Next Article in Journal
Reprogrammed Lipid Metabolism and the Lipid-Associated Hallmarks of Colorectal Cancer
Next Article in Special Issue
Circulating Cell-Free DNA in Renal Cell Carcinoma: The New Era of Precision Medicine
Previous Article in Journal
COVID-19 and Adult Acute Leukemia: Our Knowledge in Progress
Previous Article in Special Issue
The Promise of Circulating Tumor DNA in Head and Neck Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Extracellular Nucleic Acids in the Diagnosis and Progression of Colorectal Cancer

1
Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
2
Comenius University Science Park, Comenius University, 841 04 Bratislava, Slovakia
3
Geneton Ltd., 841 04 Bratislava, Slovakia
4
Department of Human Genetics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
5
Department of Surgery, Transplantation and Gastroenterology, Semmelweis University, 1082 Budapest, Hungary
6
Medirex Group Academy, n.p.o., 949 05 Nitra, Slovakia
7
Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, 842 05 Bratislava, Slovakia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(15), 3712; https://doi.org/10.3390/cancers14153712
Submission received: 16 June 2022 / Revised: 21 July 2022 / Accepted: 26 July 2022 / Published: 29 July 2022
(This article belongs to the Special Issue Cell-Free DNA as Prognostic and Predictive Biomarker in Solid Cancers)

Abstract

:

Simple Summary

Colorectal cancer (CRC) is a disease that usually shows no evident clinical symptoms in the early stages, often leading to late diagnosis. Over the past few years, a new approach based on liquid biopsy has gained far-reaching applications in less-invasive CRC diagnosis and management, allowing for the use of extracellular nucleic acids as promising biomarkers to detect CRC at an early stage and monitor disease recurrence. That is why an up-to-date review and discussion of in-depth liquid biopsy-derived DNA and RNA biomarkers is essential. We hereby offer an overview of known predisposing genetic factors for developing sporadic and hereditary CRC, and an extensive repertoire of available extracellular DNA/RNA molecules with their potential clinical applications and shortcomings. Our review may be of value to experts dealing with CRC at the molecular level as well as to clinical professionals aiming for a better understanding of state-of-the-art techniques in CRC diagnosis and management.

Abstract

Colorectal cancer (CRC) is the 3rd most common malignant neoplasm worldwide, with more than two million new cases diagnosed yearly. Despite increasing efforts in screening, many cases are still diagnosed at a late stage, when mortality is high. This paper briefly reviews known genetic causes of CRC (distinguishing between sporadic and familial forms) and discusses potential and confirmed nucleic acid biomarkers obtainable from liquid biopsies, classified by their molecular features, focusing on clinical relevance. We comment on advantageous aspects such as better patient compliance due to blood sampling being minimally invasive, the possibility to monitor mutation characteristics of sporadic and hereditary CRC in a disease showing genetic heterogeneity, and using up- or down-regulated circulating RNA markers to reveal metastasis or disease recurrence. Current difficulties and thoughts on some possible future directions are also discussed. We explore current evidence in the field pointing towards the introduction of personalized CRC management.

1. Introduction

Colorectal cancer (CRC) is one of the most threatening types of cancer in developed countries [1]. It is the 3rd most common malignant disease overall and the 2nd leading cause of death, with 2.3 million new cases per year registered worldwide [2]. Aging population, unfavorable eating habits, and other cumulative risk factors contribute to the growth of case numbers, while about 3–10% of cases are associated with inherited cancer predisposition (see Section 2). Based on tissue characteristics, five subtypes are known: adenocarcinoma (the most common subtype), carcinoid tumor, lymphoma, sarcoma [3] and gastrointestinal stromal tumor [4].
Screening families at-risk and the general population is essential, as early detection greatly improves prognosis. Available data clearly show the importance of early diagnosis, supported by the overall survival (OS) rate of CRC patients depending on the stage at which the primary tumor is diagnosed. Based on data from a retrospective population study [5], 5-year OS is 78.85% for the localized stage (stages I–II), 63.25% for the regional stage (stage III), and 20.31% for the metastatic stage (stage IV). Another study evaluating stage-dependent OS in CRC found 94%, 82%, 67%, and 11% for stages I, II, III, and IV, respectively [6,7].
Numerous traditional screening methods are available for CRC, including fecal occult blood test (FOBT) and colonoscopy. The latter is considered gold-standard, especially for inherited CRC syndromes such as Lynch syndrome (LS). Still, patient compliance is often poor, and a lack of quality standards results in missed cases [8]. FOBT is suggested for screening the general population and at-risk patients who reject colonoscopy, but its limitations include high false-positive rates and patient compliance dropping below 50% after 5–10 years [9,10]. Only about 40% of the population for whom screening is suggested will proceed with CRC testing [11]. Compliance would surely rise if more patient-friendly and less-invasive strategies were available.
Methods introduced more recently include stool DNA testing (a highly sensitive method unaffected by the proximal location of the tumor but involving costs and technical difficulties associated with large-volume stool collection and transport, making it an unlikely candidate for a frequently performed mainstream screening method in the general population [12]) and ELISA-based screening tests for CRC antigens from blood samples. Testing of peripheral blood is a promising new direction, having the advantage of being much less invasive than colonoscopy and more convenient than stool testing, resulting in a much higher rate of patient compliance [13]. Nucleic acids are also readily detectable from blood; however, input costs must continue to be reduced, a process critical to the competitiveness of such strategies compared to conventionally used protein analyses [14]. Additionally, some mutations relevant for CRC therapy, such as the ones reported in the human epidermal growth factor receptor 2 (HER2) gene, are too small to be detectable at the protein level [15].
Some of the traditional tests are not invasive but are perceived as unpleasant by patients, suggesting the need for novel approaches to CRC screening. Liquid biopsy is acceptable for most patients and seems to be the best candidate in this context. In this review, we offer a brief overview of CRC genetics and discuss current liquid biopsy-derived DNA and RNA biomarkers showing promise in the screening of CRC in the general population and families at risk.

2. Genetics of CRC

Based on the etiology and the genetics of the disease, CRC is generally categorized into three groups: (i) sporadic, (ii) familial, and (iii) hereditary [16,17]. Sporadic cases represent about 75% of all incidences. Genetic factors still play a role, but only somatic mutations are present, and family members of affected individuals do not have an increased risk of developing the disease [18]. Familial CRC is not classified as hereditary and is frequently regarded as sporadic, as no causative genes have been identified yet. First-degree relatives of known patients are at higher risk compared to the general population [18,19]. Hereditary CRC is caused by a known mutation in the germ-line; the most common example is LS, but many other (rare) syndromic forms of CRC have been reported [17,20]. They are outlined in Table 1.

2.1. Hereditary CRC

LS is the most common hereditary colon cancer syndrome, also manifesting in other cancer types. It is responsible for 2–4% of CRC cases [37] and shows an autosomal dominant pattern of inheritance, caused by one of the DNA mismatch repair (MMR) genes being affected by a heterozygous germline mutation, or a deletion in the EPCAM gene [38], and the corresponding protein losing its function (and becoming undetectable by immunohistochemistry) following a somatic loss of heterozygosity in the colon tissue. The genes MutS homolog 2 (MSH2), MutL Homolog 1 (MLH1) and PMS1 Homolog 2 (PMS2) are involved in this type of repair mechanism [39], with MSH2 and MLH1 mutations having the greatest contribution to the development of LS-associated malignancies [40]. Tumors associated with LS show a high level of microsatellite instability (MSI-H), a feature not seen in the majority of sporadic CRC cases (see below) [41].
Other hereditary CRC syndromes worth noting include Peutz–Jeghers syndrome (PJS) caused by a mutation in Serine/threonine kinase 11 (STK11), usually presenting as multiple benign hamartomatous polyps [42,43], and MUTYH-associated polyposis (MAP) inherited in an autosomal recessive pattern characterized by biallelic germline mutations in MutY DNA glycosylase (MUTYH), a gene having a role in base excision repair (BER) [26,44]. Familial adenomatous polyposis (FAP), a well-studied disease with an autosomal dominant inheritance, has been reported to be caused by insertions and deletions in the adenomatous polyposis coli (APC) tumor suppressor gene [45]. Germline mutations of oncogene-induced senescence pathway genes cause serrated polyposis syndrome [46,47], usually presenting as multiple serrated polyps of the colon [46].

2.2. Sporadic CRC

Twelve percent of sporadic tumors are MSI-H, while the rest are classified as microsatellite stable (MSS) or show low level of microsatellite instability (MSI-L). Most sporadic MSI-H cases are associated with somatic MMR deficiency caused by the CpG Island Methylator phenotype (CIMP), in which the MLH1 promoter shows biallelic somatic methylation with B-Raf Proto-Oncogene, Serine/Threonine Kinase (BRAF) mutations usually present in the background (almost never seen in LS) [41]. MSI-H tumors are thought to occur more proximally and are less differentiated, and usually show a better prognosis due to being sensitive to anti-programmed death receptor 1 (PD-1) therapy [48].
MSS tumors typically display chromosomal instability (CIN): numerical and structural alterations in chromosomes apart from a large variety of mutations in oncogenes and tumor suppressor genes [17]. Progressive telomere shortening occurs in the intestinal epithelium during aging in humans, and telomere dysfunction (anaphase bridging) has been documented in the adenoma-carcinoma transition, indicating that a telomere-based crisis may play a role in driving CIN in the early stages of human CRC [49]. Cancer progression is associated with telomerase reactivation, which is present in 85–90% of all cancer types, including CRC [50].
Mutations in APC, KRAS proto-oncogene GTPase (KRAS), and Tumor Protein 53 (TP53) are associated with lymph node metastasis [51]. KRAS mutations are found in about half of MSS/MSI-L tumors and make them resistant against anti-epidermal growth factor receptor (EGFR) antibody therapy. However, a fraction of wild-type KRAS tumors also display a poor response. The simultaneous presence of KRAS and BRAF mutations is rarely detected (most such cases are MSS CRC) [52].

3. Cell-Free Nucleic Acids as CRC Biomarkers

Liquid biopsy-based methods of cancer detection have undergone a substantial increase in popularity in recent years. Painless and quick sampling may help involve more average-risk people in cancer screening programs. As the carcinogenesis of CRC takes years, there is a relatively wide window available for early detection, but blood biopsies are likely to prove equally useful in the follow-up of non-metastatic and metastatic CRC (mCRC) [53,54]. On the other hand, circulating tumor DNA (ctDNA) assays do not provide insight into cell differentiation (tumor grade) or cancer stage (e.g., TNM classification), but suggest the overall tumor burden and may reveal the tumor’s molecular subtype and heterogeneity. So, liquid biopsy may not replace a tissue-based diagnosis, but rather provides alternate sampling for molecular tests.
There are five main sources of cell-free nucleic acids (cfNAs) in the circulation described until now: (i) apoptosis, (ii) necrosis, (iii) netosis, (iv) active secretion and (v) release by microbiota (Figure 1). cfNAs may freely circulate in body fluids, or may be bound to protein complexes or encapsulated in extracellular membrane vesicles (EMVs) (e.g., apoptotic bodies, microvesicles, or exosomes). EMVs mediate intercellular communication and were shown to play essential roles in the pathogenesis of CRC, as they are implicated in tumorigenesis, CRC progression, chemotherapy resistance, and metastasis [55,56]. Details of individual cfNA types and their release mechanisms were comprehensively covered in previous works [57,58,59,60,61] and are not the focus [55,56,57,58,59] of this review.

3.1. cfDNA Biomarkers

In response to the current status quo in the continuum of CRC management, the Colon and Rectal–Anal Task Forces of the United States National Cancer Institute recently convened a panel of multidisciplinary experts to summarize current data on the utility of ctDNA. They provide guidance and promote the efficient development and integration of this technology into clinical care. The panel focused on key areas in which ctDNA has the potential to change clinical practice [62].
The concentration of tumor-derived cfDNA, also known as ctDNA in the plasma of rectal and colon cancer patients, was compared in multiple studies, with varying results. Frattini et al. reported colon cancer patients having higher ctDNA concentrations than patients with rectal cancer (colon: 500 ng/mL, rectal: 250 ng/mL in plasma) [63], while Cassinotti et al. observed the opposite [64]. However, there is consensus on the usefulness of ctDNA biomarkers in CRC, as they show promise in the initial diagnosis, monitoring minimal residual disease, evaluation of treatment response in metastasis, identifying drivers of treatment sensitivity and resistance, and guiding therapeutic strategies to overcome resistance (Table 2) [53,65].
It has been demonstrated that cfDNA fragmentation profiles are different between healthy individuals and cancer patients [73] and also vary between tumor types [68]. Thus, accumulating evidence suggests fragmentomic cfDNA features as a potential cancer biomarker (Table 2). Zhitnyuk et al. were able to reveal the presence of early-stage colorectal and renal cancer with an area under the ROC curve (AUC) of 0.94 by deep targeted profiling of cfDNA end distributions and sequence motifs [66]. Moreover, since fragmentation is related to nucleosomal patterns, it may be useful for determining the source of tumor-derived cfDNA [67]. However, cfDNA fragmentation may not always be suitable to distinguish cancer from other forms of tissue damage. In this context, trauma-induced cfDNA was studied, while the concentration of short but not long cfDNA fragments was shown to be increased postoperatively in colorectal and bladder cancer [74].
CRC-specific mutations, including the ones in APC, BRAF, KRAS and TP53, seem to be equally detectable from cancer tissue and blood plasma [69]. Monitoring the presence of mutations observed at the time of diagnosis from ctDNA during the postoperative period may be used to predict CRC recurrence irrespective of the type of chemotherapy being applied (Table 2) [75].
Vidal et al. have demonstrated an assay offering a minimally invasive and highly sensitive method for RAS assessment in the plasma of mCRC patients, which may be readily implemented into routine clinical practice to perform baseline diagnosis to select candidate patients for anti-EGFR therapy. Moreover, a potential use in assessing the dynamics of RAS to monitor response and resistance to treatment has been suggested (Table 2) [71]. Another example is HER2, a well-known oncogenic driver in different tumor types [76]. Although its alterations are not common in CRC (3–5% of mCRC cases), when present, anti-HER2 therapy is an option [15]. Nakamura et al. reported that baseline ctDNA genotyping of HER2 copy number may stratify patients according to the efficiency of therapy with an accuracy comparable to tissue genotyping. Since ctDNA genotyping can identify patients who benefit from dual-HER2 blockade as well as monitor treatment response, they emphasize its usefulness for HER2-amplified mCRC, which may benefit patients especially in the first salvage-line treatment [72].

3.2. Methylation Status

DNA methylation is a marker that is relatively easy to detect and seems relevant for CRC diagnosis and prognosis (Table 3). Barták et al. noticed that methylated cfDNA fragments are more stable in the circulation, and methylation-related alterations are present in about 65 to 100% of tumor samples (more frequent than mutations: 5–75%) [77]. It was demonstrated that a hypermethylated Septin 9 gene (SEPT9) in circulating DNA is a specific CRC biomarker [78,79]. The SEPT9 assay reveals hypermethylation of CpG island 3 in the SEPT9 promoter [80]. While methylated SEPT9 levels in tissue and plasma samples are not strongly correlated, methylated SEPT9 is significantly higher in the plasma of patients with CRC than in patients with no evidence of the disease [81].
Hypermethylated SEPT9 ctDNA disappears after 3 months following surgery, suggesting that this molecule may be the first non-invasive biomarker for postsurgical follow-up [82,83]. The plasma-based SEPT9 gene methylation assay is currently an FDA-approved non-invasive CRC screening test known as Epi proColon® 1.0 [79,84]. In Europe and some other countries (e.g., China), a second-generation test called Epi proColon® 2.0 CE is available for early-stage CRC screening [85]. The main difference between the generations is that the original Epi proColon algorithm requires only one positive PCR reaction out of two PCRs, emphasizing sensitivity, while the Epi proColon 2.0 CE algorithm requires at least two positive PCR results, placing a greater emphasis on test specificity [86]. In a Chinese opportunistic screening study, a different SEPT9 gene methylation assay called SensiColon was validated, showing 76.6% specificity and 95.9% sensitivity for the detection of early CRC stages [78]. The overall performance of proposed SEPT9 gene methylation tests and their comparison with other CRC screening assays have been well reviewed in the study of Song et al. [86].
It should be noted that tests based on the methylation status of SEPT9 may serve as a competitive option for CRC screening and early detection, as it has been demonstrated to have a higher compliance than protein FIT tests and colonoscopy. It could be applied in asymptomatic population screening even if the screening assay does not exhibit sensitivity and specificity equivalent to FIT and FIT-DNA [87], as the uptake rate by the population seems to be a critical aspect of introducing novel screening strategies.
Among the stool-DNA tests, Cologuard™ was the first FDA-approved in vitro diagnostic assay for both left- and right-sided CRCs and pre-malignant neoplasia [85]. Cologuard™ aims to detect 11 distinct biomarkers, classified in three categories: DNA methylation biomarkers in gene promoter regions, such as the specific methylation of N-Myc Downstream-Regulated Gene 4 (NDRG4) and Bone Morphogenic Protein (BMP3), seven mutational markers in the gene KRAS, and the presence of occult hemoglobin. Additionally, the beta-actin gene (ACTB) is used as a reference for confirming the total amount of human DNA. In a pivotal case–control study (with colonoscopy as the reference method) involving 10,000 individuals aged 50 to 84 years at average risk, the Cologuard™ DNA test and the FIT (fecal immunochemical test) were used for detecting all CRC stages (I–IV) and showed a sensitivity of 92.3% and 73.8%, respectively [88]. In the detection of CRC stages I–II, DNA testing displays a 70% detection capability [89] and a 42% detection rate of advanced precancerous lesions compared to FIT [88].
Table 3. List of methylated cfDNA biomarkers in CRC.
Table 3. List of methylated cfDNA biomarkers in CRC.
DNASourceFunctionTechniqueRef.
Methylated SEPT9Plasmaspecific non-invasive CRC biomarker for postsurgical follow-upqPCR[78,82,83]
CpG island methylation in the INHBB promoterSerum/stoolbiomarker of poor prognosis in CRCBisulfite sequencing, qPCR[90]
Methylation of APC/MGMT/RASSF2A/Wif-1PlasmabiomarkerqPCR[91]
Methylation of BMP3/NDRG4/VIM/TFPI2/mutant KRAS/ACTBStoolbiomarkerQuARTS[92]
Ref.—reference; ↑—upregulated; ↓—downregulated; MPS—massively parallel sequencing; QuARTS—quantitative allele-specific real-time target and signal amplification.
There are multiple signaling pathways participating in cancer progression, the best studied ones being the mitogen-activated protein kinase (MAPK), p53 and transforming growth factor-beta (TGF-β) pathways. DNA methylation changes affect genes that are involved in these pathways and are considered to be potential biomarkers of CRC. Inhibin subunit beta B (INHBB), SPARC related modular calcium binding 2 (SMOC2), brain derived neurotrophic factor (BDNF), and transforming growth factor beta regulator 4 (TBRG4) are highly deregulated by methylation and are involved in CRC metastasis development. Promoter methylation in the above genes is detectable from liquid biopsy samples, but drawbacks of the gold-standard method bisulfite conversion-PCR (cost and labor intensity) currently limit their application in non-invasive screening [93]. In addition to being a diagnostic biomarker, CpG island methylation in the INHBB promoter (detected in serum or stool) was reported as a marker of poor prognosis in CRC [90].
Laugsand et al. performed a meta-analysis of available literature sources to find out which methylation-related changes may be used as biomarkers, comparing results from plasma, stool, urine and CRC tissue [94]. The panel APC/MGMT/RASSF2A/Wif-1 (sensitivity 87%; specificity 92%) appeared to be the most useful in plasma, and BMP3/NDRG4/VIM/TFPI2/mutant KRAS/ACTB in stool samples.

3.3. Genometastasis

There is evidence for the presence of horizontal (cell-to-cell) DNA transfer not only in bacteria, but in mammals as well [95]. The large quantity of tumor-derived cfDNAs in the blood of cancer patients suggests their possible function as carriers of certain “tumorigenic” properties to normal cells [96], supported by the observation that the concentration of cfDNA is five times higher in CRC cases than in healthy individuals [69]. The term “genometastasis” is now used by some authors after some successful experiments on malignized normal cells with tumor-derived cfDNA, such as the transfer of the tumor specific KRAS mutation by adding the serum of a CRC patient to a healthy cell line [97]. cfDNA also contains fragments of oncogenes and may behave as an oncovirus, directly participating in metastasis formation [95].
It is worth mentioning that, as CRC carcinogenesis takes several years, some patients may have already donated blood by the time of their diagnosis. As genometastasis may be considered as a possibility, it would be interesting to follow up these donors and their recipients.

3.4. mtDNA

The presence of mtDNA in the circulation (cf-mtDNA) opens up new possibilities for the non-invasive analysis of tumor profiles. Several features of the mitochondrial genome can be analyzed, such as mtDNA mutations, mtDNA copy number alterations, heteroplasmy, or cf-mtDNA fragment length distribution. Since mtDNA exhibits characteristics distinct from the nuclear genome, including high copy numbers, high mutation frequencies, and heteroplasmy, it may be considered in some novel applications of liquid biopsies [98]. Recently, some papers have become available on the putative role of cf-mtDNA in CRC screening and follow-up (Table 4).
Haupts et al. reported a higher cf-mtDNA copy number in the plasma of healthy subjects compared to CRC patients [98]. Copy number of mitochondrially encoded NADH ubiquinone oxidoreductase core subunit 1 (MT-ND1) in blood plasma has been suggested as a marker of early CRC [102]. Recently, Zhou et al. published results on cf-mtDNA content in urine of CRC patients, finding aberrant fragmentation and mutation profiles with diagnostic potential [101]. mtDNA mutations have been identified in almost every cancer type, including CRC, and are thought to contribute to the development of cancer phenotypes. However, it is not clear if such alterations are the cause or consequence of the changes that take place in tumor cells. Further studies are needed to clarify their role in carcinogenesis.

3.5. cfRNA Biomarkers

3.5.1. mRNA

Circulating cell-free mRNA (cf-mRNA) is prone to quick degradation and low abundance [103]. Accordingly, reports of cf-mRNA biomarkers in CRC are relatively scarce in comparison to non-coding RNAs (see below). A loss of glycogen synthase kinase 3 alpha (GSK3A) and RAS homolog family member A (RHOA) expression in plasma may function as a biomarker of colorectal adenoma, a precancerous lesion of CRC [104]. Another promising transcript from this group seems to be synaptophysin-like 1 (SYPL1), detected from stool or plasma samples. This mRNA may potentially be a CRC biomarker rivaling FOBT, carcinoembryonic antigen (CEA) and cancer antigen 19-9 (CA19-9). Moreover, a correlation with tumor size and the clinical stage was observed [105].
Of all CRC patients, approximately 20% are diagnosed with liver metastases at the time of primary tumor diagnosis, and up to 60% develop metachronous metastases [106]. The liver is the most frequent distant location of these metastases and in many cases the only organ affected, being the most common cause of mortality from CRC [107]. The clinical outcome for patients with colorectal liver metastases may be significantly improved by surgical resection. Nevertheless, 50–75% of patients experience recurrence after hepatectomy, most of which occur within 2 years [108,109]. A study by Pun et al. [110] has revealed that plasma levels of the Bmi1 transcript may be used as a biomarker in mCRC patients for non-invasive monitoring of occult metastases and anticipating the emergence of distant metastases. mRNA produced from the genes prostaglandin-endoperoxide synthase 2 (PTGS2), guanylate cyclase 2C (GUCY2C) and jagged canonical notch ligand 1 (JAG1) were reported to be upregulated in mCRC, while the serum expression of GUCY2C and GUCY2C/PTGS2 showed correlation with the therapeutic response [111]. However, a definitive prognostic cf-mRNA biomarker for response to therapy and survival of mCRC patients has yet to emerge.

3.5.2. miRNA

miRNAs are a class of short, non-coding RNAs known to play oncogenic and tumor suppressor roles in various malignancies [112], including CRC, by regulating mRNA targets [113]. They have been reported to circulate in the blood (cf-miRNA), providing diagnostic and prognostic usefulness in oncology [114]. In the past few years, extensive research has been conducted on miRNAs as clinically relevant biomarkers, since miRNAs are present in CRC tumor tissue, feces, and various body fluids, avoiding degradation [115]. The usefulness of various cf-miRNAs as biomarkers is constantly proven from liquid biopsy samples (Table 5).
According to Nassar et al., miRNA panels display better prognostic value than individual miRNAs [117]. Many research groups have reported diagnostic biomarkers for CRC based on blood serum/plasma miRNAs, as they are easy to handle, low-cost, and obtainable with minimal invasiveness. It is worth a note that blood is not the only form of liquid biopsy relevant for CRC management. However, samples such as saliva and urine are still not well studied in large cohorts. In recent years, salivary miRNAs have inspired some growth in interest. Sazanov et al. made measurements of miR-21 obtained from saliva samples of CRC patients. They revealed a significantly increased miR-21 expression compared to healthy individuals with an estimated sensitivity of 97% and specificity of 91% [128].
One of the first large-scale salivary miRNA characterizations in CRC samples has proposed a panel of five miRNAs (miR-186-5p, miR-29a-3p, miR-29c-3p, miR-766-3p, and miR-491-5p) (from 22 miRNAs showing dysregulated patterns), enabling discrimination of CRC patients from healthy controls with a sensitivity of 72% and a specificity of 67% [129]. Urinary miR-129-1-3p and miR-566 have been suggested as biomarkers for early CRC [130].

3.5.3. lncRNA

Long non-coding RNAs (lncRNAs) measure over 200 nucleotides in length and are emerging as biomarkers in many cancer types [131]. They are thought to contribute to CRC development and progression by affecting wingless-related integration site/beta-catenin (WNT/β-catenin), phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt), EGFR, NOTCH, mammalian target of rapamycin (mTOR) and TP53 signaling [132]. They play pivotal roles in the regulation of gene expression in apoptosis and cell proliferation.
Circulating lncRNA (cf-lncRNA) biomarkers were recently proposed in CRC [133]. Despite their emerging role in cancer, to date, few studies have focused on cf-lncRNA biomarkers in the non-invasive diagnosis and management of CRC patients (Table 6). Other lncRNAs implicated in CRC include plasmacytoma variant translocation 1 (PVT1), H19 imprinted maternally expressed transcript (H19), metastasis associated lung adenocarcinoma transcript 1 (MALAT1), small nucleolar RNA host gene 1, 7 and 15 (SNHG1, SNHG7, SNHG15) taurine upregulated 1 (TUG1), X inactive specific transcript (XIST), regulator of reprogramming (ROR) and ZEB-1 antisense RNA 1 (ZEB1-AS1) [134].

3.5.4. circRNA

Circular RNA (circRNA) is a type of single-stranded RNA forming a covalently bound loop. Various circRNAs are thought to play important roles in cancer development, affecting processes such as apoptosis, cell-cycle regulation, cell proliferation, migration, and drug resistance development [147,148]. One of the unique features of these RNAs is their covalently closed cyclic structure, making them resistant to digestion by exonucleases. For this reason, their expression in cells is stable, and their half-life is prolonged, particularly in cell-free samples [149]. It has been shown that circRNAs act as miRNA sponges, containing binding sites for miRNA molecules [150]. By this mechanism, circRNAs affect the expression level of various target genes by absorbing miRNAs, participating in the occurrence, development, and progression of CRC (Table 7) [151,152].
Hon et al. demonstrated the transfer of drug resistance to sensitive cells via exosomes. They found 105 upregulated and 34 downregulated circRNAs in the FOLFOX-resistant HCT11-R colon cancer cell line. Authors concluded that hsa_circ_0000338 isolated from exosomes could serve as an early predictor of chemoresistance development [166].
Circ-0053277 was shown to sponge miR-2467-3p, promoting proliferation, migration, and epithelial-mesenchymal transition in CRC. Its upregulation was detected in CRC tissue [167], but to our knowledge, it has not been studied yet in liquid biopsies. As implied in Table 7, there are many circRNAs reported as relevant in CRC tissue but not yet detected in the circulation, providing potential targets for future liquid biopsy-based studies. Upregulation of circ_0026416 was reported in plasma, with the authors suggesting that it may act as an oncogene of CRC by sponging miR-136 [168]. Other circRNA biomarkers successfully detected from plasma or serum samples are outlined in Table 8.

3.6. Nucleic Acids Released by Gut Microbiota

The human intestinal tract hosts about 1014 bacteria representing more than a thousand species. Intestinal microbiota is generally stable during adult life, but a number of diseases show correlation with altered microbial composition in the gut [174]. Studying the human gut microbiome became very popular recently, with some promising results. Rezasoltani et al. proposed that the most ubiquitous environmental factor in epigenetic modifications may be the gut microbiota [175]. It has been reported that reduced butyrate production, implicated in the development of CRC, may be due to an altered bacterial flora [176]. Establishing the representative pattern of a healthy gut microbiome offers the possibility to associate pattern changes with various diseases.
Components of the gut microbiome release a tremendous amount of nucleic acids and other molecules into the host’s stool and circulation. Studies are emerging to investigate the utility of circulating bacterial DNA (cbDNA) in CRC diagnostics [61]. Some authors have already reported that colorectal neoplasia patients may be distinguished from healthy individuals on this basis [177]. The role of circulating extracellular nucleic acids produced by the gut microbiome is an exciting new direction for research in CRC. However, the difficulty to collect synchronized samples and the lack of standardization protocols represent extra challenges.

4. Conclusions

A large number of potential and confirmed cfDNA- and cfRNA-based markers are now available to monitor from peripheral blood, and some even from urine. However, we are still far away from using these biomarkers efficiently in personalized and preventive medical approaches. At this point, liquid biopsy cannot replace a tissue-based histological examination, which remains the essential step to provide basic information on tumor cell properties. Research on the correlation of tumor stage and/or grade with cfNA features is emerging and seems to be the next step for the successful application of liquid biopsy in this context. Additionally, as shown by many of the recent papers we have reviewed, the scientific community still seems to be focused on proposing further novel biomarkers rather than the standardization of methods and quality controls. Despite the availability of a few validated and approved tests such as blood-based Epi proColon and stool-based Cologuard, there is still plenty of potential to improve on these testing strategies in a variety of ways. This is likely to take a lot of time and effort, as new, more efficient methods are still being developed for the detection of low-concentration and fragmented nucleic acids. CRC is a disease that, while deadly in late stages, takes long enough to develop and is often fully curable when caught early. This gives us hope and reason to believe that CRC death tolls may be greatly reduced once cost-efficient liquid biopsy-based methods make the transition into routine medical practice.

Author Contributions

J.S.: Conceptualization, Investigation, Writing—original draft preparation, Visualization, Supervision. G.B.: Conceptualization, Investigation, Writing—original draft preparation, Visualization, Supervision. O.P.: Conceptualization, Writing—review and editing, Supervision. Á.C.: Writing—original draft preparation, Supervision. B.S.: Writing—original draft preparation. P.L.: Writing—review and editing. V.R.: Supervision. B.N.: Supervision, Funding acquisition, Writing—original draft preparation. T.S.: Supervision, Funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by OP Integrated Infrastructure for the projects (1) “Long term strategic research and development focused on the occurrence of lynch syndrome in the Slovak population and possibilities of prevention of tumors associated with this syndrome”, ITMS2014+: 313011V578 (50% of costs); and (2) “Integrative strategy in the development of personalized medicine of selected malignant tumors and its impact on quality of life”, ITMS2014+: 313011V446 (50% of costs), co-financed by the European Regional Development Fund.

Conflicts of Interest

J.S., O.P. and T.S. are employees of Geneton Ltd., participating in a research and development activity that does not conflict with the manuscript’s content. V.R. is an employee of Medirex Group Academy n.p.o., and participated in a research and project activity with no potential conflict of interest to the proposed manuscript. All remaining authors declare no conflict of interest.

References

  1. Currais, P.; Rosa, I.; Claro, I. Colorectal Cancer Carcinogenesis: From Bench to Bedside. World J. Gastrointest. Oncol. 2022, 14, 654–663. [Google Scholar] [CrossRef] [PubMed]
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  3. Wong, H.H.; Chu, P. Immunohistochemical Features of the Gastrointestinal Tract Tumors. J. Gastrointest. Oncol. 2012, 3, 262–284. [Google Scholar] [PubMed]
  4. Reddy, R.M.; Fleshman, J.W. Colorectal Gastrointestinal Stromal Tumors: A Brief Review. Clin. Colon Rectal Surg. 2006, 19, 69–77. [Google Scholar] [CrossRef] [Green Version]
  5. Alyabsi, M.; Sabatin, F.; Ramadan, M.; Jazieh, A.R. Colorectal Cancer Survival among Ministry of National Guard-Health Affairs (MNG-HA) Population 2009-2017: Retrospective Study. BMC Cancer 2021, 21, 954. [Google Scholar] [CrossRef]
  6. Lansdorp-Vogelaar, I.; van Ballegooijen, M.; Zauber, A.G.; Habbema, J.D.F.; Kuipers, E.J. Effect of Rising Chemotherapy Costs on the Cost Savings of Colorectal Cancer Screening. J. Natl. Cancer Inst. 2009, 101, 1412–1422. [Google Scholar] [CrossRef] [Green Version]
  7. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer Statistics, 2018. CA Cancer J. Clin. 2018, 68, 7–30. [Google Scholar] [CrossRef]
  8. Latchford, A. How Should Colonoscopy Surveillance in Lynch Syndrome Be Performed? Gastroenterology 2020, 158, 818–819. [Google Scholar] [CrossRef]
  9. Delcò, F.; Sonnenberg, A. Limitations of the Faecal Occult Blood Test in Screening for Colorectal Cancer. Ital. J. Gastroenterol. Hepatol. 1999, 31, 119–126. [Google Scholar]
  10. Lee, J.K.; Reis, V.; Liu, S.; Conn, L.; Groessl, E.J.; Ganiats, T.G.; Ho, S.B. Improving Fecal Occult Blood Testing Compliance Using a Mailed Educational Reminder. J. Gen. Intern. Med. 2009, 24, 1192–1197. [Google Scholar] [CrossRef] [Green Version]
  11. Bretthauer, M.; Kaminski, M.F.; Løberg, M.; Zauber, A.G.; Regula, J.; Kuipers, E.J.; Hernán, M.A.; McFadden, E.; Sunde, A.; Kalager, M.; et al. Population-Based Colonoscopy Screening for Colorectal Cancer: A Randomized Clinical Trial. JAMA Intern. Med. 2016, 176, 894–902. [Google Scholar] [CrossRef]
  12. Dhaliwal, A.; Vlachostergios, P.J.; Oikonomou, K.G.; Moshenyat, Y. Fecal DNA Testing for Colorectal Cancer Screening: Molecular Targets and Perspectives. World J. Gastrointest. Oncol. 2015, 7, 178–183. [Google Scholar] [CrossRef]
  13. Fan, C.-W.; Kuo, Y.-B.; Lin, G.-P.; Chen, S.-M.; Chang, S.-H.; Li, B.-A.; Chan, E.-C. Development of a Multiplexed Tumor-Associated Autoantibody-Based Blood Test for the Detection of Colorectal Cancer. Clin. Chim. Acta 2017, 475, 157–163. [Google Scholar] [CrossRef]
  14. Ladabaum, U.; Allen, J.; Wandell, M.; Ramsey, S. Colorectal Cancer Screening with Blood-Based Biomarkers: Cost-Effectiveness of Methylated Septin 9 DNA versus Current Strategies. Cancer Epidemiol. Biomarkers Prev. 2013, 22, 1567–1576. [Google Scholar] [CrossRef] [Green Version]
  15. Ross, J.S.; Fakih, M.; Ali, S.M.; Elvin, J.A.; Schrock, A.B.; Suh, J.; Vergilio, J.-A.; Ramkissoon, S.; Severson, E.; Daniel, S.; et al. Targeting HER2 in Colorectal Cancer: The Landscape of Amplification and Short Variant Mutations in ERBB2 and ERBB3. Cancer 2018, 124, 1358–1373. [Google Scholar] [CrossRef] [Green Version]
  16. Sameer, A.S. Colorectal Cancer: Molecular Mutations and Polymorphisms. Front. Oncol. 2013, 3, 114. [Google Scholar] [CrossRef] [Green Version]
  17. Nojadeh, J.N.; Behrouz Sharif, S.; Sakhinia, E. Microsatellite Instability in Colorectal Cancer. EXCLI J. 2018, 17, 159–168. [Google Scholar]
  18. Arvelo, F.; Sojo, F.; Cotte, C. Biology of Colorectal Cancer. Ecancermedicalscience 2015, 9, 520. [Google Scholar] [CrossRef] [Green Version]
  19. Lin, O.S. Colorectal Cancer Screening in Patients at Moderately Increased Risk due to Family History. World J. Gastrointest. Oncol. 2012, 4, 125–130. [Google Scholar] [CrossRef]
  20. Daca Alvarez, M.; Quintana, I.; Terradas, M.; Mur, P.; Balaguer, F.; Valle, L. The Inherited and Familial Component of Early-Onset Colorectal Cancer. Cells 2021, 10, 710. [Google Scholar] [CrossRef]
  21. Buglyó, G.; Styk, J.; Pös, O.; Csók, Á.; Repiska, V.; Soltész, B.; Szemes, T.; Nagy, B. Liquid Biopsy as a Source of Nucleic Acid Biomarkers in the Diagnosis and Management of Lynch Syndrome. Int. J. Mol. Sci. 2022, 23, 4284. [Google Scholar] [CrossRef] [PubMed]
  22. Nejadtaghi, M.; Jafari, H.; Farrokhi, E.; Samani, K.G. Familial Colorectal Cancer Type X (FCCTX) and the Correlation with Various Genes-A Systematic Review. Curr. Probl. Cancer 2017, 41, 388–397. [Google Scholar] [CrossRef] [PubMed]
  23. Nieminen, T.T.; O’Donohue, M.-F.; Wu, Y.; Lohi, H.; Scherer, S.W.; Paterson, A.D.; Ellonen, P.; Abdel-Rahman, W.M.; Valo, S.; Mecklin, J.-P.; et al. Germline Mutation of RPS20, Encoding a Ribosomal Protein, Causes Predisposition to Hereditary Nonpolyposis Colorectal Carcinoma without DNA Mismatch Repair Deficiency. Gastroenterology 2014, 147, 595–598.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Khattab, A.; Monga, D.K. Turcot Syndrome. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
  25. Nielsen, M.; Aretz, S. Familial Adenomatous Polyposis or APC-Associated Polyposis. Hered. Colorectal Cancer 2018, 99–111. [Google Scholar] [CrossRef]
  26. Magrin, L.; Fanale, D.; Brando, C.; Corsini, L.R.; Randazzo, U.; Di Piazza, M.; Gurrera, V.; Pedone, E.; Bazan Russo, T.D.; Vieni, S.; et al. MUTYH-Associated Tumor Syndrome: The Other Face of MAP. Oncogene 2022, 41, 2531–2539. [Google Scholar] [CrossRef]
  27. Palles, C.; Latchford, A.; Valle, L. Adenomatous Polyposis Syndromes: Polymerase Proofreading-Associated Polyposis. Hered. Colorectal Cancer 2018, 113–134. [Google Scholar] [CrossRef]
  28. Weren, R.D.A.; Ligtenberg, M.J.L.; Kets, C.M.; de Voer, R.M.; Verwiel, E.T.P.; Spruijt, L.; van Zelst-Stams, W.A.G.; Jongmans, M.C.; Gilissen, C.; Hehir-Kwa, J.Y.; et al. A Germline Homozygous Mutation in the Base-Excision Repair Gene NTHL1 Causes Adenomatous Polyposis and Colorectal Cancer. Nat. Genet. 2015, 47, 668–671. [Google Scholar] [CrossRef]
  29. Wimmer, K.; Kratz, C.P.; Vasen, H.F.A.; Caron, O.; Colas, C.; Entz-Werle, N.; Gerdes, A.-M.; Goldberg, Y.; Ilencikova, D.; Muleris, M.; et al. Diagnostic Criteria for Constitutional Mismatch Repair Deficiency Syndrome: Suggestions of the European Consortium “Care for CMMRD” (C4CMMRD). J. Med. Genet. 2014, 51, 355–365. [Google Scholar] [CrossRef] [Green Version]
  30. Wagner, A.; Aretz, S.; Auranen, A.; Bruno, M.J.; Cavestro, G.M.; Crosbie, E.J.; Goverde, A.; Jelsig, A.M.; Latchford, A.; van Leerdam, M.E.; et al. The Management of Peutz-Jeghers Syndrome: European Hereditary Tumour Group (EHTG) Guideline. J. Clin. Med. Res. 2021, 10, 473. [Google Scholar] [CrossRef]
  31. Pilarski, R. Hamartoma Tumor Syndrome: A Clinical Overview. Cancers 2019, 11, 844. [Google Scholar] [CrossRef] [Green Version]
  32. Dal Buono, A.; Gaiani, F.; Poliani, L.; Laghi, L. Juvenile Polyposis Syndrome: An Overview. Best Pract. Res. Clin. Gastroenterol. 2022, 101799. [Google Scholar] [CrossRef]
  33. Lieberman, S.; Walsh, T.; Schechter, M.; Adar, T.; Goldin, E.; Beeri, R.; Sharon, N.; Baris, H.; Ben Avi, L.; Half, E.; et al. Features of Patients With Hereditary Mixed Polyposis Syndrome Caused by Duplication of GREM1 and Implications for Screening and Surveillance. Gastroenterology 2017, 152, 1876–1880.e1. [Google Scholar] [CrossRef]
  34. Ballester-Vargas, V.; Tomlinson, I. Hereditary Mixed Polyposis Syndrome. Intest. Polyposis Syndr. 2016, 165–171. [Google Scholar] [CrossRef]
  35. Taupin, D.; Lam, W.; Rangiah, D.; McCallum, L.; Whittle, B.; Zhang, Y.; Andrews, D.; Field, M.; Goodnow, C.C.; Cook, M.C. A Deleterious RNF43 Germline Mutation in a Severely Affected Serrated Polyposis Kindred. Hum. Genome Var. 2015, 2, 15013. [Google Scholar] [CrossRef] [Green Version]
  36. Quintana, I.; Mejías-Luque, R.; Terradas, M.; Navarro, M.; Piñol, V.; Mur, P.; Belhadj, S.; Grau, E.; Darder, E.; Solanes, A.; et al. Evidence Suggests That Germline Mutations Are a Rare Cause of Serrated Polyposis. Gut 2018, 67, 2230–2232. [Google Scholar] [CrossRef]
  37. Yurgelun, M.B.; Kulke, M.H.; Fuchs, C.S.; Allen, B.A.; Uno, H.; Hornick, J.L.; Ukaegbu, C.I.; Brais, L.K.; McNamara, P.G.; Mayer, R.J.; et al. Cancer Susceptibility Gene Mutations in Individuals With Colorectal Cancer. J. Clin. Oncol. 2017, 35, 1086–1095. [Google Scholar] [CrossRef]
  38. Tutlewska, K.; Lubinski, J.; Kurzawski, G. Germline Deletions in the EPCAM Gene as a Cause of Lynch Syndrome—Literature Review. Hered. Cancer Clin. Pract. 2013, 11, 9. [Google Scholar] [CrossRef] [Green Version]
  39. Lynch, H.T.; Lynch, P.M.; Lanspa, S.J.; Snyder, C.L.; Lynch, J.F.; Boland, C.R. Review of the Lynch Syndrome: History, Molecular Genetics, Screening, Differential Diagnosis, and Medicolegal Ramifications. Clin. Genet. 2009, 76, 1–18. [Google Scholar] [CrossRef]
  40. Lynch, H.T.; Shaw, T.G. Practical Genetics of Colorectal Cancer. Chin. Clin. Oncol. 2013, 2, 12. [Google Scholar]
  41. Balaguer, F.; Moreira, L.; Lozano, J.J.; Link, A.; Ramirez, G.; Shen, Y.; Cuatrecasas, M.; Arnold, M.; Meltzer, S.J.; Syngal, S.; et al. Colorectal Cancers with Microsatellite Instability Display Unique miRNA Profiles. Clin. Cancer Res. 2011, 17, 6239–6249. [Google Scholar] [CrossRef] [Green Version]
  42. Giardiello, F.M.; Trimbath, J.D. Peutz-Jeghers Syndrome and Management Recommendations. Clin. Gastroenterol. Hepatol. 2006, 4, 408–415. [Google Scholar] [CrossRef] [PubMed]
  43. Chae, H.-D.; Jeon, C.-H. Peutz-Jeghers Syndrome with Germline Mutation of STK11. Ann. Surg. Treat. Res. 2014, 86, 325–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Goodenberger, M.; Lindor, N.M. Lynch Syndrome and MYH-Associated Polyposis: Review and Testing Strategy. J. Clin. Gastroenterol. 2011, 45, 488–500. [Google Scholar] [CrossRef]
  45. Bogaert, J.; Prenen, H. Molecular Genetics of Colorectal Cancer. Ann. Gastroenterol. Hepatol. 2014, 27, 9–14. [Google Scholar]
  46. Sweetser, S.; Smyrk, T.C.; Sinicrope, F.A. Serrated Colon Polyps as Precursors to Colorectal Cancer. Clin. Gastroenterol. Hepatol. 2013, 11, 760–767, quiz e54–e55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Gala, M.K.; Mizukami, Y.; Le, L.P.; Moriichi, K.; Austin, T.; Yamamoto, M.; Lauwers, G.Y.; Bardeesy, N.; Chung, D.C. Germline Mutations in Oncogene-Induced Senescence Pathways Are Associated with Multiple Sessile Serrated Adenomas. Gastroenterology 2014, 146, 520–529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S.; et al. Mismatch Repair Deficiency Predicts Response of Solid Tumors to PD-1 Blockade. Science 2017, 357, 409–413. [Google Scholar] [CrossRef] [Green Version]
  49. Rudolph, K.L.; Millard, M.; Bosenberg, M.W.; DePinho, R.A. Telomere Dysfunction and Evolution of Intestinal Carcinoma in Mice and Humans. Nat. Genet. 2001, 28, 155–159. [Google Scholar] [CrossRef]
  50. Ayiomamitis, G.D.; Notas, G.; Zaravinos, A.; Zizi-Sermpetzoglou, A.; Georgiadou, M.; Sfakianaki, O.; Kouroumallis, E. Differences in Telomerase Activity between Colon and Rectal Cancer. Can. J. Surg. 2014, 57, 199–208. [Google Scholar] [CrossRef] [Green Version]
  51. Wang, J.-Y.; Hsieh, J.-S.; Chang, M.-Y.; Huang, T.-J.; Chen, F.-M.; Cheng, T.-L.; Alexandersen, K.; Huang, Y.-S.; Tzou, W.-S.; Lin, S.-R. Molecular Detection of APC, K-Ras, and p53 Mutations in the Serum of Colorectal Cancer Patients as Circulating Biomarkers. World J. Surg. 2004, 28, 721–726. [Google Scholar] [CrossRef]
  52. Midthun, L.; Shaheen, S.; Deisch, J.; Senthil, M.; Tsai, J.; Hsueh, C.-T. Concomitant and Mutations in Colorectal Cancer. J. Gastrointest. Oncol. 2019, 10, 577–581. [Google Scholar] [CrossRef] [PubMed]
  53. Mauri, G.; Vitiello, P.P.; Sogari, A.; Crisafulli, G.; Sartore-Bianchi, A.; Marsoni, S.; Siena, S.; Bardelli, A. Liquid Biopsies to Monitor and Direct Cancer Treatment in Colorectal Cancer. Br. J. Cancer 2022. [Google Scholar] [CrossRef] [PubMed]
  54. Zygulska, A.L.; Pierzchalski, P. Novel Diagnostic Biomarkers in Colorectal Cancer. Int. J. Mol. Sci. 2022, 23, 852. [Google Scholar] [CrossRef] [PubMed]
  55. Umwali, Y.; Yue, C.-B.; Gabriel, A.N.A.; Zhang, Y.; Zhang, X. Roles of Exosomes in Diagnosis and Treatment of Colorectal Cancer. World J. Clin. Cases 2021, 9, 4467–4479. [Google Scholar] [CrossRef]
  56. Ruiz-López, L.; Blancas, I.; Garrido, J.M.; Mut-Salud, N.; Moya-Jódar, M.; Osuna, A.; Rodríguez-Serrano, F. The Role of Exosomes on Colorectal Cancer: A Review. J. Gastroenterol. Hepatol. 2018, 33, 792–799. [Google Scholar] [CrossRef] [Green Version]
  57. Pös, O.; Biró, O.; Szemes, T.; Nagy, B. Circulating Cell-Free Nucleic Acids: Characteristics and Applications. Eur. J. Hum. Genet. 2018, 26, 937–945. [Google Scholar] [CrossRef] [Green Version]
  58. Szilágyi, M.; Pös, O.; Márton, É.; Buglyó, G.; Soltész, B.; Keserű, J.; Penyige, A.; Szemes, T.; Nagy, B. Circulating Cell-Free Nucleic Acids: Main Characteristics and Clinical Application. Int. J. Mol. Sci. 2020, 21, 6827. [Google Scholar] [CrossRef]
  59. Aucamp, J.; Bronkhorst, A.J.; Badenhorst, C.P.S.; Pretorius, P.J. The Diverse Origins of Circulating Cell-Free DNA in the Human Body: A Critical Re-Evaluation of the Literature. Biol. Rev. Camb. Philos. Soc. 2018, 93, 1649–1683. [Google Scholar] [CrossRef]
  60. Hu, Z.; Chen, H.; Long, Y.; Li, P.; Gu, Y. The Main Sources of Circulating Cell-Free DNA: Apoptosis, Necrosis and Active Secretion. Crit. Rev. Oncol. Hematol. 2021, 157, 103166. [Google Scholar] [CrossRef]
  61. Glyn, T.; Purcell, R. Circulating Bacterial DNA: A New Paradigm for Cancer Diagnostics. Front. Med. 2022, 9, 831096. [Google Scholar] [CrossRef]
  62. Dasari, A.; Morris, V.K.; Allegra, C.J.; Atreya, C.; Benson, A.B., 3rd; Boland, P.; Chung, K.; Copur, M.S.; Corcoran, R.B.; Deming, D.A.; et al. ctDNA Applications and Integration in Colorectal Cancer: An NCI Colon and Rectal-Anal Task Forces Whitepaper. Nat. Rev. Clin. Oncol. 2020, 17, 757–770. [Google Scholar] [CrossRef]
  63. Frattini, M.; Gallino, G.; Signoroni, S.; Balestra, D.; Lusa, L.; Battaglia, L.; Sozzi, G.; Bertario, L.; Leo, E.; Pilotti, S.; et al. Quantitative and Qualitative Characterization of Plasma DNA Identifies Primary and Recurrent Colorectal Cancer. Cancer Lett. 2008, 263, 170–181. [Google Scholar] [CrossRef]
  64. Cassinotti, E.; Boni, L.; Segato, S.; Rausei, S.; Marzorati, A.; Rovera, F.; Dionigi, G.; David, G.; Mangano, A.; Sambucci, D.; et al. Free Circulating DNA as a Biomarker of Colorectal Cancer. Int. J. Surg. 2013, 11 (Suppl. 1), S54–S57. [Google Scholar] [CrossRef] [Green Version]
  65. Tie, J.; Cohen, J.D.; Lahouel, K.; Lo, S.N.; Wang, Y.; Kosmider, S.; Wong, R.; Shapiro, J.; Lee, M.; Harris, S.; et al. Circulating Tumor DNA Analysis Guiding Adjuvant Therapy in Stage II Colon Cancer. N. Engl. J. Med. 2022, 386, 2261–2272. [Google Scholar] [CrossRef]
  66. Zhitnyuk, Y.V.; Koval, A.P.; Alferov, A.A.; Shtykova, Y.A.; Mamedov, I.Z.; Kushlinskii, N.E.; Chudakov, D.M.; Shcherbo, D.S. Deep cfDNA Fragment End Profiling Enables Cancer Detection. Mol. Cancer 2022, 21, 26. [Google Scholar]
  67. Cristiano, S.; Leal, A.; Phallen, J.; Fiksel, J.; Adleff, V.; Bruhm, D.C.; Jensen, S.Ø.; Medina, J.E.; Hruban, C.; White, J.R.; et al. Genome-Wide Cell-Free DNA Fragmentation in Patients with Cancer. Nature 2019, 570, 385–389. [Google Scholar] [CrossRef]
  68. Mouliere, F.; Chandrananda, D.; Piskorz, A.M.; Moore, E.K.; Morris, J.; Ahlborn, L.B.; Mair, R.; Goranova, T.; Marass, F.; Heider, K.; et al. Enhanced Detection of Circulating Tumor DNA by Fragment Size Analysis. Sci. Transl. Med. 2018, 10, eaat4921. [Google Scholar] [CrossRef] [Green Version]
  69. Flamini, E.; Mercatali, L.; Nanni, O.; Calistri, D.; Nunziatini, R.; Zoli, W.; Rosetti, P.; Gardini, N.; Lattuneddu, A.; Verdecchia, G.M.; et al. Free DNA and Carcinoembryonic Antigen Serum Levels: An Important Combination for Diagnosis of Colorectal Cancer. Clin. Cancer Res. 2006, 12, 6985–6988. [Google Scholar] [CrossRef] [Green Version]
  70. Kidess, E.; Heirich, K.; Wiggin, M.; Vysotskaia, V.; Visser, B.C.; Marziali, A.; Wiedenmann, B.; Norton, J.A.; Lee, M.; Jeffrey, S.S.; et al. Mutation Profiling of Tumor DNA from Plasma and Tumor Tissue of Colorectal Cancer Patients with a Novel, High-Sensitivity Multiplexed Mutation Detection Platform. Oncotarget 2015, 6, 2549–2561. [Google Scholar] [CrossRef] [Green Version]
  71. Vidal, J.; Muinelo, L.; Dalmases, A.; Jones, F.; Edelstein, D.; Iglesias, M.; Orrillo, M.; Abalo, A.; Rodríguez, C.; Brozos, E.; et al. Plasma ctDNA RAS Mutation Analysis for the Diagnosis and Treatment Monitoring of Metastatic Colorectal Cancer Patients. Ann. Oncol. 2017, 28, 1325–1332. [Google Scholar] [CrossRef]
  72. Nakamura, Y.; Okamoto, W.; Kato, T.; Esaki, T.; Kato, K.; Komatsu, Y.; Yuki, S.; Masuishi, T.; Nishina, T.; Ebi, H.; et al. Circulating Tumor DNA-Guided Treatment with Pertuzumab plus Trastuzumab for HER2-Amplified Metastatic Colorectal Cancer: A Phase 2 Trial. Nat. Med. 2021, 27, 1899–1903. [Google Scholar] [CrossRef]
  73. Mouliere, F.; Robert, B.; Arnau Peyrotte, E.; Del Rio, M.; Ychou, M.; Molina, F.; Gongora, C.; Thierry, A.R. High Fragmentation Characterizes Tumour-Derived Circulating DNA. PLoS ONE 2011, 6, e23418. [Google Scholar] [CrossRef]
  74. Henriksen, T.V.; Reinert, T.; Christensen, E.; Sethi, H.; Birkenkamp-Demtröder, K.; Gögenur, M.; Gögenur, I.; Zimmermann, B.G.; IMPROVE Study Group; Dyrskjøt, L.; et al. The Effect of Surgical Trauma on Circulating Free DNA Levels in Cancer Patients-Implications for Studies of Circulating Tumor DNA. Mol. Oncol. 2020, 14, 1670–1679. [Google Scholar] [CrossRef]
  75. Osumi, H.; Shinozaki, E.; Yamaguchi, K.; Zembutsu, H. Clinical Utility of Circulating Tumor DNA for Colorectal Cancer. Cancer Sci. 2019, 110, 1148–1155. [Google Scholar] [CrossRef] [Green Version]
  76. Ahcene Djaballah, S.; Daniel, F.; Milani, A.; Ricagno, G.; Lonardi, S. HER2 in Colorectal Cancer: The Long and Winding Road From Negative Predictive Factor to Positive Actionable Target. Am. Soc. Clin. Oncol. Educ. Book 2022, 42, 1–14. [Google Scholar] [CrossRef]
  77. Barták, B.K.; Nagy, Z.B.; Spisák, S.; Tulassay, Z.; Dank, M.; Igaz, P.; Molnár, B. In vivo analysis of circulating cell-free DNA release and degradation. Orv. Hetil. 2018, 159, 223–233. [Google Scholar] [CrossRef] [Green Version]
  78. Wu, D.; Zhou, G.; Jin, P.; Zhu, J.; Li, S.; Wu, Q.; Wang, G.; Sheng, J.; Wang, J.; Song, L.; et al. Detection of Colorectal Cancer Using a Simplified SEPT9 Gene Methylation Assay Is a Reliable Method for Opportunistic Screening. J. Mol. Diagn. 2016, 18, 535–545. [Google Scholar] [CrossRef] [Green Version]
  79. Song, L.; Li, Y.; Jia, J.; Zhou, G.; Wang, J.; Kang, Q.; Jin, P.; Sheng, J.; Cai, G.; Cai, S.; et al. Algorithm Optimization in Methylation Detection with Multiple RT-qPCR. PLoS ONE 2016, 11, e0163333. [Google Scholar] [CrossRef] [Green Version]
  80. Wasserkort, R.; Kalmar, A.; Valcz, G.; Spisak, S.; Krispin, M.; Toth, K.; Tulassay, Z.; Sledziewski, A.Z.; Molnar, B. Aberrant Septin 9 DNA Methylation in Colorectal Cancer Is Restricted to a Single CpG Island. BMC Cancer 2013, 13, 398. [Google Scholar] [CrossRef] [Green Version]
  81. Tóth, K.; Wasserkort, R.; Sipos, F.; Kalmár, A.; Wichmann, B.; Leiszter, K.; Valcz, G.; Juhász, M.; Miheller, P.; Patai, Á.V.; et al. Detection of Methylated Septin 9 in Tissue and Plasma of Colorectal Patients with Neoplasia and the Relationship to the Amount of Circulating Cell-Free DNA. PLoS ONE 2014, 9, e115415. [Google Scholar] [CrossRef]
  82. Leon Arellano, M.; García-Arranz, M.; Ruiz, R.; Olivera, R.; Magallares, S.; Olmedillas-Lopez, S.; Valdes-Sanchez, T.; Guadalajara, H.; García-Olmo, D. A First Step to a Biomarker of Curative Surgery in Colorectal Cancer by Liquid Biopsy of Methylated Septin 9 Gene. Dis. Markers 2020, 2020, 9761406. [Google Scholar] [CrossRef] [PubMed]
  83. Wills, B.; Gorse, E.; Lee, V. Role of Liquid Biopsies in Colorectal Cancer. Curr. Probl. Cancer 2018, 42, 593–600. [Google Scholar] [CrossRef] [PubMed]
  84. Provenzale, D.; Gupta, S.; Ahnen, D.J.; Markowitz, A.J.; Chung, D.C.; Mayer, R.J.; Regenbogen, S.E.; Blanco, A.M.; Bray, T.; Cooper, G.; et al. NCCN Guidelines Insights: Colorectal Cancer Screening, Version 1.2018. J. Natl. Compr. Cancer Netw. 2018, 16, 939–949. [Google Scholar] [CrossRef] [PubMed]
  85. Lamb, Y.N.; Dhillon, S. Epi proColon 2.0 CE: A Blood-Based Screening Test for Colorectal Cancer. Mol. Diagn. Ther. 2017, 21, 225–232. [Google Scholar] [CrossRef]
  86. Song, L.; Jia, J.; Peng, X.; Xiao, W.; Li, Y. The Performance of the SEPT9 Gene Methylation Assay and a Comparison with Other CRC Screening Tests: A Meta-Analysis. Sci. Rep. 2017, 7, 3032. [Google Scholar] [CrossRef] [Green Version]
  87. Adler, A.; Geiger, S.; Keil, A.; Bias, H.; Schatz, P.; deVos, T.; Dhein, J.; Zimmermann, M.; Tauber, R.; Wiedenmann, B. Improving Compliance to Colorectal Cancer Screening Using Blood and Stool Based Tests in Patients Refusing Screening Colonoscopy in Germany. BMC Gastroenterol. 2014, 14, 183. [Google Scholar] [CrossRef] [Green Version]
  88. Imperiale, T.F.; Ransohoff, D.F.; Itzkowitz, S.H.; Levin, T.R.; Lavin, P.; Lidgard, G.P.; Ahlquist, D.A.; Berger, B.M. Multitarget Stool DNA Testing for Colorectal-Cancer Screening. N. Engl. J. Med. 2014, 370, 1287–1297. [Google Scholar] [CrossRef] [Green Version]
  89. Ahlquist, D.A. Multi-Target Stool DNA Test: A New High Bar for Noninvasive Screening. Dig. Dis. Sci. 2015, 60, 623–633. [Google Scholar] [CrossRef]
  90. Mayor, R.; Casadomé, L.; Azuara, D.; Moreno, V.; Clark, S.J.; Capellà, G.; Peinado, M.A. Long-Range Epigenetic Silencing at 2q14.2 Affects Most Human Colorectal Cancers and May Have Application as a Non-Invasive Biomarker of Disease. Br. J. Cancer 2009, 100, 1534–1539. [Google Scholar] [CrossRef] [Green Version]
  91. Lee, B.B.; Lee, E.J.; Jung, E.H.; Chun, H.-K.; Chang, D.K.; Song, S.Y.; Park, J.; Kim, D.-H. Aberrant Methylation of APC, MGMT, RASSF2A, and Wif-1 Genes in Plasma as a Biomarker for Early Detection of Colorectal Cancer. Clin. Cancer Res. 2009, 15, 6185–6191. [Google Scholar] [CrossRef] [Green Version]
  92. Ahlquist, D.A.; Taylor, W.R.; Mahoney, D.W.; Zou, H.; Domanico, M.; Thibodeau, S.N.; Boardman, L.A.; Berger, B.M.; Lidgard, G.P. The Stool DNA Test Is More Accurate than the Plasma Septin 9 Test in Detecting Colorectal Neoplasia. Clin. Gastroenterol. Hepatol. 2012, 10, 272–277.e1. [Google Scholar] [CrossRef] [Green Version]
  93. Gutierrez, A.; Demond, H.; Brebi, P.; Ili, C.G. Novel Methylation Biomarkers for Colorectal Cancer Prognosis. Biomolecules 2021, 11, 1722. [Google Scholar] [CrossRef]
  94. Laugsand, E.A.; Brenne, S.S.; Skorpen, F. DNA Methylation Markers Detected in Blood, Stool, Urine, and Tissue in Colorectal Cancer: A Systematic Review of Paired Samples. Int. J. Colorectal Dis. 2021, 36, 239–251. [Google Scholar] [CrossRef]
  95. Trejo-Becerril, C.; Pérez-Cárdenas, E.; Taja-Chayeb, L.; Anker, P.; Herrera-Goepfert, R.; Medina-Velázquez, L.A.; Hidalgo-Miranda, A.; Pérez-Montiel, D.; Chávez-Blanco, A.; Cruz-Velázquez, J.; et al. Cancer Progression Mediated by Horizontal Gene Transfer in an in Vivo Model. PLoS ONE 2012, 7, e52754. [Google Scholar] [CrossRef] [Green Version]
  96. Alekseeva, L.; Mironova, N. Role of Cell-Free DNA and Deoxyribonucleases in Tumor Progression. Int. J. Mol. Sci. 2021, 22, 2246. [Google Scholar] [CrossRef]
  97. Thierry, A.R.; El Messaoudi, S.; Gahan, P.B.; Anker, P.; Stroun, M. Origins, Structures, and Functions of Circulating DNA in Oncology. Cancer Metastasis Rev. 2016, 35, 347–376. [Google Scholar] [CrossRef] [Green Version]
  98. Haupts, A.; Vogel, A.; Foersch, S.; Hartmann, M.; Maderer, A.; Wachter, N.; Huber, T.; Kneist, W.; Roth, W.; Lang, H.; et al. Comparative Analysis of Nuclear and Mitochondrial DNA from Tissue and Liquid Biopsies of Colorectal Cancer Patients. Sci. Rep. 2021, 11, 16745. [Google Scholar] [CrossRef]
  99. Meddeb, R.; Dache, Z.A.A.; Thezenas, S.; Otandault, A.; Tanos, R.; Pastor, B.; Sanchez, C.; Azzi, J.; Tousch, G.; Azan, S.; et al. Quantifying Circulating Cell-Free DNA in Humans. Sci. Rep. 2019, 9, 5220. [Google Scholar] [CrossRef] [Green Version]
  100. Xu, Y.; Zhou, J.; Yuan, Q.; Su, J.; Li, Q.; Lu, X.; Zhang, L.; Cai, Z.; Han, J. Quantitative Detection of Circulating MT-ND1 as a Potential Biomarker for Colorectal Cancer. Bosn. J. Basic Med. Sci. 2021, 21, 577–586. [Google Scholar] [CrossRef]
  101. Zhou, K.; Liu, Y.; Yuan, Q.; Lai, D.; Guo, S.; Wang, Z.; Su, L.; Zhang, H.; Wang, X.; Guo, W.; et al. Next-Generation Sequencing-Based Analysis of Urine Cell-Free mtDNA Reveals Aberrant Fragmentation and Mutation Profile in Cancer Patients. Clin. Chem. 2022, 68, 561–573. [Google Scholar] [CrossRef]
  102. Thyagarajan, B.; Guan, W.; Fedirko, V.; Barcelo, H.; Tu, H.; Gross, M.; Goodman, M.; Bostick, R.M. No Association between Mitochondrial DNA Copy Number and Colorectal Adenomas. Mol. Carcinog. 2016, 55, 1290–1296. [Google Scholar] [CrossRef] [Green Version]
  103. Rapisuwon, S.; Vietsch, E.E.; Wellstein, A. Circulating Biomarkers to Monitor Cancer Progression and Treatment. Comput. Struct. Biotechnol. J. 2016, 14, 211–222. [Google Scholar] [CrossRef] [Green Version]
  104. Xue, V.W.; Cheung, M.T.; Chan, P.T.; Luk, L.L.Y.; Lee, V.H.; Au, T.C.; Yu, A.C.; Cho, W.C.S.; Tsang, H.F.A.; Chan, A.K.; et al. Non-Invasive Potential Circulating mRNA Markers for Colorectal Adenoma Using Targeted Sequencing. Sci. Rep. 2019, 9, 12943. [Google Scholar] [CrossRef]
  105. Shu, T.; Wu, K.; Guo, Y.; He, Q.; Song, X.; Shan, J.; Wu, L.; Liu, J.; Wang, Z.; Liu, L.; et al. Evaluation of Fecal SYPL1 as a Diagnostic Biomarker in Colorectal Cancer. Clin. Biochem. 2022, 103, 8–15. [Google Scholar] [CrossRef] [PubMed]
  106. Engstrand, J.; Nilsson, H.; Strömberg, C.; Jonas, E.; Freedman, J. Colorectal Cancer Liver Metastases—A Population-Based Study on Incidence, Management and Survival. BMC Cancer 2018, 18, 78. [Google Scholar] [CrossRef] [PubMed]
  107. Favoriti, P.; Carbone, G.; Greco, M.; Pirozzi, F.; Pirozzi, R.E.M.; Corcione, F. Worldwide Burden of Colorectal Cancer: A Review. Updates Surg. 2016, 68, 7–11. [Google Scholar] [CrossRef] [PubMed]
  108. Battula, N.; Tsapralis, D.; Mayer, D.; Isaac, J.; Muiesan, P.; Sutcliffe, R.P.; Bramhall, S.; Mirza, D.; Marudanayagam, R. Repeat Liver Resection for Recurrent Colorectal Metastases: A Single-Centre, 13-Year Experience. HPB 2014, 16, 157–163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Imai, K.; Allard, M.-A.; Benitez, C.C.; Vibert, E.; Sa Cunha, A.; Cherqui, D.; Castaing, D.; Bismuth, H.; Baba, H.; Adam, R. Early Recurrence After Hepatectomy for Colorectal Liver Metastases: What Optimal Definition and What Predictive Factors? Oncologist 2016, 21, 887–894. [Google Scholar] [CrossRef] [Green Version]
  110. Pun, J.C.-S.; Chan, J.Y.-J.; Chun, B.K.-M.; Ng, K.-W.; Tsui, S.Y.-K.; Wan, T.M.-H.; Lo, O.; Poon, J.T.-C.; Ng, L.; Pang, R. Plasma Bmi1 mRNA as a Potential Prognostic Biomarker for Distant Metastasis in Colorectal Cancer Patients. Mol. Clin. Oncol. 2014, 2, 817–820. [Google Scholar] [CrossRef] [Green Version]
  111. Jimenez-Luna, C.; González-Flores, E.; Ortiz, R.; Martínez-González, L.J.; Antúnez-Rodríguez, A.; Expósito-Ruiz, M.; Melguizo, C.; Caba, O.; Prados, J. Circulating PTGS2, JAG1, GUCY2C and PGF mRNA in Peripheral Blood and Serum as Potential Biomarkers for Patients with Metastatic Colon Cancer. J. Clin. Med. Res. 2021, 10, 2248. [Google Scholar] [CrossRef]
  112. Kong, Y.W.; Ferland-McCollough, D.; Jackson, T.J.; Bushell, M. microRNAs in Cancer Management. Lancet Oncol. 2012, 13, e249–e258. [Google Scholar] [CrossRef]
  113. Peng, Y.; Croce, C.M. The Role of MicroRNAs in Human Cancer. Signal Transduct. Target. Ther. 2016, 1, 15004. [Google Scholar] [CrossRef] [Green Version]
  114. Sohel, M.M.H. Circulating microRNAs as Biomarkers in Cancer Diagnosis. Life Sci. 2020, 248, 117473. [Google Scholar] [CrossRef]
  115. Turchinovich, A.; Weiz, L.; Langheinz, A.; Burwinkel, B. Characterization of Extracellular Circulating microRNA. Nucleic Acids Res. 2011, 39, 7223–7233. [Google Scholar] [CrossRef]
  116. Kang, E.; Jung, S.C.; Nam, S.K.; Park, Y.; Seo, S.H.; Park, K.U.; Oh, H.-K.; Kim, D.-W.; Kang, S.-B.; Lee, H.S. Tissue miR-200c-3p and Circulating miR-1290 as Potential Prognostic Biomarkers for Colorectal Cancer. Sci. Rep. 2022, 12, 2295. [Google Scholar] [CrossRef]
  117. Nassar, F.J.; Msheik, Z.S.; Itani, M.M.; Helou, R.E.; Hadla, R.; Kreidieh, F.; Bejjany, R.; Mukherji, D.; Shamseddine, A.; Nasr, R.R.; et al. Circulating miRNA as Biomarkers for Colorectal Cancer Diagnosis and Liver Metastasis. Diagnostics (Basel) 2021, 11, 341. [Google Scholar] [CrossRef]
  118. Chen, E.; Li, Q.; Wang, H.; Yang, F.; Min, L.; Yang, J. MiR-92a Promotes Tumorigenesis of Colorectal Cancer, a Transcriptomic and Functional Based Study. Biomed. Pharmacother. 2018, 106, 1370–1377. [Google Scholar] [CrossRef]
  119. Huang, Z.; Huang, D.; Ni, S.; Peng, Z.; Sheng, W.; Du, X. Plasma microRNAs Are Promising Novel Biomarkers for Early Detection of Colorectal Cancer. Int. J. Cancer 2010, 127, 118–126. [Google Scholar] [CrossRef]
  120. Min, L.; Chen, L.; Liu, S.; Yu, Y.; Guo, Q.; Li, P.; Zhu, S. Loss of Circulating Exosomal miR-92b Is a Novel Biomarker of Colorectal Cancer at Early Stage. Int. J. Med. Sci. 2019, 16, 1231–1237. [Google Scholar] [CrossRef] [Green Version]
  121. Fu, F.; Jiang, W.; Zhou, L.; Chen, Z. Circulating Exosomal miR-17-5p and miR-92a-3p Predict Pathologic Stage and Grade of Colorectal Cancer. Transl. Oncol. 2018, 11, 221–232. [Google Scholar] [CrossRef]
  122. Zou, S.-L.; Chen, Y.-L.; Ge, Z.-Z.; Qu, Y.-Y.; Cao, Y.; Kang, Z.-X. Downregulation of Serum Exosomal miR-150-5p Is Associated with Poor Prognosis in Patients with Colorectal Cancer. Cancer Biomark. 2019, 26, 69–77. [Google Scholar] [CrossRef] [PubMed]
  123. Sun, L.; Liu, X.; Pan, B.; Hu, X.; Zhu, Y.; Su, Y.; Guo, Z.; Zhang, G.; Xu, M.; Xu, X.; et al. Serum Exosomal miR-122 as a Potential Diagnostic and Prognostic Biomarker of Colorectal Cancer with Liver Metastasis. J. Cancer 2020, 11, 630–637. [Google Scholar] [CrossRef]
  124. Imaoka, H.; Toiyama, Y.; Fujikawa, H.; Hiro, J.; Saigusa, S.; Tanaka, K.; Inoue, Y.; Mohri, Y.; Mori, T.; Kato, T.; et al. Circulating microRNA-1290 as a Novel Diagnostic and Prognostic Biomarker in Human Colorectal Cancer. Ann. Oncol. 2016, 27, 1879–1886. [Google Scholar] [CrossRef]
  125. Peng, X.; Wang, J.; Zhang, C.; Liu, K.; Zhao, L.; Chen, X.; Huang, G.; Lai, Y. A Three-miRNA Panel in Serum as a Noninvasive Biomarker for Colorectal Cancer Detection. Int. J. Biol. Markers 2020, 35, 74–82. [Google Scholar] [CrossRef]
  126. Zhang, Y.; Li, M.; Ding, Y.; Fan, Z.; Zhang, J.; Zhang, H.; Jiang, B.; Zhu, Y. Serum MicroRNA Profile in Patients with Colon Adenomas or Cancer. BMC Med. Genom. 2017, 10, 23. [Google Scholar] [CrossRef] [Green Version]
  127. Maminezhad, H.; Ghanadian, S.; Pakravan, K.; Razmara, E.; Rouhollah, F.; Mossahebi-Mohammadi, M.; Babashah, S. A Panel of Six-Circulating miRNA Signature in Serum and Its Potential Diagnostic Value in Colorectal Cancer. Life Sci. 2020, 258, 118226. [Google Scholar] [CrossRef]
  128. Sazanov, A.A.; Kiselyova, E.V.; Zakharenko, A.A.; Romanov, M.N.; Zaraysky, M.I. Plasma and Saliva miR-21 Expression in Colorectal Cancer Patients. J. Appl. Genet. 2017, 58, 231–237. [Google Scholar] [CrossRef]
  129. Rapado-González, Ó.; Majem, B.; Álvarez-Castro, A.; Díaz-Peña, R.; Abalo, A.; Suárez-Cabrera, L.; Gil-Moreno, A.; Santamaría, A.; López-López, R.; Muinelo-Romay, L.; et al. A Novel Saliva-Based miRNA Signature for Colorectal Cancer Diagnosis. J. Clin. Med. Res. 2019, 8, 2029. [Google Scholar] [CrossRef] [Green Version]
  130. Iwasaki, H.; Shimura, T.; Kitagawa, M.; Yamada, T.; Nishigaki, R.; Fukusada, S.; Okuda, Y.; Katano, T.; Horike, S.-I.; Kataoka, H. A Novel Urinary miRNA Biomarker for Early Detection of Colorectal Cancer. Cancers 2022, 14, 461. [Google Scholar] [CrossRef]
  131. Taniue, K.; Akimitsu, N. The Functions and Unique Features of LncRNAs in Cancer Development and Tumorigenesis. Int. J. Mol. Sci. 2021, 22, 632. [Google Scholar] [CrossRef] [PubMed]
  132. Schwarzmueller, L.; Bril, O.; Vermeulen, L.; Léveillé, N. Emerging Role and Therapeutic Potential of lncRNAs in Colorectal Cancer. Cancers 2020, 12, 3843. [Google Scholar] [CrossRef] [PubMed]
  133. Galamb, O.; Barták, B.K.; Kalmár, A.; Nagy, Z.B.; Szigeti, K.A.; Tulassay, Z.; Igaz, P.; Molnár, B. Diagnostic and Prognostic Potential of Tissue and Circulating Long Non-Coding RNAs in Colorectal Tumors. World J. Gastroenterol. 2019, 25, 5026–5048. [Google Scholar] [CrossRef] [PubMed]
  134. Chen, S.; Shen, X. Long Noncoding RNAs: Functions and Mechanisms in Colon Cancer. Mol. Cancer 2020, 19, 167. [Google Scholar] [CrossRef] [PubMed]
  135. Wu, Y.; Yang, L.; Zhao, J.; Li, C.; Nie, J.; Liu, F.; Zhuo, C.; Zheng, Y.; Li, B.; Wang, Z.; et al. Nuclear-Enriched Abundant Transcript 1 as a Diagnostic and Prognostic Biomarker in Colorectal Cancer. Mol. Cancer 2015, 14, 191. [Google Scholar] [CrossRef] [Green Version]
  136. Dai, M.; Chen, X.; Mo, S.; Li, J.; Huang, Z.; Huang, S.; Xu, J.; He, B.; Zou, Y.; Chen, J.; et al. Meta-Signature LncRNAs Serve as Novel Biomarkers for Colorectal Cancer: Integrated Bioinformatics Analysis, Experimental Validation and Diagnostic Evaluation. Sci. Rep. 2017, 7, 46572. [Google Scholar] [CrossRef]
  137. Shaker, O.G.; Senousy, M.A.; Elbaz, E.M. Association of rs6983267 at 8q24, HULC rs7763881 Polymorphisms and Serum lncRNAs CCAT2 and HULC with Colorectal Cancer in Egyptian Patients. Sci. Rep. 2017, 7, 16246. [Google Scholar] [CrossRef]
  138. Wang, L.; Duan, W.; Yan, S.; Xie, Y.; Wang, C. Circulating Long Non-Coding RNA Colon Cancer-Associated Transcript 2 Protected by Exosome as a Potential Biomarker for Colorectal Cancer. Biomed. Pharmacother. 2019, 113, 108758. [Google Scholar] [CrossRef]
  139. Liu, T.; Zhang, X.; Gao, S.; Jing, F.; Yang, Y.; Du, L.; Zheng, G.; Li, P.; Li, C.; Wang, C. Exosomal Long Noncoding RNA CRNDE-H as a Novel Serum-Based Biomarker for Diagnosis and Prognosis of Colorectal Cancer. Oncotarget 2016, 7, 85551–85563. [Google Scholar] [CrossRef]
  140. Yang, Y.-N.; Zhang, R.; Du, J.-W.; Yuan, H.-H.; Li, Y.-J.; Wei, X.-L.; Du, X.-X.; Jiang, S.-L.; Han, Y. Predictive Role of UCA1-Containing Exosomes in Cetuximab-Resistant Colorectal Cancer. Cancer Cell Int. 2018, 18, 164. [Google Scholar] [CrossRef] [Green Version]
  141. Yu, M.; Song, X.-G.; Zhao, Y.-J.; Dong, X.-H.; Niu, L.-M.; Zhang, Z.-J.; Shang, X.-L.; Tang, Y.-Y.; Song, X.-R.; Xie, L. Circulating Serum Exosomal Long Non-Coding RNAs FOXD2-AS1, NRIR, and XLOC_009459 as Diagnostic Biomarkers for Colorectal Cancer. Front. Oncol. 2021, 11, 618967. [Google Scholar] [CrossRef]
  142. Abedini, P.; Fattahi, A.; Agah, S.; Talebi, A.; Beygi, A.H.; Amini, S.M.; Mirzaei, A.; Akbari, A. Expression Analysis of Circulating Plasma Long Noncoding RNAs in Colorectal Cancer: The Relevance of lncRNAs ATB and CCAT1 as Potential Clinical Hallmarks. J. Cell. Physiol. 2019, 234, 22028–22033. [Google Scholar] [CrossRef]
  143. Zhao, W.; Song, M.; Zhang, J.; Kuerban, M.; Wang, H. Combined Identification of Long Non-Coding RNA CCAT1 and HOTAIR in Serum as an Effective Screening for Colorectal Carcinoma. Int. J. Clin. Exp. Pathol. 2015, 8, 14131–14140. [Google Scholar]
  144. Liu, Y.; Chen, X.; Chen, X.; Liu, J.; Gu, H.; Fan, R.; Ge, H. Long Non-Coding RNA HOTAIR Knockdown Enhances Radiosensitivity through Regulating microRNA-93/ATG12 Axis in Colorectal Cancer. Cell Death Dis. 2020, 11, 175. [Google Scholar] [CrossRef]
  145. Hu, D.; Zhan, Y.; Zhu, K.; Bai, M.; Han, J.; Si, Y.; Zhang, H.; Kong, D. Plasma Exosomal Long Non-Coding RNAs Serve as Biomarkers for Early Detection of Colorectal Cancer. Cell. Physiol. Biochem. 2018, 51, 2704–2715. [Google Scholar] [CrossRef]
  146. Zhu, M.; Luo, Y.; Xu, A.; Xu, X.; Zhong, M.; Ran, Z. Long Noncoding RNA TCONS_00026334 Is Involved in Suppressing the Progression of Colorectal Cancer by Regulating miR-548n/TP53INP1 Signaling Pathway. Cancer Med. 2020, 9, 8639–8649. [Google Scholar] [CrossRef]
  147. Xiao, W.; Li, J.; Hu, J.; Wang, L.; Huang, J.-R.; Sethi, G.; Ma, Z. Circular RNAs in Cell Cycle Regulation: Mechanisms to Clinical Significance. Cell Prolif. 2021, 54, e13143. [Google Scholar] [CrossRef]
  148. Ameli-Mojarad, M.; Ameli-Mojarad, M.; Hadizadeh, M.; Young, C.; Babini, H.; Nazemalhosseini-Mojarad, E.; Bonab, M.A. The Effective Function of Circular RNA in Colorectal Cancer. Cancer Cell Int. 2021, 21, 496. [Google Scholar] [CrossRef]
  149. Memczak, S.; Jens, M.; Elefsinioti, A.; Torti, F.; Krueger, J.; Rybak, A.; Maier, L.; Mackowiak, S.D.; Gregersen, L.H.; Munschauer, M.; et al. Circular RNAs Are a Large Class of Animal RNAs with Regulatory Potency. Nature 2013, 495, 333–338. [Google Scholar] [CrossRef]
  150. Ren, S.; Lin, P.; Wang, J.; Yu, H.; Lv, T.; Sun, L.; Du, G. Circular RNAs: Promising Molecular Biomarkers of Human Aging-Related Diseases via Functioning as an miRNA Sponge. Mol. Ther. Methods Clin. Dev. 2020, 18, 215–229. [Google Scholar] [CrossRef]
  151. Liu, K.; Mou, Y.; Shi, X.; Liu, T.; Chen, Z.; Zuo, X. Circular RNA 100146 Promotes Colorectal Cancer Progression by the MicroRNA 149/HMGA2 Axis. Mol. Cell. Biol. 2021, 41, e00445-20. [Google Scholar] [CrossRef]
  152. Li, S.; Yan, G.; Liu, W.; Li, C.; Wang, X. Circ0106714 Inhibits Tumorigenesis of Colorectal Cancer by Sponging miR-942-5p and Releasing DLG2 via Hippo-YAP Signaling. Mol. Carcinog. 2020, 59, 1323–1342. [Google Scholar] [CrossRef] [PubMed]
  153. Xu, X.-W.; Zheng, B.-A.; Hu, Z.-M.; Qian, Z.-Y.; Huang, C.-J.; Liu, X.-Q.; Wu, W.-D. Circular RNA hsa_circ_000984 Promotes Colon Cancer Growth and Metastasis by Sponging miR-106b. Oncotarget 2017, 8, 91674–91683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Yu, C.; Li, D.; Yan, Q.; Wang, Y.; Yang, X.; Zhang, S.; Zhang, Y.; Zhang, Z. Circ_0005927 Inhibits the Progression of Colorectal Cancer by Regulating miR-942-5p/BATF2 Axis. Cancer Manag. Res. 2021, 13, 2295–2306. [Google Scholar] [CrossRef] [PubMed]
  155. Shang, A.; Gu, C.; Wang, W.; Wang, X.; Sun, J.; Zeng, B.; Chen, C.; Chang, W.; Ping, Y.; Ji, P.; et al. Exosomal circPACRGL Promotes Progression of Colorectal Cancer via the miR-142-3p/miR-506-3p- TGF-β1 Axis. Mol. Cancer 2020, 19, 117. [Google Scholar] [CrossRef]
  156. Geng, Y.; Zheng, X.; Hu, W.; Wang, Q.; Xu, Y.; He, W.; Wu, C.; Zhu, D.; Wu, C.; Jiang, J. Hsa_circ_0009361 Acts as the Sponge of miR-582 to Suppress Colorectal Cancer Progression by Regulating APC2 Expression. Clin. Sci. 2019, 133, 1197–1213. [Google Scholar] [CrossRef]
  157. Hsiao, K.-Y.; Lin, Y.-C.; Gupta, S.K.; Chang, N.; Yen, L.; Sun, H.S.; Tsai, S.-J. Noncoding Effects of Circular RNA CCDC66 Promote Colon Cancer Growth and Metastasis. Cancer Res. 2017, 77, 2339–2350. [Google Scholar] [CrossRef] [Green Version]
  158. Lu, H.; Yao, B.; Wen, X.; Jia, B. FBXW7 Circular RNA Regulates Proliferation, Migration and Invasion of Colorectal Carcinoma through NEK2, mTOR, and PTEN Signaling Pathways in Vitro and in Vivo. BMC Cancer 2019, 19, 918. [Google Scholar] [CrossRef] [Green Version]
  159. Ren, C.; Zhang, Z.; Wang, S.; Zhu, W.; Zheng, P.; Wang, W. Circular RNA hsa_circ_0001178 Facilitates the Invasion and Metastasis of Colorectal Cancer through Upregulating ZEB1 via Sponging Multiple miRNAs. Biol. Chem. 2020, 401, 487–496. [Google Scholar] [CrossRef]
  160. Ren, T.-J.; Liu, C.; Hou, J.-F.; Shan, F.-X. CircDDX17 Reduces 5-Fluorouracil Resistance and Hinders Tumorigenesis in Colorectal Cancer by Regulating miR-31-5p/KANK1 Axis. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 1743–1754. [Google Scholar]
  161. Wang, X.; Zhang, H.; Yang, H.; Bai, M.; Ning, T.; Deng, T.; Liu, R.; Fan, Q.; Zhu, K.; Li, J.; et al. Exosome-Delivered circRNA Promotes Glycolysis to Induce Chemoresistance through the miR-122-PKM2 Axis in Colorectal Cancer. Mol. Oncol. 2020, 14, 539–555. [Google Scholar] [CrossRef]
  162. Jian, X.; He, H.; Zhu, J.; Zhang, Q.; Zheng, Z.; Liang, X.; Chen, L.; Yang, M.; Peng, K.; Zhang, Z.; et al. Hsa_circ_001680 Affects the Proliferation and Migration of CRC and Mediates Its Chemoresistance by Regulating BMI1 through miR-340. Mol. Cancer 2020, 19, 20. [Google Scholar] [CrossRef]
  163. Xu, Y.; Qiu, A.; Peng, F.; Tan, X.; Wang, J.; Gong, X. Exosomal Transfer of Circular RNA FBXW7 Ameliorates the Chemoresistance to Oxaliplatin in Colorectal Cancer by Sponging miR-18b-5p. Neoplasma 2021, 68, 108–118. [Google Scholar] [CrossRef]
  164. Xi, L.; Liu, Q.; Zhang, W.; Luo, L.; Song, J.; Liu, R.; Wei, S.; Wang, Y. Circular RNA circCSPP1 Knockdown Attenuates Doxorubicin Resistance and Suppresses Tumor Progression of Colorectal Cancer via miR-944/FZD7 Axis. Cancer Cell Int. 2021, 21, 153. [Google Scholar] [CrossRef]
  165. Zhao, K.; Cheng, X.; Ye, Z.; Li, Y.; Peng, W.; Wu, Y.; Xing, C. Exosome-Mediated Transfer of circ_0000338 Enhances 5-Fluorouracil Resistance in Colorectal Cancer through Regulating MicroRNA 217 (miR-217) and miR-485-3p. Mol. Cell. Biol. 2021, 41, e00517-20. [Google Scholar] [CrossRef]
  166. Hon, K.W.; Ab-Mutalib, N.S.; Abdullah, N.M.A.; Jamal, R.; Abu, N. Extracellular Vesicle-Derived Circular RNAs Confers Chemoresistance in Colorectal Cancer. Sci. Rep. 2019, 9, 16497. [Google Scholar] [CrossRef] [Green Version]
  167. Yu, C.-Y.; Kuo, H.-C. The Emerging Roles and Functions of Circular RNAs and Their Generation. J. Biomed. Sci. 2019, 26, 29. [Google Scholar] [CrossRef]
  168. Liang, Y.; Shi, J.; He, Q.; Sun, G.; Gao, L.; Ye, J.; Tang, X.; Qu, H. Hsa_circ_0026416 Promotes Proliferation and Migration in Colorectal Cancer via miR-346/NFIB Axis. Cancer Cell Int. 2020, 20, 494. [Google Scholar] [CrossRef]
  169. Mohammadi, D.; Zafari, Y.; Estaki, Z.; Mehrabi, M.; Moghbelinejad, S. Evaluation of Plasma circ_0006282 as a Novel Diagnostic Biomarker in Colorectal Cancer. J. Clin. Lab. Anal. 2022, 36, e24147. [Google Scholar] [CrossRef]
  170. Li, J.; Song, Y.; Wang, J.; Huang, J. Plasma Circular RNA Panel Acts as a Novel Diagnostic Biomarker for Colorectal Cancer Detection. Am. J. Transl. Res. 2020, 12, 7395–7403. [Google Scholar]
  171. Ye, D.-X.; Wang, S.-S.; Huang, Y.; Chi, P. A 3-Circular RNA Signature as a Noninvasive Biomarker for Diagnosis of Colorectal Cancer. Cancer Cell Int. 2019, 19, 276. [Google Scholar] [CrossRef]
  172. Pan, B.; Qin, J.; Liu, X.; He, B.; Wang, X.; Pan, Y.; Sun, H.; Xu, T.; Xu, M.; Chen, X.; et al. Identification of Serum Exosomal Hsa-Circ-0004771 as a Novel Diagnostic Biomarker of Colorectal Cancer. Front. Genet. 2019, 10, 1096. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Zhang, J.; Cai, A.; Zhao, Y. Three CircRNAs Function as Potential Biomarkers for Colorectal Cancer. Clin. Lab. 2020, 66. [Google Scholar] [CrossRef] [PubMed]
  174. Sobhani, I.; Tap, J.; Roudot-Thoraval, F.; Roperch, J.P.; Letulle, S.; Langella, P.; Corthier, G.; Tran Van Nhieu, J.; Furet, J.P. Microbial Dysbiosis in Colorectal Cancer (CRC) Patients. PLoS ONE 2011, 6, e16393. [Google Scholar] [CrossRef] [PubMed]
  175. Rezasoltani, S.; Asadzadeh-Aghdaei, H.; Nazemalhosseini-Mojarad, E.; Dabiri, H.; Ghanbari, R.; Zali, M.R. Gut Microbiota, Epigenetic Modification and Colorectal Cancer. Iran. J. Microbiol. 2017, 9, 55–63. [Google Scholar] [PubMed]
  176. Marchesi, J.R.; Dutilh, B.E.; Hall, N.; Peters, W.H.M.; Roelofs, R.; Boleij, A.; Tjalsma, H. Towards the Human Colorectal Cancer Microbiome. PLoS ONE 2011, 6, e20447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Xiao, Q.; Lu, W.; Kong, X.; Shao, Y.W.; Hu, Y.; Wang, A.; Bao, H.; Cao, R.; Liu, K.; Wang, X.; et al. Alterations of Circulating Bacterial DNA in Colorectal Cancer and Adenoma: A Proof-of-Concept Study. Cancer Lett. 2021, 499, 201–208. [Google Scholar] [CrossRef]
Figure 1. Origin of extracellular nucleic acids in circulation. Tumor cells release extracellular cfNAs through a combination of (i) cell death such as apoptosis, necrosis, and netosis or (ii) active secretion. Circulating bacterial DNA (cbDNA) may also be detected in blood samples from cancer patients. The loss of membrane integrity in necrosis results in releasing intracellular contents, including fragments of DNA, into the circulation. In netosis, activated neutrophils release neutrophil extracellular traps, webs of chromatin, and mitochondrial DNA (mtDNA). cfDNA may exist in the peripheral blood as either free or linked to proteins in the form of nucleosomal DNA, or associated with extracellular membrane vesicles (EMVs) such as exosomes or microvesicles secreted by cells. Methylated DNA and various types of RNA are present as well (created with Biorender.com, accessed on 21 July 2022).
Figure 1. Origin of extracellular nucleic acids in circulation. Tumor cells release extracellular cfNAs through a combination of (i) cell death such as apoptosis, necrosis, and netosis or (ii) active secretion. Circulating bacterial DNA (cbDNA) may also be detected in blood samples from cancer patients. The loss of membrane integrity in necrosis results in releasing intracellular contents, including fragments of DNA, into the circulation. In netosis, activated neutrophils release neutrophil extracellular traps, webs of chromatin, and mitochondrial DNA (mtDNA). cfDNA may exist in the peripheral blood as either free or linked to proteins in the form of nucleosomal DNA, or associated with extracellular membrane vesicles (EMVs) such as exosomes or microvesicles secreted by cells. Methylated DNA and various types of RNA are present as well (created with Biorender.com, accessed on 21 July 2022).
Cancers 14 03712 g001
Table 1. Classification and genetic causes of hereditary CRC syndromes.
Table 1. Classification and genetic causes of hereditary CRC syndromes.
SyndromeGene(s)InheritanceRef.
Lynch Syndrome (LS)Heterozygous mutations in MMR genes MLH1, MSH2 (or EPCAM deletions), MSH6 and PMS2AD[21]
Familial Colorectal Cancer Type X (FCCTX)BRCA2, SEMA4, NTS, RASSF9, GALNT12, KRAS, BRAF, APC, BMPR1A, and RPS20-[22,23]
Turcot Syndrome (TS)MMR genes (MLH1 and PMS2) or APC AD/AR[24]
Familial Adenomatous Polyposis (FAP)/APC-Associated PolyposisAPCAD[25]
MUTYH-Associated Polyposis (MAP)MUTYHAR[26]
Polymerase Proofreading-Associated Polyposis (PPAP)POLD1/POLEAD[27]
NTHL1-Tumor SyndromeNTHL1AR[28]
Constitutional MMR Deficiency Syndrome (CMMRD) Biallelic mutations in MMR genes MLH1, MSH2, MSH6 and PMS2AR[29]
Peutz–Jeghers Syndrome (PJS)STK11/LKB1AD[30]
PTEN Hamartoma Tumor Syndrome (PHTS)PTENAD [31]
Juvenile Polyposis Syndrome (JPS)SMAD4/BMPR1AAD[32]
Hereditary Mixed Polyposis Syndrome (HMPS)GREM1AD[33,34]
RNF43-associated Serrated PolyposisRNF43AD[35,36]
Ref.—reference; AD—autosomal dominant; AR—autosomal recessive; MMR—mismatch repair.
Table 2. List of cfDNA/ctDNA biomarkers in mutation analysis and CRC management.
Table 2. List of cfDNA/ctDNA biomarkers in mutation analysis and CRC management.
Study SourceBiomarker/FunctionTechniqueAccuracy
Zhitnyuk et al. [66]ATAC-seq datasetcfDNA fragment end profiles/reveal the presence of early-stage colorectal and renal cancersAnchored multiplex PCR followed by MPSAUC = 0.94
Cristiano et al. [67]Plasmagenome-wide fragmentation features/detection of seven cancer types *Low coverage WGS AUC = 0.94
Mouliere et al. [68]Plasmafragment length and copy number analysis/distinguish cancer from healthy individualsLow coverage WGS (0.4×)AUC > 0.99
Flamini et al. [69]Serumcarriers of certain “tumorigenic” propertiesqPCRAUC = 0.86
Kidess et al. [70]Plasma and tissueBRAF, EGFR, KRAS, PIK3CASCODA followed by MPSNA
Vidal et al. [71]PlasmaRAS/diagnosis and anti-EGFR treatment monitoring of mCRCOncoBEAM RAS ctDNA assayNA
Nakamura et al. [72]PlasmaHER2/monitor anti-HER2 therapy response in mCRCGuardant360AUC = 0.53
NA—not available; AUC—area under the ROC curve; MPS—massively parallel sequencing; WGS—whole genome sequencing; * breast, colorectal, lung, ovarian, pancreatic, gastric, and bile duct cancer.
Table 4. List of cf-mtDNA copy number changes in CRC.
Table 4. List of cf-mtDNA copy number changes in CRC.
StudySourceExp.FunctionTechnique
Haupts et al. [98]Plasmapotential diagnostic biomarker for CRC screeningMPS
Meddeb et al. [99]Plasmadiagnostic and prognostic biomarker in metastatic CRC patientsqRT-PCR
Xu et al. [100]Plasma/tissuebiomarker of early CRC, prediction of tumor response and progressionddPCR, MPS
Zhou et al. [101]Urinemonitoring of aberrant fragmentation and mutation profiles MPS
Exp.—expression; ↑—upregulated; MPS—massively parallel sequencing; ddPCR—droplet digital PCR.
Table 5. List of cf-miRNAs and their role in CRC. Several miRNAs have been present in EMVs that may be extracted from a serum/plasma source.
Table 5. List of cf-miRNAs and their role in CRC. Several miRNAs have been present in EMVs that may be extracted from a serum/plasma source.
cf-miRNAsSourceExp.TargetsBiomarker/FunctionRef.
miR-1290Plasmaepithelial-mesenchymal transition (EMT) markersprognostic/poor overall survival, advanced TNM stage[116]
miR-21 miR-145 miR-203 miR-155 miR-210 miR-31
miR-345
Plasmavarious downstream targets (e.g., PTEN, PDCD4 genes; WNT/β-Catenin signaling pathway, etc.)diagnostic/differentiation of surgery- naïve CRC patients, diagnosis of liver metastases[117]
miR-92aPlasma/
EMVs
signaling pathways-BMPs/SMAD; WNT/β-Catenin; PTEN/AKT/FoxO; genes-DKK3, KLF4, SMAD7diagnostic/distinguishing advanced neoplasia CRC patients, early CRC screening[118,119]
miR-92bPlasma/
EMVs
NAdiagnostic/early CRC detection[120]
miR-17-5p
mmiR-92a-3p
Serum/
EMVs
NAprognostic/primary and mCRC diagnosis, correlation with stages and grades of CRC[121]
miR-150-5pSerum/
EMVs
ZEB1diagnostic and prognostic/poor differentiation, positive lymph node metastasis, TNM stage[122]
miR-122Serum/
EMVs
PKM2prognostic/differentiation of CRC patients with liver metastasis[123]
miR-1290Serumvarious tumor suppressors (e.g., Forkhead box protein-A1, N-acetyltransferase etc.)diagnostic/early CRC detection, recurrence monitoring, tumor aggressivity[124]
miR-30e-3p,
mmiR-146a-5p/
mmiR-148a-3p
Serum↑|↓miR-146a-5p via carboxypeptidase M/src-FAKdiagnostic[125]
miR-1247-5p miR-1293 miR-548at-5p
miR-107 miR-139-3p
Serumvarious downstream targetsdiagnostic/detection of precancerous polyps and early CRC stages[126]
miR-19a miR-20a miR-150 let-7a | miR-143 miR-145Serum↑|↓various downstream targetsdiagnostic, prognostic/CRC screening, TNM staging and LNM status determination[127]
miR-21Plasma/salivaMAPK, WNT/β-Catenin signaling/PTEN, PDCD, DKK2diagnostic and prognostic/CRC screening[128]
miR-186-5p miR-29a-3p miR-29c-3p miR-766-3p miR-491-5pSalivavarious downstream targetsdiagnostic and prognostic/distinguishing CRC from healthy controls, predicting disease outcome in advanced stages[129]
miR-129-1-3p
mmiR-566
UrineNAdiagnostic/early CRC detection[130]
Exp.—expression; Ref.—references; ↑—upregulated; ↓—downregulated; NA—not available; EMVs—extracellular membrane vesicles.
Table 6. List of cf-lncRNAs and their role in CRC. Several lncRNAs are present in EMVs that may be extracted from serum/plasma samples.
Table 6. List of cf-lncRNAs and their role in CRC. Several lncRNAs are present in EMVs that may be extracted from serum/plasma samples.
cf-lncRNAsSourceExp.Biomarker/FunctionRef.
NEAT1 variant 1/variant 2Whole blooddiagnostic[135]
BLACAT1Serumdiagnostic/distinguishing CRC patients, non-cancer patients and healthy individuals[136]
CCAT2, HULCSerumdiagnostic/screening of CRC or adenomatous polyps[137]
CCAT2Serum/EMVsdiagnostic[138]
CRNDE-hSerum/EMVsprognostic and diagnostic/low overall survival of CRC patients[139]
UCA1Serum/EMVspredictive/resistance to cetuximab[140]
FOXD2-AS1
NRIR
XLOC_0009459
EMVsdiagnostic/early-stage CRC diagnosis[141]
ATB, CCAT1Plasmadiagnostic[142]
CCAT1, HOTAIR|p21Plasma↑|↓prognostic[143]
HOTAIRPlasmaprognostic/increase radiosensitivity via miRNA-93/ATG12 axis[144]
LNCV6_116109/98390/38772/108266/84003/98602Plasma/EMVsdiagnostic/early-stage CRC diagnosis[145]
TCONS_00026334NAtumor suppressor/suppress CRC progression via miR-548n/TP53ONP1 axis[146]
Exp.—expression; Ref.—references; ↑—upregulated; ↓—downregulated; EMVs—extracellular membrane vesicles.
Table 7. List of circRNAs relevant for CRC progression and clinical/biological features (not all have been studied in liquid biopsies).
Table 7. List of circRNAs relevant for CRC progression and clinical/biological features (not all have been studied in liquid biopsies).
circRNATarget Molecules/GenesFunctionRef.
hsa-circ-000984miR-106b/↑ of CDK6proliferation, metastasis[153]
hsa-circ-0005927miR-942-5p/↓ of BATF2cell colony-forming ability, apoptosis, migration [154]
circPACRGLmiR-142-3p, miR-506-3p/↑ of TGF-β1 proliferation, metastasis (migration and invasion) [155]
hsa-circ-0009361miR-582-3p/↓ of APC2/WNT/β-catenin signaling pathsuppress cell growth and metastasis[156]
circCCDC66various oncogenesproliferation, migration, invasion[157]
circ-FBXW7NEK2, mTOR, PTENproliferation, migration, invasion[158]
hsa-circ-0001178miR-382/587/616/↑ of ZEB1metastasis, invasion[159]
hsa-circ-DDX17miR-31-5p/↓ of KANK1promotes sensitivity to 5-FU[160]
ciRS-122miR-122/↑ of PKM2promotes resistance to oxaliplatin[161]
hsa-circ-001680miR-340/↑ of BMI1promotes chemoresistance to irinotecan[162]
circ-FBXW7miR-18b-5p ameliorates chemoresistance to oxaliplatin[163]
circ-CSPP1miR-944/↓ of FZD7enhanced doxorubicin sensitivity[164]
circ-0000338miR-217, miR-485-3penhanced 5-FU resistance[165]
Ref.—references; ↑—upregulated; ↓—downregulated.
Table 8. List of circRNAs described as biomarkers for CRC diagnosis and screening (from liquid biopsies).
Table 8. List of circRNAs described as biomarkers for CRC diagnosis and screening (from liquid biopsies).
circRNASourceExp.Biomarker/FunctionRef.
hsa-circ-0006282Plasmadiagnostic; improving the detection and monitoring of CRCs in combination with carcinoembryonic antigen (CEA) and carbohydrate antigen199 (CA199)[169]
hsa-circ-0001900
hsa-circ-0001178
hsa-circ-0005927
Plasmadiagnostic/improving the detection of CAE-negative CRC[170]
hsa-circ-0082182
hsa-circ-0000370|
hsa-circ-0035455
Plasma↑|↓diagnostic[171]
hsa-circ-0004771Serum/
EMVs
diagnostic/differentiation of benign intestinal diseases, stage I/II CRCs, and CRCs from healthy individuals[172]
circ-FMN2
circ-LMNB1
circ-ZNF609
Serumdiagnostic and prognostic/correlation with histopathological grade, lymph node metastasis, TNM stages[173]
Ref.—references; ↑—upregulated; ↓—downregulated; EMVs—extracellular membrane vesicles.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Styk, J.; Buglyó, G.; Pös, O.; Csók, Á.; Soltész, B.; Lukasz, P.; Repiská, V.; Nagy, B.; Szemes, T. Extracellular Nucleic Acids in the Diagnosis and Progression of Colorectal Cancer. Cancers 2022, 14, 3712. https://doi.org/10.3390/cancers14153712

AMA Style

Styk J, Buglyó G, Pös O, Csók Á, Soltész B, Lukasz P, Repiská V, Nagy B, Szemes T. Extracellular Nucleic Acids in the Diagnosis and Progression of Colorectal Cancer. Cancers. 2022; 14(15):3712. https://doi.org/10.3390/cancers14153712

Chicago/Turabian Style

Styk, Jakub, Gergely Buglyó, Ondrej Pös, Ádám Csók, Beáta Soltész, Peter Lukasz, Vanda Repiská, Bálint Nagy, and Tomáš Szemes. 2022. "Extracellular Nucleic Acids in the Diagnosis and Progression of Colorectal Cancer" Cancers 14, no. 15: 3712. https://doi.org/10.3390/cancers14153712

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop