous blood draining the tumour area at op-eration. Ugeskr Laeger 1955; 117(25): 822-3.
[PMID: 13247497]
[75]
Pruitt JC, Hilberg AW, Kaiser RF. Malignant cells in peripheral blood. N Engl J Med 1958; 259(24): 1161-4.
[http://dx.doi.org/10.1056/NEJM195812112592404] [PMID: 13613483]
[76]
Roberts S, Jonasson O, Long L, McGrath R, McGrew EA, Cole WH. Clinical significance of cancer cells in the circulating blood: Two to five-year surviv-al. Ann Surg 1961; 154(3): 362-71.
[http://dx.doi.org/10.1097/00000658-196109000-00006] [PMID: 13742183]
[77]
Chen CJ, Sung WW, Chen HC, et al. Early assessment of colorectal cancer by quantifying circulating tumor cells in peripheral blood: ECT2 in diagnosis of colo-rectal cancer. Int J Mol Sci 2017; 18(4): 743.
[http://dx.doi.org/10.3390/ijms18040743] [PMID: 28362321]
[78]
Yang Y, Li J, Jin L, et al. Independent correlation between Ki67 index and circulating tumor cells in the diag-nosis of colorectal cancer. Anticancer Res 2017; 37(8): 4693-700.
[PMID: 28739773]
[79]
Lyberopoulou A, Aravantinos G, Efstatho-poulos EP, et al. Mutational analysis of circulating tu-mor cells from colorectal cancer patients and correlation with primary tumor tissue. PLoS One 2015; 10(4)e0123902
[http://dx.doi.org/10.1371/journal.pone.0123902] [PMID: 25902072]
[80]
Mulcahy HE, Lyautey J, Lederrey C, et al. A prospective study of K-ras mutations in the plasma of pan-creatic cancer patients. Clin Cancer Res 1998; 4(2): 271-5.
[PMID: 9516910]
[81]
Kopreski MS, Benko FA, Kwee C, et al. Detection of mutant K-ras DNA in plasma or serum of pa-tients with colorectal cancer. Br J Cancer 1997; 76(10): 1293-9.
[http://dx.doi.org/10.1038/bjc.1997.551] [PMID: 9374374]
[82]
Wang JY, Hsieh JS, Chang MY, et al. Molecu-lar detection of APC, K- ras, and p53 mutations in the serum of colorectal cancer patients as circulating biomarkers. World J Surg 2004; 28(7): 721-6.
[http://dx.doi.org/10.1007/s00268-004-7366-8] [PMID: 15185002]
[83]
Lauschke H, Caspari R, Friedl W, et al. Detec-tion of APC and k-ras mutations in the serum of patients with colorectal cancer. Cancer Detect Prev 2001; 25(1): 55-61.
[PMID: 11270422]
[84]
Thierry AR, Mouliere F, El Messaoudi S, et al. Clinical validation of the detection of KRAS and BRAF mutations from circulating tumor DNA. Nat Med 2014; 20(4): 430-5.
[http://dx.doi.org/10.1038/nm.3511] [PMID: 24658074]
[85]
Lan YT, Chen MH, Fang WL, et al. Clinical relevance of cell-free DNA in gastrointestinal tract malignan-cy. Oncotarget 2017; 8(2): 3009-17.
[http://dx.doi.org/10.18632/oncotarget.13821] [PMID: 27936467]
[86]
Wang X, Shi XQ, Zeng PW, Mo FM, Chen ZH. Circulating cell free DNA as the diagnostic marker for colorectal cancer: A systematic review and meta-analysis. Oncotarget 2018; 9(36): 24514-24.
[http://dx.doi.org/10.18632/oncotarget.25314] [PMID: 29849957]
[87]
Arko-Boham B, Aryee NA, Blay RM, et al. Circulating cell-free DNA integrity as a diagnostic and prog-nostic marker for breast and prostate cancers. Cancer Genet 2019; 235-236: 65-71.
[http://dx.doi.org/10.1016/j.cancergen.2019.04.062] [PMID: 31105051]
[88]
Leng S, Zheng J, Jin Y, et al. Plasma cell-free DNA level and its integrity as biomarkers to distinguish non-small cell lung cancer from tuberculosis. Clin Chim Acta 2018; 477: 160-5.
[http://dx.doi.org/10.1016/j.cca.2017.11.003] [PMID: 29113814]
[89]
Feng J, Gang F, Li X, et al. Plasma cell-free DNA and its DNA integrity as biomarker to distinguish prostate cancer from benign prostatic hyperplasia in patients with in-creased serum prostate-specific antigen. Int Urol Nephrol 2013; 45(4): 1023-8.
[http://dx.doi.org/10.1007/s11255-013-0491-2] [PMID: 23779229]
[90]
Leszinski G, Lehner J, Gezer U, Holden-rieder S. Increased DNA integrity in colorectal cancer. In Vivo 2014; 28(3): 299-303.
[PMID: 24815830]
[91]
Bedin C, Enzo MV, Del Bianco P, Pucci-arelli S, Nitti D, Agostini M. Diagnostic and prog-nostic role of cell‐free DNA testing for colorectal cancer pa-tients. Int J Cancer 2017; 140(8): 1888-98.
[http://dx.doi.org/10.1002/ijc.30565] [PMID: 27943272]
[92]
Flamini E, Mercatali L, Nanni O, et al. Free DNA and carcinoembryonic antigen serum levels: An im-portant combination for diagnosis of colorectal cancer. Clin Cancer Res 2006; 12(23): 6985-8.
[http://dx.doi.org/10.1158/1078-0432.CCR-06-1931] [PMID: 17145818]
[93]
Hao TB, Shi W, Shen XJ, et al. Circulating cell-free DNA in serum as a biomarker for diagnosis and prog-nostic prediction of colorectal cancer. Br J Cancer 2014; 111(8): 1482-9.
[http://dx.doi.org/10.1038/bjc.2014.470] [PMID: 25157833]
[94]
Church TR, Wandell M, Lofton-Day C, et al. Prospective evaluation of methylated SEPT9 in plasma for detection of asymptomatic colorectal cancer. Gut 2014; 63(2): 317-25.
[http://dx.doi.org/10.1136/gutjnl-2012-304149] [PMID: 23408352]
[95]
Petit J, Carroll G, Gould T, Pockney P, Dun M, Scott RJ. Cell-free DNA as a diagnostic blood-based biomarker for colorectal cancer: A systematic review. J Surg Res 2019; 236: 184-97.
[http://dx.doi.org/10.1016/j.jss.2018.11.029] [PMID: 30694754]
[96]
Ladabaum U, Allen J, Wandell M, Ramsey S. Colorectal cancer screening with blood-based bi-omarkers: Cost-effectiveness of methylated septin 9 DNA versus current strategies. Cancer Epidemiol Biomarkers Prev 2013; 22(9): 1567-76.
[http://dx.doi.org/10.1158/1055-9965.EPI-13-0204] [PMID: 23796793]
[97]
Ladabaum U, Alvarez-Osorio L, Rösch T, Brueggenjuergen B. Cost-effectiveness of colorectal cancer screening in Germany: Current endoscopic and fecal testing strategies versus plasma methylated Septin 9 DNA. Endosc Int Open 2014; 2(2): E96-E104.
[http://dx.doi.org/10.1055/s-0034-1377182] [PMID: 26135268]
[98]
Xu F, Yu S, Han J, et al. Detection of circulating tumor DNA Methylation in diagnosis of colorectal cancer. Clin Transl Gastroenterol 2021; 12(8)e00386
[http://dx.doi.org/10.14309/ctg.0000000000000386] [PMID: 34382948]
[99]
Lin WH, Xiao J, Ye ZY, et al. Circulating tumor DNA methylation marker MYO1-G for diagnosis and moni-toring of colorectal cancer. Clin Epigenetics 2021; 13(1): 232.
[http://dx.doi.org/10.1186/s13148-021-01216-0] [PMID: 34961566]
[100]
Ng EKO, Chong WWS, Jin H, et al. Differen-tial expression of microRNAs in plasma of patients with col-orectal cancer: A potential marker for colorectal cancer screening. Gut 2009; 58(10): 1375-81.
[http://dx.doi.org/10.1136/gut.2008.167817] [PMID: 19201770]
[101]
Zekri ARN, Youssef ASED, Lotfy MM, et al. Circulating serum miRNAs as diagnostic markers for colorectal cancer. PLoS One 2016; 11(5)e0154130
[http://dx.doi.org/10.1371/journal.pone.0154130] [PMID: 27135244]
[102]
Liu GH, Zhou ZG, Chen R, et al. Serum miR-21 and miR-92a as biomarkers in the diagnosis and prognosis of colorectal cancer. Tumour Biol 2013; 34(4): 2175-81.
[http://dx.doi.org/10.1007/s13277-013-0753-8] [PMID: 23625654]
[103]
Liang G, Yanliang Z. Engineered exosomes for targeted co-delivery of miR-21 inhibitor and chemotherapeu-tics to reverse drug resistance in colon cancer. J Nanobiotechnology 2020; 18(1): 1-15.
[http://dx.doi.org/10.1186/s12951-019-0560-5] [PMID: 31898555]
[104]
Ogata-Kawata H, Izumiya M, Kurioka D, et al. Circulating exosomal microRNAs as biomarkers of co-lon cancer. PLoS One 2014; 9(4)e92921
[http://dx.doi.org/10.1371/journal.pone.0092921] [PMID: 24705249]
[105]
Farace F, Massard C, Vimond N, et al. A direct comparison of CellSearch and ISET for circulating tu-mour-cell detection in patients with metastatic carcinomas. Br J Cancer 2011; 105(6): 847-53.
[http://dx.doi.org/10.1038/bjc.2011.294] [PMID: 21829190]
[106]
Kanaan Z, Rai SN, Eichenberger MR, et al. Plasma MiR-21. Ann Surg 2012; 256(3): 544-51.
[http://dx.doi.org/10.1097/SLA.0b013e318265bd6f] [PMID: 22868372]
[107]
Pan C, Yan X, Li H, et al. Systematic literature review and clinical validation of circulating microRNAs as diagnostic biomarkers for colorectal cancer. Oncotarget 2017; 8(40): 68317-28.
[http://dx.doi.org/10.18632/oncotarget.19344] [PMID: 28978119]
[108]
Huang Z, Huang D, Ni S, Peng Z, Sheng W, Du X. Plasma microRNAs are promising novel bi-omarkers for early detection of colorectal cancer. Int J Cancer 2010; 127(1): 118-26.
[http://dx.doi.org/10.1002/ijc.25007] [PMID: 19876917]
[109]
Wang Q, Huang Z, Ni S, et al. Plasma miR-601 and miR-760 are novel biomarkers for the early detection of colorectal cancer. PLoS One 2012; 7(9)e44398
[http://dx.doi.org/10.1371/journal.pone.0044398] [PMID: 22970209]
[110]
Kanaan Z, Roberts H, Eichenberger MR, et al. A plasma microRNA panel for detection of colorectal adenomas: A step toward more precise screening for colorec-tal cancer. Ann Surg 2013; 258(3): 400-8.
[http://dx.doi.org/10.1097/SLA.0b013e3182a15bcc] [PMID: 24022433]
[111]
Zou SL, Chen YL, Ge ZZ, Qu YY, Cao Y, Kang ZX. Downregulation of serum exosomal miR-150-5p is associated with poor prognosis in patients with colorec-tal cancer. Cancer Biomark 2019; 26(1): 69-77.
[http://dx.doi.org/10.3233/CBM-190156] [PMID: 31306108]
[112]
Matsumura T, Sugimachi K, Iinuma H, et al. Exosomal microRNA in serum is a novel biomarker of re-currence in human colorectal cancer. Br J Cancer 2015; 113(2): 275-81.
[http://dx.doi.org/10.1038/bjc.2015.201] [PMID: 26057451]
[113]
Wang F, Ma YL, Zhang P, et al. SP1 mediates the link between methylation of the tumour suppressor miR-149 and outcome in colorectal cancer. J Pathol 2013; 229(1): 12-24.
[http://dx.doi.org/10.1002/path.4078] [PMID: 22821729]
[114]
Luo J, Xiong Y, Fu P, et al. Exosomal long non-coding RNAs: Biological properties and therapeutic potential in cancer treatment. J Zhejiang Univ Sci B 2019; 20(6): 488-95.
[http://dx.doi.org/10.1631/jzus.B1900039] [PMID: 31090274]
[115]
Bhan A, Soleimani M, Mandal SS. Long noncoding RNA and cancer: A new paradigm. Cancer Res 2017; 77(15): 3965-81.
[http://dx.doi.org/10.1158/0008-5472.CAN-16-2634] [PMID: 28701486]
[116]
Oehme F, Krahl S, Gyorffy B, et al. Low level of exosomal long non-coding RNA HOTTIP is a prognostic biomarker in colorectal cancer. RNA Biol 2019; 16(10): 1339-45.
[http://dx.doi.org/10.1080/15476286.2019.1637697] [PMID: 31251124]
[117]
Xu W, Zhou G, Wang H, et al. Circulating lncRNA SNHG11 as a novel biomarker for early diagnosis and prognosis of colorectal cancer. Int J Cancer 2020; 146(10): 2901-12.
[http://dx.doi.org/10.1002/ijc.32747] [PMID: 31633800]
[118]
Ye C, Shen Z, Wang B, et al. A novel long non-coding RNA lnc-GNAT1-1 is low expressed in colorectal cancer and acts as a tumor suppressor through regulating RKIP-NF-κB-Snail circuit. J Exp Clin Cancer Res 2016; 35(1): 187.
[http://dx.doi.org/10.1186/s13046-016-0467-z] [PMID: 27912775]
[119]
Gong W, Tian M, Qiu H, Yang Z. Elevated serum level of lncRNA-HIF1A-AS1 as a novel diagnostic predictor for worse prognosis in colorectal carcinoma. Cancer Biomark 2017; 20(4): 417-24.
[http://dx.doi.org/10.3233/CBM-170179] [PMID: 28946548]
[120]
Liu L, Meng T, Yang XH, et al. Prognostic and predictive value of long non-coding RNA GAS5 and mircoRNA-221 in colorectal cancer and their effects on colo-rectal cancer cell proliferation, migration and invasion. Cancer Biomark 2018; 22(2): 283-99.
[http://dx.doi.org/10.3233/CBM-171011] [PMID: 29630521]
[121]
Li Y, Zheng Q, Bao C, et al. Circular RNA is enriched and stable in exosomes: A promising biomarker for cancer diagnosis. Cell Res 2015; 25(8): 981-4.
[http://dx.doi.org/10.1038/cr.2015.82] [PMID: 26138677]
[122]
Lin J, Cai D, Li W, et al. Plasma circular RNA pan-el acts as a novel diagnostic biomarker for colorectal cancer. Clin Biochem 2019; 74: 60-8.
[http://dx.doi.org/10.1016/j.clinbiochem.2019.10.012] [PMID: 31669510]
[123]
Ye D, Wang S, Huang Y, Chi P. A 3-circular RNA signature as a noninvasive biomarker for diagnosis of colorectal cancer. Cancer Cell Int 2019; 19(1): 276.
[http://dx.doi.org/10.1186/s12935-019-0995-7] [PMID: 31700498]
[124]
Chen HY, Li XN, Ye CX, Chen ZL, Wang ZJ. Circular RNA circHUWE1 is upregulated and promotes cell proliferation, migration and invasion in colorectal cancer by sponging miR-486. OncoTargets Ther 2020; 13: 423-34.
[http://dx.doi.org/10.2147/OTT.S233338]
[125]
Li XN, Wang ZJ, Ye CX, Zhao BC, Huang XX, Yang L. Circular RNA circVAPA is up-regulated and exerts oncogenic properties by sponging miR-101 in colorectal cancer. Biomed Pharmacother 2019; 112108611
[http://dx.doi.org/10.1016/j.biopha.2019.108611] [PMID: 30797148]
[126]
Tsai WS, Chen JS, Shao HJ, et al. Circulating tumor cell count correlates with colorectal neoplasm progres-sion and is a prognostic marker for distant metastasis in non-metastatic patients. Sci Rep 2016; 6(1): 24517.
[http://dx.doi.org/10.1038/srep24517] [PMID: 27075165]
[127]
Douard R, Wind P, Sales JP, et al. Long-term prognostic value of detection of circulating colorectal cancer cells using CGM2 reverse transcriptase-polymerase chain re-action assay. Surgery 2006; 139(4): 556-62.
[http://dx.doi.org/10.1016/j.surg.2005.09.025] [PMID: 16627067]
[128]
Lu Y, Wang P, Peng J, Wang X, Zhu Y, Shen N. Meta-analysis reveals the prognostic value of circulating tumour cells detected in the peripheral blood in patients with non-metastatic colorectal cancer. Sci Rep 2017; 7(1): 905.
[http://dx.doi.org/10.1038/s41598-017-01066-y] [PMID: 28424486]
[129]
Lin C-C. Circulating tumor cells correlates with adverse outcomes for non-metastatic colorectal cancer patients both before and after surgery. J Cancer 2015; 112(8): 1306-13.
[http://dx.doi.org/10.21203/rs.3.rs-1752037/v1]
[130]
El Messaoudi S, Mouliere F, Du Manoir S, et al. Circulating DNA as a strong multimarker prognostic tool for metastatic colorectal cancer patient management care. Clin Cancer Res 2016; 22(12): 3067-77.
[http://dx.doi.org/10.1158/1078-0432.CCR-15-0297] [PMID: 26847055]
[131]
Perdyan A, Spychalski P, Kacperczyk J, Rostkowska O, Kobiela J. Circulating tumor DNA in KRAS positive colorectal cancer patients as a prognostic factor-a systematic review and meta-analysis. Crit Rev Oncol Hematol 2020; 154103065
[http://dx.doi.org/10.1016/j.critrevonc.2020.103065] [PMID: 32763752]
[132]
Lin JK, Lin PC, Lin CH, et al. Clinical relevance of alterations in quantity and quality of plasma DNA in colo-rectal cancer patients: Based on the mutation spectra detected in primary tumors. Ann Surg Oncol 2014; 21 (Suppl. 4): 680-6.
[http://dx.doi.org/10.1245/s10434-014-3804-5] [PMID: 24841357]
[133]
Lee HS, Hwang SM, Kim TS, et al. Circulating methylated septin 9 nucleic Acid in the plasma of patients with gastrointestinal cancer in the stomach and colon. Transl Oncol 2013; 6(3): 290-IN4.
[http://dx.doi.org/10.1593/tlo.13118] [PMID: 23730408]
[134]
Liu Y, Chew MH, Tham CK, Tang CL, Ong SY, Zhao Y. Methylation of serum SST gene is an inde-pendent prognostic marker in colorectal cancer. Am J Cancer Res 2016; 6(9): 2098-108.
[PMID: 27725914]
[135]
Lin PC, Lin JK, Lin CH, et al. Clinical relevance of plasma DNA methylation in colorectal cancer patients identified by using a genome-wide high-resolution array. Ann Surg Oncol 2015; 22 (Suppl. 3): 1419-27.
[http://dx.doi.org/10.1245/s10434-014-4277-2] [PMID: 25472652]
[136]
Peng ZY, Gu RH, Yan B. Downregulation of exosomeencapsulated miR‐548c‐5p is associated with poor prognosis in colorectal cancer. J Cell Biochem 2019; 120(2): 1457-63.
[http://dx.doi.org/10.1002/jcb.27291] [PMID: 30171732]
[137]
Sun Y, Yang B, Lin M, Yu H, Chen H, Zhang Z. Identification of serum miR-30a-5p as a diag-nostic and prognostic biomarker in colorectal cancer. Cancer Biomark 2019; 24(3): 299-305.
[http://dx.doi.org/10.3233/CBM-182129] [PMID: 30829615]
[138]
Yuan D, Li K, Zhu K, Yan R, Dang C. Plas-ma miR-183 predicts recurrence and prognosis in patients with colorectal cancer. Cancer Biol Ther 2015; 16(2): 268-75.
[http://dx.doi.org/10.1080/15384047.2014.1002327] [PMID: 25629978]
[139]
Kou CH, Zhou T, Han XL, Zhuang HJ, Qi-an HX. Downregulation of mir-23b in plasma is associat-ed with poor prognosis in patients with colorectal cancer. Oncol Lett 2016; 12(6): 4838-44.
[http://dx.doi.org/10.3892/ol.2016.5265] [PMID: 28101227]
[140]
Takano Y, Masuda T, Iinuma H, et al. Circu-lating exosomal microRNA-203 is associated with metastasis possibly via inducing tumor-associated macrophages in colo-rectal cancer. Oncotarget 2017; 8(45): 78598-613.
[http://dx.doi.org/10.18632/oncotarget.20009] [PMID: 29108252]
[141]
Yan S, Han B, Gao S, et al. Exosome-encapsulated microRNAs as circulating biomarkers for colo-rectal cancer. Oncotarget 2017; 8(36): 60149-58.
[http://dx.doi.org/10.18632/oncotarget.18557] [PMID: 28947960]
[142]
Yan S, Jiang Y, Liang C, et al. Exosomal miR‐6803‐5p as potential diagnostic and prognostic marker in colorectal cancer. J Cell Biochem 2018; 119(5): 4113-9.
[http://dx.doi.org/10.1002/jcb.26609] [PMID: 29240249]
[143]
Yan S, Liu G, Jin C, et al. MicroRNA‐6869‐5p acts as a tumor suppressor via targeting TLR4/NF-κB signaling pathway in colorectal cancer. J Cell Physiol 2018; 233(9): 6660-8.
[http://dx.doi.org/10.1002/jcp.26316] [PMID: 29206292]
[144]
Gao T, Liu X, He B, et al. Exosomal lncRNA 91H is associated with poor development in colorectal cancer by modifying HNRNPK expression. Cancer Cell Int 2018; 18(1): 11.
[http://dx.doi.org/10.1186/s12935-018-0506-2] [PMID: 29410604]
[145]
Liu T, Zhang X, Gao S, et al. Exosomal long noncoding RNA CRNDE-h as a novel serum-based bi-omarker for diagnosis and prognosis of colorectal cancer. Oncotarget 2016; 7(51): 85551-63.
[http://dx.doi.org/10.18632/oncotarget.13465] [PMID: 27888803]
[146]
Luan Y, Li X, Luan Y, et al. Circulating lncRNA UCA1 promotes malignancy of colorectal cancer via the miR-143/MYO6 axis. Mol Ther Nucleic Acids 2020; 19: 790-803.
[http://dx.doi.org/10.1016/j.omtn.2019.12.009] [PMID: 31955010]
[147]
Wang Y, Zhang D, Zhang C, Sun Y. The diag-nostic and prognostic value of serum lncRNA NEAT1 in colorectal cancer. Cancer Manag Res 2020; 12: 10985-92.
[http://dx.doi.org/10.2147/CMAR.S269978] [PMID: 33173332]
[148]
Pan B, Qin J, Liu X, et al. Identification of serum exosomal hsa-circ-0004771 as a novel diagnostic biomarker of colorectal cancer. Front Genet 2019; 10: 1096.
[http://dx.doi.org/10.3389/fgene.2019.01096] [PMID: 31737058]
[149]
Lu C, Fu L, Qian X, Dou L, Cang S. Knock-down of circular RNA circ-FARSA restricts colorectal cancer cell growth through regulation of miR-330-5p/LASP1 axis. Arch Biochem Biophys 2020; 689108434
[http://dx.doi.org/10.1016/j.abb.2020.108434] [PMID: 32473899]
[150]
Wang R, Wang J, Chen Y, et al. Circular RNA circLDLR facilitates cancer progression by altering the miR-30a-3p/SOAT1 axis in colorectal cancer. Cell Death Discov 2022; 8(1): 314.
[http://dx.doi.org/10.1038/s41420-022-01110-5] [PMID: 35821230]
[151]
Chambers AE, Frick J, Tanner N, Gerkin R, Kundranda M, Dragovich T. Chemotherapy re-challenge response rate in metastatic colorectal cancer. J Gastrointest Oncol 2018; 9(4): 679-86.
[http://dx.doi.org/10.21037/jgo.2018.04.08] [PMID: 30151264]
[152]
Zhang D, Zhao L, Zhou P, et al. Circulating tumor microemboli (CTM) and vimentin+ circulating tumor cells (CTCs) detected by a size-based platform predict worse prognosis in advanced colorectal cancer patients during chemotherapy. Cancer Cell Int 2017; 17(1): 6.
[http://dx.doi.org/10.1186/s12935-016-0373-7] [PMID: 28070168]
[153]
Lankiewicz S, Zimmermann S, Hollmann C, Hillemann T, Greten TF. Circulating tumour cells as a predictive factor for response to systemic chemotherapy in patients with advanced colorectal cancer. Mol Oncol 2008; 2(4): 349-55.
[http://dx.doi.org/10.1016/j.molonc.2008.09.001] [PMID: 19383356]
[154]
Huang X, Gao P, Song Y, et al. Relationship between circulating tumor cells and tumor response in colo-rectal cancer patients treated with chemotherapy: A meta-analysis. BMC Cancer 2014; 14(1): 976.
[http://dx.doi.org/10.1186/1471-2407-14-976] [PMID: 25519477]
[155]
Cao H, Liu X, Chen Y, et al. Circulating tumor DNA is capable of monitoring the therapeutic response and resistance in advanced colorectal cancer patients undergoing combined target and chemotherapy. Front Oncol 2020; 10: 466.
[http://dx.doi.org/10.3389/fonc.2020.00466] [PMID: 32318348]
[156]
Sun X, Yuan W, Hao F, Zhuang W, Re-search C. Promoter methylation of RASSF1A indicates prognosis for patients with stage II and III colorectal cancer treated with oxaliplatin-based chemotherapy. Med Sci Monit 2017; 23: 5389-95.
[http://dx.doi.org/10.12659/MSM.903927] [PMID: 29128865]
[157]
Barault L, Amatu A, Siravegna G, et al. Dis-covery of methylated circulating DNA biomarkers for com-prehensive non-invasive monitoring of treatment response in metastatic colorectal cancer. Gut 2018; 67(11): 1995-2005.
[http://dx.doi.org/10.1136/gutjnl-2016-313372] [PMID: 28982739]
[158]
Barault L, Amatu A, Bleeker FE, et al. Digital PCR quantification of MGMT methylation refines prediction of clinical benefit from alkylating agents in glioblastoma and metastatic colorectal cancer. Ann Oncol 2015; 26(9): 1994-9.
[http://dx.doi.org/10.1093/annonc/mdv272] [PMID: 26113646]
[159]
Chen Q, Xia HW, Ge XJ, Zhang YC, Tang QL, Bi F. Serum miR-19a predicts resistance to FOLFOX chemotherapy in advanced colorectal cancer cases. Asian Pac J Cancer Prev 2013; 14(12): 7421-6.
[http://dx.doi.org/10.7314/APJCP.2013.14.12.7421] [PMID: 24460313]
[160]
Liu C, Eng C, Shen J, et al. Serum exosomal miR-4772-3p is a predictor of tumor recurrence in stage II and III colon cancer. Oncotarget 2016; 7(46): 76250-60.
[http://dx.doi.org/10.18632/oncotarget.12841] [PMID: 27788488]
[161]
Hu J, Cai G, Xu Y, Cai S. The Plasma microRNA miR-1914* and -1915 suppresses chemoresistant in colorec-tal cancer patients by down-regulating NFIX. Curr Mol Med 2016; 16(1): 70-82.
[http://dx.doi.org/10.2174/1566524016666151222144656] [PMID: 26695693]
[162]
Zhang J, Zhang K, Bi M, Jiao X, Zhang D, Dong Q. Circulating microRNA expressions in colorectal cancer as predictors of response to chemotherapy. Anticancer Drugs 2014; 25(3): 346-52.
[http://dx.doi.org/10.1097/CAD.0000000000000049] [PMID: 24304648]
[163]
Svoboda M, Sana J, Fabian P, et al. MicroRNA expression profile associated with response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer patients. Radiat Oncol 2012; 7(1): 195.
[http://dx.doi.org/10.1186/1748-717X-7-195] [PMID: 23167930]
[164]
Xu Y, Zhu M, Oncology T. Novel exosomal miR-46146 transfer oxaliplatin chemoresistance in colorectal can-cer. Clin Transl Oncol 2020; 22(7): 1105-16.
[http://dx.doi.org/10.1007/s12094-019-02237-1] [PMID: 31728833]
[165]
Yang Y, Zhang R, Du J, et al. Predictive role of UCA1-containing exosomes in cetuximab-resistant colorectal cancer. Cancer Cell Int 2018; 18(1): 164.
[http://dx.doi.org/10.1186/s12935-018-0660-6] [PMID: 30377411]
[166]
Xiao Y, Yurievich UA, Yosypovych SV. Long noncoding RNA XIST is a prognostic factor in colorec-tal cancer and inhibits 5-fluorouracil-induced cell cytotoxici-ty through promoting thymidylate synthase expression. Oncotarget 2017; 8(47): 83171-82.
[http://dx.doi.org/10.18632/oncotarget.20487] [PMID: 29137332]
[167]
Li L, Shang J, Zhang Y, et al. MEG3 is a prog-nostic factor for CRC and promotes chemosensitivity by en-hancing oxaliplatin-induced cell apoptosis. Oncol Rep 2017; 38(3): 1383-92.
[http://dx.doi.org/10.3892/or.2017.5828] [PMID: 28731151]
[168]
Sun F, Liang W, Qian J. The identification of CRNDE, H19, UCA1 and HOTAIR as the key lncRNAs in-volved in oxaliplatin or irinotecan resistance in the chemo-therapy of colorectal cancer based on integrative bioinfor-matics analysis. Mol Med Rep 2019; 20(4): 3583-96.
[http://dx.doi.org/10.3892/mmr.2019.10588] [PMID: 31432188]
[169]
Deng X, Ruan H, Zhang X, et al. Long noncod-ing RNA CCAL transferred from fibroblasts by exosomes promotes chemoresistance of colorectal cancer cells. Int J Cancer 2020; 146(6): 1700-16.
[http://dx.doi.org/10.1002/ijc.32608] [PMID: 31381140]
[170]
Hon KW, Ab-Mutalib NS, Abdullah NMA, Jamal R, Abu N. Extracellular vesicle-derived circular RNAs confers chemoresistance in colorectal cancer. Sci Rep 2019; 9(1): 16497.
[http://dx.doi.org/10.1038/s41598-019-53063-y] [PMID: 31712601]
[171]
Xu Y, Qiu A, Peng F, Tan X, Wang J, Gong XJN. Exosomal transfer of circular RNA FBXW7 amelio-rates the chemoresistance to oxaliplatin in colorectal cancer by sponging miR-18b-5p. Neoplasma 2020; 68(11): 108-18.
[PMID: 33147048]
[172]
Lai M, Liu G, Li R, et al. Hsa_circ_0079662 induces the resistance mechanism of the chemotherapy drug oxaliplatin through the TNF-α pathway in human colon cancer. J Cell Mol Med 2020; 24(9): 5021-7.
[http://dx.doi.org/10.1111/jcmm.15122] [PMID: 32243061]
[173]
Jiang Z, Hou Z, Liu W, Yu Z, Liang Z, Chen S. Circular RNA protein tyrosine kinase 2 (circPTK2) promotes colorectal cancer proliferation, migra-tion, invasion and chemoresistance. Bioengineered 2022; 13(1): 810-23.
[http://dx.doi.org/10.1080/21655979.2021.2012952] [PMID: 34974791]
[174]
Harb W, Fan A, Tran T, et al. Mutational analysis of circulating tumor cells using a novel microfluidic collec-tion device and qPCR assay. Transl Oncol 2013; 6(5): 528-IN1.
[http://dx.doi.org/10.1593/tlo.13367] [PMID: 24151533]
[175]
Lu NN, Xie M, Wang J, et al. Biotin-triggered decomposable immunomagnetic beads for capture and re-lease of circulating tumor cells. ACS Appl Mater Interfaces 2015; 7(16): 8817-26.
[http://dx.doi.org/10.1021/acsami.5b01397] [PMID: 25853336]
[176]
Mikolajczyk SD. Detection of EpCAM-negative and cytokeratin-negative circulating tumor cells in peripheral blood. J Oncol 2011; 2011: 1-10.
[http://dx.doi.org/10.1155/2011/252361]
[177]
Talasaz AH, Powell AA, Huber DE, et al. Isolating highly enriched populations of circulating epithelial cells and other rare cells from blood using a magnetic sweep-er device. Proc Natl Acad Sci USA 2009; 106(10): 3970-5.
[http://dx.doi.org/10.1073/pnas.0813188106] [PMID: 19234122]
[178]
Xiong K, Wei W, Jin Y, et al. Biomimetic immuno-magnetosomes for high-performance enrichment of circulating tumor cells. Adv Mater 2016; 28(36): 7929-35.
