Liquid biopsy of cancer: a multimodal diagnostic tool in clinical oncology

Raffaele Palmirotta, Domenica Lovero, Paola Cafforio, Claudia Felici, Francesco Mannavola, Eleonora Pellè, Davide Quaresmini, Marco Tucci, Franco Silvestris, Raffaele Palmirotta, Domenica Lovero, Paola Cafforio, Claudia Felici, Francesco Mannavola, Eleonora Pellè, Davide Quaresmini, Marco Tucci, Franco Silvestris

Abstract

Over the last decades, the concept of precision medicine has dramatically renewed the field of medical oncology; the introduction of patient-tailored therapies has significantly improved all measurable outcomes. Liquid biopsy is a revolutionary technique that is opening previously unexpected perspectives. It consists of the detection and isolation of circulating tumor cells, circulating tumor DNA and exosomes, as a source of genomic and proteomic information in patients with cancer. Many technical hurdles have been resolved thanks to newly developed techniques and next-generation sequencing analyses, allowing a broad application of liquid biopsy in a wide range of settings. Initially correlated to prognosis, liquid biopsy data are now being studied for cancer diagnosis, hopefully including screenings, and most importantly for the prediction of response or resistance to given treatments. In particular, the identification of specific mutations in target genes can aid in therapeutic decisions, both in the appropriateness of treatment and in the advanced identification of secondary resistance, aiming to early diagnose disease progression. Still application is far from reality but ongoing research is leading the way to a new era in oncology. This review summarizes the main techniques and applications of liquid biopsy in cancer.

Keywords: cancer; circulating tumor DNA; circulating tumor cells; exosomes; liquid biopsy; targeted therapy.

Conflict of interest statement

Conflict of interest statement: The authors declare that there is no conflict of interest.

Figures

Figure 1.
Figure 1.
Molecular applications of circulating tumor cells (CTCs), circulating tumor DNA (ctDNA) and exosomes as liquid biopsy for personalized medicine.
Figure 2.
Figure 2.
Flow chart of combined methods to isolate circulating tumor cells (CTCs). (a) Peripheral blood samples are subjected to density gradient stratification and leukocyte depletion is assessed by an immunomagnetic method using anti-CD45 and anti-glycophorin conjugated microbeads (AUTOMACS (Miltenyi Biotec GmbH, Bergisch Gladbach, Germany)). (b) The CTC-enriched fraction is stained by specific fluorochrome conjugated antibodies and loaded in a dedicated cartridge which then is subjected to dielectric forces (DEPArray (Menarini Silicon Biosystems, Firenze, Italy)). (c) CTCs are visualized by dedicated software and selected by positive fluorescence for tumor-specific markers and negativity for CD45 leukocyte marker. 4’,6-diaminidino-2-phenylindole (DAPI) is used to counterstain nuclei. The CTCs are moved into a parking area and recovered as single or grouped cells in a buffer drop.
Figure 3.
Figure 3.
The DEPArray technology is based on the use of a dielectrophoretic field (DEP) generated by electrodes in a matrix underlying a liquid layer of cells. (a) The DEPArray constellation for the creation of DEP is determined to be a determinant of the entire intrapolarization. (b) The whole cell is isolated from the individual software, and (c) after computational imaging, individual cells or groups of cells are moved and recovered by a drop of buffer in a specific tube.
Figure 4.
Figure 4.
Mutational analysis performed on circulating tumor cells (CTCs) isolated by DEPArray using next-generation sequencing, Sanger sequencing or digital polymerase chain reaction (PCR). DAPI , 4’,6-diaminidino-2-phenylindole.
Figure 5.
Figure 5.
Exosomes biogenesis and content. Exosomes are end products of the recycling endosomal pathway and originate from inward budding of the plasma membrane, leading to the formation of multi vesicular bodies (MVBs). Following active packaging of signaling molecules into MVBs, they fuse with the plasma membrane and release their contents into the extracellular space in the form of exosomes. Exosomes consist of a lipid bilayer which contains both transmembrane and nonmembrane proteins, as well as noncoding RNAs, mRNAs and either single-stranded or double-stranded DNA. They also express a conserved set of proteins independently by cellular origin, including CD63, CD81, and CD9 tetraspanins, while those from cancer cells are rich in tumor-associated antigens. MHC, major histocompatibility complex.

References

    1. McCarthy JJ, McLeod HL, Ginsburg GS. Genomic medicine: a decade of successes, challenges, and opportunities. Sci Transl Med 2013; 5: 189sr4.
    1. Hodson R. Precision medicine. Nature 2016; 537: S49.
    1. Savonarola A, Palmirotta R, Guadagni F, Silvestris F. Pharmacogenetics and pharmacogenomics: role of mutational analysis in anti-cancer targeted therapy. Pharmacogenomics J 2012; 12: 277–286.
    1. Wang J, Chang S, Li G, Sun Y. Application of liquid biopsy in precision medicine: opportunities and challenges. Front Med 2017; 11: 522–527.
    1. Perakis S, Speicher MR. Emerging concepts in liquid biopsies. BMC Med 2017; 15: 75.
    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.
    1. Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A. Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol 2013; 10: 472–484.
    1. Buder A, Tomuta C, Filipits M. The potential of liquid biopsies. Curr Opin Oncol 2016; 28: 130–134.
    1. Zhang W, Xia W, Lv Z, Ni C, Xin Y, Yang L. Liquid Biopsy for Cancer: Circulating Tumor Cells, Circulating Free DNA or Exosomes? Cell Physiol Biochem 2017; 41: 755–768.
    1. Ashworth T. A case of cancer in which cells similar to those in the tumors were seen in the blood after death. Med J Aust 1869; 14: 146–147.
    1. Parkinson DR, Dracopoli N, Petty BG, et al. Considerations in the development of circulating tumor cell technology for clinical use. J Transl Med 2012; 10: 138.
    1. Young R, Pailler E, Billiot F, et al. Circulating tumor cells in lung cancer. Acta Cytologica 2012; 56: 655–660.
    1. Krebs MG, Metcalf RL, Carter L, et al. Molecular analysis of circulating tumour cells-biology and biomarkers. Nat Rev Clin Oncol 2014; 11: 129–144.
    1. Labelle M, Begum S, Hynes RO. Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis. Cancer Cell. 2011; 20: 576–590.
    1. Le Gal K, Ibrahim MX, Wiel C, et al. Antioxidants can increase melanoma metastasis in mice. Sci Transl Med 2015; 7: 308re8.
    1. van der Toom EE, Verdone JE, Gorin MA, et al. Technical challenges in the isolation and analysis of circulating tumor cells. Oncotarget. 2016; 7: 62754–62766.
    1. Gascoyne PR, Shim S. Isolation of circulating tumor cells by dielectrophoresis. Cancers. 2014; 6: 545–579.
    1. Sollier E, Go DE, Che J, et al. Size-selective collection of circulating tumor cells using Vortex technology. Lab Chip 2014; 14: 63–77.
    1. Au SH, Storey BD, Moore JC, et al. Clusters of circulating tumor cells traverse capillary-sized vessels. Proc Natl Acad Sci U S A 2016; 113: 4947–4952.
    1. Park ES, Jin C, Guo Q, et al. Continuous flow deformability-based separation of circulating tumor cells using microfluidic ratchets. Small 2016; 12: 1909–1919.
    1. Zheng S, Lin HK, Lu B, et al. 3D microfilter device for viable circulating tumor cell (CTC) enrichment from blood. Biomed Microdevices 2011; 13: 203–213.
    1. Lopez-Riquelme N, Minguela A, Villar-Permuy F, et al. Imaging cytometry for counting circulating tumor cells: comparative analysis of the CellSearch vs ImageStream systems. APMIS 2013; 121: 1139–1143.
    1. Li P, Mao Z, Peng Z, et al. Acoustic separation of circulating tumor cells. Proc Natl Acad Sci U S A 2015; 112: 4970–4975.
    1. Arya SK, Lim B, Rahman AR. Enrichment, detection and clinical significance of circulating tumor cells. Lab Chip 2013; 13: 1995–2027.
    1. Abonnenc M, Manaresi N, Borgatti M, et al. Programmable interactions of functionalized single bioparticles in a dielectrophoresis-based microarray chip. Anal Chem 2013; 85: 8219–8224.
    1. Schneck H, Gierke B, Uppenkamp F, et al. EpCAM-independent enrichment of circulating tumor cells in metastatic breast cancer. PLoS One. 2015; 10: e0144535.
    1. de Wit S, van Dalum G, Terstappen LW. Detection of circulating tumor cells. Scientifica 2014; 2014: 819362.
    1. Alix-Panabieres C, Pantel K. Clinical applications of circulating tumor cells and circulating tumor DNA as liquid biopsy. Cancer Discov 2016; 6: 479–491.
    1. Viswanath B, Kim S, Lee K. Recent insights into nanotechnology development for detection and treatment of colorectal cancer. Int J Nanomedicine 2016; 11: 2491–2504.
    1. Lin M, Chen JF, Lu YT, et al. Nanostructure embedded microchips for detection, isolation, and characterization of circulating tumor cells. Acc Chem Res 2014; 47: 2941–2950.
    1. Ming Y, Li Y, Xing H, et al. Circulating tumor cells: from theory to nanotechnology-based detection. Front Pharmacol 2017; 8: 35.
    1. Palmirotta R, Lovero D, Silvestris E, et al. Next-generation Sequencing (NGS) analysis on single Circulating Tumor Cells (CTCs) with no need of Whole-genome Amplification (WGA). Cancer Genomics Proteomics 2017; 14: 173–179.
    1. Cristofanilli M, Budd GT, Ellis MJ, et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med 2004; 351: 781–791.
    1. Messaritakis I, Stoltidis D, Kotsakis A, et al. TTF-1- and/or CD56-positive circulating tumor cells in patients with small cell lung cancer (SCLC). Sci Rep 2017; 7: 45351.
    1. Lu SH, Tsai WS, Chang YH, et al. Identifying cancer origin using circulating tumor cells. Cancer Biol Ther 2016; 17: 430–438.
    1. Beije N, Onstenk W, Kraan J, et al. Prognostic impact of HER2 and ER status of circulating tumor cells in metastatic breast cancer patients with a HER2-negative primary tumor. Neoplasia. 2016; 18: 647–653.
    1. Yadavalli S, Jayaram S, Manda SS, et al. Data-Driven Discovery of Extravasation Pathway in Circulating Tumor Cells. Sci Rep 2017; 7: 43710.
    1. Bulfoni M, Gerratana L, Del Ben F, et al. In patients with metastatic breast cancer the identification of circulating tumor cells in epithelial-to-mesenchymal transition is associated with a poor prognosis. Breast Cancer Res 2016; 18: 30.
    1. Zhao R, Cai Z, Li S, et al. Expression and clinical relevance of epithelial and mesenchymal markers in circulating tumor cells from colorectal cancer. Oncotarget 2017; 8: 9293–9302.
    1. Satelli A, Batth I, Brownlee Z, et al. EMT circulating tumor cells detected by cell-surface vimentin are associated with prostate cancer progression. Oncotarget 2017; 8: 49329–49337.
    1. Chebouti I, Kasimir-Bauer S, Buderath P, et al. EMT-like circulating tumor cells in ovarian cancer patients are enriched by platinum-based chemotherapy. Oncotarget 2017; 8: 48820–48831.
    1. Li S, Chen Q, Li H, Wu Y, Feng J, Yan Y. Mesenchymal circulating tumor cells (CTCs) and OCT4 mRNA expression in CTCs for prognosis prediction in patients with non-small-cell lung cancer. Clin Transl Oncol 2017; 19: 1147–1153.
    1. Correnti M, Raggi C. Stem-like plasticity and heterogeneity of circulating tumor cells: current status and prospect challenges in liver cancer. Oncotarget 2017; 8: 7094–7115.
    1. Aktas B, Tewes M, Fehm T, et al. Stem cell and epithelial-mesenchymal transition markers are frequently overexpressed in circulating tumor cells of metastatic breast cancer patients. Breast Cancer Res 2009; 11: R46.
    1. Paterlini-Brechot P, Benali NL. Circulating tumor cells (CTC) detection: clinical impact and future directions. Cancer Lett 2007; 253: 180–204.
    1. Esmaeilsabzali H, Beischlag TV, Cox ME, Parameswaran AM, Park EJ. Detection and isolation of circulating tumor cells: principles and methods. Biotechnol Adv 2013; 31: 1063–1084.
    1. Friedlander TW, Premasekharan G, Paris PL. Looking back, to the future of circulating tumor cells. Pharmacol Ther 2014; 142: 271–280.
    1. Giuliano M, Giordano A, Jackson S, et al. Circulating tumor cells as early predictors of metastatic spread in breast cancer patients with limited metastatic dissemination. Breast Cancer Res 2014; 16: 440.
    1. de Bono JS, Scher HI, Montgomery RB, et al. Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin Cancer Res. 2008; 14: 6302–6309.
    1. Normanno N, De Luca A, Gallo M, et al. The prognostic role of circulating tumor cells in lung cancer. Expert Rev Anticancer Ther 2016; 16: 859–867.
    1. Tsai WS, Chen JS, Shao HJ, et al. Circulating tumor cell count correlates with colorectal neoplasm progression and is a prognostic marker for distant metastasis in non-metastatic patients. Sci Rep 2016; 6: 24517.
    1. Ilie M, Hofman V, Long-Mira E, et al. ‘Sentinel‘ circulating tumor cells allow early diagnosis of lung cancer in patients with chronic obstructive pulmonary disease. PLoS One 2014; 9: e111597.
    1. Yan WT, Cui X, Chen Q, et al. Circulating tumor cell status monitors the treatment responses in breast cancer patients: a meta-analysis. Sci Rep 2017; 7: 43464.
    1. Paoletti C, Muniz MC, Thomas DG, et al. Development of circulating tumor cell-endocrine therapy index in patients with hormone receptor-positive breast cancer. Clin Cancer Res 2015; 21: 2487–2498.
    1. Smerage JB, Barlow WE, Hortobagyi GN, et al. Circulating tumor cells and response to chemotherapy in metastatic breast cancer: SWOG S0500. J Clin Oncol 2014; 32: 3483–3489.
    1. Miyamoto DT, Lee RJ, Stott SL, et al. Androgen receptor signaling in circulating tumor cells as a marker of hormonally responsive prostate cancer. Cancer Disc 2012; 2: 995–1003.
    1. Antonarakis ES, Lu C, Wang H, et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med 2014; 371: 1028–1038.
    1. Thadani-Mulero M, Portella L, Sun S, et al. Androgen receptor splice variants determine taxane sensitivity in prostate cancer. Cancer Res 2014; 74: 2270–2282.
    1. Luo X, Mitra D, Sullivan RJ, et al. Isolation and molecular characterization of circulating melanoma cells. Cell Rep 2014; 7: 645–653.
    1. Sakaizawa K, Goto Y, Kiniwa Y, et al. Mutation analysis of BRAF and KIT in circulating melanoma cells at the single cell level. Br J Cancer 2012; 106: 939–946.
    1. Maheswaran S, Sequist LV, Nagrath S, et al. Detection of mutations in EGFR in circulating lung-cancer cells. N Engl J Med 2008; 359: 366–377.
    1. Gasch C, Bauernhofer T, Pichler M, et al. Heterogeneity of epidermal growth factor receptor status and mutations of KRAS/PIK3CA in circulating tumor cells of patients with colorectal cancer. Clin Chem 2013; 59: 252–260.
    1. Kondo Y, Hayashi K, Kawakami K, et al. KRAS mutation analysis of single circulating tumor cells from patients with metastatic colorectal cancer. BMC Cancer 2017; 17: 311.
    1. Nicolazzo C, Raimondi C, Mancini M, et al. Monitoring PD-L1 positive circulating tumor cells in non-small cell lung cancer patients treated with the PD-1 inhibitor nivolumab. Sci Rep 2016; 6: 31726.
    1. Ito H, Sato J, Tsujino Y, et al. Long-term prognostic impact of circulating tumour cells in gastric cancer patients. World J Gastroenterol 2016; 22: 10232–10241.
    1. Pimienta M, Edderkaoui M, Wang R, et al. The potential for circulating tumor cells in pancreatic cancer management. Front Physiol 2017; 8: 381.
    1. Wu XL, Tu Q, Faure G, et al. Diagnostic and prognostic value of circulating tumor cells in head and neck squamous cell carcinoma: a systematic review and meta-analysis. Sci Rep 2016; 6: 20210.
    1. Khan MS, Kirkwood A, Tsigani T, et al. Circulating tumor cells as prognostic markers in neuroendocrine tumors. J Clin Oncol 2013; 31: 365–372.
    1. Satelli A, Mitra A, Cutrera JJ, et al. Universal marker and detection tool for human sarcoma circulating tumor cells. Cancer Res 2014; 74: 1645–1650.
    1. Krebs MG, Sloane R, Priest L, et al. Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer. J Clin Oncol 2011; 29: 1556–1563.
    1. Sefrioui D, Blanchard F, Toure E, et al. Diagnostic value of CA19.9, circulating tumour DNA and circulating tumour cells in patients with solid pancreatic tumours. Br J Cancer 2017; 117: 1017–1025.
    1. Mascalchi M, Maddau C, Sali L, et al. Circulating tumor cells and microemboli can differentiate malignant and benign pulmonary lesions. J Cancer 2017; 8: 2223–2230.
    1. Pixberg CF, Schulz WA, Stoecklein NH, Neves RP. Characterization of DNA methylation in circulating tumor cells. Genes 2015; 6: 1053–1075.
    1. Chimonidou M, Strati A, Tzitzira A, et al. DNA methylation of tumor suppressor and metastasis suppressor genes in circulating tumor cells. Clin Chem 2011; 57: 1169–1177.
    1. Friedlander TW, Ngo VT, Dong H, et al. Detection and characterization of invasive circulating tumor cells derived from men with metastatic castration-resistant prostate cancer. Int J Cancer 2014; 134: 2284–2293.
    1. Lyberopoulou A, Galanopoulos M, Aravantinos G, et al. Identification of methylation profiles of cancer-related genes in circulating tumor cells population. Anticancer Res 2017; 37: 1105–1112.
    1. Mastoraki S, Strati A, Tzanikou E, et al. ESR1 Methylation: A liquid biopsy-based epigenetic assay for the follow-up of patients with metastatic breast cancer receiving endocrine treatment. Clin Cancer Res 2018; 24: 1500–1510.
    1. Chimonidou M, Strati A, Malamos N, et al. Direct comparison study of DNA methylation markers in EpCAM-positive circulating tumour cells, corresponding circulating tumour DNA, and paired primary tumours in breast cancer. Oncotarget 2017; 8: 72054–72068.
    1. Pixberg CF, Raba K, Muller F, et al. Analysis of DNA methylation in single circulating tumor cells. Oncogene 2017; 36: 3223–3231.
    1. Micalizzi DS, Maheswaran S, Haber DA. A conduit to metastasis: circulating tumor cell biology. Genes Dev 2017; 31: 1827–1840.
    1. Manicone M, Poggiana C, Facchinetti A, et al. Critical issues in the clinical application of liquid biopsy in non-small cell lung cancer. J Thorac Dis 2017; 9: S1346-S1358.
    1. Pestrin M, Salvianti F, Galardi F, et al. Heterogeneity of PIK3CA mutational status at the single cell level in circulating tumor cells from metastatic breast cancer patients. Mol Oncol 2015; 9: 749–757.
    1. Sundaresan TK, Sequist LV, Heymach JV, et al. Detection of T790M, the Acquired Resistance EGFR Mutation, by Tumor Biopsy versus Noninvasive Blood-Based Analyses. Clin Cancer Res 2016; 22: 1103–1110.
    1. Jordan NV, Bardia A, Wittner BS, et al. HER2 expression identifies dynamic functional states within circulating breast cancer cells. Nature 2016; 537: 102–106.
    1. Markou A, Lazaridou M, Paraskevopoulos P, et al. Multiplex gene expression profiling of in vivo isolated circulating tumor cells in high-risk prostate cancer patients. Clin Chem 2018; 64: 297–306.
    1. Mandel P, Metais P. [Not Available]. Comptes rendus des seances de la Societe de biologie et de ses filiales. 1948; 142: 241–243.
    1. Tan EM, Schur PH, Carr RI, Kunkel HG. Deoxybonucleic acid (DNA) and antibodies to DNA in the serum of patients with systemic lupus erythematosus. J Clin Invest 1966; 45: 1732–1740.
    1. Leon SA, Shapiro B, Sklaroff DM, Yaros MJ. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res 1977; 37: 646–650.
    1. Sorenson GD, Pribish DM, Valone FH, et al. Soluble normal and mutated DNA sequences from single-copy genes in human blood. Cancer Epidemiol Biomarkers Prev 1994; 3: 67–71.
    1. Stroun M, Lyautey J, Lederrey C, et al. About the possible origin and mechanism of circulating DNA apoptosis and active DNA release. Clin Chim Acta 2001; 313: 139–142.
    1. Jahr S, Hentze H, Englisch S, et al. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res 2001; 61: 1659–1665.
    1. Mouliere F, Robert B, Arnau Peyrotte E, et al. High fragmentation characterizes tumour-derived circulating DNA. PLoS One 2011; 6: e23418.
    1. Bergsmedh A, Szeles A, Henriksson M, et al. Horizontal transfer of oncogenes by uptake of apoptotic bodies. Proc Natl Acad Sci U S A 2001; 98: 6407–6411.
    1. Fleischhacker M, Schmidt B. Circulating nucleic acids (CNAs) and cancer–a survey. Biochim Biophys Acta 2007; 1775: 181–232.
    1. Lo YM, Zhang J, Leung TN, et al. Rapid clearance of fetal DNA from maternal plasma. Am J Hum Genet 1999; 64: 218–224.
    1. Yao W, Mei C, Nan X, Hui L. Evaluation and comparison of in vitro degradation kinetics of DNA in serum, urine and saliva: a qualitative study. Gene 2016; 590: 142–148.
    1. Atamaniuk J, Vidotto C, Tschan H, et al. Increased concentrations of cell-free plasma DNA after exhaustive exercise. Clin Chem 2004; 50: 1668–70.
    1. Diehl F, Schmidt K, Choti MA, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med 2008; 14: 985–990.
    1. Underhill HR, Kitzman JO, Hellwig S, et al. Fragment length of circulating tumor DNA. PLoS Genet 2016; 12: e1006162.
    1. Chan KC, Yeung SW, Lui WB, et al. Effects of preanalytical factors on the molecular size of cell-free DNA in blood. Clin Chem 2005; 51: 781–784.
    1. Su YH, Wang M, Aiamkitsumrit B, et al. Detection of a K-ras mutation in urine of patients with colorectal cancer. Cancer Biomark 2005; 1: 177–182.
    1. Wang Y, Springer S, Mulvey CL, et al. Detection of somatic mutations and HPV in the saliva and plasma of patients with head and neck squamous cell carcinomas. Sci Transl Med 2015; 7: 293ra104.
    1. De Mattos-Arruda L, Mayor R, Ng CK, et al. Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nat Commun 2015; 6: 8839.
    1. Barra GB, Santa Rita TH, de Almeida Vasques J, et al. EDTA-mediated inhibition of DNases protects circulating cell-free DNA from ex vivo degradation in blood samples. Clin Biochem 2015; 48: 976–981.
    1. El Messaoudi S, Rolet F, Mouliere F, Thierry AR. Circulating cell free DNA: Preanalytical considerations. Clin Chim Acta 2013; 424: 222–230.
    1. Parpart-Li S, Bartlett B, Popoli M, et al. The Effect of Preservative and Temperature on the Analysis of Circulating Tumor DNA. Clin Cancer Res 2017; 23: 2471–2477.
    1. Han X, Wang J, Sun Y. Circulating Tumor DNA as Biomarkers for Cancer Detection. Genomics Proteomics Bioinformatics 2017; 15: 59–72.
    1. Parsons HA, Beaver JA, Park BH. Circulating Plasma Tumor DNA. Adv Exp Med Biol 2016; 882: 259–276.
    1. Wan JCM, Massie C, Garcia-Corbacho J, et al. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat Reviews Cancer 2017; 17: 223–238.
    1. Diehl F, Li M, He Y, et al. BEAMing: single-molecule PCR on microparticles in water-in-oil emulsions. Nat Method 2006; 3: 551–559.
    1. Li M, Diehl F, Dressman D, et al. BEAMing up for detection and quantification of rare sequence variants. Nat Method 2006; 3: 95–97.
    1. Rothe F, Laes JF, Lambrechts D, et al. Plasma circulating tumor DNA as an alternative to metastatic biopsies for mutational analysis in breast cancer. Ann Oncol 2014; 25: 1959–1965.
    1. Forshew T, Murtaza M, Parkinson C, et al. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med 2012; 4: 136ra68.
    1. Newman AM, Bratman SV, To J, et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med 2014; 20: 548–554.
    1. Kinde I, Wu J, Papadopoulos N, et al. Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci U S A 2011; 108: 9530–9535.
    1. Shu Y, Wu X, Tong X, et al. Circulating tumor DNA mutation profiling by targeted next generation sequencing provides guidance for personalized treatments in multiple cancer types. Sci Rep 2017; 7: 583.
    1. Chen S, Liu M, Zhou Y. Bioinformatics analysis for cell-free tumor DNA sequencing data. Methods Mol Biol 2018; 1754: 67–95.
    1. Leary RJ, Kinde I, Diehl F, et al. Development of personalized tumor biomarkers using massively parallel sequencing. Sci Transl Med 2010; 2: 20ra14.
    1. Herman JG, Graff JR, Myohanen S, et al. Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci U S A 1996; 93: 9821–9826.
    1. Licchesi JD, Herman JG. Methylation-specific PCR. Methods Mol Biol 2009; 507: 305–323.
    1. Fackler MJ, Sukumar S. Quantitation of DNA Methylation by Quantitative Multiplex Methylation-Specific PCR (QM-MSP) Assay. Methods Mol Biol 2018; 1708: 473–496.
    1. Fackler MJ, Lopez Bujanda Z, Umbricht C, et al. Novel methylated biomarkers and a robust assay to detect circulating tumor DNA in metastatic breast cancer. Cancer Res 2014; 74: 2160–2170.
    1. Bailey VJ, Zhang Y, Keeley BP, et al. Single-tube analysis of DNA methylation with silica superparamagnetic beads. Clin Chem 2010; 56: 1022–1025.
    1. Bailey VJ, Keeley BP, Zhang Y, et al. Enzymatic incorporation of multiple dyes for increased sensitivity in QD-FRET sensing for DNA methylation detection. Chembiochem 2010; 11: 71–74.
    1. Chan KC, Jiang P, Chan CW, et al. Noninvasive detection of cancer-associated genome-wide hypomethylation and copy number aberrations by plasma DNA bisulfite sequencing. Proc Natl Acad Sci U S A 2013; 110: 18761–18768.
    1. 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: 1250–1264.
    1. Beck J, Urnovitz HB, Riggert J, Clerici M, Schutz E. Profile of the circulating DNA in apparently healthy individuals. Clin Chem 2009; 55: 730–738.
    1. Mao L, Hruban RH, Boyle JO, Tockman M, Sidransky D. Detection of oncogene mutations in sputum precedes diagnosis of lung cancer. Cancer Res 1994; 54: 1634–1637.
    1. Gormally E, Vineis P, Matullo G, et al. TP53 and KRAS2 mutations in plasma DNA of healthy subjects and subsequent cancer occurrence: a prospective study. Cancer Res 2006; 66: 6871–6876.
    1. Fernandez-Cuesta L, Perdomo S, Avogbe PH, et al. Identification of circulating tumor DNA for the early detection of small-cell lung cancer. EBioMed 2016; 10: 117–123.
    1. Genovese G, Kahler AK, Handsaker RE, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med 2014; 371: 2477–2487.
    1. Sozzi G, Conte D, Leon M, et al. Quantification of free circulating DNA as a diagnostic marker in lung cancer. J Clin Oncol 2003; 21: 3902–3908.
    1. Kato S, Lippman SM, Flaherty KT, Kurzrock R. The conundrum of genetic ‘Drivers’ in benign conditions. J Natl Cancer Inst 2016; 108.
    1. Kim K, Shin DG, Park MK, et al. Circulating cell-free DNA as a promising biomarker in patients with gastric cancer: diagnostic validity and significant reduction of cfDNA after surgical resection. Ann Surg Treat Res 2014; 86: 136–142.
    1. Frattini M, Gallino G, Signoroni S, et al. Quantitative and qualitative characterization of plasma DNA identifies primary and recurrent colorectal cancer. Cancer Lett 2008; 263: 170–181.
    1. Tie J, Wang Y, Tomasetti C, et al. Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer. Sci Transl Med 2016; 8: 346ra92.
    1. Garcia-Murillas I, Schiavon G, Weigelt B, et al. Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Sci Transl Med 2015; 7: 302ra133.
    1. Olsson E, Winter C, George A, et al. Serial monitoring of circulating tumor DNA in patients with primary breast cancer for detection of occult metastatic disease. EMBO Mol Med 2015; 7: 1034–1047.
    1. Bardelli A, Pantel K. Liquid Biopsies, What We Do Not Know (Yet). Cancer Cell 2017; 31: 172–179.
    1. Garcia JM, Garcia V, Silva J, et al. Extracellular tumor DNA in plasma and overall survival in breast cancer patients. Genes Chromosomes Cancer 2006; 45: 692–701.
    1. Castells A, Puig P, Mora J, et al. K-ras mutations in DNA extracted from the plasma of patients with pancreatic carcinoma: diagnostic utility and prognostic significance. J Clin Oncol 1999; 17: 578–584.
    1. Dianxu F, Shengdao Z, Tianquan H, et al. A prospective study of detection of pancreatic carcinoma by combined plasma K-ras mutations and serum CA19–9 analysis. Pancreas 2002; 25: 336–341.
    1. Kopreski MS, Benko FA, Borys DJ, et al. Somatic mutation screening: identification of individuals harboring K-ras mutations with the use of plasma DNA. J Natl Cancer Inst 2000; 92: 918–923.
    1. Shinozaki M, O’Day SJ, Kitago M, et al. Utility of circulating B-RAF DNA mutation in serum for monitoring melanoma patients receiving biochemotherapy. Clin Cancer Res 2007; 13: 2068–2074.
    1. Dawson SJ, Tsui DW, Murtaza M, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med 2013; 368: 1199–1209.
    1. Misale S, Arena S, Lamba S, et al. Blockade of EGFR and MEK intercepts heterogeneous mechanisms of acquired resistance to anti-EGFR therapies in colorectal cancer. Sci Transl Med 2014; 6: 224ra26.
    1. Diaz LA, Jr., Williams RT, Wu J, et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 2012; 486: 537–540.
    1. Thress KS, Brant R, Carr TH, et al. EGFR mutation detection in ctDNA from NSCLC patient plasma: a cross-platform comparison of leading technologies to support the clinical development of AZD9291. Lung Cancer 2015; 90: 509–515.
    1. Sequist LV, Yang JC, Yamamoto N, et al. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol 2013; 31: 3327–3334.
    1. Douillard JY, Ostoros G, Cobo M, et al. First-line gefitinib in Caucasian EGFR mutation-positive NSCLC patients: a phase-IV, open-label, single-arm study. Br J Cancer 2014; 110: 55–62.
    1. Wu YL, Zhou C, Liam CK, et al. First-line erlotinib versus gemcitabine/cisplatin in patients with advanced EGFR mutation-positive non-small-cell lung cancer: analyses from the phase III, randomized, open-label, ENSURE study. Ann Oncol 2015; 26: 1883–189.
    1. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol Mol Sci. 2013; 200: 373–383.
    1. Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol 2002; 2: 569–579.
    1. Pan BT, Johnstone RM. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor. Cell 1983; 33: 967–978.
    1. Harding CV, Heuser JE, Stahl PD. Exosomes: looking back three decades and into the future. J Cell Biol Mol Sci. 2013; 200: 367–371.
    1. Weidle UH, Birzele F, Kollmorgen G, Ruger R. The Multiple Roles of Exosomes in Metastasis. Cancer Genomics Proteomics 2017; 14: 1–15.
    1. Tucci M, Mannavola F, Passarelli A, Stucci LS, Cives M, Silvestris F. Exosomes in melanoma: a role in tumor progression, metastasis and impaired immune system activity. Oncotarget 2018; 9: 20826–20837.
    1. Giallombardo M, Chacartegui Borras J, Castiglia M, et al. Exosomal miRNA analysis in non-small cell lung cancer (NSCLC) patients’ plasma through qPCR: a feasible liquid biopsy tool. J Vis Exp 2016.
    1. Thakur BK, Zhang H, Becker A, et al. Double-stranded DNA in exosomes: a novel biomarker in cancer detection. Cell Res 2014; 24: 766–769.
    1. Logozzi M, De Milito A, Lugini L, et al. High levels of exosomes expressing CD63 and caveolin-1 in plasma of melanoma patients. PLoS One 2009; 4: e5219.
    1. Nilsson J, Skog J, Nordstrand A, et al. Prostate cancer-derived urine exosomes: a novel approach to biomarkers for prostate cancer. Br J Cancer 2009; 100: 1603–1607.
    1. Skog J, Wurdinger T, van Rijn S, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol 2008; 10: 1470–1476.
    1. Li P, Kaslan M, Lee SH, et al. Progress in exosome isolation techniques. Theranostics. 2017; 7: 789–804.
    1. Thery C, Amigorena S, Raposo G, et al. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol. 2006; Chapter 3: Unit 3 22.
    1. Lane RE, Korbie D, Anderson W, Vaidyanathan R, Trau M. Analysis of exosome purification methods using a model liposome system and tunable-resistive pulse sensing. Sci Rep 2015; 5: 7639.
    1. Peterson MF, Otoc N, Sethi JK, Gupta A, Antes TJ. Integrated systems for exosome investigation. Methods 2015; 87: 31–45.
    1. Costa-Silva B, Aiello NM, Ocean AJ, et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol 2015; 17: 816–826.
    1. Mitchell PJ, Welton J, Staffurth J, et al. Can urinary exosomes act as treatment response markers in prostate cancer? J Transl Med 2009; 7: 4.
    1. Kim Y, Ignatchenko V, Yao CQ, et al. Identification of differentially expressed proteins in direct expressed prostatic secretions of men with organ-confined versus extracapsular prostate cancer. Mol Cell Proteomics 2012; 11: 1870–1884.
    1. Huang X, Yuan T, Tschannen M, et al. Characterization of human plasma-derived exosomal RNAs by deep sequencing. BMC Genomics 2013; 14: 319.
    1. Madhavan B, Yue S, Galli U, et al. Combined evaluation of a panel of protein and miRNA serum-exosome biomarkers for pancreatic cancer diagnosis increases sensitivity and specificity. Int J Cancer 2015; 136: 2616–2627.
    1. Lunavat TR, Cheng L, Einarsdottir BO, et al. BRAF(V600) inhibition alters the microRNA cargo in the vesicular secretome of malignant melanoma cells. Proc Natl Acad Sci U S A 2017; 114: E5930-E5939.
    1. Kahlert C, Melo SA, Protopopov 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.
    1. Tucci M, Passarelli A, Mannavola F, et al. Serum exosomes as predictors of clinical response to ipilimumab in metastatic melanoma. Oncoimmunology 2018; 7: e1387706.
    1. Inamdar S, Nitiyanandan R, Rege K. Emerging applications of exosomes in cancer therapeutics and diagnostics. Bioeng Transl Med 2017; 2: 70–80.
    1. Jamal-Hanjani M, Wilson GA, McGranahan N, et al. Tracking the Evolution of Non-Small-Cell Lung Cancer. N Engl J Med 2017; 376: 2109–2121.
    1. Harouaka R, Kang Z, Zheng SY, et al. Circulating tumor cells: advances in isolation and analysis, and challenges for clinical applications. Pharmacol Ther 2014; 141: 209–221.
    1. Mohan S, Chemi F, Brady G. Challenges and unanswered questions for the next decade of circulating tumour cell research in lung cancer. Transl Lung Cancer Res 2017; 6: 454–472.
    1. Wu T, Cheng B, Fu L. Clinical applications of circulating tumor cells in pharmacotherapy: challenges and perspectives. Mol Pharmacol 2017; 92: 232–239.
    1. Verlingue L, Alt M, Kamal M, et al. Challenges for the implementation of high-throughput testing and liquid biopsies in personalized medicine cancer trials. Pers Med 2014; 11: 545–558.
    1. Castro-Giner F, Gkountela S, Donato C, et al. Cancer Diagnosis Using a Liquid Biopsy: Challenges and Expectations. Diagnostics 2018; 8.
    1. Salvianti F, Pazzagli M, Pinzani P. Single circulating tumor cell sequencing as an advanced tool in cancer management. Expert Rev Mol Diagn 2016; 16: 51–63.

Source: PubMed

3
Prenumerera