Skip to main content
International Journal of Hematology-Oncology and Stem Cell Research logoLink to International Journal of Hematology-Oncology and Stem Cell Research
. 2018 Jul 1;12(3):235–248.

Liquid Biopsy in Thyroid Cancer: New Insight

Fatemeh Khatami 1, Seyed Mohammad Tavangar 1,2
PMCID: PMC6305265  PMID: 30595827

Abstract

Thyroid cancer, one of the most widespread malignancies of the endocrine-related system that over the past three decades, has a vivid increasing rate. The diagnosis and management of it is dependent on the tumor type and stage. Thyroid cancer is divided into four main types, including PTC (papillary thyroid carcinoma), FTC (follicular thyroid carcinoma), MTC (medullarly thyroid carcinoma), and ATC (anaplastic thyroid carcinoma). The development of the noninvasive diagnostic tool for plasma genotyping, also known as “liquid biopsy”, brings a new insight for cancer diagnosis and prognosis. It is mainly containing circulating tumor DNA (ctDNA), circulating tumor cell (CTC), exosomes and extrachromosomal circular DNA (ecDNA). Liquid biopsy as a new plasma genotyping source brings a new prospective of tumor monitoring and therapy. It beneficially reduces the need of tissue biopsy and made early recognition of relapse as well. This article summarizes its components characteristics and their benefit in diagnosis and treatment of thyroid cancer.

Key Words: Biopsy, Carcinoma, Thyroid cancer, Endocrine system diseases, cfDNA, CTCs

Introduction

Main components of liquid biopsy

It is needless to say that tissue biopsies have some weak points like being invasive and useless in understanding metastatic risk, disease progression, and treatment effectiveness more than being hard for repeating 1. Over the past few decades, the new real-time diagnostic tool which is referred as ‘‘liquid biopsy’’ has been considered in different type of cancer enormously 2-4. In contrary to analysis of solid tumors requirement as an invasive procedures, blood tests are easy and safe to carry out and several samples can be taken over time. Actually, the concept of liquid biopsy is composed of circulating tumor DNA (ctDNA), circulating tumor cells (CTCs) and exosomes (Figure1) which will be considered in this review in details.

Figure1.

Figure1

Main components of liquid biopsy for genetic and epigenetic analysis of thyroid tumors.

Circulating tumor DNA (ctDNA)

It was in 1940 that for the first time the presence of extracellular or cell-free nucleic acids was recognized by Mandel and Metais 5, after that the rheumatologic literature talked about the existence ctDNA in the 19806. Finally, patients with cancer were identified to have high levels of cell-free DNA (cfDNA) in their plasma like patients with benign diseases including inflammatory bowel disease7. In fact, ctDNA are calculated to be presented in blood with the length of 160 to 200 base pairs (bp), predominantly 166 bp long that are released by tumor cells into the bloodstream 8-10. They are a genetic representative of tumor which contains the exact genetic defects identical to their original tumor cells. Interestingly, all molecular variations, including point mutations, rearrangements, amplifications and gene copy variations are easily detectable in plasma's ctDNAs. Cell free DNAs are not completely limited to cancer cells for the reasons that live cells naturally shedding DNA fragments as a part of a homeostatic process 11-14 On the other hand, cancer patients usually have far elevated amount of ctDNA than healthy individuals from 0.01% to more than 90%15-17. The logic of this variable amount of ctDNA levels in cancer patients can be connected to the tumor burden of tumor, tumor stage, and efficacy of treatments 17, 18. Although the exact mechanism of coming off ctDNAs into plasma is not clear completely, some suggesting biological processes could be involved, including apoptosis and necrosis from dying cells, or active release from viable tumor cells 12, 19-22. The cfDNA molecular alterations in plasma can reveal the status of the human body in a timely manner, therefore a study designed to check the background somatic mutations in white blood cells (WBC) and cfDNA for healthy controls 23. In order to realize the pattern and source cfDNA mutations, a panel of 50 cancer-associated genes was analyzed in both WBC and cfDNA groups23. It was shown that most of mutations in cfDNA originated from WBC and NPM1 gene was the most frequently mutant gene in both WBC and cfDNA22,23.

In normal physiologic conditions, apoptotic and necrotic cells are removed through phagocytes, so ctDNA levels in serum or plasma are quite low, but this mechanism is not applicable in tumor cells. It is possible that in solid tumors ctDNA release through necrosis, autophagy, and other physiologic 16, 24. It should be kept in mind that unlike apoptosis, necrosis DNA fragments are larger because of incomplete and random DNA digestion 25. By far, the most interesting spectacle which is brought to the science of oncology by ctDNA is called horizontal tumor gene transfer phenomenon mediated by circulating DNA 2, 26, 27. It explains that the impact of some other molecules than DNA in tumor formation cannot be ruled out; this is a pure fact that ctDNAs are biologically active DNA to raise tumor progression26. In fact, ctDNA represents all genetic alterations which exist in the tumoral genomic DNA, so ctDNA carries genomic and epigenomic alterations such as point mutations, loss of heterozygosity (LOH), rearranged genomic sequences, microsatellite instability (MSI), copy number variation (CNV) and DNA methylation28-30. The results of whole-genome sequencing analysis of ctDNA made it clear that copy number variation (CNV) and Single-nucleotide polymorphisms (SNPs) were noticed in all malignant tumors, but not in healthy individuals31. Chan et al. applied shotgun massively parallel sequencing approach in the plasma of cancer patients and successfully completed the whole genome-wide sequencing of CNVs and point mutations32. In 1996, recognition of microsatellite instability and loss of heterozygosity in ctDNA were first described by Nawroz et al 33. DNA methylation as epigenetic change plays crucial roles in gene expression regulation and genetic alteration34,35. In fact, alteration of DNA methylation in the non-coding and promoter region of genes can be connected with tumor formation, tumor development, and metastatic spread 36, 37. In 1999, unusual DNA methylations were noticed in the plasma and serum of lung 38, 39, breast 40 and liver cancers41. Afterwards, several researches have pointed out that ctDNA methylation can be considered as an excellent candidate for diagnostic and prognostic of cancer 29,42-44. Methylation profiling of ctDNA in esophageal cancer patients mentioned the highly significant differences in the methylation status between ctDNA and equivalent tumor tissues45. It should be kept in mind that contrary to ctDNA free, RNA molecules are not able to survive in the bloodstream. There is an exception about cell-free microRNAs that can be noticed in plasma or serum of cancer patients46. Indeed, detecting RNA molecules could be possible through extracellular vesicles such as exosomes (both coding and non-coding) in platelets47,48. More than cfDNA and cfRNA there are some extrachromosomal circular DNA (ecDNA) which are newly suggested to be presented in blood as a liquid biopsy component 49.

Circulating Tumor Cells (CTCs)

The key reason of cancer-associated death is tumor metastasis, unfortunately, the knowledge of this procedure has not completed yet. In fact, dissemination regularly occurs through the blood, so circulating tumor cells (CTCs) as a candidate circulating element are interesting50. Circulating tumor cells (CTCs) are circulating cells in the vasculature or lymphatic’s which are released from primary tumors 51, 52. CTCs have the leading role in metastasis which is a key step in the progression of tumors in other distant organs and responsible for the majority of cancer-related deaths53. Although for the first time in 1869 CTCs were observed by Thomas Ashworth in the blood of a man with metastatic cancer 54, the value of CTCs in modern cancer research instigated in the mid of 1990s by [J. Uhr, UT-Dallas, L. Terstappen and P. Liberti, Immunicon, Philadelphia]. Afterwards, some cancer researches have confirmed that CTCs are derived from primary tumor55. Moreover, noteworthy efforts in understanding the biological properties of CTCs have confirmed their critical role in the metastatic spread of carcinoma56. Up to now, several technologies with the essential sensitivity and reproducibility to identify CTCs in patients with metastatic disease have recently been developed 57-63. Several studies have shown that the detection of CTCs in the peripheral blood of patients with lung cancer may have prognostic and predicting efficacy in treatment with chemotherapy39, 64-66.

The ‘seed and soil’ theory which is related to tumor invasion and dissemination was launched in 1889 67. According to this theory, the basic properties of the tumor cells as a seed and host microenvironment as a soil are main determinants of tumor formation sites56,68,69. Without a doubt, the hypothesis that some CTCs direct ‘tumor-initiating’ process has been supposed because CTCs are proficient to seed detached metastatic disease70, 71. Some reviews of the metastatic process supposed that a reversible epithelial-to-mesenchymal transition (EMT) as a crucial step of metastasis is completely dependent on CTC 72-74. For diagnosis and treatment of breast cancer, CTCs are among the most extensively studied ones75,76. There is a definite correlation between CTCs and breast cancer prognosis and survival76. CTCs have been reported to harbor many types of mutations and transformations, but, according to the result of a systematic review, the clinical implication of CTCs molecular characteristics, including Her2, EGFR, CEA, CA15-3, CK19, Ki67, PIK3CA, TGF-β and CXCL1 is more truthful than enumeration of CTCs before and during treatment, especially for making the best personalized treatment decision76-79. Moreover, the change in the number of CTCs in the field of treatment strategies and drug development could be valuable because patients with a remarkable reduction in CTC count after treatment usually show better outcomes80-83.

Exosomes

It was established that exosomes are cell-derived nucleic-acid- and protein-rich nanoparticles which are floating in almost all bodily fluids 84, 85. Actually, exosomes are small particles with a diameter of 30 - 100 nm, which is larger than low-density lipoproteins (LDL) and much smaller than red blood cells. The presence of membranous vesicles outside cells in eukaryotic fluids, including blood and urine, was acknowledged 50 years ago although at that time they were assumed as useless products releasing from plasma membrane86, 87. Exosomes can exist in various biological fluids, such as plasma and urine 88. At first, exosomes were taken in to account for having role in the removal of needless molecules, after a while some valuable studies clarified exosomes’ complex function in tumor progression and metastasis88. They are released from eukaryotic cells when multi-vesicular bodies are fused with the plasma membrane or when they can straightly release from the plasma membrane89. The potential of exosomes as a cancer diagnostic tool has been tested for lung cancer90 and prostate cancer91. Interestingly, the advantage of exosomes is that they are predominant in the bloodstream than CTCs92.

Liquid biopsy detection and characterizations methods

The most important step for liquid biopsy analysis is detection and characterization of them in cancer patients. Thanks to recent developments in sequencing technologies like the digital polymerase chain reaction (dPCR) and next-generation sequencing (NGS), now it is easily possible to be detected in blood93-95. Nowadays, numerous dPCR systems which are droplet-based platforms such as QX200 Droplet Digital PCR System (Bio-Rad Laboratories), RainDrop Digital PCR System (RainDance Technologies) with very high sensitivity are industrialized96,97. Moreover, NGS techniques can analyze multiple, broad regions of target ctDNA94,98,99.Other foremost techniques for detection of mutations in specific genomic regions of ctDNA are “Ion AmpliSeq Technology (Thermo Fisher Scientific)” and “Ion Personal Genome Machine (Ion PGM)”100,101. Also, there are some target capture-based platforms like Sure Select Target Enrichment System (Agilent Technologies) which is generally active for targeted sequencing in combination with the Illumina paired-end sequencing102,103. Interestingly, it was described that Personalized Profiling deep Sequencing of Rearranged Ends can help to the finding of personalized cancer biomarkers 104, 105.

Techniques for detecting Circulating tumor cells are mostly related to the enrichment of CTCs according to different properties of CTCs that discriminate them from other normal hematopoietic cells. Some physical properties are dimensions, density, electric charges, and some biological characteristics are cell surface molecular markers. Epithelial-marker based approaches are the most common practical strategies for CTC detection based on epithelial markers like Cell Search system which is the only FDA-approved platform for CTC detection in clinical practice on patients with breast, prostate, and colorectal cancers106. Moreover, presentation of a mixture of different epithelial markers could be helpful to recover additional epithelium-originating tumor cells107. Low blood volume as limiting step can also be solved by Cell Collector which used EpCAM antibody-coated wire to capture CTCs in vivo108. There is also the chip-based platform CTC-iChip that is an excellent combination of size-based selection and label-dependent enrichment109. Additional sized-based approaches are ISET110, Screen Cell and Can Patrol111, Parsotix112 and JETTA 93 systems.

Over the past few decades, many techniques have been developed in order to characterization of exosomes from biological fluids. Usually, biophysical methods are zoomed on the exosomal size range like optical particle tracking which is a method that quantify the size of exosomes from 10 nm to 2 µm and the velocity of the particles113-119. Additionally, some microfluidic-based methodologies could be used for exosomal characterization as well120-122. More than exosomal size, the exosome specific molecular markers like proteins and nucleic acids are suitable markers for tumor tracking. As a matter of fact, exosomes are released through both normal and cancerous cells and include several membrane and cytoplasmic proteins. Consequently, its proteins like Enolase 1, Heat shock protein 8 (HSPA8), α (cytosolic), and class A member 1 (HSP90AA1) can be important in clinical diagnostics123, 124. Generally, it could be said that exosomal proteins are allocated to the different functional categories such as tetraspanins (CD9, CD63 and CD81), heat shock proteins (HSC70 and HSC90), membrane transporters (GTPases) and lipid-bound proteins125. Not only exosomes are involved in the pathogenesis of cancers but also they are involved in neurodegenerative pathologies, including Alzheimer’s, Parkinson’s and Creutzfeldt-Jakob diseases126. Exosomal microRNAs can be useful for diagnostic of several cancer types, for example, some miRNAs were distinguished to be particular biomarkers of ovarian cancer127,128. In patients with lung adenocarcinoma, prostate cancer and esophageal squamous cell cancer (ESCC), the levels of exosomal miRNAs have increased 129-131. Also, exosomal microRNAs may be possible indicative biomarkers for renal fibrosis 132 and heart failure133. Several companies have improved different technologies for ctDNA, CTCs and detection and characterization of exosomes (Table1).

Table 1.

The liquid biopsy detection and characterization techniques in experimental applications

Technique Descriptions
CTC-Chip Capture CTCs by using EpCAM- coated microposts under strict manipulation of velocity and shear force
CTC-iChip The CTC-iChip is composed of two separate microfluidic devices that house three different microfluidic
components engineered for inline operation: DLD to remove nucleated cells from whole blood by size-based
deflection by using a specially designed array of posts performed in CTC-iChip1, inertial focusing to line up cells
to prepare for precise magnetic separation and magnetophoresis for sensitive separation of bead-labeled WBCs
and unlabeled CTCs, which are performed in CTC-iChip2. PLTs, platelets
Adna Test Adna Test has a combination of antibodies that bind with high specificity and affinity to epitopes or antigens on
the relevant cancer cells. After magnetic separation, the enriched cells are lysed and purified several time to
make the relevant tumor cell information available in the form of mRNA.
EPISPOT( Epithelial Immuno SPOT) CTCs are enriched by negative depletion and subsequently cultured on a membrane coated with antibodies that
capture the secreted proteins. Afterward, the proteins are readily identifiable by immune fluorescence
microscopy using fluorochrome-labeled secondary antibodies targeting the protein of interest.
Photoacoustic flowmetry Making use of the broadband absorption spectrum of melanin, it has been tested to detect melanoma cells and
has been combined with nanoparticles targeting cell surface antigens to broaden its applicability in CTC
detection.
 
Affinity based assays
Cell Search
The only FDA-approved technology for CTC detection is based on immune magnetic enrichment. It employs an
immunomagnetic enrichment step to isolate cells that express the epithelial cells’ adhesion molecule (EpCAM).
Additionally, to be identified as a CTC, the cellmust contain a nucleus, express cytoplasmic cytokeratin, and
have a diameter larger than 5μm. This technology has demonstrated the prognostic utility of enumerating and
monitoringCTC counts in patients with metastatic breast, prostate, and colorectal cancers. Semi-automated
analyzer enriches CTCs with ferrofluid nanoparticles coated with anti-EpCAM antibodies, then CD45-, CK8+,
CK18+ and CK19+ cells are counted by a four-color semi-automated fluorescence microscope
DEPArray (SiliconBiosystems) DEPArray™ technology is based on the ability of a non-uniform electric field to exert forces on neutral,
polarizable particles, such as cells, that are suspended in a liquid. This electrokinetic principle, called
dielectrophoresis (DEP), can be used to trap cells in DEP “cages” by creating an electric field above a subset of
electrodes in an array that is in counter phase with the electric field of adjacent electrodes. When a DEP cage is
moved by a change in the electric field pattern, the trapped cell moves with it.
MagSweeper A magnetic stir bar coated with an antibody to EpCAM. The device can process 9 mL of blood per hour and
purified cells of interest can be individually selected for subsequent molecular analysis, since the MagSweeper
technology preserves cell function and does not perturb gene expression.
Telomescan A novel cancer detection platform that measures telomerase activity from viable CTCs captured on a parylene-C
slot microfilter. Using a constant low pressure delivery system, the new microfilter platform is capable of cell
capture from 1 mL of whole blood in less than 5 min, achieving 90% capture efficiency. Addition of an
adenovirus-containing GFP to peripheral blood assay, incubation with cancer cells allows precise enumeration
and visualization of CTCs.

Thyroid Cancer

Thyroid cancer is the most common malignancy of the endocrine system with the remarkable increasing incidence rate over the last three decades134,135. According to the National Cancer Institute, the incidence of thyroid cancer has gotten higher with annually death rate of 0.8% from 2002 to 2011136-138. More often than not, thyroid cancer is diagnosed through Fine Needle Aspiration (FNA) biopsy, and tissue biopsy is classified into four main types, including 70% to 80% of thyroid cancers, papillary thyroid carcinoma (PTC) which is the least aggressive type of cancer139-143, follicular thyroid carcinoma (FTC), which is more aggressive than PTC, medullary thyroid carcinoma (MTC) that develops from C cells in the thyroid gland, and is more aggressive and less differentiated than papillary or follicular cancers and sometimes is associated with multiple endocrine neoplasia 2 (MEN2) and anaplastic thyroid carcinoma (ATC) that is the most dangerous form of thyroid cancer with the high capacity of metastasis to the adjacent lymph nodes and distant sites140,144. Treatment options for thyroid cancer, depending on its type and stage, are surgery, radioactive iodine (131I) therapy, and molecular-targeted therapies with a number of tyrosine kinase inhibitors (TKIs) 145. Several genetic and epigenetic alterations could have leading role for thyroid cancer like mutations leading to the activation of the MAPK and PI3K–AKT signaling pathways146, MMP2, caspase3147-149, survivin150 and nm23151. Point mutations of BRAF and RAS genes as well as RET/PTC and PAX8/PPARγ chromosomal rearrangements were found in thyroid cancer 146, 152-154. In addition to genetic mutations and rearrangements, there are epigenetic modifications which are suggested as important factors for thyroid cancer initiation and progression149, 155.

Liquid biopsy applications in thyroid cancer management

In order to real time monitoring of thyroid cancer from diagnosis to post treatment steps, some molecular markers of a noninvasive repeatable biopsy is needed, which means liquid biopsy can be the best candidate. Choosing plasma or serum as a source of cfDNA is challenging because serum apparently contains a greater quantity of free circulating DNA than plasma156. The underlying reason for this is unclear, but important because it may have clinical implications in interpreting results and using the appropriate resource156. Actually, high levels of circulating cell-free DNA (cf-DNA) have been established to associate with cancer diagnosis and progression. In 2013, it was shown by Mariangela Zane that hypermethylation of SLC5A8 and SLC26A4 genes that are both involved in the iodine metabolism and BRAFV600E mutation in ctDNA have valuable diagnostic value in thyroid cancer patients 149, 157.

Serum DNA methylation assessment as a novel diagnostic tool for thyroid cancer was introduced in 2006158. In that research, the evaluation of methylation status of five genes (CALCA, CDH1, TIMP3, DAPK, and RARβ2) been done by real-time quantitative methylation-specific PCR. Finally, they have confirmed the potential efficacy of serum DNA methylation markers as an innovative diagnostic marker for both patients with thyroid nodules and thyroid cancer recurrence in earlier treated patients158. Afterwards, the detectable free circulating BRAF in patients with PTC was mentioned as a possible determinant of tumor clinical implication159. Moreover, it was explained that decreasing levels of BRAFV600cfDNA were associated with longer tumor treating field 160. A higher amount of circulating mutant BRAFV600 in plasma was reported as a definite related factor with shorter overall survival in patients who were under BRAF/MEK inhibitors treatment160. ATC is so aggressive that needs rapid diagnosis and multimodality management. The University of Texas MD Anderson Cancer Center, between August 2015 and April 2016, run a research in which next-generation sequencing was used in twenty-three patients with ATC161. Based on those data, both tumor-based and cfDNA analysis usage in the setting of clinical-trial development and application was suggested161. Another aggressive thyroid tumor is medullary thyroid carcinoma which is triggered by activating mutations of the RET proto-oncogene receptor (RETM918T mutation) 162, 163. A cohort study was done by Caitlin Evers on 145 plasma samples from 98 patients (45 RETM918T tumor positive, 25 RETM918T tumor negative and 28 unidentified tumor mutation condition) by using Amplification Refractory Mutation System PCR (ARMS) and the Bio-Rad QX200™ Droplet Digital™ PCR system (ddPCR) (Bio-Rad Laboratories, Hercules, CA). Both ARMS and ddPCR are recommended for plasma DNA analysis in the way of mutation detection during disease progression162. For thyroid cancer, personalized medicine approach, interestingly the result of a research had revealed that Vemurafenib have its anti-tumor activity in patients with progressive, circulating BRAFV600E mutation positive refractory to radioactive iodine that had not been treated with a multi-kinase inhibitor drugs 164.

Not only circulating DNAs can be valuable source for real-time thyroid tumor tracking but also circulating RNAs have this potential. So, there are some studies which are focused on circulating RNAs in plasma of cancer patients. For example, BRAFV600E as an ordinary mutation of PTC is associated with insistent features of disease165. For evaluation of the viability and accuracy of a novel RNA-based blood assay to discriminate individuals with a high-risk tumor mutation in patients with PTC, circulating BRAFV600Elevels were compared with surgical pathologic DNA-based tissue BRAF mutation assays 165. The correlation of the RNA-based blood assay and tissue BRAF status was reported, so this RNA-based blood assay was described as an excellent biomarker for prognosis, surveillance, clinical decision making compared to BRAF-targeted therapies165. Additionally, exploring the plasma Long Non-Coding RNA (lncRNAs) for the finding of non-131I-avid lung metastases of PTC has been done 166. It was shown that two lncRNAs (ENST00000462717 and ENST00000415582) were up regulated and two (TCONS_00024700 and NR_028494) were down regulated in the non-131I-avid lung metastases of PTC166.

An interesting case report had illustrated that circulating epithelial cells (CECs) enumeration simplifies the identification and follow-up of a patient with early stage PTCs167. A panel of CEC quantification with serum thyroglobulin testing could be a valuable diagnostic marker for monitoring of thyroid cancer patients167. Some data make it evident that collective analysis of serum thyroglobulin with CECs, which are EpCAM positive, is completely applicable for patients at disease-free status and the patients with distant metastasis distinguishing 168. Therefore, CEC testing thereby can supplement the current standard methods for monitoring disease status of PTC167. High-resolution imaging for the detection and characterization of CTCs was used in patients with esophageal, hepatocellular, thyroid and ovarian cancers by Barry M. Dent in January 2016, which resulted in more numbers of CTC detection in the blood of the cancer patient with known metastatic disease169. In detail, CTCs were detected in 3 of 6 thyroid cancer patients and most of these tumor cells expressed cytokeratin, thyroglobulin and Sodium: Iodide Symporter (NIS) 169. The presence of more than or equal to five CTCs per 7.5 ml of blood in patients with metastatic modularlyTC (metMTC) is associated with inferior overall survival 170. Additional research had shown that in metastatic PTC patients CTCs were characterized by aneuploidy, with higher levels of CTCs in metastatic PTC in comparison with controls171. Interestingly, the designed probes of lung cancer were suitable for detecting genetic aberrations in metastatic PTC patients’ CTCs that logically could explain the similar lineage-specific chromosomal changes in thyroid and lung malignant progenitor cells170.

Exosomes, 30–120 nm endocytic membrane-derived vesicles, are important for inter-and intra -cellular communication as well as protein and RNA delivery. Because of their role, they have a variety of proteins, nucleic acids, and lipids172, 173. It has been proved frequently that molecular components of exosomes, including exosomal proteins and microRNAs (miRNAs) could be suitable non-invasive biomarkers for clinical diagnosis of tumors 174-179. Very recently, a study revealed that PTC is connected with specific changes in exosomal miRNA profiles180. Actually, miRNA-31 was found to be over-represented in the plasma exosomes of PTC compared to benign tumors, while miRNA-21 was helpful for FTC benign tumors discrimination180. MiRNA-21 and miRNA-181a-5p were both expressed equally in the exosomes of patients with PTC and FTC; therefore, their assessment will be beneficial to decide between PTC and FTC with 100 % sensitivity and 77 % specificity180. Moreover, tumor levels of miR-222 and miR-146b were coupled to the PTC recurrence, whereas miR-222 and miR-146b levels in the circulation were linked to the presence of PTC181. Some studies were evidence for exosomes and their cancer-derived miRNAs, which regulated the proliferation of recipient cells. For example, PTC-derived exosomes contain miR-146b and miR-222, which alter proliferation of other cells in a malignant behavior 182.

Conclusion

Taking everything into consideration now is the exact time to be focused on liquid biopsy for thyroid cancer management. It is really important that liquid biopsy will improve the thyroid cancer diagnostic and prognostic strategies in the minimally non-invasive way.

Acknowledgments

This work was supported by the Endocrinology and Metabolism Population Sciences Institute. This article was a part of a superior project which was granted by the National Institute for Medical Research Development (NIMAD, Grant number: 957222).

CONFLICT OF INTEREST

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.

References

  • 1.Marrinucci D, Bethel K, Luttgen M, et al. Circulating tumor cells from well-differentiated lung adenocarcinoma retain cytomorphologic features of primary tumor type. Arch Pathol Lab Med. 2009;33(9):1468–71. doi: 10.1043/1543-2165-133.9.1468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Alix-Panabières C, Pantel K. Clinical Applications of Circulating Tumor Cells and Circulating Tumor DNA as Liquid Biopsy. Cancer Discov. 2016;6(5):479–91. doi: 10.1158/2159-8290.CD-15-1483. [DOI] [PubMed] [Google Scholar]
  • 3.Bardelli A, Pantel K. Liquid Biopsies, What We Do Not Know (Yet) Cancer Cell. 2017;31(2):172–179. doi: 10.1016/j.ccell.2017.01.002. [DOI] [PubMed] [Google Scholar]
  • 4.Jr LAD, Bardelli A. Liquid Biopsies: Genotyping Circulating Tumor DNA. J Clin Oncol. 2014;32(6):579–86. doi: 10.1200/JCO.2012.45.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Mandel P, Metais P. Les acides nucléiques du plasma sanguin chez l'homme. C R Seances Soc Biol Fil. 1948;142(3-4):241–3. [PubMed] [Google Scholar]
  • 6.Leon SA, Ehrlich GE, Shapiro B, et al. Free DNA in the serum of rheumatoid arthritis patients. J Rheumatol. 1977;4(2):139–43. [PubMed] [Google Scholar]
  • 7.Shapiro B, Chakrabarty M, Cohn EM, et al. Determination of circulating DNA levels in patients with benign or malignant gastrointestinal disease. Cancer. 1983;51(11):2116–20. doi: 10.1002/1097-0142(19830601)51:11<2116::aid-cncr2820511127>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
  • 8.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(4):1659–65. [PubMed] [Google Scholar]
  • 9.Chan KCA, Zhang J, Hui ABY, et al. Size Distributions of Maternal and Fetal DNA in Maternal Plasma. Clin Chem. 2004;50(1):88–92. doi: 10.1373/clinchem.2003.024893. [DOI] [PubMed] [Google Scholar]
  • 10.Mouliere F, Robert B, Arnau Peyrotte E, et al. High Fragmentation Characterizes Tumour-Derived Circulating DNA. PLoS One. 2011;6(9):e23418. doi: 10.1371/journal.pone.0023418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.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(1–2):139–42. doi: 10.1016/s0009-8981(01)00665-9. [DOI] [PubMed] [Google Scholar]
  • 12.Anker P, Stroun M, Maurice PA. Spontaneous release of DNA by human blood lymphocytes as shown in an in vitro system. Cancer Res. 1975;35(9):2375–82. [PubMed] [Google Scholar]
  • 13.Stroun M, Maurice P, Vasioukhin V, et al. The Origin and Mechanism of Circulating DNA. Ann N Y Acad Sci. 2000;906:161–8. doi: 10.1111/j.1749-6632.2000.tb06608.x. [DOI] [PubMed] [Google Scholar]
  • 14.Stroun M, Lyautey J, Lederrey C, et al. Alu Repeat Sequences Are Present in Increased Proportions Compared to a Unique Gene in Plasma/Serum DNA. Ann N Y Acad Sci. 2001;945(1):258–64. doi: 10.1111/j.1749-6632.2001.tb03894.x. [DOI] [PubMed] [Google Scholar]
  • 15.Stroun M, Anker P, Maurice P, et al. Neoplastic Characteristics of the DNA Found in the Plasma of Cancer Patients. Oncology. 1989;46(5):318–22. doi: 10.1159/000226740. [DOI] [PubMed] [Google Scholar]
  • 16.Delgado PO, Alves BC, Gehrke Fde S, et al. Characterization of cell-free circulating DNA in plasma in patients with prostate cancer. Tumour Biol. 2013;34(2):983–6. doi: 10.1007/s13277-012-0634-6. [DOI] [PubMed] [Google Scholar]
  • 17.Diehl F, Schmidt K, Choti MA, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14(9):985–90. doi: 10.1038/nm.1789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kohler C, Barekati Z, Radpour R, et al. Cell-free DNA in the circulation as a potential cancer biomarker. Anticancer Res. 2011;31(8):2623–8. [PubMed] [Google Scholar]
  • 19.Schwarzenbach H, Hoon DSB, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 2011;11(6):426–37. doi: 10.1038/nrc3066. [DOI] [PubMed] [Google Scholar]
  • 20.Stroun M, Anker P. Nucleic acids spontaneously released by living frog auricles. Biochem J. 1972;128(3):100p–101p. doi: 10.1042/bj1280100pb. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.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(1-2):139–42. doi: 10.1016/s0009-8981(01)00665-9. [DOI] [PubMed] [Google Scholar]
  • 22.Kajbafzadeh A-M, Payabvash S, Salmasi AH, et al. Smooth muscle cell apoptosis and defective neural development in congenital ureteropelvic junction obstruction. J Urol. 2006;176(2):718–23. doi: 10.1016/j.juro.2006.03.041. [DOI] [PubMed] [Google Scholar]
  • 23.Xia L, Li Z, Zhou B, et al. Baseline mutation profiling of 1134 samples of circulating cell-free DNA and blood cells from healthy individuals. BioRxiv. 2016:089813. [Google Scholar]
  • 24.Roninson IB, Broude EV, Chang BD. If not apoptosis, then what? Treatment-induced senescence and mitotic catastrophe in tumor cells. Drug Resist Updat. 2001;4(5):303–13. doi: 10.1054/drup.2001.0213. [DOI] [PubMed] [Google Scholar]
  • 25.Wang BG, Huang HY, Chen YC, et al. Increased plasma DNA integrity in cancer patients. Cancer Res. 2003;63(14):3966–8. [PubMed] [Google Scholar]
  • 26.Trejo-Becerril C, Perez-Cardenas E, Taja-Chayeb L, et al. Cancer progression mediated by horizontal gene transfer in an in vivo model. PLoS One. 2012;7(12):e52754. doi: 10.1371/journal.pone.0052754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ansari J, Yun JW, Kompelli AR, et al. The liquid biopsy in lung cancer. Genes Cancer. 2016;7(11-12):355. doi: 10.18632/genesandcancer.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Qin Z, Ljubimov VA, Zhou C, et al. Cell-free circulating tumor DNA in cancer. Chin J Cancer. 2016;35:36. doi: 10.1186/s40880-016-0092-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Marzese DM, Hirose H, Hoon DS. Diagnostic and prognostic value of circulating tumor-related DNA in cancer patients. Expert Rev Mol Diagn. 2013;13(8):827–44. doi: 10.1586/14737159.2013.845088. [DOI] [PubMed] [Google Scholar]
  • 30.Saffar H, Sanii S, Heshmat R, et al. Expression of galectin-3, nm-23, and cyclooxygenase-2 could potentially discriminate between benign and malignant pheochromocytoma. Am J Clin Pathol. 2011;135(3):454–60. doi: 10.1309/AJCPI8AJLUZ3CZLN. [DOI] [PubMed] [Google Scholar]
  • 31.Leary RJ, Sausen M, Kinde I, et al. Detection of Chromosomal Alterations in the Circulation of Cancer Patients with Whole-Genome Sequencing. Sci Transl Med. 2012;4(162):162ra154. doi: 10.1126/scitranslmed.3004742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Chan KCA, Jiang P, Zheng YWL, et al. Cancer Genome Scanning in Plasma: Detection of Tumor-Associated Copy Number Aberrations, Single-Nucleotide Variants, and Tumoral Heterogeneity by Massively Parallel Sequencing. Clin Chem. 2013;59(1):211–24. doi: 10.1373/clinchem.2012.196014. [DOI] [PubMed] [Google Scholar]
  • 33.Nawroz H, Koch W, Anker P, et al. Microsatellite alterations in serum DNA of head and neck cancer patients. Nat Med. 1996;2(9):1035–7. doi: 10.1038/nm0996-1035. [DOI] [PubMed] [Google Scholar]
  • 34.Khatami F, Noorinayer B, Ghiasi S, et al. Lack of effects of single nucleotide polymorphisms of the DNA methyltransferase 1 gene on gastric cancer in Iranian patients: a case control study. Asian Pac J Cancer Prev. 2009;10(6):1177–82. [PubMed] [Google Scholar]
  • 35.Khatami F, Larijani B, Heshmat R, et al. Meta-analysis of promoter methylation in eight tumor-suppressor genes and its association with the risk of thyroid cancer. PloS one. 2017;12(9):e0184892. doi: 10.1371/journal.pone.0184892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Khatami F, Mohebi SR, Ghiasi S, et al. Effects of amino acid substitution polymorphisms of two DNA methyltransferases on susceptibility to sporadic colorectal cancer. Asian Pac J Cancer Prev. 2009;10(6):1183–8. [PubMed] [Google Scholar]
  • 37.Heyn H, Esteller M. DNA methylation profiling in the clinic: applications and challenges. Nat Rev Genet. 2012;13(10):679–92. doi: 10.1038/nrg3270. [DOI] [PubMed] [Google Scholar]
  • 38.Esteller M, Sanchez-Cespedes M, Rosell R, et al. Detection of aberrant promoter hypermethylation of tumor suppressor genes in serum DNA from non-small cell lung cancer patients. Cancer Res. 1999;59(1):67–70. [PubMed] [Google Scholar]
  • 39.NASSERI‐MOGHADDAM S, Malekzadeh R, Sotoudeh M, et al. Lower esophagus in dyspeptic Iranian patients: a prospective study. J Gastroenterol Hepatol. 2003;18(3):315–21. doi: 10.1046/j.1440-1746.2003.02969.x. [DOI] [PubMed] [Google Scholar]
  • 40.Silva J, Dominguez G, Villanueva M, et al. Aberrant DNA methylation of the p16INK4a gene in plasma DNA of breast cancer patients. Br J Cancer. 1999;80(8):1262–4. doi: 10.1038/sj.bjc.6690495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wong IH, Lo YD, Zhang J, et al. Detection of aberrant p16 methylation in the plasma and serum of liver cancer patients. Cancer Res. 1999;59(1):71–3. [PubMed] [Google Scholar]
  • 42.Kawakami K, Brabender J, Lord RV, et al. Hypermethylated APC DNA in plasma and prognosis of patients with esophageal adenocarcinoma. J Natl Cancer Inst. 2000;92(22):1805–11. doi: 10.1093/jnci/92.22.1805. [DOI] [PubMed] [Google Scholar]
  • 43.Lecomte T, Berger A, Zinzindohoué F, et al. Detection of free‐circulating tumor‐associated DNA in plasma of colorectal cancer patients and its association with prognosis. Int J Cancer. 2002;100(5):542–8. doi: 10.1002/ijc.10526. [DOI] [PubMed] [Google Scholar]
  • 44.Warton K, Samimi G. Methylation of cell-free circulating DNA in the diagnosis of cancer. Front Mol Biosci. 2015;2:13. doi: 10.3389/fmolb.2015.00013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Zhai R, Zhao Y, Su L, et al. Genome-wide DNA methylation profiling of cell-free serum DNA in esophageal adenocarcinoma and Barrett esophagus. Neoplasia. 2012;14(1):29–33. doi: 10.1593/neo.111626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Schwarzenbach H, Nishida N, Calin GA, et al. Clinical relevance of circulating cell-free microRNAs in cancer. Nat Rev Clin Oncol. 2014;11(3):145–56. doi: 10.1038/nrclinonc.2014.5. [DOI] [PubMed] [Google Scholar]
  • 47.Best Myron G, Sol N, Kooi I, et al. RNA-Seq of Tumor-Educated Platelets Enables Blood-Based Pan-Cancer, Multiclass, and Molecular Pathway Cancer Diagnostics. Cancer Cell. 2015;28(5):666–76. doi: 10.1016/j.ccell.2015.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Joosse Simon A, Pantel K. Tumor-Educated Platelets as Liquid Biopsy in Cancer Patients. Cancer Cell. 2015;28(5):552–4. doi: 10.1016/j.ccell.2015.10.007. [DOI] [PubMed] [Google Scholar]
  • 49.Khatami F, Larijani B, Tavangar SM. The presence of tumor extrachomosomal circular DNA (ecDNA) as a component of liquid biopsy in blood. Medical Hypotheses. Med Hypotheses. 2018;114:5–7. doi: 10.1016/j.mehy.2018.02.018. [DOI] [PubMed] [Google Scholar]
  • 50.Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science. 2011;331(6024):1559–64. doi: 10.1126/science.1203543. [DOI] [PubMed] [Google Scholar]
  • 51.Riquet M, Rivera C, Gibault L, et al. [Lymphatic spread of lung cancer: anatomical lymph node chains unchained in zones] Rev Pneumol Clin. 2014;70(1-2):16–25. doi: 10.1016/j.pneumo.2013.07.001. [DOI] [PubMed] [Google Scholar]
  • 52.Plaks V, Koopman CD, Werb Z, Cancer Circulating Tumor Cells. Science. 2013;341(6151):1186–8. doi: 10.1126/science.1235226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Gupta GP, Massague J. Cancer metastasis: building a framework. Cell. 2006;127(4):679–95. doi: 10.1016/j.cell.2006.11.001. [DOI] [PubMed] [Google Scholar]
  • 54.Ashworth T. A case of cancer in which cells similar to those in the tumours were seen in the blood after death. Aust Med J. 1869;14(3):146–9. [Google Scholar]
  • 55.Fehm T, Sagalowsky A, Clifford E, et al. Cytogenetic evidence that circulating epithelial cells in patients with carcinoma are malignant. Clin Cancer Res. 2002;8(7):2073–84. [PubMed] [Google Scholar]
  • 56.Fidler IJ. The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis revisited. Nat Rev Cancer. 2003;3(6):453–8. doi: 10.1038/nrc1098. [DOI] [PubMed] [Google Scholar]
  • 57.Yu M, Ting DT, Stott SL, et al. RNA sequencing of pancreatic circulating tumour cells implicates WNT signalling in metastasis. Nature. 2012;487(7408):510–3. doi: 10.1038/nature11217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Sleijfer S, Gratama JW, Sieuwerts AM, et al. Circulating tumour cell detection on its way to routine diagnostic implementation? Eur J Cancer. 2007;43(18):2645–50. doi: 10.1016/j.ejca.2007.09.016. [DOI] [PubMed] [Google Scholar]
  • 59.Hayes DF, Smerage J. Is there a role for circulating tumor cells in the management of breast cancer? Clin Cancer Res. 2008;14(12):3646–50. doi: 10.1158/1078-0432.CCR-07-4481. [DOI] [PubMed] [Google Scholar]
  • 60.Pantel K, Riethdorf S. Pathology: are circulating tumor cells predictive of overall survival? Nat Rev Clin Oncol. 2009;6(4):190–1. doi: 10.1038/nrclinonc.2009.23. [DOI] [PubMed] [Google Scholar]
  • 61.Panteleakou Z, Lembessis P, Sourla A, et al. Detection of circulating tumor cells in prostate cancer patients: methodological pitfalls and clinical relevance. Mol Med. 2009;15(3-4):101–114. doi: 10.2119/molmed.2008.00116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Esmaeilsabzali H, Beischlag TV, Cox ME, et al. Detection and isolation of circulating tumor cells: principles and methods. Biotechnol Adv. 2013;31(7):1063–84. doi: 10.1016/j.biotechadv.2013.08.016. [DOI] [PubMed] [Google Scholar]
  • 63.Nieva J, Wendel M, Luttgen MS, et al. High-definition imaging of circulating tumor cells and associated cellular events in non-small cell lung cancer patients: a longitudinal analysis. Phys Biol. 2012;9(1):016004. doi: 10.1088/1478-3975/9/1/016004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hou JM, Krebs M, Ward T, et al. Circulating tumor cells as a window on metastasis biology in lung cancer. Am J Pathol. 2011;178(3):989–96. doi: 10.1016/j.ajpath.2010.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.O'Flaherty JD, Gray S, Richard D, et al. Circulating tumour cells, their role in metastasis and their clinical utility in lung cancer. Lung Cancer. 2012;76(1):19–25. doi: 10.1016/j.lungcan.2011.10.018. [DOI] [PubMed] [Google Scholar]
  • 66.Gallo M, De Luca A, Maiello MR, et al. Clinical utility of circulating tumor cells in patients with non-small-cell lung cancer. Transl Lung Cancer Res. 2017;6(4):486–498. doi: 10.21037/tlcr.2017.05.07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Fidler IJ, Poste G. The "seed and soil" hypothesis revisited. Lancet Oncol. 2008;9(8):808. doi: 10.1016/S1470-2045(08)70201-8. [DOI] [PubMed] [Google Scholar]
  • 68.Krebs MG, Hou J-M, Ward TH, et al. Circulating tumour cells: their utility in cancer management and predicting outcomes. Ther Adv Med Oncol. 2010;2(6):351–365. doi: 10.1177/1758834010378414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Coman DR, de LR, Mcc UM. Studies on the mechanisms of metastasis; the distribution of tumors in various organs in relation to the distribution of arterial emboli. Cancer Res. 1951;11(8):648–51. [PubMed] [Google Scholar]
  • 70.Theodoropoulos PA, Polioudaki H, Agelaki S, et al. Circulating tumor cells with a putative stem cell phenotype in peripheral blood of patients with breast cancer. Cancer Lett. 2010;288(1):99–106. doi: 10.1016/j.canlet.2009.06.027. [DOI] [PubMed] [Google Scholar]
  • 71.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(4):R46. doi: 10.1186/bcr2333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Thiery JP, Sleeman JP. Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol. 2006;7(2):131–42. doi: 10.1038/nrm1835. [DOI] [PubMed] [Google Scholar]
  • 73.Thiery JP. Epithelial-mesenchymal transitions in development and pathologies. Curr Opin Cell Biol. 2003;15(6):740–6. doi: 10.1016/j.ceb.2003.10.006. [DOI] [PubMed] [Google Scholar]
  • 74.Yang J, Mani SA, Weinberg RA. Exploring a new twist on tumor metastasis. Cancer Res. 2006;66(9):4549–52. doi: 10.1158/0008-5472.CAN-05-3850. [DOI] [PubMed] [Google Scholar]
  • 75.Aceto N, Bardia A, Miyamoto DT, et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell. 2014;158(5):1110–1122. doi: 10.1016/j.cell.2014.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Khatami F, Aghayan HR, Sanaei M, et al. The Potential of Circulating Tumor Cells in Personalized Management of Breast Cancer: A Systematic Review. Acta Med Iran. 2017;55(3):175–93. [PubMed] [Google Scholar]
  • 77.Rack B, Schindlbeck C, Jückstock J, et al. Circulating tumor cells predict survival in early average-to-high risk breast cancer patients. J Natl Cancer Inst. 2014;106(5):dju066. doi: 10.1093/jnci/dju066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Tavangar SM, Shojaee A, Tabriz HM, et al. Immunohistochemical expression of Ki67, c-erbB-2, and c-kit antigens in benign and malignant pheochromocytoma. Pathol Res Pract. 2010;206(5):305–9. doi: 10.1016/j.prp.2010.01.007. [DOI] [PubMed] [Google Scholar]
  • 79.Omidfar K, Moinfar Z, Sohi AN, et al. Expression of EGFRvIII in thyroid carcinoma: immunohistochemical study by camel antibodies. Immunol Invest. 2009;38(2):165–80. doi: 10.1080/08820130902735998. [DOI] [PubMed] [Google Scholar]
  • 80.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(19):6302–9. doi: 10.1158/1078-0432.CCR-08-0872. [DOI] [PubMed] [Google Scholar]
  • 81.de Bono JS, Logothetis CJ, Molina A, et al. Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med. 2011;364(21):1995–2005. doi: 10.1056/NEJMoa1014618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Yap TA, Olmos D, Brunetto AT, et al. Phase I trial of a selective c-MET inhibitor ARQ 197 incorporating proof of mechanism pharmacodynamic studies. J Clin Oncol. 2011;29(10):1271–9. doi: 10.1200/JCO.2010.31.0367. [DOI] [PubMed] [Google Scholar]
  • 83.Bianchini D, Omlin A, Pezaro C, et al. First-in-human Phase I study of EZN-4176, a locked nucleic acid antisense oligonucleotide to exon 4 of the androgen receptor mRNA in patients with castration-resistant prostate cancer. Br J Cancer. 2013;109(10):2579–86. doi: 10.1038/bjc.2013.619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.van der Pol E, Boing AN, Harrison P, et al. Classification, functions, and clinical relevance of extracellular vesicles. Pharmacol Rev. 2012;64(3):676–705. doi: 10.1124/pr.112.005983. [DOI] [PubMed] [Google Scholar]
  • 85.Keller S, Sanderson MP, Stoeck A, et al. Exosomes: from biogenesis and secretion to biological function. Immunol Lett. 2006;107(2):102–8. doi: 10.1016/j.imlet.2006.09.005. [DOI] [PubMed] [Google Scholar]
  • 86.Edgar JR. Q&A: What are exosomes, exactly? BMC Biol. 2016;14:46. doi: 10.1186/s12915-016-0268-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Harding C, Heuser J, Stahl P. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J Cell Biol. 1983;97(2):329–39. doi: 10.1083/jcb.97.2.329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Zhang W, Xia W, Lv Z, et al. Liquid Biopsy for Cancer: Circulating Tumor Cells, Circulating Free DNA or Exosomes? Cell Physiol Biochem. 2017;41(2):755–768. doi: 10.1159/000458736. [DOI] [PubMed] [Google Scholar]
  • 89.Raposo G, Nijman HW, Stoorvogel W, et al. B lymphocytes secrete antigen-presenting vesicles. J Exp Med. 1996;183(3):1161–72. doi: 10.1084/jem.183.3.1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Sheridan C. Exosome cancer diagnostic reaches market. Nat Biotechnol. 2016;34(4):359–60. doi: 10.1038/nbt0416-359. [DOI] [PubMed] [Google Scholar]
  • 91.Mouritzen P, Fredsøe JC, Blondal T, et al. AACR; 2016. Abstract B40: A two-microRNA signature in urinary exosomes for diagnosis of prostate cancer. [Google Scholar]
  • 92.Webb S. The cancer bloodhounds. Nat Biotechnol. 2016;34(11):1090–1094. doi: 10.1038/nbt.3717. [DOI] [PubMed] [Google Scholar]
  • 93.Riahi R, Gogoi P, Sepehri S, et al. A novel microchannel-based device to capture and analyze circulating tumor cells (CTCs) of breast cancer. Int J Oncol. 2014;44(6):1870–8. doi: 10.3892/ijo.2014.2353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Murtaza M, Dawson SJ, Tsui DW, et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497(7447):108–12. doi: 10.1038/nature12065. [DOI] [PubMed] [Google Scholar]
  • 95.Ignatiadis M, Lee M, Jeffrey SS. Circulating Tumor Cells and Circulating Tumor DNA: Challenges and Opportunities on the Path to Clinical Utility. Clin Cancer Res. 2015;21(21):4786–800. doi: 10.1158/1078-0432.CCR-14-1190. [DOI] [PubMed] [Google Scholar]
  • 96.Li M, Diehl F, Dressman D, et al. BEAMing up for detection and quantification of rare sequence variants. Nat Methods. 2006;3(2):95–7. doi: 10.1038/nmeth850. [DOI] [PubMed] [Google Scholar]
  • 97.Baker M. Digital PCR hits its stride. Nat Methods. 2012;9(6):541. [Google Scholar]
  • 98.Leary RJ, Sausen M, Kinde I, et al. Detection of chromosomal alterations in the circulation of cancer patients with whole-genome sequencing. Sci Transl Med. 2012;4(162):162ra54. doi: 10.1126/scitranslmed.3004742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Chan KC, Jiang P, Zheng YW, et al. Cancer genome scanning in plasma: detection of tumor-associated copy number aberrations, single-nucleotide variants, and tumoral heterogeneity by massively parallel sequencing. Clin Chem. 2013;59(1):211–24. doi: 10.1373/clinchem.2012.196014. [DOI] [PubMed] [Google Scholar]
  • 100.Yeo ZX, Chan M, Yap YS, et al. Improving indel detection specificity of the Ion Torrent PGM benchtop sequencer. PLoS One. 2012;7(9):e45798. doi: 10.1371/journal.pone.0045798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Sabetkish S, Kajbafzadeh AM, Sabetkish N, et al. Whole‐organ tissue engineering: Decellularization and recellularization of three‐dimensional matrix liver scaffolds. Annu Rev Biomed Eng. 2011;13:27–53. doi: 10.1146/annurev-bioeng-071910-124743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Loman NJ, Misra RV, Dallman TJ, et al. Performance comparison of benchtop high-throughput sequencing platforms. Nat Biotechnol. 2012;30(5):434–9. doi: 10.1038/nbt.2198. [DOI] [PubMed] [Google Scholar]
  • 103.Takai E, Totoki Y, Nakamura H, et al. Clinical utility of circulating tumor DNA for molecular assessment in pancreatic cancer. Adv Exp Med Biol. 2016;924:13–17. doi: 10.1007/978-3-319-42044-8_3. [DOI] [PubMed] [Google Scholar]
  • 104.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. doi: 10.1038/nm.3519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Leary RJ, Kinde I, Diehl F, et al. Development of personalized tumor biomarkers using massively parallel sequencing. Sci Transl Med. 2010;2(20):20ra14. doi: 10.1126/scitranslmed.3000702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Riethdorf S, Fritsche H, Muller V, et al. Detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer: a validation study of the CellSearch system. Clin Cancer Res. 2007;13(3):920–8. doi: 10.1158/1078-0432.CCR-06-1695. [DOI] [PubMed] [Google Scholar]
  • 107.Thege FI, Lannin TB, Saha TN, et al. Microfluidic immunocapture of circulating pancreatic cells using parallel EpCAM and MUC1 capture: characterization, optimization and downstream analysis. Lab Chip. 2014;14(10):1775–84. doi: 10.1039/c4lc00041b. [DOI] [PubMed] [Google Scholar]
  • 108.Saucedo-Zeni N, Mewes S, Niestroj R, et al. A novel method for the in vivo isolation of circulating tumor cells from peripheral blood of cancer patients using a functionalized and structured medical wire. Int J Oncol. 2012;41(4):1241–50. doi: 10.3892/ijo.2012.1557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Ozkumur E, Shah AM, Ciciliano JC, et al. Inertial focusing for tumor antigen-dependent and -independent sorting of rare circulating tumor cells. Sci Transl Med. 2013;5(179):179ra47. doi: 10.1126/scitranslmed.3005616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Vona G, Sabile A, Louha M, et al. Isolation by size of epithelial tumor cells: a new method for the immunomorphological and molecular characterization of circulatingtumor cells. Am J Pathol. 2000;156(1):57–63. doi: 10.1016/S0002-9440(10)64706-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Wu S, Liu Z, Liu S, et al. Enrichment and enumeration of circulating tumor cells by efficient depletion of leukocyte fractions. Clin Chem Lab Med. 2014;52(2):243–51. doi: 10.1515/cclm-2013-0558. [DOI] [PubMed] [Google Scholar]
  • 112.Joosse SA, Gorges TM, Pantel K. Biology, detection, and clinical implications of circulating tumor cells. EMBO Mol Med. 2015;7(1):1–11. doi: 10.15252/emmm.201303698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Dragovic RA, Gardiner C, Brooks AS, et al. Sizing and phenotyping of cellular vesicles using Nanoparticle Tracking Analysis. Nanomedicine. 2011;7(6):780–8. doi: 10.1016/j.nano.2011.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Lim J, Yeap SP, Che HX, et al. Characterization of magnetic nanoparticle by dynamic light scattering. Nanoscale Res Lett. 2013;8(1):381. doi: 10.1186/1556-276X-8-381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Graham MD. The Coulter principle: Imaginary origins. Cytometry A. 2013;83(12):1057–1061. doi: 10.1002/cyto.a.22398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Sharma S, Gillespie BM, Palanisamy V, et al. Quantitative nanostructural and single-molecule force spectroscopy biomolecular analysis of human-saliva-derived exosomes. Langmuir. 2011;27(23):14394–400. doi: 10.1021/la2038763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Aras O, Shet A, Bach RR, et al. Induction of microparticle- and cell-associated intravascular tissue factor in human endotoxemia. Blood. 2004;103(12):4545–53. doi: 10.1182/blood-2003-03-0713. [DOI] [PubMed] [Google Scholar]
  • 118.Petersen KE, Manangon E, Hood JL, et al. A review of exosome separation techniques and characterization of B16-F10 mouse melanoma exosomes with AF4-UV-MALS-DLS-TEM. Anal Bioanal Chem. 2014;406(30):7855–66. doi: 10.1007/s00216-014-8040-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Schachermeyer S, Ashby J, Zhong W. Advances in field-flow fractionation for the analysis of biomolecules: instrument design and hyphenation. Anal Bioanal Chem. 2012;404(4):1151–8. doi: 10.1007/s00216-012-6069-5. [DOI] [PubMed] [Google Scholar]
  • 120.Pospichalova V, Svoboda J, Dave Z, et al. Simplified protocol for flow cytometry analysis of fluorescently labeled exosomes and microvesicles using dedicated flow cytometer. J Extracell Vesicles. 2015;4:25530. doi: 10.3402/jev.v4.25530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Smith ZJ, Lee C, Rojalin T, et al. Single exosome study reveals subpopulations distributed among cell lines with variability related to membrane content. J Extracell Vesicles. 2015;4:28533. doi: 10.3402/jev.v4.28533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.He M, Crow J, Roth M, et al. Integrated immunoisolation and protein analysis of circulating exosomes using microfluidic technology. Lab Chip. 2014;14(19):3773–80. doi: 10.1039/c4lc00662c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Mathivanan S, Simpson RJ. ExoCarta: A compendium of exosomal proteins and RNA. Proteomics. 2009;9(21):4997–5000. doi: 10.1002/pmic.200900351. [DOI] [PubMed] [Google Scholar]
  • 124.Alimoghaddam K, Shariftabrizi A, Tavangar M, et al. Anti-leukemic and anti-angiogenesis efficacy of arsenic trioxide in new cases of acute promyelocytic leukemia. Leuk Lymphoma. 2006;47(1):81–8. doi: 10.1080/10428190500300373. [DOI] [PubMed] [Google Scholar]
  • 125.Carrasco-Ramírez P, Greening DW, Andrés G, et al. Podoplanin is a component of extracellular vesicles that reprograms cell-derived exosomal proteins and modulates lymphatic vessel formation. Oncotarget. 2016;7(13):16070–89. doi: 10.18632/oncotarget.7445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Howitt J, Hill AF. Exosomes in the Pathology of Neurodegenerative Diseases. J Biol Chem. 2016;291(52):26589–26597. doi: 10.1074/jbc.R116.757955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Valadi H, Ekstrom K, Bossios A, et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9(6):654–9. doi: 10.1038/ncb1596. [DOI] [PubMed] [Google Scholar]
  • 128.Taylor DD, Gercel-Taylor C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol Oncol. 2008;110(1):13–21. doi: 10.1016/j.ygyno.2008.04.033. [DOI] [PubMed] [Google Scholar]
  • 129.Rabinowits G, Gercel-Taylor C, Day JM, et al. Exosomal microRNA: a diagnostic marker for lung cancer. Clin Lung Cancer. 2009;10(1):42–6. doi: 10.3816/CLC.2009.n.006. [DOI] [PubMed] [Google Scholar]
  • 130.Brase JC, Johannes M, Schlomm T, et al. Circulating miRNAs are correlated with tumor progression in prostate cancer. Int J Cancer. 2011;128(3):608–16. doi: 10.1002/ijc.25376. [DOI] [PubMed] [Google Scholar]
  • 131.Tanaka Y, Kamohara H, Kinoshita K, et al. Clinical impact of serum exosomal microRNA-21 as a clinical biomarker in human esophageal squamous cell carcinoma. Cancer. 2013;119(6):1159–67. doi: 10.1002/cncr.27895. [DOI] [PubMed] [Google Scholar]
  • 132.Lv LL, Cao YH, Ni HF, et al. MicroRNA-29c in urinary exosome/microvesicle as a biomarker of renal fibrosis. Am J Physiol Renal Physiol. 2013;305(8):F1220–7. doi: 10.1152/ajprenal.00148.2013. [DOI] [PubMed] [Google Scholar]
  • 133.Kuwabara Y, Ono K, Horie T, et al. Increased microRNA-1 and microRNA-133a levels in serum of patients with cardiovascular disease indicate myocardial damage. Circ Cardiovasc Genet. 2011;4(4):446–54. doi: 10.1161/CIRCGENETICS.110.958975. [DOI] [PubMed] [Google Scholar]
  • 134.Dhir M, McCoy KL, Ohori NP, et al. Correct extent of thyroidectomy is poorly predicted preoperatively by the guidelines of the American Thyroid Association for low and intermediate risk thyroid cancers. Surgery. 2018;163(1):81–87. doi: 10.1016/j.surg.2017.04.029. [DOI] [PubMed] [Google Scholar]
  • 135.Haghpanah V, Soliemanpour B, Heshmat R, et al. Endocrine cancer in Iran: based on cancer registry system. Indian J Cancer. 2006;43(2):80–5. doi: 10.4103/0019-509x.25889. [DOI] [PubMed] [Google Scholar]
  • 136.National Cancer Institute. "SEER stat fact sheets: thyroid cancer." Surveillance, Epidemiology, and End Results Program website. 2016.
  • 137.Larijani B, Shirzad M, Mohagheghi M, et al. Epidemiologic feature of thyroid cancer based on cancer registry data system. Iranian Journal of Public Health. 2005;34(4):1–7. [Google Scholar]
  • 138.Larijani B, Shirzad M, Mohagheghi M, et al. Epidemiologic analysis of the Tehran cancer institute data system registry (TCIDSR) Asian Pac J Cancer Prev. 2004;5(1):36–9. [PubMed] [Google Scholar]
  • 139.Cooper DS, Doherty GM, Haugen BR, et al. Revised American Thyroid Association management guidelines for patients with thyroid nodules and differentiated thyroid cancer. Thyroid. 2009;19(11):1167–214. doi: 10.1089/thy.2009.0110. [DOI] [PubMed] [Google Scholar]
  • 140.Nikiforov YE, Yip L, Nikiforova MN. New strategies in diagnosing cancer in thyroid nodules: impact of molecular markers. Clin Cancer Res. 2013;19(9):2283–8. doi: 10.1158/1078-0432.CCR-12-1253. [DOI] [PubMed] [Google Scholar]
  • 141.Tavangar S, Monajemzadeh M, Larijani B, et al. Immunohistochemical study of oestrogen receptors in 351 human thyroid glands. Singapore Med J. 2007;48(8):744–7. [PubMed] [Google Scholar]
  • 142.Haddadi-Nezhad S, Larijani B, Tavangar SM, et al. Comparison of fine-needle-nonaspiration with fine-needle-aspiration technique in the cytologic studies of thyroid nodules. Endocr Pathol. 2003;14(4):369–73. doi: 10.1385/ep:14:4:369. [DOI] [PubMed] [Google Scholar]
  • 143.Shirzad M, Larijani B, Hedayat A, et al. Diagnostic value of frozen section examination in thyroid nodule-surgery at the shariati hospital (1997–2000) Endocr Pathol. 2003;14(3):263–8. doi: 10.1007/s12022-003-0019-x. [DOI] [PubMed] [Google Scholar]
  • 144.Katoh H, Yamashita K, Enomoto T, et al. Classification and general considerations of thyroid cancer. Ann Clin Pathol. 2015;3(1):1045. [Google Scholar]
  • 145.Gild ML, Bullock M, Robinson BG, et al. Multikinase inhibitors: a new option for the treatment of thyroid cancer. Nat Rev Endocrinol. 2011;7(10):617–24. doi: 10.1038/nrendo.2011.141. [DOI] [PubMed] [Google Scholar]
  • 146.Nikiforov YE, Nikiforova MN. Molecular genetics and diagnosis of thyroid cancer. Nat Rev Endocrinol. 2011;7(10):569–80. doi: 10.1038/nrendo.2011.142. [DOI] [PubMed] [Google Scholar]
  • 147.Saffar H, Sanii S, Emami B, et al. Evaluation of MMP2 and Caspase-3 expression in 107 cases of papillary thyroid carcinoma and its association with prognostic factors. Pathol Res Pract. 2013;209(3):195–9. doi: 10.1016/j.prp.2012.06.011. [DOI] [PubMed] [Google Scholar]
  • 148.Sanii S, Saffar H, Tabriz HM, et al. Expression of matrix metalloproteinase-2, but not caspase-3, facilitates distinction between benign and malignant thyroid follicular neoplasms. Asian Pac J Cancer Prev. 2012;13(5):2175–8. doi: 10.7314/apjcp.2012.13.5.2175. [DOI] [PubMed] [Google Scholar]
  • 149.Mohammadi-asl J, Larijani B, Khorgami Z, et al. Qualitative and quantitative promoter hypermethylation patterns of the P16, TSHR, RASSF1A and RARβ2 genes in papillary thyroid carcinoma. Med Oncol. 2011;28(4):1123–8. doi: 10.1007/s12032-010-9587-z. [DOI] [PubMed] [Google Scholar]
  • 150.Haghpanah V, Shooshtarizadeh P, Heshmat R, et al. Immunohistochemical analysis of survivin expression in thyroid follicular adenoma and carcinoma. Appl Immunohistochem Mol Morphol. 2006;14(4):422–5. doi: 10.1097/01.pai.0000213100.88074.b8. [DOI] [PubMed] [Google Scholar]
  • 151.Tabriz HM, Adabi K, Lashkari A, et al. Immunohistochemical analysis of nm23 protein expression in thyroid papillary carcinoma and follicular neoplasm. Pathol Res Pract. 2009;205(2):83–7. doi: 10.1016/j.prp.2008.08.007. [DOI] [PubMed] [Google Scholar]
  • 152.Mohammadi-Asl J, Larijani B, Khorgami Z, et al. Prevalence of BRAFV600E mutation in Iranian patients with papillary thyroid carcinoma: a single-center study. J Appl Sci. 2009;9(19):3593–7. [Google Scholar]
  • 153.Khatami F, Larijani B, Tavangar SM. Circulating Tumor BRAF Mutation and Personalized Thyroid Cancer Treatment. Asian Pac J Cancer Prev. 2017;18(2):293–294. doi: 10.22034/APJCP.2017.18.2.293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Amoli MM, Yazdani N, Amiri P, et al. HLA-DR association in papillary thyroid carcinoma. Dis Markers. 2010;28(1):49–53. doi: 10.3233/DMA-2010-0683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Sarmadi S, Izadi-Mood N, Sotoudeh K, et al. Altered PTEN expression; a diagnostic marker for differentiating normal, hyperplastic and neoplastic endometrium. Diagn Pathol. 2009;4:41. doi: 10.1186/1746-1596-4-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Umetani N, Hiramatsu S, Hoon DS. Higher amount of free circulating DNA in serum than in plasma is not mainly caused by contaminated extraneous DNA during separation. Ann N Y Acad Sci. 2006;1075:299–307. doi: 10.1196/annals.1368.040. [DOI] [PubMed] [Google Scholar]
  • 157.Zane M, Agostini M, Enzo MV, et al. Circulating cell-free DNA, SLC5A8 and SLC26A4 hypermethylation, BRAFV600E: A non-invasive tool panel for early detection of thyroid cancer. Biomed Pharmacother. 2013;67(8):723–30. doi: 10.1016/j.biopha.2013.06.007. [DOI] [PubMed] [Google Scholar]
  • 158.Hu S, Ewertz M, Tufano RP, et al. Detection of serum deoxyribonucleic acid methylation markers: a novel diagnostic tool for thyroid cancer. J Clin Endocrinol Metab. 2006;91(1):98–104. doi: 10.1210/jc.2005-1810. [DOI] [PubMed] [Google Scholar]
  • 159.Chuang TC, Chuang AY, Poeta L, et al. Detectable BRAF mutation in serum DNA samples from patients with papillary thyroid carcinomas. Head Neck. 2010;32(2):229–34. doi: 10.1002/hed.21178. [DOI] [PubMed] [Google Scholar]
  • 160.Janku F, Huang HJ, Claes B, et al. BRAF mutation testing in cell-free DNA from the plasma of patients with advanced cancers using a rapid, automated molecular diagnostics system. Mol Cancer Ther. 2016;15(6):1397–404. doi: 10.1158/1535-7163.MCT-15-0712. [DOI] [PubMed] [Google Scholar]
  • 161.Sandulache VC, Williams MD, Lai SY, et al. Real-time genomic characterization utilizing circulating cell-free DNA in patients with anaplastic thyroid carcinoma. Thyroid. 2017;27(1):81–87. doi: 10.1089/thy.2016.0076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Evers C, Duose DY, Mehrotra M, et al. Liquid Biopsy: Comparison of Mutation Detection Methods for Measurement of RET M918T Circulating Cell-Free DNA in Medullary Thyroid Cancer Patients. Cancer Genetics. 2016;209(6):287. [Google Scholar]
  • 163.Khatami F, Tavangar SM. Genetic and Epigenetic of Medullary Thyroid Cancer. Iran Biomed J. 2018;22(3):142–50. doi: 10.22034/ibj.22.3.142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Brose MS, Cabanillas ME, Cohen EE, et al. Vemurafenib in patients with BRAF(V600E)-positive metastatic or unresectable papillary thyroid cancer refractory to radioactive iodine: a non-randomised, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016;17(9):1272–82. doi: 10.1016/S1470-2045(16)30166-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Lubitz CC, Parangi S, Holm TM, et al. Detection of Circulating BRAF V600E in Patients with Papillary Thyroid Carcinoma. J Mol Diagn. 2016;18(1):100–8. doi: 10.1016/j.jmoldx.2015.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Qiu Z-L, Shen C-T, Sun Z, et al. Circulating Long Non-Coding RNAs Act as Biomarkers for Predicting 131I Uptake and Mortality in Papillary Thyroid Cancer Patients with Lung Metastases. Cell Physiol Biochem. 2016;40(6):1377–1390. doi: 10.1159/000453190. [DOI] [PubMed] [Google Scholar]
  • 167.Hsieh C-H, Lin H-C, Huang S-B, et al. Circulating epithelial cell enumeration facilitates the identification and follow-up of a patient with early stage papillary thyroid microcarcinoma: A case report. Clin Chim Acta. 2016;454:107–11. doi: 10.1016/j.cca.2015.12.030. [DOI] [PubMed] [Google Scholar]
  • 168.Tseng C-P, Lin J-D, Lin H-C, et al. Combined analysis of circulating epithelial cell count and serum thyroglobulin for differentiating disease status of the patients with papillary thyroid carcinoma. Oncotarget. 2016;7(13):17242–17253. doi: 10.18632/oncotarget.6587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Dent BM, Ogle LF, O'Donnell RL, et al. High‐resolution imaging for the detection and characterisation of circulating tumour cells from patients with oesophageal, hepatocellular, thyroid and ovarian cancers. Int J Cancer. 2016;138(1):206–16. doi: 10.1002/ijc.29680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Xu JY, Handy B, Michaelis CL, et al. Detection and prognostic significance of circulating tumor cells in patients with metastatic thyroid cancer. J Clin Endocrinol Metab. 2016;101(11):4461–4467. doi: 10.1210/jc.2016-2567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Xu JY, Zaidi T, Cote GJ, et al. Circulating Tumor Cells (CTCs) in Metastatic Papillary Thyroid Cancer: Report of a Case-Control Pilot Study. Thyroid Neoplasia: Endocrine Society. 2016 p. PP22-3-PP-3. [Google Scholar]
  • 172.Lakkaraju A, Rodriguez-Boulan E. Itinerant exosomes: emerging roles in cell and tissue polarity. Trends Cell Biol. 2008;18(5):199–209. doi: 10.1016/j.tcb.2008.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Van Niel G, Porto-Carreiro I, Simoes S, et al. Exosomes: a common pathway for a specialized function. J Biochem. 2006;140(1):13–21. doi: 10.1093/jb/mvj128. [DOI] [PubMed] [Google Scholar]
  • 174.Lin J, Li J, Huang B, et al. Exosomes: Novel Biomarkers for Clinical Diagnosis. ScientificWorldJournal. 2015;2015:657086. doi: 10.1155/2015/657086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Mitchell PS, Parkin RK, Kroh EM, et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci U S A. 2008 29;105(30):10513–8. doi: 10.1073/pnas.0804549105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Hunter MP, Ismail N, Zhang X, et al. Detection of microRNA expression in human peripheral blood microvesicles. PLoS One. 2008;3(11):e3694. doi: 10.1371/journal.pone.0003694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Rabinowits G, Gerçel-Taylor C, Day JM, et al. Exosomal microRNA: a diagnostic marker for lung cancer. Clin Lung Cancer. 2009;10(1):42–6. doi: 10.3816/CLC.2009.n.006. [DOI] [PubMed] [Google Scholar]
  • 178.Tanaka Y, Kamohara H, Kinoshita K, et al. Clinical impact of serum exosomal microRNA‐21 as a clinical biomarker in human esophageal squamous cell carcinoma. Cancer. 2013;119(6):1159–67. doi: 10.1002/cncr.27895. [DOI] [PubMed] [Google Scholar]
  • 179.Takeshita N, Hoshino I, Mori M, et al. Serum microRNA expression profile: miR-1246 as a novel diagnostic and prognostic biomarker for oesophageal squamous cell carcinoma. Br J Cancer. 2013;108(3):644–52. doi: 10.1038/bjc.2013.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Samsonov R, Burdakov V, Shtam T, et al. Plasma exosomal miR-21 and miR-181a differentiates follicular from papillary thyroid cancer. Tumour Biol. 2016;37(9):12011–12021. doi: 10.1007/s13277-016-5065-3. [DOI] [PubMed] [Google Scholar]
  • 181.Lee JC, Zhao JT, Clifton‐Bligh RJ, et al. MicroRNA‐222 and MicroRNA‐146b are tissue and circulating biomarkers of recurrent papillary thyroid cancer. Cancer. 2013;119(24):4358–65. doi: 10.1002/cncr.28254. [DOI] [PubMed] [Google Scholar]
  • 182.Lee JC, Zhao JT, Gundara J, et al. Papillary thyroid cancer-derived exosomes contain miRNA-146b and miRNA-222. J Surg Res. 2015;196(1):39–48. doi: 10.1016/j.jss.2015.02.027. [DOI] [PubMed] [Google Scholar]

Articles from International Journal of Hematology-Oncology and Stem Cell Research are provided here courtesy of Tehran University of Medical Sciences

RESOURCES