[http://dx.doi.org/10.1002/adma.201601643] [PMID: 27376951]
[179]
Zhu DM, Wu L, Suo M, et al. Engineered red blood cells for capturing circulating tumor cells with high performance. Nanoscale 2018; 10(13): 6014-23.
[http://dx.doi.org/10.1039/C7NR08032H] [PMID: 29542756]
[180]
He W, Kularatne SA, Kalli KR, et al. Quantita-tion of circulating tumor cells in blood samples from ovarian and prostate cancer patients using tumor-specific fluorescent ligands. Int J Cancer 2008; 123(8): 1968-73.
[http://dx.doi.org/10.1002/ijc.23717] [PMID: 18661519]
[181]
Sequist LV, Nagrath S, Toner M, Haber DA, Lynch TJ. The CTC-chip: An exciting new tool to detect circulating tumor cells in lung cancer patients. J Thorac Oncol 2009; 4(3): 281-3.
[http://dx.doi.org/10.1097/JTO.0b013e3181989565] [PMID: 19247082]
[182]
Stott SL, Hsu CH, Tsukrov DI, et al. Isolation of circulating tumor cells using a microvortex-generating her-ringbone-chip. Proc Natl Acad Sci USA 2010; 107(43): 18392-7.
[http://dx.doi.org/10.1073/pnas.1012539107] [PMID: 20930119]
[183]
Gleghorn JP, Pratt ED, Denning D, et al. Capture of circulating tumor cells from whole blood of pros-tate cancer patients using geometrically enhanced differential immunocapture (GEDI) and a prostate-specific antibody. Lab Chip 2010; 10(1): 27-9.
[http://dx.doi.org/10.1039/B917959C] [PMID: 20024046]
[184]
Adams AA, Okagbare PI, Feng J, et al. High-ly efficient circulating tumor cell isolation from whole blood and label-free enumeration using polymer-based microfluid-ics with an integrated conductivity sensor. J Am Chem Soc 2008; 130(27): 8633-41.
[http://dx.doi.org/10.1021/ja8015022] [PMID: 18557614]
[185]
Jan YJ, Chen JF, Zhu Y, et al. NanoVelcro rare-cell assays for detection and characterization of circulating tumor cells. Adv Drug Deliv Rev 2018; 125: 78-93.
[http://dx.doi.org/10.1016/j.addr.2018.03.006] [PMID: 29551650]
[186]
Park MH, Reátegui E, Li W, et al. Enhanced isolation and release of circulating tumor cells using nano-particle binding and ligand exchange in a microfluidic chip. J Am Chem Soc 2017; 139(7): 2741-9.
[http://dx.doi.org/10.1021/jacs.6b12236] [PMID: 28133963]
[187]
Yoon HJ, Kim TH, Zhang Z, et al. Sensitive capture of circulating tumour cells by functionalized gra-phene oxide nanosheets. Nat Nanotechnol 2013; 8(10): 735-41.
[http://dx.doi.org/10.1038/nnano.2013.194] [PMID: 24077027]
[188]
Wang S, Wang H, Jiao J, et al. Three-dimensional nanostructured substrates toward efficient cap-ture of circulating tumor cells. Angew Chem Int Ed 2009; 48(47): 8970-3.
[http://dx.doi.org/10.1002/anie.200901668] [PMID: 19847834]
[189]
Loeian MS, Mehdi Aghaei S, Farhadi F, et al. Liquid biopsy using the nanotube-CTC-chip: Capture of invasive CTCs with high purity using preferential adherence in breast cancer patients. Lab Chip 2019; 19(11): 1899-915.
[http://dx.doi.org/10.1039/C9LC00274J] [PMID: 31049504]
[190]
Harouaka RA, Zhou MD, Yeh YT, et al. Flexible micro spring array device for high-throughput en-richment of viable circulating tumor cells. Clin Chem 2014; 60(2): 323-33.
[http://dx.doi.org/10.1373/clinchem.2013.206805] [PMID: 24132944]
[191]
Zhou MD, Hao S, Williams AJ, et al. Separable bilayer microfiltration device for viable label-free enrichment of circulating tumour cells. Sci Rep 2014; 4(1): 7392.
[http://dx.doi.org/10.1038/srep07392] [PMID: 25487434]
[192]
Kim TH, Lim M, Park J, et al. FAST: Size-selective, clog-free isolation of rare cancer cells from whole blood at a liquid–liquid interface. Anal Chem 2017; 89(2): 1155-62.
[http://dx.doi.org/10.1021/acs.analchem.6b03534] [PMID: 27958721]
[193]
Mastoraki S, Strati A, Tzanikou E, et al. ESR1 Methylation: A liquid biopsy-based epigenetic assay for the follow-up of pa-tients with metastatic breast cancer receiving endocrine treatment. Clin Cancer Res 2018; 24(6): 1500-10.
[http://dx.doi.org/10.1158/1078-0432.CCR-17-1181] [PMID: 29284708]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy