Next Article in Journal
Current and Future Trends on Diagnosis and Prognosis of Glioblastoma: From Molecular Biology to Proteomics
Previous Article in Journal
Importance of the Androgen Receptor Signaling in Gene Transactivation and Transrepression for Pubertal Maturation of the Testis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Prospect and Challenges to the Flow of Liquid Biopsy in Africa

by
Dada Oluwaseyi Temilola
1,2,
Martha Wium
1,
Tangbadioa Herve Coulidiati
1,3,
Henry Ademola Adeola
4,
Giuseppina Maria Carbone
5,
Carlo Vittorio Catapano
5 and
Luiz Fernando Zerbini
1,*
1
International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town 7925, South Africa
2
Integrative Biomedical Sciences Division, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
3
Training and Research unit in Sciences and Technology, University Norbert Zongo, P.O. Box 376, Koudougou 376, Burkina Faso
4
Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town 7925, South Africa
5
Institute of Oncology Research, Università della Svizzera Italiana, Via Vincenzo Vela 6, CH-6500 Bellinzona, Switzerland
*
Author to whom correspondence should be addressed.
Cells 2019, 8(8), 862; https://doi.org/10.3390/cells8080862
Submission received: 16 July 2019 / Revised: 30 July 2019 / Accepted: 3 August 2019 / Published: 9 August 2019

Abstract

:
Liquid biopsy technologies have the potential to transform cancer patient management as it offers non-invasive diagnosis and real-time monitoring of disease progression and treatment responses. The use of liquid biopsy for non-invasive cancer diagnosis can have pivotal importance for the African continent where access to medical infrastructures is limited, as it eliminates the need for surgical biopsies. To apply liquid biopsy technologies in the African setting, the influence of environmental and population genetic factors must be known. In this review, we discuss the use of circulating tumor cells, cell-free nucleic acids, extracellular vesicles, protein, and other biomolecules in liquid biopsy technology for cancer management with special focus on African studies. We discussed the prospect, barriers, and other aspects that pose challenges to the use of liquid biopsy in the African continent.

1. Introduction

Cancer is a growing public health threat globally. GLOBOCAN 2018 data showed an overall increase in cancer cases worldwide with 18.1 million new cases and 9.6 million cancer deaths in 2018 [1]. Africa and Asia were showed to have a higher proportion of cancer mortality in relation to the proportion of incident cases when compared with other regions of the world [1].
The incidence and mortality rate of cancer differ across regions and between sexes. Globally, lung cancer had the highest incidence among males in 2018, with prostate cancer having the highest mortality burden among African men. Breast cancer still has the highest incidence and mortality burden among women worldwide [1]. The incidence and mortality rate of breast cancer have remained relatively unchanged over the years in many developed countries. In many parts of Africa, Asia, and South America the incidence of breast cancer is, however increasing rapidly, with Africa having the highest age-standardized mortality rate globally [1,2,3,4].
The rising burden of cancer in Africa has been attributed to factors such as inadequate health care facilities, poor access to quality and affordable health care, as well as inadequate infrastructure to support African-based research [5]. Furthermore, most cancers are diagnosed late in Africa which in turn worsen the prognosis [6,7].
Tissue biopsy, the established method of cancer diagnosis, is invasive and can be accompanied by various surgical complications. Tissue biopsy reflects a small section of the tissue and may miss important diagnostic details. It may be inadequate for a complete genomic profile of a patient’s tumors because regions within and between primary and metastatic tumors can have different genomic mutations [8]. In a liquid biopsy, cancer is diagnosed or monitored by analyzing body fluids such as blood, urine, or saliva [9]. Liquid biopsy is based on detecting tumor cells or tumor-derived molecules (DNA, RNA, exosomes, and protein) that were released from tumors into circulation (Figure 1). Improved diagnosis, early detection, and better monitoring of disease progression and treatment response are imperative in Africa due to the overall rising burden of cancer throughout the continent. Invasive diagnostic procedures are a barrier to overcome due to surgical risk, costs, limited access, and poor compliance by the population. Therefore, development and implementation of non-invasive liquid biopsy methodologies for cancer management are a top priority for the next decades for basic and clinical scientists in Africa. In addition to being used in cancer management, liquid biopsy tests are also clinically used to detect fetal chromosomal abnormalities during pregnancies and monitor organ transplants [10].
There is presently an increasing number of studies on circulating tumor molecules in diagnosis and prognosis of cancers. Studies on the role of circulating molecules in cancer diagnosis started globally in the late 1990s [11,12,13,14] but African-based studies started only in late 2000 (Table 1). The majority of African-based studies were done in Egypt, with a few other studies from Tunisia, South Africa, Gambia, Cameroon, and Senegal (Table 1). Importantly, the causes of cancer differ in different populations. Distinct pathogens, carcinogens, dietary habits, social conditions, and genetic background may influence tumorigenesis depending on population and geographical settings. The genetic and epigenetic variation from population to population may lead to ample variations in natural history and clinical outcome across different populations. For example, some cancers, such as prostate cancers, are more aggressive in the African population [15]. Also, more cancers in Africa and Asia are related to infective pathogens than in other continents. This requires that more African-based studies are done to validate the applicability of circulating biomarkers and liquid biopsy technologies in diagnosis and treatment of cancer in Africa. Host genetics, tumor genetics, and epigenetic variations need to be explored and taken into account to identify population-specific cancer biomarkers in liquid biopsy adapted and optimized for diagnostic use in African countries. The process of optimization of these cutting-edge technologies should also imperatively aim at reducing costs and increasing affordable access throughout the African continent.
There are three main types of circulating molecules investigated as tumor biomarkers through liquid biopsy procedures: Circulating tumor cells (CTC), tumor-released nucleic acids like DNA and RNA, and small extracellular vesicles or exosomes, Table 2. Here we aim at reviewing studies on the role of circulating tumor molecules in the diagnosis and treatment of cancer, with a particular focus in the African continent. This review will also discuss the prospect and challenges associated with the use of circulating tumor molecules in liquid biopsy for diagnosis and treatment of cancer in Africa.

2. Component of Liquid Biopsy

2.1. Circulating Tumor Cells

Cancer deaths are mostly due to tumor metastasis [77]. Tumor cells dislodge from the primary tumor and use the blood and lymph system as highways to travel to other parts of the body, where they invade and form metastatic tumors. Our understanding of tumor cell migration is far from complete. It is associated with epithelial-to-mesenchymal transition (EMT) and governed by numerous genes and proteins in which genetic aberrations can accelerate or decelerate cancer progression. The potential of these circulating tumor cells (CTCs) in cancer diagnosis and treatment is twofold. Firstly, it can be used as a diagnostic marker to predict disease progression and survival in metastatic cancer [78,79,80], indicating treatment failure [81,82], distinguish benign from malignant growth [83], and in early-stage cancer diagnosis [84,85]. Secondly, with improvements in sequencing technologies, it also provides a window into the genetic landscape of diseases which could be used to stratify patient treatment [86].
Several methods are available for the detection of CTCs in the blood (reviewed in [87]). The only US Food and Drug Administration (FDA) approved method to quantify CTCs is the CellSearch system (Verdex LLC, San Diego, CA, USA). This method has been approved to monitor treatment effectiveness in metastatic breast, prostate, and colorectal cancer patients. The CellSearch system is sensitive, robust, and can detect a single CTCs in 7.5 mL of blood. CTCs are stable for about 96 h at room temperature when blood is collected in CellSave preservative tubes [88,89], making shipment at room temperature from remote locations viable. A limitation of this method is that the enrichment step captures cells expressing the epithelial cell adhesion molecule (EpCAM) on their surface and not all CTCs express EpCAM. Some CTCs exhibit stem cell-like or normal-like characteristics with no EpCAM surface protein while others undergo EMT transition with reduce or loss of EpCAM surface protein [90,91,92]. These CTCs will be undetectable by the CellSearch technology. Another concern is false positive results, circulating cells that are EpCAM-, CK8-, CK18-, and CK19 -positive have been detected in patients with benign inflammatory colon diseases, based on nuclear morphology these cells were consistent with benign gland [93]. It should be noted that no CTCs were detected in healthy control patients. In blood spiking experiments with carcinoma cells that have high levels of EpCAM, the sensitivity to detect CTCs is higher than 85% [94,95], but with cells that express low levels of EpCAM, the sensitivity is about 42% [96].
Using the CellSearch method, 5 or more CTCs for metastatic breast and prostate cancer and 3 or more in metastatic colorectal cancer were associated with shorter progression-free and overall survival [78,79,80]. CTC count is a more reproducible predictor of overall survival than traditional methods such as radiology [97] or prostate specific antigen (PSA) in prostate cancer [98].
Cancer treatment decisions are complicated by the disparity in responses observed between patients. Discontinuing ineffective treatment earlier may decrease morbidity due to toxicity, allow alternative treatment, and reduce treatment cost. Cancer treatment leads to a reduction in CTC count [99]. Assessing the CTC count before and after treatment can predict the outcome of treatment in castration-resistant prostate cancer [81]. CTC count is superior to traditional radiology because it can detect treatment failure earlier and more accurately [97]. A phase II trial on advanced colorectal cancer has shown that CTC count can also be used in the stratification of treatment. Patients with high CTC count may benefit most from an intensive multidrug regimen, which is usually associated with high toxicity. This can be avoided in patients with a low CTC count [100].
CTC count has also been recently used to differentiate between benign and malignant tumors. Lung lesions on a PET/CT-scan (positron emission tomography/computed tomography scan) can be benign or malignant and a tissue biopsy is needed for diagnose. A recent study found that it is possible to distinguish benign lesions from lung cancer lesions using CTCs as a marker [83].
Studies report that CTCs are present in 10% to 55% of early-stage (stage I to III) breast cancer patients and are associated with poorer outcomes [101,102]. CTC count can potentially be used for early diagnosis of cancer in patients that have an increased risk for cancer [85]. Patients with chronic obstructive pulmonary disease (COPD) have an increased risk of lung cancer [103]. It is possible to detect CTCs in COPD patients 1 to 4 years before the tumor was detectable with a CT-scan [85].
Blood contains CTCs from all tumor sites in the body and it has been suggested that this is a molecular proxy for the overall disease [104], in contrast to a tissue biopsy which only represents cancer at a particular site. This may open the possibility to use CTCs to assess the current tumor biology in order to monitor genetic aberrations that may influence treatment choices and personalize cancer treatment. CTCs are difficult to isolate compared to circulating tumor DNA (ctDNA) but can provide information on the genome as well as the transcriptome and proteome of cancer. Recent advances in whole genome amplification (WGA) have made it possible to interrogate a single CTCs with microarray-based comparative genomic hybridization (array-CGH) and high-throughput [105] and single-cell sequencing (SCS) [86,106,107].
Using amplicon-based sequencing it was found that aberrations in CTCs correlate with that found in the primary tumor, including mutations and amplifications in druggable genes [108]. CTC RNA sequencing data show that in breast cancer regulation patterns differ based on the location of the metastatic tumors [109]. Gulbahce, et al. [86] showed that genomic sequencing of CTCs can be used to analyze tumor heterogeneity and the detected somatic alterations provide information that may be used in stratify treatment. It is also possible to culture CTCs ex vivo, enabling drug sensitivity testing [110]. Clinical trials utilizing genomic aberrations in CTCs or expanded CTC drug sensitivity testing to stratify patient treatment are needed and may shed some light on its feasibility in the clinical setting.
The CTC studies done in Africa are listed in Table 1. Bahnassy, et al. [19] showed that the melanoma antigen-encoding gene 1 (MAGE1) and MAGE3 are expressed on the surface of CTCs from hepatocellular carcinoma (HCC) patients but not in healthy volunteers or chronic hepatitis C patients. Sayed et al. [18] evaluated the uses of CTC and cancer stem cells (CSCs) during the treatment of breast cancer. They found that CTC count at diagnosis can predict overall survival and CTC count after chemotherapy can predict disease-free survival and overall survival. High levels of CD44+/CD24− CSCs that remain after treatment was an indicator for recurrence [18]. Increased levels of CD133, a marker for stem cells and cancer stem cells, were associated with higher stage tumors and poor prognosis in HCC patients [20].

2.2. Circulating Tumor DNA

Mandel [111] first identified cell-free DNA (cfDNA) in 1948 and about 3 decades later Leon, et al. [112] found an increased level of cfDNA in the circulation of cancer patients. The increased level of cfDNA cannot only distinguish cancer patients from healthy individuals [113] but can also differentiate patients with malignant tumors from those with benign tumors (prognostic ability) [114,115].
In addition to being found in the blood, cfDNA is also found in urine, cerebrospinal fluid, saliva, and breast milk [116]. cfDNA is highly fragmented, it measures between 150 and 200 bp with an average length of 167 bp [117,118]. Apoptosis and/or necrosis are considered to be the main sources of cfDNA, although the full mechanisms by which cfDNA is released into circulation is not completely understood [118,119]. DNA fragments released from tumor cells are referred to as circulating tumor DNA (ctDNA). ctDNA has a longer fragment size than DNA fragment released from normal cells [120,121]. The fragment size of cfDNA is used to calculate the DNA integrity index. DNA integrity index is the ratio of long to short DNA fragments [122]. DNA integrity index is used along with cfDNA levels to diagnose cancer, monitor treatment response, and predict tumor stages [123].
DNA fragment size between different tumor types also differs largely due to metabolic and biological differences among tumors [124]. For example, the DNA fragment size from brain cancers reflects the filtration effect of the blood–brain barrier [124].
The half-life of cfDNA is approximately 2 h after which it is cleared from the circulation [125]. This means that cfDNA analysis can provide a real-time view of the genetic landscape that includes all the tumors in a patient (primary and metastatic). Studies have shown that DNA fragments released from tumors into circulation harbor tumor-specific aberrations, including mutations in tumor suppressors and oncogenes, microsatellite instability, DNA methylation, loss of heterozygosity and copy number variation [126,127,128,129,130]. Next-generation sequencing (NGS) can be used to identify all known and unknown genomic aberrations but is time-consuming and expensive. PCR-based techniques, such as real-time PCR or droplet PCR, is less expensive and faster but can only be used to assess known aberrations [131].
Wyatt, et al. [132] showed that driver DNA mutations found in metastatic tissue biopsies were concurrently present in the cfDNA. They concluded that for most patients, analyzing genetic alterations in cfDNA is sufficient to identify driver DNA alterations for managing metastatic castration-resistant prostate cancer [132]. Genetic mutations detected in CTC and ctDNA from the same patient show high concordance (<73%), although complimentary assessment may be beneficial to assess the dynamic tumor profile [133].
The FDA has approved two tests based on cfDNA for cancer management, the first is the Cobas EGFR Mutation Test v2 (Roche Molecular Diagnostics, Basel, Switzerland). The Cobas EGFR Mutation Test is a real-time PCR test that can detect and quantify 42 mutations on the epidermal growth factor receptor (EGFR) gene [134]. This test was approved to guide treatment decisions in non-small cell lung cancer (NSCLC). When compared to tissue-derived DNA results, the sensitivity of this cfDNA test is 72.1% and the specificity is 97.9% [135]. The second is the Epi proColon test, a real-time PCR test that detects hypermethylation in the promoter region of the Septin 9 gene (SEPT9) [136]. This test is the first approved blood-based screening test for colorectal cancer. The sensitivity of the test is between 71.1% and 95.6% and the specificity is between 81.5% and 99% [137].
Additionally, the AmoyDx Super-ARMS EGFR mutation test has been approved by the Chinese FDA for the detection of EGFR mutations in lung cancer. Four cfDNA-based tests have been approved for the EU market. They are the Qiagen Therascreen EGFR RGQ Plasma PCR kit (for detection of EGFR del19 and EGFRL858R in lung cancer); Sysmex Inostics OncoBEAM RAS CRC Kit (for detection of KRAS and NRAS mutations in colorectal cancer); Idylla™ ctKRAS Mutation Test (for detecting KRAS mutations in metastatic colorectal cancer patients), and the Idylla™ ctNRAS-BRAF mutation test (for detecting NRAS and BRAF mutations in metastatic colorectal cancer patients) [138]. A number of studies have investigated cfDNA in cancer management in Africa (Table 1). Ibrahim, et al. [30] found that both qualitative (fragment size) and quantitative aspects of cfDNA are associated with prognosis, metastasis, and treatment responses of Egyptian breast cancer patient. Fawzy, et al. [29] studied the role of cfDNA and DNA integrity in patients with metastatic prostate cancer and found cfDNA to be a potential non-invasive biomarker for screening and monitoring metastasis in prostate cancer patients. A recent study by Marchio, et al. [26] used droplet digital PCR technique to detect TP53 R249S mutants in cfDNA of HCC patients from the Central African Republic and Cameroon. The study suggested that the technique may be used for diagnosis and to conduct public health surveys on populations at risk of HCC [26].

2.3. Circulating Tumor RNAs

For decades, the concept of extracellular RNA has been one of the major focuses of scientific research. One of the most important studies was reported by Stroun and co-workers in 1978, who demonstrated that RNAs are released from cells into the culture medium [139]. Following this study, other studies have demonstrated that free-circulating RNAs can be released in the bloodstream of healthy people or cancer patients within particles-associated vesicles such as exosomes, microvesicles, and apoptotic bodies [123,140]. These vesicles protect the free-circulating RNAs from ribonucleases degradation and confer their stability [141]. Circulating RNAs do not result from random degradation but regulated cleavage and may play a specific role in cell physiology and also in cell-to-cell communication [142,143]. Circulating RNAs are detectable in human body fluids such as plasma, serum, and urine and have also been implicated in some disease outcomes like cancers [123]. Sensitive techniques like droplet digital PCR and RNA sequencing have also been recently developed and are used for circulating RNA detection [144]. These characteristics make circulating RNAs a potential biomarker in cancer management. Circulating RNAs include coding RNAs and non-coding RNAs (long non-coding RNAs and microRNAs) will be discussed in the sections below.

2.3.1. Circulating Coding RNA

Circulating messenger RNAs (mRNAs) have been detected in body fluids of various cancer patients. These circulating mRNAs are associated with several cancer types such as breast, gastric, prostate, and colon cancers [145,146,147,148]. Many studies have shown the use of these circulating mRNAs as potential biomarkers in cancer patients for early detection and cancer progression monitoring [149,150].
In Africa, only a few studies have been reported. An Egyptian study investigated the role of transforming growth factor-beta 1 (TGF-β1) and Golgi protein 73 (GP73) circulating cell-free mRNAs as a potential biomarker for HCC [70]. The authors reported that TGF-β1 and GP73 mRNA expression was elevated in the serum of HCC patients compared to the control group. They also found that alpha-fetoprotein (AFP), which is usually measured for monitoring of patients with high HCC risk, showed a lower expression level than TGF-β1 and GP73. Abdelgawad, et al. [69] reported that the GPC3 level was elevated in the serum of all HCC patients compared to control subjects. The study also showed that measurement of the GPC3 level in serum of Egyptian patients with HCC is more sensitive than the currently used marker AFP.

2.3.2. Long Non-Coding RNAs

Long non-coding RNAs (lncRNAs) represent a class of nucleic acid transcripts that are longer than 200 nucleotides in length and are not coding for any protein [151]. They have the potential to regulate various biological events such as cell differentiation, proliferation, migration, and invasion [152]. They are implicated in tumorigenesis with oncogene or tumor suppressor roles [152]. lncRNA profiles are more organ- and tumor-specific than other RNA entities and it was reported that circulating lncRNAs could reflect the pathological and physiological change of cancer patients [153,154]. A study by Xie, et al. [155] showed that lncRNAs SOX2OT and ANRIL were upregulated in lung cancer patients in comparison to healthy controls and therefore postulated that they could be good circulating markers for non-small cell lung cancer prognosis [155]. Lv, et al. [156] also demonstrated that high level of lncRNA HOTAIR in the serum of breast cancer patients induces less response to neoadjuvant chemotherapy, hence highlighting HOTAIR as a biomarker for the monitoring of breast cancer treatment.
The only non-coding RNA clinically approved test is the PROGENSA® PCA3 urine test for prostate cancer screening in patients with one or more negative biopsies with a sensitivity and specificity of 48.4% and 78.6%, respectively [157]. This test detects the non-coding RNA prostate cancer antigen 3 (PCA3) in post-digital rectal exam first catch urine [158].
Only a few studies (Table 1) have investigated the role of lncRNAs as biomarkers for cancer diagnosis in Africa. Zidan, et al. [71] reported that MALAT1 (metastasis associated lung adenocarcinoma transcript 1) expression is increased in serum of Egyptian breast cancer patients. The study found MALAT1 to be more sensitive than CA15-3 which is the current marker for breast cancer [71]. Hashad, et al. [74] showed higher expression level of lncRNA H19 in the serum of gastric cancer patients when compared with the healthy control group and highlighted the possibility of using lncRNA H19 as a biomarker for gastric cancer diagnosis [74]. El-Tawdi, et al. [73] showed lncRNA-CTBP as a biomarker for HCC diagnosis. The study proposed the use of a panel of markers including lncRNA-CTBP, miR-16-2, miR-21-5p, and LAMP2 for best sensitivity and specificity of HCC diagnosis [73].

2.3.3. Circulating microRNAs

MicroRNAs (miRNAs) are endogenous non-coding RNA transcripts with a length around 18 to 24 nucleotides [159] which play an important role in cell physiology by modulating gene expression at post-transcriptional level. They recognize their target messenger RNA by binding completely or incompletely to the 3′-untranslated region (3′-UTR) and induce their effect either via translational repression or mRNA degradation [160]. MicroRNAs are actively released from cells into various human body fluids like plasma, serum, urine, and saliva and their expression level is correlated with disease progression and physiological states [159,161,162].
Circulating microRNAs are packed within vesicles or associated with RNA binding proteins and are stable in bio-fluids [163]. In extracellular vesicles about half of the RNA are microRNAs [164]. Previous studies have proposed circulating miRNAs as non-invasive biomarkers for human diseases like cancer, mainly due to their high stability in bio-fluids and their expression level that correlated with the disease stage [165,166]. By comparing the profile of miRNAs in healthy subjects and cancer patients, many studies have observed alterations of miRNAs expression in many human tumors with the potential role of determining cancer outcome [167,168,169].
In the context of Africa, Motawi, et al. [170] reported that circulating miR-21 and miR-221 can be used to discriminate between breast cancer patients and healthy subjects in Egyptian women. The study showed that the expression of the two circulating miRNAs is higher in breast cancer cases compared to healthy persons. A study by Fattah, et al. [171] confirmed circulating miR-21 as a potential diagnostic marker for breast cancer in the Egyptian population. Circulating miRNA has also been used as a diagnostic marker for HCC. Alnoanmany, et al. [62] demonstrated circulating miR-21 was highly expressed in Egyptian HCC patients compared to the control group. They found that its use as a diagnostic tool for HCC is more sensitive than AFP, the current diagnostic marker for HCC [62]. Elhamamsy, et al. [55] in their study on Egyptian acute myeloid leukemia (AML) patients, showed that the expression level of circulating miR-92a, miR-143, and miR-342 were downregulated compared to control individuals. The study demonstrated that these circulating miRNAs have sensitivity and specificity high enough to be good markers for AML [55]. Several circulating miRNAs were identified as biomarkers for colorectal carcinoma (CRC). Zekri, et al. [61] compared the miRNA profile between CRC patients and healthy controls and found that circulating miR-17, miR-19a, miR-20a, and miR-223 were up-regulated in CRC patients. The circulating miRNAs found by Zekri, et al. [61] were shown to demonstrate high diagnostic performance which may be useful biomarkers for the screening of CRC and monitoring tumor dynamics.

2.4. Other Circulating Molecules

2.4.1. Exosomes

The three main groups of extracellular vesicles released by cells are microvesicles, apoptotic bodies, and exosomes [172]. Exosomes are the best characterized and the most studied of these three types of extracellular vesicles [173]. Exosomes are small round vesicles with a size range of 40–150 nm in diameter [174,175]. Exosomes originate from endosomes and are actively produced and secreted by different types of cells including tumor cells [174,175]. Exosomes have been identified in blood, urine, cerebrospinal fluid, saliva, breast milk, pleural effusions, and nasal secretions [176,177,178].
Exosomes are employed by cells as vehicles to transmit molecular messages between homotypic and heterotypic cells and affect the phenotype of the recipient cells [175,179]. The exosome cargo includes a wide range of molecules such as proteins, lipids, mRNA, non-coding RNA, miRNA, and DNA. Plasma from cancer patients generally contains higher levels of exosomes compared to healthy control individuals, sustaining that tumor cells secrete more exosomes than normal cells [175]. Tumor-released exosomes exert their functions through cell–cell communications and impact malignant transformation, angiogenesis, immune response, and metastatic spread [180,181].
Accordingly, exosomes have a strong potential as blood or urine-based biomarkers for diagnostic prognostic and therapeutic management of cancer [175,179]. Recent studies highlight the relevance of tumor-secreted exosomes for cancer progression, response to therapy, and the possibility that different tumor types may secrete exosomes with unique cargos reflecting the tumor phenotype and clinical behavior. Furthermore, the exosome content in terms of DNA, RNA, and proteins gives insights into the intrinsic characteristic of tumors and can be exploited for personalized medicine approaches.
Despite their high potential, few studies have been done on the role of exosomes in the diagnosis and prognosis of cancer in Africa. An Egyptian study by Khalil, et al. [76] showed that exosomal lncRNA-RP11-510M2.10 can be used as a diagnostic and prognostic marker for lung cancer. Abd El Gwad, et al. [75] in their study showed that the accuracy of early diagnosis of HCC can be significantly improved using serum exosomal miR-1262, lncRNA-RP11-513I15.6, and AFP.

2.4.2. Circulating Proteins and Peptides

Recent discoveries on new protein and peptide biomarkers have marked a new horizon in their use in non-invasive cancer diagnosis [182,183]. Several protein markers such as CA19-9, AFP, CEA, CA15-3, PSA, CA125, are in use clinically to boost the diagnosis and monitoring of cancer. Not all of these protein markers have a high specificity to cancer and the need to discover novel biomarkers that are highly specific and sensitive remains.
Numerous studies have evaluated the use of protein biomarkers in African populations; with some aiming at the discovery of novel protein biomarkers [184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200]. A study by Adeola, et al. [15] identified 73 urinary proteins as potential biomarkers for prostate cancer in the South African population. Abdel Wahab et al. [184] identified 33 deregulated proteins in Egyptian cohort that could be used as a prognostic signature for hepatitis C-virus related HCC. A recent review by Adeola, et al. [201] highlights the prospects of using proteomic biomarkers in Africa.

3. Challenges to Implementation of Liquid Biopsy Technology in Africa

Even though liquid biopsy techniques have been used for the diagnosis of various cancer types in the advanced western world [201], its routine clinical use has not received wide coverage in African healthcare settings. This can be due to limiting factors such as lack of skilled personnel, poor health infrastructure, and poor government policies [201]. As a relatively new technology, a potential drawback of liquid biopsy in Africa is that it’s market products would not be affordable to the majority of low-income countries; and it may not be easily available for the general population. A typical example is Kenya which recently became the third African country after South Africa and Tunisia in which the use of liquid biopsy is commercially made available to the public [202]. The estimated cost for each test was considered to be seventy thousand Kenyan Shillings (about $7000) which is practically unaffordable to middle- and low-income earners that form the majority of the country’s population [202].
In addition, it may be difficult to ascertain the sensitivity, specificity, and efficacy of liquid biopsy methods in African patients, as most of the available products have not been tested in African populations. Also, most of the technology used for detecting genetic mutations and alterations are developed using non-African populations. This is particularly concerning because of the wide geographic/latitudinal variations in pathogenesis and natural history of cancers in Africa [203]. Therefore, ad hoc designed clinical studies in African populations are needed to address these questions and give insight on population-specific biomarkers for early diagnosis and monitoring.
Generally, parallel detection of various liquid biopsy analytes may be challenging from a small volume of blood [204]; and this integrated liquid biopsy investigation has to be significantly improved to provide in-depth information on tumor genotypes/phenotypes. The issue of low-frequency mutant allele in the analysis of cell-free nucleic acid is also a huge challenge due to variability among early-stage tumors [205]. Thus, adequate, cutting-edge, basic, translational, and clinical research in the African continent should be supported in this important and emerging field, which has enormous potential and social impact for improving public health at sustainable costs. For this technology to emerge in Africa, great capacity needs to be built in terms of specialized personnel training and infrastructure building, not only in the domain of molecular biology and clinical research but also in computational biology and bioinformatics, to standardize and validate potential biomarkers [204,206].

4. The Prospect for Liquid Biopsy in Africa

In spite of the challenges, liquid biopsy has emerged as a rapid, reliable, and minimally invasive cancer screening solution, with high specificity and sensitivity for cancer diagnosis and monitoring [205,206,207,208]. In western world settings, the use of liquid biopsy options like CTCs and ctDNA has been a cutting-edge technology that has improved the detection and monitoring of cancer in a small amount of blood samples [209,210,211,212,213]. It proved that real-time monitoring of disease progression can be used to personalize treatment, which can increase effectiveness and reduce the cost of treatment. Indeed, new technologies for detection of gene mutations, genetic rearrangements, and additional cancer-specific biomarkers applied to liquid biopsy promise further cost-cutting.
Cancer in Africa is a public health menace. Most cancer cases are diagnosed late in Africa [214]. For example, in Zimbabwe 80% of cervical cancer patients presented with advanced disease [215] and in Tanzania, more than 70% of breast cancer patient were diagnosed at stage III and above [216]. Factors such as limited knowledge of signs and symptoms of cancer, limited screening facilities, and fear of surgery are some of the major barriers to early presentation and cancer diagnosis in Africa [217,218,219]. Liquid biopsy could help in addressing these challenges as it is non-invasive and does not require surgical facilities not always available in African settings.
As shown in the developed world, liquid biopsy can be an implementable and promising concept that would non-invasively boost the prompt identification of cancers and reduce the morbidities and mortalities associated with the late/end-stage diagnosis of cancer. The initial burden and high costs due of the introduction of this high-tech, cutting-edge technology should be sustained by national and international collaborative research and health care programs aimed at reducing disparities both among and within African countries. Eventually, health care costs in African countries would be reduced by implementing policies and technologies leading to easier access to diagnostic procedures, early detection, and effective clinical predictive measures. Additionally, liquid biopsy approaches would promote the practice of evidence-based precision medicine in Africa, applying therapies when needed, and avoiding the human and societal costs of under- and over-treatment. Ultimately, it is imperative that more resources are channeled towards African-based studies on the molecular basis of cancer, enabling technologies and personnel training with the goal of developing genomic, proteomic, and cell-based biomarkers that are specific to the African population. This will make easy and early diagnosis a reality and help to improve the management of cancer in the African continent.

Author Contributions

Conceptualization, D.O.T., M.W., L.F.Z.; Writing of original draft, D.O.T., M.W., T.H.C., H.A.A.; Writing—review and editing, D.O.T., M.W., T.H.C., H.A.A., G.M.C., C.V.C., L.F.Z.; Supervision, G.M.C., C.V.C., L.F.Z.; Funding acquisition, G.M.C., C.V.C., L.F.Z.

Funding

Support by The International Centre for Genetic Engineering and Biotechnology, ICGEB (LFZ); ICGEB Arturo Falaschi fellowship (D.O.T.); Joint Research Grant South Africa/Switzerland Research Partnership Programme Bilateral Agreement (L.F.Z., C.V.C., G.M.C.). Funded by South African Medical Research Council (SAMRC) for a mid-career scientist and Self-initiated research grant; and the South African National Research Foundation (NRF) for incentive and research development grants for rated researchers (H.A.A).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed]
  2. Jemal, A.; Center, M.M.; DeSantis, C.; Ward, E.M. Global patterns of cancer incidence and mortality rates and trends. Cancer Epidem. Biomar. 2010, 19, 1893–1907. [Google Scholar] [CrossRef] [PubMed]
  3. Shin, H.R.; Boniol, M.; Joubert, C.; Hery, C.; Haukka, J.; Autier, P.; Nishino, Y.; Sobue, T.; Chen, C.J.; You, S.L.; et al. Secular trends in breast cancer mortality in five east asian populations: Hong kong, japan, korea, singapore and taiwan. Cancer Sci. 2010, 101, 1241–1246. [Google Scholar] [CrossRef] [PubMed]
  4. Youlden, D.R.; Cramb, S.M.; Dunn, N.A.; Muller, J.M.; Pyke, C.M.; Baade, P.D. The descriptive epidemiology of female breast cancer: An international comparison of screening, incidence, survival and mortality. Cancer Epidemiol. 2012, 36, 237–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Vorobiof, D.A.; Abratt, R. The cancer burden in africa. S. Afr. Med. J. 2007, 97, 937–939. [Google Scholar] [PubMed]
  6. Ibrahim, N.A.; Oludara, M.A. Socio-demographic factors and reasons associated with delay in breast cancer presentation: A study in nigerian women. Breast 2012, 21, 416–418. [Google Scholar] [CrossRef] [PubMed]
  7. Ouasmani, F.; Hanchi, Z.; Haddou Rahou, B.; Bekkali, R.; Ahid, S.; Mesfioui, A. Determinants of patient delay in seeking diagnosis and treatment among moroccan women with cervical cancer. Obstet. Gynecol. Int. 2016, 2016, 4840762. [Google Scholar] [CrossRef]
  8. Gerlinger, M.; Rowan, A.J.; Horswell, S.; Math, M.; Larkin, J.; Endesfelder, D.; Gronroos, E.; Martinez, P.; Matthews, N.; Stewart, A.; et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 2012, 366, 883–892. [Google Scholar] [CrossRef]
  9. Cheung, A.H.; Chow, C.; To, K.F. Latest development of liquid biopsy. J. Thorac. Dis. 2018, 10, S1645–S1651. [Google Scholar] [CrossRef]
  10. Wong, A.I.; Lo, Y.D. Noninvasive fetal genomic, methylomic, and transcriptomic analyses using maternal plasma and clinical implications. Trends Mol. Med. 2015, 21, 98–108. [Google Scholar] [CrossRef]
  11. Esteller, M.; Sanchez-Cespedes, M.; Rosell, R.; Sidransky, D.; Baylin, S.B.; Herman, J.G. Detection of aberrant promoter hypermethylation of tumor suppressor genes in serum DNA from non-small cell lung cancer patients. Cancer Res. 1999, 59, 67–70. [Google Scholar] [PubMed]
  12. Kopreski, M.S.; Benko, F.A.; Kwee, C.; Leitzel, K.E.; Eskander, E.; Lipton, A.; Gocke, C.D. Detection of mutant k-ras DNA in plasma or serum of patients with colorectal cancer. Br. J. Cancer 1997, 76, 1293–1299. [Google Scholar] [CrossRef] [PubMed]
  13. Sanchez-Cespedes, M.; Monzo, M.; Rosell, R.; Pifarre, A.; Calvo, R.; Lopez-Cabrerizo, M.P.; Astudillo, J. Detection of chromosome 3p alterations in serum DNA of non-small-cell lung cancer patients. Ann. Oncol. 1998, 9, 113–116. [Google Scholar] [CrossRef] [PubMed]
  14. Yamada, T.; Nakamori, S.; Ohzato, H.; Oshima, S.; Aoki, T.; Higaki, N.; Sugimoto, K.; Akagi, K.; Fujiwara, Y.; Nishisho, I.; et al. Detection of k-ras gene mutations in plasma DNA of patients with pancreatic adenocarcinoma: Correlation with clinicopathological features. Clin. Cancer Res. 1998, 4, 1527–1532. [Google Scholar] [PubMed]
  15. Adeola, H.A.; Soares, N.C.; Paccez, J.D.; Kaestner, L.; Blackburn, J.M.; Zerbini, L.F. Discovery of novel candidate urinary protein biomarkers for prostate cancer in a multiethnic cohort of south african patients via label-free mass spectrometry. Proteomics Clin. Appl. 2015, 9, 597–609. [Google Scholar] [CrossRef] [PubMed]
  16. Montaser, L.M.; Sonbol, A.A.E.; EL-Gammal, A.S.; El-Yazeed, A.S.A. Role of circulating endothelial progenitor cells in patients with breast cancer. Int. J. Curr. Res. 2018, 10, 65250–65256. [Google Scholar]
  17. Elnagdy, M.H.; Farouk, O.; Seleem, A.K.; Nada, H.A. Tff1 and tff3 mrnas are higher in blood from breast cancer patients with metastatic disease than those without. J. Oncol. 2018, 2018, 4793498. [Google Scholar] [CrossRef]
  18. Sayed, M.; Zahran, A.M.; Hassan, M.S.F.; Mohamed, D.O. Circulating tumor cells and cancer stem cells: Clinical implications in nonmetastatic breast cancer. Breast Cancer Basic Clin. Res. 2016, 10, 197. [Google Scholar] [CrossRef]
  19. Bahnassy, A.A.; Zekri, A.R.; El-Bastawisy, A.; Fawzy, A.; Shetta, M.; Hussein, N.; Omran, D.; Ahmed, A.A.; El-Labbody, S.S. Circulating tumor and cancer stem cells in hepatitis c virus-associated liver disease. World J. Gastroenterol. 2014, 20, 18240–18248. [Google Scholar] [CrossRef]
  20. Mansour, A.H.; Elkhodary, T.R.; Anwar, R.; Habeeb, M.R.; Mohammed, M.A. Regulation of cancer stem cell marker (cd133) by transforming growth factor beta in hepatocellular carcinoma. Int. J. Cancer 2014, 10, 65–73. [Google Scholar]
  21. Zedan, A.; Zahran, A.M.; Maximos, D.W.; Hassan, M.F.S. The role of circulating tumor cells (ctcs) in predicting the response of primary (neoadjuvant) chemotherapy and its impact as a prognostic factor in early breast cancer. SECI Oncol. 2014, 2, 68–75. [Google Scholar] [CrossRef]
  22. Ebeed, S.A.; El-Moneim, N.; Saad, A.; Zaher, E.R.; Yassin, O.G.; Khamis, S.A. Diagnostic and prognostic value of circulating tumor cells in female breast cancer patients. Alexandria Med. J. 2012, 48, 197–206. [Google Scholar] [CrossRef] [Green Version]
  23. Teama, S.H.; Agwa, S.H.A. Detection of circulating tumor cells by nested rt-pcr targeting egfr/cea/ck20mrnas in colorectal carcinoma patients. Egypt. J. Med. Hum. Genet. 2010, 11, 173–180. [Google Scholar] [CrossRef]
  24. Zehentner, B.K.; Persing, D.H.; Deme, A.; Toure, P.; Hawes, S.E.; Brooks, L.; Feng, Q.; Hayes, D.C.; Critichlow, C.W.; Houghton, R.L.; et al. Mammaglobin as a novel breast cancer biomarker: Multigene reverse transcription-pcr assay and sandwich elisa. Clin. Chem. 2004, 50, 2069–2076. [Google Scholar] [CrossRef] [PubMed]
  25. Hanekom, G.; Stubbings, H.; Johnson, C.; Kidson, S. The detection of circulating melanoma cells correlates with tumour thickness and ulceration but is not predictive of metastasis for patients with primary melanoma. Melanoma Res. 1999, 9, 465–473. [Google Scholar] [CrossRef] [PubMed]
  26. Marchio, A.; Amougou Atsama, M.; Bere, A.; Komas, N.P.; Noah Noah, D.; Atangana, P.J.A.; Camengo-Police, S.M.; Njouom, R.; Bekondi, C.; Pineau, P. Droplet digital pcr detects high rate of tp53 r249s mutants in cell-free DNA of middle african patients with hepatocellular carcinoma. Clin. Exp. Med. 2018, 18, 421–431. [Google Scholar] [CrossRef]
  27. Hussein, N.A.; Mohamed, S.N.; Ahmed, M.A. Plasma alu-247, alu-115, and cfdna integrity as diagnostic and prognostic biomarkers for breast cancer. Appl. Biochem. Biotech. 2018, 187, 1028–1045. [Google Scholar] [CrossRef]
  28. Soliman, S.E.-S.; Alhanafy, A.M.; Habib, M.S.E.d.; Hagag, M.; Ibrahem, R.A.L. Serum circulating cell free DNA as potential diagnostic and prognostic biomarker in non small cell lung cancer. BB Rep. 2018, 15, 45–51. [Google Scholar] [CrossRef]
  29. Fawzy, A.; Sweify, K.M.; El-Fayoumy, H.M.; Nofal, N. Quantitative analysis of plasma cell-free DNA and its DNA integrity in patients with metastatic prostate cancer using alu sequence. J. Egypt. Natl. Canc. Inst. 2016, 28, 235–242. [Google Scholar] [CrossRef]
  30. Ibrahim, I.H.; Kamel, M.M.; Ghareeb, M. Circulating DNA in egyptian women with breast cancer. Asian Pac. J. Cancer Prev. 2016, 17, 2989–2993. [Google Scholar]
  31. El-Gayar, D.; El-Abd, N.; Hassan, N.; Ali, R. Increased free circulating DNA integrity index as a serum biomarker in patients with colorectal carcinoma. Asian Pac. J. Cancer Prev. 2016, 17, 939–944. [Google Scholar] [CrossRef] [PubMed]
  32. Mahmoud, E.H.; Fawzy, A.; Ahmad, O.K.; Ali, A.M. Plasma circulating cell-free nuclear and mitochondrial DNA as potential biomarkers in the peripheral blood of breast cancer patients. Asian Pac. J. Cancer Prev. 2015, 16, 8299–8305. [Google Scholar] [CrossRef] [PubMed]
  33. Zaher, E.R.; Anwar, M.M.; Kohail, H.M.; El-Zoghby, S.M.; Abo-El-Eneen, M.S. Cell-free DNA concentration and integrity as a screening tool for cancer. Indian J. Cancer 2013, 50, 175–183. [Google Scholar] [CrossRef] [PubMed]
  34. Hashad, D.; Sorour, A.; Ghazal, A.; Talaat, I. Free circulating tumor DNA as a diagnostic marker for breast cancer. J. Clin. Lab. Anal. 2012, 26, 467–472. [Google Scholar] [CrossRef] [PubMed]
  35. El-Shazly, S.F.; Eid, M.A.; El-Sourogy, H.A.; Attia, G.F.; Ezzat, S.A. Evaluation of serum DNA integrity as a screening and prognostic tool in patients with hepatitis c virus-related hepatocellular carcinoma. Int. J. Biol. Markers 2010, 25, 79–86. [Google Scholar] [CrossRef] [PubMed]
  36. Iyer, P.; Zekri, A.R.; Hung, C.W.; Schiefelbein, E.; Ismail, K.; Hablas, A.; Seifeldin, I.A.; Soliman, A.S. Concordance of DNA methylation pattern in plasma and tumor DNA of egyptian hepatocellular carcinoma patients. Exp. Mol. Pathol. 2010, 88, 107–111. [Google Scholar] [CrossRef] [PubMed]
  37. Hosny, G.; Farahat, N.; Hainaut, P. Tp53 mutations in circulating free DNA from egyptian patients with non-hodgkin’s lymphoma. Cancer Lett. 2009, 275, 234–239. [Google Scholar] [CrossRef] [PubMed]
  38. Hosny, G.; Farahat, N.; Tayel, H.; Hainaut, P. Ser-249 tp53 and ctnnb1 mutations in circulating free DNA of egyptian patients with hepatocellular carcinoma versus chronic liver diseases. Cancer Lett. 2008, 264, 201–208. [Google Scholar] [CrossRef] [PubMed]
  39. Kirk, G.D.; Lesi, O.A.; Mendy, M.; Szymanska, K.; Whittle, H.; Goedert, J.J.; Hainaut, P.; Montesano, R. 249(ser) tp53 mutation in plasma DNA, hepatitis b viral infection, and risk of hepatocellular carcinoma. Oncogene 2005, 24, 5858–5867. [Google Scholar] [CrossRef]
  40. Szymanska, K.; Lesi, O.A.; Kirk, G.D.; Sam, O.; Taniere, P.; Scoazec, J.Y.; Mendy, M.; Friesen, M.D.; Whittle, H.; Montesano, R.; et al. Ser-249tp53 mutation in tumour and plasma DNA of hepatocellular carcinoma patients from a high incidence area in the gambia, west Africa. Int. J. Cancer 2004, 110, 374–379. [Google Scholar] [CrossRef]
  41. Van der Vaart, M.; Semenov, D.V.; Kuligina, E.V.; Richter, V.A.; Pretorius, P.J. Characterisation of circulating DNA by parallel tagged sequencing on the 454 platform. Clin. Chim. Acta 2009, 409, 21–27. [Google Scholar] [CrossRef] [PubMed]
  42. Van der Vaart, M.; Pretorius, P.J. A method for characterization of total circulating DNA. Ann. N. Y. Acad. Sci. 2008, 1137, 92–97. [Google Scholar] [CrossRef] [PubMed]
  43. Abd-El-Fattah, A.A.; Sadik, N.A.; Shaker, O.G.; Aboulftouh, M.L. Differential micrornas expression in serum of patients with lung cancer, pulmonary tuberculosis, and pneumonia. Cell Biochem. Biophys. 2013, 67, 875–884. [Google Scholar] [CrossRef] [PubMed]
  44. Ali, L.H.; Higazi, A.M.; Moness, H.M.; Farag, N.M.; Saad, Z.M.; Moukareb, H.A.; Soliman, W.; El Sagheer, G.; Abd El Hamid, S.R.; Abdl Hamid, H. Clinical significances and diagnostic utilities of both mir-215 and squamous cell carcinoma antigen-igm versus alpha-fetoprotein in egyptian patients with hepatitis c virus-induced hepatocellular carcinoma. Clin. Exp. Gastroenterol. 2019, 12, 51–66. [Google Scholar] [CrossRef] [PubMed]
  45. Ezzat, W.M.; Amr, K.S.; Elhosary, Y.A.; Hegazy, A.E.; Fahim, H.H.; Eltaweel, N.H.; Kamel, R.R. Detection of DNA methylated micrornas in hepatocellular carcinoma. Gene 2019, 702, 153–157. [Google Scholar] [CrossRef] [PubMed]
  46. Hetta, H.F.; Zahran, A.M.; Shafik, E.A.; El-Mahdy, R.I.; Mohamed, N.A.; Nabil, E.E.; Esmaeel, H.M.; Alkady, O.A.; Elkady, A.; Mohareb, D.A.; et al. Circulating mirna-21 and mirna-23a expression signature as potential biomarkers for early detection of non-small-cell lung cancer. Microrna 2019, 8, 206–215. [Google Scholar] [CrossRef] [PubMed]
  47. Mahmoud, E.H.; Fawzy, A.; Elshimy, R.A.A. Serum microrna-21 negatively relates to expression of programmed cell death-4 in patients with epithelial ovarian cancer. Asian Pac. J. Cancer Prev. 2018, 19, 33–38. [Google Scholar] [PubMed]
  48. Sabry, D.; El-Deek, S.E.M.; Maher, M.; El-Baz, M.A.H.; El-Bader, H.M.; Amer, E.; Hassan, E.A.; Fathy, W.; El-Deek, H.E.M. Role of mirna-210, mirna-21 and mirna-126 as diagnostic biomarkers in colorectal carcinoma: Impact of hif-1alpha-vegf signaling pathway. Mol. Cell. Biochem. 2019, 454, 177–189. [Google Scholar] [CrossRef] [PubMed]
  49. Swellam, M.; Mahmoud, M.S.; Hashim, M.; Hassan, N.M.; Sobeih, M.E.; Nageeb, A.M. Clinical aspects of circulating mirna-335 in breast cancer patients: A prospective study. J. Cell. Biochem. 2019, 120, 8975–8982. [Google Scholar] [CrossRef] [PubMed]
  50. Swellam, M.; El Magdoub, H.M.; Hassan, N.M.; Hefny, M.M.; Sobeih, M.E. Potential diagnostic role of circulating mirnas in breast cancer: Implications on clinicopathological characters. Clin. Biochem. 2018, 56, 47–54. [Google Scholar] [CrossRef] [PubMed]
  51. Toraih, E.A.; Alghamdi, S.A.; El-Wazir, A.; Hosny, M.M.; Hussein, M.H.; Khashana, M.S.; Fawzy, M.S. Dual biomarkers long non-coding rna gas5 and microrna-34a co-expression signature in common solid tumors. PLoS ONE 2018, 13, e0198231. [Google Scholar] [CrossRef] [PubMed]
  52. Zekri, A.N.; El-Sisi, E.R.; Youssef, A.S.E.; Kamel, M.M.; Nassar, A.; Ahmed, O.S.; El Kassas, M.; Barakat, A.B.; Abd El-Motaleb, A.I.; Bahnassy, A.A. Microrna signatures for circulating cd133-positive cells in hepatocellular carcinoma with hcv infection. PLoS ONE 2018, 13, e0193709. [Google Scholar] [CrossRef] [PubMed]
  53. Demerdash, H.M.; Hussien, H.M.; Hassouna, E.; Arida, E.A. Detection of microrna in hepatic cirrhosis and hepatocellular carcinoma in hepatitis c genotype-4 in egyptian patients. Biomed. Res. Int. 2017, 2017, 1806069. [Google Scholar] [CrossRef] [PubMed]
  54. Elemeery, M.N.; Badr, A.N.; Mohamed, M.A.; Ghareeb, D.A. Validation of a serum microrna panel as biomarkers for early diagnosis of hepatocellular carcinoma post-hepatitis c infection in egyptian patients. World J. Gastroenterol. 2017, 23, 3864. [Google Scholar] [CrossRef] [PubMed]
  55. Elhamamsy, A.R.; El Sharkawy, M.S.; Zanaty, A.F.; Mahrous, M.A.; Mohamed, A.E.; Abushaaban, E.A. Circulating mir-92a, mir-143 and mir-342 in plasma are novel potential biomarkers for acute myeloid leukemia. Int. J. Mol. Cell. Med. 2017, 6, 77–86. [Google Scholar]
  56. Elshafei, A.; Shaker, O.; Abd El-Motaal, O.; Salman, T. The expression profiling of serum mir-92a, mir-375, and mir-760 in colorectal cancer: An egyptian study. Tumour Biol. 2017, 39. [Google Scholar] [CrossRef] [PubMed]
  57. Khairy, A.; Hamza, I.; Shaker, O.; Yosry, A. Serum mirna panel in egyptian patients with chronic hepatitis c related hepatocellular carcinoma. Asian Pac. J. Cancer Prev. 2016, 17, 2699–2703. [Google Scholar]
  58. Motawi, T.K.; Rizk, S.M.; Ibrahim, T.M.; Ibrahim, I.A. Circulating micrornas, mir-92a, mir-100 and mir-143, as non-invasive biomarkers for bladder cancer diagnosis. Cell Biochem. Funct. 2016, 34, 142–148. [Google Scholar] [CrossRef]
  59. Motawi, T.M.; Sadik, N.A.; Shaker, O.G.; Ghaleb, M.H. Elevated serum microrna-122/222 levels are potential diagnostic biomarkers in egyptian patients with chronic hepatitis c but not hepatic cancer. Tumour Biol. 2016, 37, 9865–9874. [Google Scholar] [CrossRef]
  60. Swellam, M.; El-Khazragy, N. Clinical impact of circulating micrornas as blood-based marker in childhood acute lymphoblastic leukemia. Tumour Biol. 2016, 37, 10571–10576. [Google Scholar] [CrossRef]
  61. Zekri, A.R.; Youssef, A.S.; Lotfy, M.M.; Gabr, R.; Ahmed, O.S.; Nassar, A.; Hussein, N.; Omran, D.; Medhat, E.; Eid, S.; et al. Circulating serum mirnas as diagnostic markers for colorectal cancer. PLoS ONE 2016, 11, e0154130. [Google Scholar] [CrossRef] [PubMed]
  62. Alnoanmany, W.; Ismail, H.A.; El-Said, H.; Obada, M.; Sakr, M.A.; Elfert, A.Y. Diagnostic potential of circulating microrna-21 in hepatocellular carcinoma. Int. J. Sci. Technol. Res. 2015, 4, 429–433. [Google Scholar]
  63. Eissa, S.; Matboli, M.; Hegazy, M.G.; Kotb, Y.M.; Essawy, N.O. Evaluation of urinary microrna panel in bladder cancer diagnosis: Relation to bilharziasis. Transl. Res. 2015, 165, 731–739. [Google Scholar] [CrossRef] [PubMed]
  64. El-Abd, N.E.; Fawzy, N.A.; El-Sheikh, S.M.; Soliman, M.E. Circulating mirna-122, mirna-199a, and mirna-16 as biomarkers for early detection of hepatocellular carcinoma in egyptian patients with chronic hepatitis c virus infection. Mol. Diagn. Ther. 2015, 19, 213–220. [Google Scholar] [CrossRef] [PubMed]
  65. Hagrass, H.A.; Sharaf, S.; Pasha, H.F.; Tantawy, E.A.; Mohamed, R.H.; Kassem, R. Circulating micrornas—A new horizon in molecular diagnosis of breast cancer. Genes Cancer 2015, 6, 281–287. [Google Scholar] [PubMed]
  66. Motawi, T.K.; Shaker, O.G.; El-Maraghy, S.A.; Senousy, M.A. Serum micrornas as potential biomarkers for early diagnosis of hepatitis c virus-related hepatocellular carcinoma in egyptian patients. PLoS ONE 2015, 10, e0137706. [Google Scholar] [CrossRef] [PubMed]
  67. Abdalla, M.A.; Haj-Ahmad, Y. Promising candidate urinary microrna biomarkers for the early detection of hepatocellular carcinoma among high-risk hepatitis c virus egyptian patients. J. Cancer 2012, 3, 19–31. [Google Scholar] [CrossRef] [PubMed]
  68. Hamdi, K.; Blancato, J.; Goerlitz, D.; Islam, M.; Neili, B.; Abidi, A.; Gat, A.; Ayed, F.B.; Chivi, S.; Loffredo, C.; et al. Circulating cell-free mirna expression and its association with clinicopathologic features in inflammatory and non-inflammatory breast cancer. Asian Pac. J. Cancer Prev. 2016, 17, 1801–1810. [Google Scholar] [CrossRef] [PubMed]
  69. Abdelgawad, I.A.; Mossallam, G.I.; Radwan, N.H.; Elzawahry, H.M.; Elhifnawy, N.M. Can glypican3 be diagnostic for early hepatocellular carcinoma among egyptian patients? Asian Pac. J. Cancer Prev. 2013, 14, 7345–7349. [Google Scholar] [CrossRef]
  70. Ibrahim, G.H.; Mahmoud, M.A.; Aly, N.M. Evaluation of circulating transforming growth factor-beta1, glypican-3 and golgi protein-73 mrnas expression as predictive markers for hepatocellular carcinoma in egyptian patients. Mol. Biol. Rep. 2013, 40, 7069–7075. [Google Scholar] [CrossRef]
  71. Zidan, H.E.; Karam, R.A.; El-Seifi, O.S.; Abd Elrahman, T.M. Circulating long non-coding rna malat1 expression as molecular biomarker in egyptian patients with breast cancer. Cancer Genet. 2018, 220, 32–37. [Google Scholar] [CrossRef] [PubMed]
  72. 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] [PubMed]
  73. El-Tawdi, A.H.; Matboli, M.; Shehata, H.H.; Tash, F.; El-Khazragy, N.; Azazy Ael, S.; Abdel-Rahman, O. Evaluation of circulatory rna-based biomarker panel in hepatocellular carcinoma. Mol. Diagn. Ther. 2016, 20, 265–277. [Google Scholar] [CrossRef] [PubMed]
  74. Hashad, D.; Elbanna, A.; Ibrahim, A.; Khedr, G. Evaluation of the role of circulating long non-coding rna h19 as a promising novel biomarker in plasma of patients with gastric cancer. J. Clin. Lab. Anal. 2016, 30, 1100–1105. [Google Scholar] [CrossRef] [PubMed]
  75. Abd El Gwad, A.; Matboli, M.; El-Tawdi, A.; Habib, E.K.; Shehata, H.; Ibrahim, D.; Tash, F. Role of exosomal competing endogenous rna in patients with hepatocellular carcinoma. J. Cell. Biochem. 2018, 119, 8600–8610. [Google Scholar] [CrossRef] [PubMed]
  76. Khalil, F.; Shehata, H.H.; Osman, N.; Abdel-Rahman, O.; Ali, M.; Mohamed, G. Evaluation of the diagnostic role of non-coding rna and exosomal related gene association in lung cancer. Egypt. J. Hosp. Med. 2018, 72, 4148–4153. [Google Scholar]
  77. Weigelt, B.; Peterse, J.L.; van’t Veer, L.J. Breast cancer metastasis: Markers and models. Nat. Rev. Cancer 2005, 5, 591–602. [Google Scholar] [CrossRef] [PubMed]
  78. Cristofanilli, M.; Budd, G.T.; Ellis, M.J.; Stopeck, A.; Matera, J.; Miller, M.C.; Reuben, J.M.; Doyle, G.V.; Allard, W.J.; Terstappen, L.W.; et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N. Engl. J. Med. 2004, 351, 781–791. [Google Scholar] [CrossRef]
  79. Cohen, S.J.; Punt, C.J.; Iannotti, N.; Saidman, B.H.; Sabbath, K.D.; Gabrail, N.Y.; Picus, J.; Morse, M.; Mitchell, E.; Miller, M.C.; et al. Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J. Clin. Oncol. 2008, 26, 3213–3221. [Google Scholar] [CrossRef]
  80. Goldkorn, A.; Ely, B.; Quinn, D.I.; Tangen, C.M.; Fink, L.M.; Xu, T.; Twardowski, P.; Van Veldhuizen, P.J.; Agarwal, N.; Carducci, M.A.; et al. Circulating tumor cell counts are prognostic of overall survival in swog s0421: A phase iii trial of docetaxel with or without atrasentan for metastatic castration-resistant prostate cancer. J. Clin. Oncol. 2014, 32, 1136–1142. [Google Scholar] [CrossRef]
  81. De Bono, J.S.; Scher, H.I.; Montgomery, R.B.; Parker, C.; Miller, M.C.; Tissing, H.; Doyle, G.V.; Terstappen, L.W.; Pienta, K.J.; Raghavan, D. Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin. Cancer Res. 2008, 14, 6302–6309. [Google Scholar] [CrossRef] [PubMed]
  82. Cristofanilli, M.; Hayes, D.F.; Budd, G.T.; Ellis, M.J.; Stopeck, A.; Reuben, J.M.; Doyle, G.V.; Matera, J.; Allard, W.J.; Miller, M.C.; et al. Circulating tumor cells: A novel prognostic factor for newly diagnosed metastatic breast cancer. J. Clin. Oncol. 2005, 23, 1420–1430. [Google Scholar] [CrossRef] [PubMed]
  83. Fiorelli, A.; Accardo, M.; Carelli, E.; Angioletti, D.; Santini, M.; Di Domenico, M. Circulating tumor cells in diagnosing lung cancer: Clinical and morphologic analysis. Ann. Thorac. Surg. 2015, 99, 1899–1905. [Google Scholar] [CrossRef] [PubMed]
  84. Rhim, A.D.; Mirek, E.T.; Aiello, N.M.; Maitra, A.; Bailey, J.M.; McAllister, F.; Reichert, M.; Beatty, G.L.; Rustgi, A.K.; Vonderheide, R.H.; et al. Emt and dissemination precede pancreatic tumor formation. Cell 2012, 148, 349–361. [Google Scholar] [CrossRef] [PubMed]
  85. Ilie, M.; Hofman, V.; Long-Mira, E.; Selva, E.; Vignaud, J.M.; Padovani, B.; Mouroux, J.; Marquette, C.H.; Hofman, P. “Sentinel” circulating tumor cells allow early diagnosis of lung cancer in patients with chronic obstructive pulmonary disease. PLoS ONE 2014, 9, e111597. [Google Scholar] [CrossRef]
  86. Gulbahce, N.; Magbanua, M.J.M.; Chin, R.; Agarwal, M.R.; Luo, X.; Liu, J.; Hayden, D.M.; Mao, Q.; Ciotlos, S.; Li, Z.; et al. Quantitative whole genome sequencing of circulating tumor cells enables personalized combination therapy of metastatic cancer. Cancer Res. 2017, 77, 4530–4541. [Google Scholar] [CrossRef]
  87. Lin, E.; Cao, T.; Nagrath, S.; King, M.R. Circulating tumor cells: Diagnostic and therapeutic applications. Annu. Rev. Biomed. Eng. 2018, 20, 329–352. [Google Scholar] [CrossRef]
  88. Beije, N.; Jager, A.; Sleijfer, S. Circulating tumor cell enumeration by the cellsearch system: The clinician’s guide to breast cancer treatment? Cancer Treat. Rev. 2015, 41, 144–150. [Google Scholar] [CrossRef]
  89. Kraan, J.; Sleijfer, S.; Strijbos, M.H.; Ignatiadis, M.; Peeters, D.; Pierga, J.Y.; Farace, F.; Riethdorf, S.; Fehm, T.; Zorzino, L.; et al. External quality assurance of circulating tumor cell enumeration using the cellsearch® system: A feasibility study. Cytometry B Clin. Cytom. 2011, 80, 112–118. [Google Scholar] [CrossRef]
  90. Gorges, T.M.; Tinhofer, I.; Drosch, M.; Rose, L.; Zollner, T.M.; Krahn, T.; von Ahsen, O. Circulating tumour cells escape from epcam-based detection due to epithelial-to-mesenchymal transition. BMC Cancer 2012, 12, 178. [Google Scholar] [CrossRef]
  91. Sieuwerts, A.M.; Kraan, J.; Bolt, J.; van der Spoel, P.; Elstrodt, F.; Schutte, M.; Martens, J.W.; Gratama, J.W.; Sleijfer, S.; Foekens, J.A. Anti-epithelial cell adhesion molecule antibodies and the detection of circulating normal-like breast tumor cells. J. Natl. Cancer Inst. 2009, 101, 61–66. [Google Scholar] [CrossRef] [PubMed]
  92. Armstrong, A.J.; Marengo, M.S.; Oltean, S.; Kemeny, G.; Bitting, R.L.; Turnbull, J.D.; Herold, C.I.; Marcom, P.K.; George, D.J.; Garcia-Blanco, M.A. Circulating tumor cells from patients with advanced prostate and breast cancer display both epithelial and mesenchymal markers. Mol. Cancer Res. 2011, 9, 997–1007. [Google Scholar] [CrossRef] [PubMed]
  93. Pantel, K.; Denève, E.; Nocca, D.; Coffy, A.; Vendrell, J.P.; Maudelonde, T.; Riethdorf, S.; Alix-Panabières, C. Circulating epithelial cells in patients with benign colon diseases. Clin. Chem. 2012, 58, 936–940. [Google Scholar]
  94. Allard, W.J.; Matera, J.; Miller, M.C.; Repollet, M.; Connelly, M.C.; Rao, C.; Tibbe, A.G.; Uhr, J.W.; Terstappen, L.W. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin. Cancer Res. 2004, 10, 6897–6904. [Google Scholar] [CrossRef] [PubMed]
  95. Riethdorf, S.; Fritsche, H.; Muller, V.; Rau, T.; Schindlbeck, C.; Rack, B.; Janni, W.; Coith, C.; Beck, K.; Janicke, F.; et al. Detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer: A validation study of the cellsearch system. Clin. Cancer Res. 2007, 13, 920–928. [Google Scholar] [CrossRef] [PubMed]
  96. Punnoose, E.A.; Atwal, S.K.; Spoerke, J.M.; Savage, H.; Pandita, A.; Yeh, R.-F.; Pirzkall, A.; Fine, B.M.; Amler, L.C.; Chen, D.S.; et al. Molecular biomarker analyses using circulating tumor cells. PLoS ONE 2010, 5, e12517. [Google Scholar] [CrossRef] [PubMed]
  97. Budd, G.T.; Cristofanilli, M.; Ellis, M.J.; Stopeck, A.; Borden, E.; Miller, M.C.; Matera, J.; Repollet, M.; Doyle, G.V.; Terstappen, L.W. Circulating tumor cells versus imaging—Predicting overall survival in metastatic breast cancer. Clin. Cancer Res. 2006, 12, 6403–6409. [Google Scholar] [CrossRef]
  98. Heller, G.; McCormack, R.; Kheoh, T.; Molina, A.; Smith, M.R.; Dreicer, R.; Saad, F.; de Wit, R.; Aftab, D.T.; Hirmand, M.; et al. Circulating tumor cell number as a response measure of prolonged survival for metastatic castration-resistant prostate cancer: A comparison with prostate-specific antigen across five randomized phase iii clinical trials. J. Clin. Oncol. 2018, 36, 572–580. [Google Scholar] [CrossRef]
  99. Yan, W.T.; Cui, X.; Chen, Q.; Li, Y.F.; Cui, Y.H.; Wang, Y.; Jiang, J. Circulating tumor cell status monitors the treatment responses in breast cancer patients: A meta-analysis. Sci. Rep. 2017, 7, 43464. [Google Scholar] [CrossRef]
  100. Krebs, M.G.; Renehan, A.G.; Backen, A.; Gollins, S.; Chau, I.; Hasan, J.; Valle, J.W.; Morris, K.; Beech, J.; Ashcroft, L.; et al. Circulating tumor cell enumeration in a phase ii trial of a four-drug regimen in advanced colorectal cancer. Clin. Colorectal Cancer 2015, 14, 115–122. [Google Scholar] [CrossRef]
  101. Molloy, T.J.; Bosma, A.J.; Baumbusch, L.O.; Synnestvedt, M.; Borgen, E.; Russnes, H.G.; Schlichting, E.; van’t Veer, L.J.; Naume, B. The prognostic significance of tumour cell detection in the peripheral blood versus the bone marrow in 733 early-stage breast cancer patients. Breast Cancer Res. 2011, 13, R61. [Google Scholar] [CrossRef] [PubMed]
  102. Daskalaki, A.; Agelaki, S.; Perraki, M.; Apostolaki, S.; Xenidis, N.; Stathopoulos, E.; Kontopodis, E.; Hatzidaki, D.; Mavroudis, D.; Georgoulias, V. Detection of cytokeratin-19 mrna-positive cells in the peripheral blood and bone marrow of patients with operable breast cancer. Br. J. Cancer 2009, 101, 589–597. [Google Scholar] [CrossRef] [PubMed]
  103. Skillrud, D.M.; Offord, K.P.; Miller, R.D. Higher risk of lung cancer in chronic obstructive pulmonary disease. A prospective, matched, controlled study. Ann. Intern. Med. 1986, 105, 503–507. [Google Scholar] [CrossRef] [PubMed]
  104. Bardelli, A.; Pantel, K. Liquid biopsies, what we do not know (yet). Cancer Cell 2017, 31, 172–179. [Google Scholar] [CrossRef] [PubMed]
  105. Fu, Y.; Li, C.; Lu, S.; Zhou, W.; Tang, F.; Xie, X.S.; Huang, Y. Uniform and accurate single-cell sequencing based on emulsion whole-genome amplification. Proc. Natl. Acad. Sci. USA 2015, 112, 11923–11928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Lohr, J.G.; Adalsteinsson, V.A.; Cibulskis, K.; Choudhury, A.D.; Rosenberg, M.; Cruz-Gordillo, P.; Francis, J.M.; Zhang, C.Z.; Shalek, A.K.; Satija, R.; et al. Whole-exome sequencing of circulating tumor cells provides a window into metastatic prostate cancer. Nat. Biotechnol. 2014, 32, 479–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. D’Avola, D.; Villacorta-Martin, C.; Martins-Filho, S.N.; Craig, A.; Labgaa, I.; von Felden, J.; Kimaada, A.; Bonaccorso, A.; Tabrizian, P.; Hartmann, B.M.; et al. High-density single cell mrna sequencing to characterize circulating tumor cells in hepatocellular carcinoma. Sci. Rep. 2018, 8, 11570. [Google Scholar] [CrossRef] [PubMed]
  108. Kong, S.L.; Liu, X.; Suhaimi, N.M.; Koh, K.J.H.; Hu, M.; Lee, D.Y.S.; Cima, I.; Phyo, W.M.; Lee, E.X.W.; Tai, J.A.; et al. Molecular characterization of circulating colorectal tumor cells defines genetic signatures for individualized cancer care. Oncotarget 2017, 8, 68026–68037. [Google Scholar] [CrossRef] [PubMed]
  109. Aceto, N.; Bardia, A.; Wittner, B.S.; Donaldson, M.C.; O’Keefe, R.; Engstrom, A.; Bersani, F.; Zheng, Y.; Comaills, V.; Niederhoffer, K.; et al. Ar expression in breast cancer ctcs associates with bone metastases. Mol. Cancer Res. 2018, 16, 720–727. [Google Scholar] [CrossRef] [PubMed]
  110. Zhang, Z.; Shiratsuchi, H.; Palanisamy, N.; Nagrath, S.; Ramnath, N. Expanded circulating tumor cells from a patient with alk-positive lung cancer present with eml4-alk rearrangement along with resistance mutation and enable drug sensitivity testing: A case study. J. Thorac. Oncol. 2017, 12, 397–402. [Google Scholar] [CrossRef]
  111. Mandel, P. Les acides nucleiques du plasma sanguin chez 1 homme. C.R. Seances Soc. Biol. Fil. 1948, 142, 241–243. [Google Scholar]
  112. Leon, S.A.; Shapiro, B.; Sklaroff, D.M.; Yaros, M.J. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res. 1977, 37, 646–650. [Google Scholar] [PubMed]
  113. Agassi, R.; Czeiger, D.; Shaked, G.; Avriel, A.; Sheynin, J.; Lavrenkov, K.; Ariad, S.; Douvdevani, A. Measurement of circulating cell-free DNA levels by a simple fluorescent test in patients with breast cancer. Am. J. Clin. Pathol. 2015, 143, 18–24. [Google Scholar] [CrossRef] [PubMed]
  114. Boddy, J.L.; Gal, S.; Malone, P.R.; Harris, A.L.; Wainscoat, J.S. Prospective study of quantitation of plasma DNA levels in the diagnosis of malignant versus benign prostate disease. Clin. Cancer Res. 2005, 11, 1394–1399. [Google Scholar] [CrossRef] [PubMed]
  115. Gordian, E.; Ramachandran, K.; Reis, I.M.; Manoharan, M.; Soloway, M.S.; Singal, R. Serum free circulating DNA is a useful biomarker to distinguish benign versus malignant prostate disease. Cancer Epidem. Biomar. 2010, 19, 1984–1991. [Google Scholar] [CrossRef] [PubMed]
  116. Fleischhacker, M.; Schmidt, B. Circulating nucleic acids (cnas) and cancer—A survey. BBA-Rev. Cancer 2007, 1775, 181–232. [Google Scholar] [CrossRef] [PubMed]
  117. Jiang, P.; Chan, C.W.; Chan, K.C.; Cheng, S.H.; Wong, J.; Wong, V.W.; Wong, G.L.; Chan, S.L.; Mok, T.S.; Chan, H.L.; et al. Lengthening and shortening of plasma DNA in hepatocellular carcinoma patients. Proc. Natl. Acad. Sci. USA 2015, 112, E1317–E1325. [Google Scholar] [CrossRef] [Green Version]
  118. Snyder, M.W.; Kircher, M.; Hill, A.J.; Daza, R.M.; Shendure, J. Cell-free DNA comprises an in vivo nucleosome footprint that informs its tissues-of-origin. Cell 2016, 164, 57–68. [Google Scholar] [CrossRef]
  119. Stroun, M.; Lyautey, J.; Lederrey, C.; Olson-Sand, A.; Anker, P. About the possible origin and mechanism of circulating DNA—Apoptosis and active DNA release. Clin. Chim. Acta 2001, 313, 139–142. [Google Scholar] [CrossRef]
  120. Umetani, N.; Giuliano, A.E.; Hiramatsu, S.H.; Amersi, F.; Nakagawa, T.; Martino, S.; Hoon, D.S. Prediction of breast tumor progression by integrity of free circulating DNA in serum. J. Clin. Oncol. 2006, 24, 4270–4276. [Google Scholar] [CrossRef]
  121. Giacona, M.B.; Ruben, G.C.; Iczkowski, K.A.; Roos, T.B.; Porter, D.M.; Sorenson, G.D. Cell-free DNA in human blood plasma: Length measurements in patients with pancreatic cancer and healthy controls. Pancreas 1998, 17, 89–97. [Google Scholar] [CrossRef]
  122. Wang, B.G.; Huang, H.Y.; Chen, Y.C.; Bristow, R.E.; Kassauei, K.; Cheng, C.C.; Roden, R.; Sokoll, L.J.; Chan, D.W.; Shih Ie, M. Increased plasma DNA integrity in cancer patients. Cancer Res. 2003, 63, 3966–3968. [Google Scholar] [PubMed]
  123. Schwarzenbach, H.; Hoon, D.S.; Pantel, K. Cell-free nucleic acids as biomarkers in cancer patients. Nat. Rev. Cancer 2011, 11, 426–437. [Google Scholar] [CrossRef] [PubMed]
  124. Mouliere, F.; Rosenfeld, N. Circulating tumor-derived DNA is shorter than somatic DNA in plasma. Proc. Natl. Acad. Sci. USA 2015, 112, 3178–3179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Diehl, F.; Schmidt, K.; Choti, M.A.; Romans, K.; Goodman, S.; Li, M.; Thornton, K.; Agrawal, N.; Sokoll, L.; Szabo, S.A.; et al. Circulating mutant DNA to assess tumor dynamics. Nat. Med. 2008, 14, 985–990. [Google Scholar] [CrossRef] [PubMed]
  126. Cheng, F.; Su, L.; Qian, C. Circulating tumor DNA: A promising biomarker in the liquid biopsy of cancer. Oncotarget 2016, 7, 48832–48841. [Google Scholar] [CrossRef]
  127. Husain, H.; Nykin, D.; Bui, N.; Quan, D.; Gomez, G.; Woodward, B.; Venkatapathy, S.; Duttagupta, R.; Fung, E.; Lippman, S.M.; et al. Cell-free DNA from ascites and pleural effusions: Molecular insights into genomic aberrations and disease biology. Mol. Cancer Ther. 2017, 16, 948–955. [Google Scholar] [CrossRef]
  128. Fujiwara, K.; Fujimoto, N.; Tabata, M.; Nishii, K.; Matsuo, K.; Hotta, K.; Kozuki, T.; Aoe, M.; Kiura, K.; Ueoka, H.; et al. Identification of epigenetic aberrant promoter methylation in serum DNA is useful for early detection of lung cancer. Clin. Cancer Res. 2005, 11, 1219–1225. [Google Scholar]
  129. Stroun, M.; Anker, P.; Maurice, P.; Lyautey, J.; Lederrey, C.; Beljanski, M. Neoplastic characteristics of the DNA found in the plasma of cancer-patients. Oncology 1989, 46, 318–322. [Google Scholar] [CrossRef]
  130. 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]
  131. Zhang, H.; Liu, R.; Yan, C.; Liu, L.; Tong, Z.; Jiang, W.; Yao, M.; Fang, W.; Chen, Z. Advantage of next-generation sequencing in dynamic monitoring of circulating tumor DNA over droplet digital pcr in cetuximab treated colorectal cancer patients. Transl. Oncol. 2019, 12, 426–431. [Google Scholar] [CrossRef]
  132. Wyatt, A.W.; Annala, M.; Aggarwal, R.; Beja, K.; Feng, F.; Youngren, J.; Foye, A.; Lloyd, P.; Nykter, M.; Beer, T.M.; et al. Concordance of circulating tumor DNA and matched metastatic tissue biopsy in prostate cancer. J. Natl. Cancer Inst. 2017, 109, djx118. [Google Scholar] [CrossRef] [PubMed]
  133. Kidess-Sigal, E.; Liu, H.E.; Triboulet, M.M.; Che, J.; Ramani, V.C.; Visser, B.C.; Poultsides, G.A.; Longacre, T.A.; Marziali, A.; Vysotskaia, V.; et al. Enumeration and targeted analysis of kras, braf and pik3ca mutations in ctcs captured by a label-free platform: Comparison to ctdna and tissue in metastatic colorectal cancer. Oncotarget 2016, 7, 85349–85364. [Google Scholar] [CrossRef] [PubMed]
  134. Malapelle, U.; Sirera, R.; Jantus-Lewintre, E.; Reclusa, P.; Calabuig-Fariñas, S.; Blasco, A.; Pisapia, P.; Rolfo, C.; Camps, C. Profile of the roche cobas® egfr mutation test v2 for non-small cell lung cancer. Expert Rev. Mol. Diagn. 2017, 17, 209–215. [Google Scholar] [CrossRef] [PubMed]
  135. Wu, Y.-L.; Lee, V.; Liam, C.-K.; Lu, S.; Park, K.; Srimuninnimit, V.; Wang, J.; Zhou, C.; Appius, A.; Button, P.; et al. Clinical utility of a blood-based egfr mutation test in patients receiving first-line erlotinib therapy in the ensure, fastact-2, and aspiration studies. Lung Cancer 2018, 126, 1–8. [Google Scholar] [CrossRef] [PubMed]
  136. Potter, N.T.; Hurban, P.; White, M.N.; Whitlock, K.D.; Lofton-Day, C.E.; Tetzner, R.; Koenig, T.; Quigley, N.B.; Weiss, G. Validation of a real-time pcr–based qualitative assay for the detection of methylated sept9 DNA in human plasma. Clin. Chem. 2014, 60, 1183–1191. [Google Scholar] [CrossRef] [PubMed]
  137. 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]
  138. Ou, S.-H.I.; Nagasaka, M.; Zhu, V.W. Liquid biopsy to identify actionable genomic alterations. ASCO Educ. Book 2018, 38, 978–997. [Google Scholar] [CrossRef] [PubMed]
  139. Stroun, M.; Anker, P.; Beljanski, M.; Henri, J.; Lederrey, C.; Ojha, M.; Maurice, P.A. Presence of rna in the nucleoprotein complex spontaneously released by human lymphocytes and frog auricles in culture. Cancer Res. 1978, 38, 3546–3554. [Google Scholar]
  140. Colombo, M.; Raposo, G.; Thery, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell. Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
  141. Tsui, N.B.; Ng, E.K.; Lo, Y.M. Stability of endogenous and added rna in blood specimens, serum, and plasma. Clin. Chem. 2002, 48, 1647–1653. [Google Scholar]
  142. Suraj, S.; Dhar, C.; Srivastava, S. Circulating nucleic acids: An analysis of their occurrence in malignancies. Biomed. Rep. 2017, 6, 8–14. [Google Scholar] [CrossRef] [PubMed]
  143. Umu, S.U.; Langseth, H.; Bucher-Johannessen, C.; Fromm, B.; Keller, A.; Meese, E.; Lauritzen, M.; Leithaug, M.; Lyle, R.; Rounge, T.B. A comprehensive profile of circulating rnas in human serum. RNA Biol. 2018, 15, 242–250. [Google Scholar] [CrossRef] [PubMed]
  144. 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] [PubMed] [Green Version]
  145. Garcia, V.; Garcia, J.M.; Pena, C.; Silva, J.; Dominguez, G.; Lorenzo, Y.; Diaz, R.; Espinosa, P.; de Sola, J.G.; Cantos, B.; et al. Free circulating mrna in plasma from breast cancer patients and clinical outcome. Cancer Lett. 2008, 263, 312–320. [Google Scholar] [CrossRef] [PubMed]
  146. March-Villalba, J.A.; Martinez-Jabaloyas, J.M.; Herrero, M.J.; Santamaria, J.; Alino, S.F.; Dasi, F. Cell-free circulating plasma htert mrna is a useful marker for prostate cancer diagnosis and is associated with poor prognosis tumor characteristics. PLoS ONE 2012, 7, e43470. [Google Scholar] [CrossRef] [PubMed]
  147. Sahengbieke, S.; Wang, J.; Li, X.; Wang, Y.; Lai, M.; Wu, J. Circulating cell-free high mobility group at-hook 2 mrna as a detection marker in the serum of colorectal cancer patients. J. Clin. Lab. Anal. 2018, 32, e22332. [Google Scholar] [CrossRef] [PubMed]
  148. Shen, J.; Kong, W.; Wu, Y.; Ren, H.; Wei, J.; Yang, Y.; Yang, Y.; Yu, L.; Guan, W.; Liu, B. Plasma mrna as liquid biopsy predicts chemo-sensitivity in advanced gastric cancer patients. J. Cancer 2017, 8, 434–442. [Google Scholar] [CrossRef] [PubMed]
  149. De Souza, M.F.; Kuasne, H.; de Camargo Barros-Filho, M.; Cilião, H.L.; Marchi, F.A.; Fuganti, P.E.; Paschoal, A.R.; Rogatto, S.R.; de Syllos Cólus, I.M. Circulating mrnas and mirnas as candidate markers for the diagnosis and prognosis of prostate cancer. PloS ONE 2017, 12, e0184094. [Google Scholar] [CrossRef] [PubMed]
  150. Tani, N.; Ichikawa, D.; Ikoma, D.; Tomita, A.; Sai, S.; Ikoma, H.; Okamoto, K.; Ochiai, T.; Ueda, Y.; Otsuji, E.; et al. Circulating cell-free mrna in plasma as a tumor marker for patients with primary and recurrent gastric cancer. Anticancer Res. 2007, 27, 1207–1212. [Google Scholar] [PubMed]
  151. Fok, E.T.; Scholefield, J.; Fanucchi, S.; Mhlanga, M.M. The emerging molecular biology toolbox for the study of long noncoding rna biology. Epigenomics 2017, 9, 1317–1327. [Google Scholar] [CrossRef] [PubMed]
  152. Bhat, S.A.; Ahmad, S.M.; Mumtaz, P.T.; Malik, A.A.; Dar, M.A.; Urwat, U.; Shah, R.A.; Ganai, N.A. Long non-coding rnas: Mechanism of action and functional utility. Non-coding RNA Res. 2016, 1, 43–50. [Google Scholar] [CrossRef] [PubMed]
  153. Zhao, M.; Wang, S.; Li, Q.; Ji, Q.; Guo, P.; Liu, X. Malat1: A long non-coding rna highly associated with human cancers. Oncol. Lett. 2018, 16, 19–26. [Google Scholar] [CrossRef] [PubMed]
  154. Chen, C.; Feng, Y.; Wang, X. Lncrna zeb1-as1 expression in cancer prognosis: Review and meta-analysis. Clin. Chim. Acta 2018, 484, 265–271. [Google Scholar] [CrossRef] [PubMed]
  155. Xie, Y.; Zhang, Y.; Du, L.; Jiang, X.; Yan, S.; Duan, W.; Li, J.; Zhan, Y.; Wang, L.; Zhang, S. Circulating long noncoding rna act as potential novel biomarkers for diagnosis and prognosis of non-small cell lung cancer. Mol. Oncol. 2018, 12, 648–658. [Google Scholar] [CrossRef] [PubMed]
  156. Lv, R.; Zhang, J.; Zhang, W.; Huang, Y.; Wang, N.; Zhang, Q.; Qu, S. Circulating hotair expression predicts the clinical response to neoadjuvant chemotherapy in patients with breast cancer. Cancer Biomark. 2018, 22, 249–256. [Google Scholar]
  157. Aubin, S.M.J.; Reid, J.; Sarno, M.J.; Blase, A.; Aussie, J.; Rittenhouse, H.; Rittmaster, R.; Andriole, G.L.; Groskopf, J. Pca3 molecular urine test for predicting repeat prostate biopsy outcome in populations at risk: Validation in the placebo arm of the dutasteride reduce trial. J. Urology 2010, 184, 1947–1952. [Google Scholar] [CrossRef] [PubMed]
  158. De La Taille, A. Progensa™ pca3 test for prostate cancer detection. Expert Rev. Mol. Diagn. 2007, 7, 491–497. [Google Scholar] [CrossRef] [PubMed]
  159. Sohel, M.H. Extracellular/circulating micrornas: Release mechanisms, functions and challenges. Achiev. Life Sci. 2016, 10, 175–186. [Google Scholar] [CrossRef]
  160. Catalanotto, C.; Cogoni, C.; Zardo, G. Microrna in control of gene expression: An overview of nuclear functions. Int. J. Mol. Sci. 2016, 17, 1712. [Google Scholar] [CrossRef]
  161. Bortoluzzi, S.; Lovisa, F.; Gaffo, E.; Mussolin, L. Small rnas in circulating exosomes of cancer patients: A minireview. High-Throughput 2017, 6, 13. [Google Scholar] [CrossRef]
  162. Iftikhar, H.; Carney, G.E. Evidence and potential in vivo functions for biofluid mirnas: From expression profiling to functional testing: Potential roles of extracellular mirnas as indicators of physiological change and as agents of intercellular information exchange. BioEssays 2016, 38, 367–378. [Google Scholar] [CrossRef] [PubMed]
  163. Kosaka, N.; Iguchi, H.; Ochiya, T. Circulating microrna in body fluid: A new potential biomarker for cancer diagnosis and prognosis. Cancer Sci. 2010, 101, 2087–2092. [Google Scholar] [CrossRef] [PubMed]
  164. Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular vesicles: Composition, biological relevance, and methods of study. Bioscience 2015, 65, 783–797. [Google Scholar] [CrossRef] [PubMed]
  165. Mitchell, P.S.; Parkin, R.K.; Kroh, E.M.; Fritz, B.R.; Wyman, S.K.; Pogosova-Agadjanyan, E.L.; Peterson, A.; Noteboom, J.; O’Briant, K.C.; Allen, A.; et al. Circulating micrornas as stable blood-based markers for cancer detection. Proc. Natl. Acad. Sci. USA 2008, 105, 10513–10518. [Google Scholar] [CrossRef] [PubMed]
  166. Roth, C.; Rack, B.; Muller, V.; Janni, W.; Pantel, K.; Schwarzenbach, H. Circulating micrornas as blood-based markers for patients with primary and metastatic breast cancer. Breast Cancer Res. 2010, 12, R90. [Google Scholar] [CrossRef] [PubMed]
  167. Cuk, K.; Zucknick, M.; Heil, J.; Madhavan, D.; Schott, S.; Turchinovich, A.; Arlt, D.; Rath, M.; Sohn, C.; Benner, A.; et al. Circulating micrornas in plasma as early detection markers for breast cancer. Int. J. Cancer 2013, 132, 1602–1612. [Google Scholar] [CrossRef]
  168. Song, C.J.; Chen, H.; Chen, L.Z.; Ru, G.M.; Guo, J.J.; Ding, Q.N. The potential of micrornas as human prostate cancer biomarkers: A meta-analysis of related studies. J. Cell. Biochem. 2018, 119, 2763–2786. [Google Scholar] [CrossRef]
  169. Wang, Y.N.; Chen, Z.H.; Chen, W.C. Novel circulating micrornas expression profile in colon cancer: A pilot study. Eur. J. Med. Res. 2017, 22, 51. [Google Scholar] [CrossRef]
  170. Motawi, T.M.; Sadik, N.A.; Shaker, O.G.; El Masry, M.R.; Mohareb, F. Study of micrornas-21/221 as potential breast cancer biomarkers in egyptian women. Gene 2016, 590, 210–219. [Google Scholar] [CrossRef]
  171. Fattah, H.I.A.; Mahmoud, N.H.; Elzoghby, D.M.; Matar, M.M.; El-Shaer, I.M.M. Clinical utility of circulating microrna-21 in breast cancer. Egypt. J. Hosp. Med. 2018, 71, 2950–2955. [Google Scholar]
  172. Fernando, M.R.; Jiang, C.; Krzyzanowski, G.D.; Ryan, W.L. New evidence that a large proportion of human blood plasma cell-free DNA is localized in exosomes. PLoS ONE 2017, 12, e0183915. [Google Scholar] [CrossRef] [PubMed]
  173. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Harding, C.V.; Heuser, J.E.; Stahl, P.D. Exosomes: Looking back three decades and into the future. J. Cell Biol. 2013, 200, 367–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Xu, R.; Rai, A.; Chen, M.; Suwakulsiri, W.; Greening, D.W.; Simpson, R.J. Extracellular vesicles in cancer—Implications for future improvements in cancer care. Nat. Rev. Clin. Oncol. 2018, 15, 617–638. [Google Scholar] [CrossRef] [PubMed]
  176. Sheridan, C. Exosome cancer diagnostic reaches market. Nat. Biotechnol. 2016, 34, 359–360. [Google Scholar] [CrossRef]
  177. Vanni, I.; Alama, A.; Grossi, F.; Dal Bello, M.G.; Coco, S. Exosomes: A new horizon in lung cancer. Drug Discov. Today 2017, 22, 927–936. [Google Scholar] [CrossRef]
  178. Witwer, K.W.; Buzas, E.I.; Bemis, L.T.; Bora, A.; Lasser, C.; Lotvall, J.; Nolte-’t Hoen, E.N.; Piper, M.G.; Sivaraman, S.; Skog, J.; et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J. Extracell. Vesicles 2013, 2, 20360. [Google Scholar] [CrossRef]
  179. Xu, R.; Greening, D.W.; Zhu, H.J.; Takahashi, N.; Simpson, R.J. Extracellular vesicle isolation and characterization: Toward clinical application. J. Clin. Investig. 2016, 126, 1152–1162. [Google Scholar] [CrossRef]
  180. Costa-Silva, B.; Aiello, N.M.; Ocean, A.J.; Singh, S.; Zhang, H.; Thakur, B.K.; Becker, A.; Hoshino, A.; Mark, M.T.; Molina, H.; et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell. Biol. 2015, 17, 816–826. [Google Scholar] [CrossRef]
  181. Skog, J.; Wurdinger, T.; van Rijn, S.; Meijer, D.H.; Gainche, L.; Sena-Esteves, M.; Curry, W.T., Jr.; Carter, B.S.; Krichevsky, A.M.; Breakefield, X.O. Glioblastoma microvesicles transport rna and proteins that promote tumour growth and provide diagnostic biomarkers. Nat. Cell. Biol. 2008, 10, 1470–1476. [Google Scholar] [CrossRef]
  182. Di Meo, A.; Pasic, M.D.; Yousef, G.M. Proteomics and peptidomics: Moving toward precision medicine in urological malignancies. Oncotarget 2016, 7, 52460–52474. [Google Scholar] [CrossRef] [PubMed]
  183. Pasic, M.D.; Yousef, G.M.; Diamandis, E.P. The proteomic revolution in laboratory medicine. Clin. Biochem. 2013, 46, 397–398. [Google Scholar] [CrossRef] [PubMed]
  184. Abdel Wahab, A.H.A.; El-Halawany, M.S.; Emam, A.A.; Elfiky, A.; Abd Elmageed, Z.Y. Identification of circulating protein biomarkers in patients with hepatocellular carcinoma concomitantly infected with chronic hepatitis c virus. Biomarkers 2017, 22, 621–628. [Google Scholar] [CrossRef] [PubMed]
  185. Alkady, M.M.; Abdel-Messeih, P.L.; Nosseir, N.M. Assessment of serum levels of the adipocytokine chemerin in colorectal cancer patients. J. Med. Biochem. 2018, 37, 313–319. [Google Scholar] [CrossRef] [PubMed]
  186. Aref, S.; Osman, E.; Mansy, S.; Omer, N.; Azmy, E.; Goda, T.; El-Sherbiny, M. Prognostic relevance of circulating matrix metalloproteinase-2 in acute myeloid leukaemia patients. Hematol. Oncol. 2007, 25, 121–126. [Google Scholar] [CrossRef] [PubMed]
  187. Ben Anes, A.; ben Nasr, H.; Hammann, P.; Kuhn, L.; Trimeche, M.; Hamrita, B.; Bougmiza, I.; Chaieb, A.; Khairi, H.; Chahed, K. Assessment of the clinical significance of antigenic and functional levels of α1-proteinase inhibitor (α1-pi) in infiltrating ductal breast carcinomas. Clin. Biochem. 2012, 45, 1421–1431. [Google Scholar] [CrossRef] [PubMed]
  188. El-Bassiouni, N.E.; Nosseir, M.M.; Madkour, M.E.; Zoheiry, M.M.; Bekheit, I.W.; Ibrahim, R.A.; Ibrahim, I.M.; El Bassiouny, A.E. Role of fibrogenic markers in chronic hepatitis c and associated hepatocellular carcinoma. Mol. Biol. Rep. 2012, 39, 6843–6850. [Google Scholar] [CrossRef] [PubMed]
  189. El-Mesallamy, H.O.; Hamdy, N.M.; Zaghloul, A.S.; Sallam, A.M. Clinical value of circulating lipocalins and insulin-like growth factor axis in pancreatic cancer diagnosis. Pancreas 2013, 42, 149–154. [Google Scholar] [CrossRef]
  190. El-Shal, A.S.; Zidan, H.E.; Rashad, N.M.; Wadea, F.M. Angiopoietin-like protein 3 and 4 expression 4 and their serum levels in hepatocellular carcinoma. Cytokine 2017, 96, 75–86. [Google Scholar] [CrossRef]
  191. Eltaher, S.M.; El-Gil, R.; Fouad, N.; Mitwali, R.; El-Kholy, H. Evaluation of serum levels and significance of soluble cd40 ligand in screening patients with hepatitis c virus-related hepatocellular carcinoma. East. Mediterr. Health J. 2016, 22, 603–610. [Google Scholar] [CrossRef]
  192. Hamdy, K.; Al Swaff, R.; Hussein, H.A.; Gamal, M. Assessment of serum adiponectin in egyptian patients with hcv-related cirrhosis and hepatocellular carcinoma. J. Endocrinol. Investig. 2015, 38, 1225–1231. [Google Scholar] [CrossRef] [PubMed]
  193. Hamrita, B.; Nasr, H.B.; Hammann, P.; Kuhn, L.; Guillier, C.L.; Chaieb, A.; Khairi, H.; Chahed, K. An elongation factor-like protein (ef-tu) elicits a humoral response in infiltrating ductal breast carcinomas: An immunoproteomics investigation. Clin. Biochem. 2011, 44, 1097–1104. [Google Scholar] [CrossRef] [PubMed]
  194. M’Hamdi, H.; Baizig, N.M.; OE, E.L.; M’Hamdi, N.; Attia, Z.; Gritli, S.; Gamoudi, A.; El May, M.V.; May, A.E. Usefulness of igf-1 serum levels as diagnostic marker of nasopharyngeal carcinoma. Immunobiology 2016, 221, 1304–1308. [Google Scholar] [CrossRef] [PubMed]
  195. Mohamed, A.A.; Soliman, H.; Ismail, M.; Ziada, D.; Farid, T.M.; Aref, A.M.; Al Daly, M.E.; Abd Elmageed, Z.Y. Evaluation of circulating adh and mic-1 as diagnostic markers in egyptian patients with pancreatic cancer. Pancreatology 2015, 15, 34–39. [Google Scholar] [CrossRef] [PubMed]
  196. Shaarawy, M.; El-Sharkawy, S.A. Biomarkers of intrinsic angiogenic and anti-angiogenic activity in patients with endometrial hyperplasia and endometrial cancer. Acta Oncol. 2001, 40, 513–518. [Google Scholar] [CrossRef] [PubMed]
  197. Shaker, O.G.; Ay El-Deen, M.A.; Abd El-Rahim, M.T.; Talaat, R.M. Gene expression of e-selectin in tissue and its protein level in serum of breast cancer patients. Tumori 2006, 92, 524–530. [Google Scholar] [CrossRef] [PubMed]
  198. Talaat, R.M.; Salem, T.A.; El-Masry, S.; Imbarek, A.; Mokhles, M.; Abdel-Aziz, A. Circulating pro-and anti-angiogenic mediators in patients infected with hepatitis c at different stages of hepatocellular carcinoma. J. Med. Virol. 2014, 86, 1120–1129. [Google Scholar] [CrossRef] [PubMed]
  199. Zekri, A.; Bakr, Y.M.; Ezzat, M.M.; Zakaria, M.; Elbaz, T.M. Circulating levels of adipocytokines as potential biomarkers for early detection of colorectal carcinoma in egyptian patients. Asian Pac. J. Cancer Prev. 2015, 16, 6923–6928. [Google Scholar] [CrossRef] [PubMed]
  200. Zohny, S.F.; Fayed, S.T. Clinical utility of circulating matrix metalloproteinase-7 (mmp-7), cc chemokine ligand 18 (ccl18) and cc chemokine ligand 11 (ccl11) as markers for diagnosis of epithelial ovarian cancer. Med. Oncol. 2010, 27, 1246–1253. [Google Scholar] [CrossRef]
  201. Adeola, H.A.; Blackburn, J.M.; Rebbeck, T.R.; Zerbini, L.F. Emerging proteomics biomarkers and prostate cancer burden in africa. Oncotarget 2017, 8, 37991–38007. [Google Scholar] [CrossRef]
  202. Kinyua, P. Kenya-third African country to use blood-based tests to detect cancer. Jamhuri News, 21 May 2018. [Google Scholar]
  203. Adeola, H.A.; Adefuye, A.O.; Jimoh, S.A. Potential latitudinal variation in orodigestive tract cancers in Africa. S. Afr. Med. J. 2018, 108, 347–351. [Google Scholar] [CrossRef] [PubMed]
  204. Neumann, M.H.D.; Bender, S.; Krahn, T.; Schlange, T. Ctdna and ctcs in liquid biopsy—Current status and where we need to progress. Comput. Struct. Biotechnol. J. 2018, 16, 190–195. [Google Scholar] [CrossRef] [PubMed]
  205. Perakis, S.; Speicher, M.R. Emerging concepts in liquid biopsies. BMC Med. 2017, 15, 75. [Google Scholar] [CrossRef] [PubMed]
  206. Arneth, B. Update on the types and usage of liquid biopsies in the clinical setting: A systematic review. BMC Cancer 2018, 18, 527. [Google Scholar] [CrossRef] [PubMed]
  207. Han, X.; Wang, J.; Sun, Y. Circulating tumor DNA as biomarkers for cancer detection. Egypt. J. Med. Hum. Genet. 2017, 15, 59–72. [Google Scholar] [CrossRef] [PubMed]
  208. Babayan, A.; Pantel, K. Advances in liquid biopsy approaches for early detection and monitoring of cancer. Genome Med. 2018, 10, 21. [Google Scholar] [CrossRef] [PubMed]
  209. Kapeleris, J.; Kulasinghe, A.; Warkiani, M.E.; Vela, I.; Kenny, L.; O’Byrne, K.; Punyadeera, C. The prognostic role of circulating tumor cells (ctcs) in lung cancer. Front. Oncol. 2018, 8, 311. [Google Scholar] [CrossRef] [PubMed]
  210. Tanos, R.; Thierry, A.R. Clinical relevance of liquid biopsy for cancer screening. Transl. Cancer Res. 2018, 7, S105–S129. [Google Scholar] [CrossRef]
  211. Alix-Panabieres, C.; Pantel, K. Real-time liquid biopsy: Circulating tumor cells versus circulating tumor DNA. Ann. Transl. Med. 2013, 1, 18. [Google Scholar] [PubMed]
  212. Gao, Y.; Zhu, Y.; Yuan, Z. Circulating tumor cells and circulating tumor DNA provide new insights into pancreatic cancer. Int. J. Med. Sci. 2016, 13, 902–913. [Google Scholar] [CrossRef]
  213. Heitzer, E.; Perakis, S.; Geigl, J.B.; Speicher, M.R. The potential of liquid biopsies for the early detection of cancer. NPJ Precis. Oncol. 2017, 1, 36. [Google Scholar] [CrossRef] [PubMed]
  214. Azubuike, S.O.; Muirhead, C.; Hayes, L.; McNally, R. Rising global burden of breast cancer: The case of sub-saharan africa (with emphasis on nigeria) and implications for regional development: A. review. World J. Surg. Oncol. 2018, 16, 63. [Google Scholar] [CrossRef] [PubMed]
  215. Chirenje, Z.M.; Rusakaniko, S.; Akino, V.; Mlingo, M. A review of cervical cancer patients presenting in harare and parirenyatwa hospitals in 1998. Cent. Afr. J. Med. 2000, 46, 264–267. [Google Scholar] [CrossRef] [PubMed]
  216. Rambau, P.F.; Chalya, P.L.; Manyama, M.M.; Jackson, K.J. Pathological features of breast cancer seen in northwestern tanzania: A nine years retrospective study. BMC Res. Notes 2011, 4, 214. [Google Scholar] [CrossRef] [PubMed]
  217. Amosu, A.M.; Degun, A.M.; Thomas, A.M.; Babalola, A.O. Assessment of awareness, perception, specific knowledge, and screening behaviour regarding breast cancer among rural women in ipokia local government area, ogun state, nigeria. Arch. Appl. Sci. Res. 2011, 3, 253–265. [Google Scholar]
  218. Clegg-Lamptey, J.; Dakubo, J.; Attobra, Y.N. Why do breast cancer patients report late or abscond during treatment in ghana? A pilot study. Ghana Med. J. 2009, 43, 127–131. [Google Scholar] [PubMed]
  219. Mbuka-Ongona, D.; Tumbo, J.M. Knowledge about breast cancer and reasons for late presentation by cancer patients seen at princess marina hospital, gaborone, botswana. Afr. J. Prim. Health Care Fam. Med. 2013, 5, 465. [Google Scholar] [CrossRef]
Figure 1. The advantages and disadvantages of a tissue biopsy in comparison with a liquid biopsy for cancer diagnosis and treatment. The illustration shows a tumor consisting of heterogeneous cells (represented by different colors). During a tissue biopsy, a small section of tissue is removed; this section may not represent the heterogeneity of the tumor. Tumor cells can undergo epithelial-to-mesenchymal transition (EMT) and enter the blood (CTC). Small molecules are also released from tumor cells into the blood, these include cfDNA, RNA, and exosomes. Tumor-specific alterations in CTCs, cfDNA, RNA, and exosomes found in blood (liquid biopsy) can be utilized to diagnose and treat cancer.
Figure 1. The advantages and disadvantages of a tissue biopsy in comparison with a liquid biopsy for cancer diagnosis and treatment. The illustration shows a tumor consisting of heterogeneous cells (represented by different colors). During a tissue biopsy, a small section of tissue is removed; this section may not represent the heterogeneity of the tumor. Tumor cells can undergo epithelial-to-mesenchymal transition (EMT) and enter the blood (CTC). Small molecules are also released from tumor cells into the blood, these include cfDNA, RNA, and exosomes. Tumor-specific alterations in CTCs, cfDNA, RNA, and exosomes found in blood (liquid biopsy) can be utilized to diagnose and treat cancer.
Cells 08 00862 g001
Table 1. African-based studies on the role of circulating molecules in cancer diagnosis.
Table 1. African-based studies on the role of circulating molecules in cancer diagnosis.
SamplesStudy DesignCancer TypeDownstream Analysis CountryReferences
CTC
Blood75 BC patients
20 healthy controls
Breast cancerCirculating endothelial progenitor cells count
CD14, CD133 and VEGFR2 expression levels (flow cytometry)
EgyptMontaser, et al. [16]
Blood50 BC patients
14 healthy controls
Breast cancermRNA expression levels (qPCR)EgyptElnagdy, et al. [17]
Blood51 BC patientsBreast cancerCTC and CSC count (flow cytometry)EgyptSayed, et al. [18]
Peripheral blood70 HCC patients
30 CHC patients
33 healthy controls
Liver cancerCTC and CSC countProtein expression levels of CK19, CD133, CD90 (flow cytometry)EgyptBahnassy, et al. [19]
Blood50 HCC patients
20 healthy controls
Liver cancerCTC count (flow cytometry)EgyptMansour, et al. [20]
Blood40 BC patientsBreast cancerCTC and CSC count (flow cytometry)EgyptZedan, et al. [21]
Blood50 BC patients
30 healthy controls
Breast cancermRNA expression levels (qPCR)EgyptEbeed, et al. [22]
Blood36 CRC patients
18 healthy controls
Colorectal cancermRNA expression levels (qPCR)EgyptTeama and Agwa [23]
Blood147 BC patients
94 healthy controls
(41 U.S. healthy volunteers)
Breast cancermRNA expression levels (qPCR)SenegalZehentner, et al. [24]
Peripheral blood143 primary melanoma patientsMelanomaThe use of qPCR to determine the presence of tyrosinase mRNA in peripheral bloodSouth AfricaHanekom, et al. [25]
cfDNA
Plasma195 HCC patients
263 CLD control patients
49 healthy controls
Liver cancercfDNA mutational analysis using droplet digital PCRCameroon, Central African RepublicMarchio, et al. [26]
Plasma40 BC patients
10 healthy controls
Breast cancercfDNA quantification and Integrity index using qPCREgyptHussein, et al. [27]
Serum60 LC patients
40 COPD patients
40 healthy controls
Lung cancercfDNA quantification and Integrity index using qPCREgyptSoliman, et al. [28]
Plasma50 PCa patients
25 BPH patients
30 healthy controls
Prostate cancercfDNA quantification and Integrity index using qPCREgyptFawzy, et al. [29]
Serum40 BC patients
40 healthy controls
Breast cancercfDNA quantification using qPCREgyptIbrahim, et al. [30]
Serum50 CRC patients
10 colonic polyps’ patients
20 healthy controls
Colorectal cancercfDNA quantification and Integrity index using qPCREgyptEl-Gayar, et al. [31]
Plasma50 BC patients
30 benign breast lesions
20 healthy controls
Breast cancerQuantification of cfDNA andmtDNA using multiplex qPCREgyptMahmoud, et al. [32]
Plasma120 cancer patients
120 patients with benign diseases
120 healthy controls
Breast, Lung, Colon and Liver cancerscfDNA quantification and Integrity index using qPCREgyptZaher, et al. [33]
Plasma42 BC patients
30 benign lesion patients
27 healthy controls
Breast cancercfDNA quantification and Integrity index using qPCREgyptHashad, et al. [34]
Serum25 HCV-related HCC patients
25 chronic HCV patients
15 healthy controls
Liver cancercfDNA quantification and Integrity index using qPCREgyptEl-Shazly, et al. [35]
Plasma28 HCC patientsLiver cancerMethylation profile determined for five genes using qPCREgyptIyer, et al. [36]
Serum20 NHL patients
20 healthy controls
non-Hodgkin’s lymphomacfDNA quantification using Fluorometric assayEgyptHosny, et al. [37]
Serum76 HCC patients
110 CLD patients
69 healthy controls
Liver cancercfDNA quantification and sequencing of the positive RFLP fragments using nested PCREgyptHosny, et al. [38]
Plasma216 HCC patients
121 liver cirrhosis patients
408 healthy controls
Liver cancercfDNA quantification and sequencing using nested PCRGambiaKirk, et al. [39]
Plasma29 HCC patientsLiver cancercfDNA quantification and sequencing using nested PCRGambiaSzymanska, et al. [40]
Plasma12 PCa patients
10 healthy controls
Prostate cancercfDNA quantification and parallel tagged sequencingSouth Africavan der Vaart, et al. [41]
Plasma1 BC patient
1 healthy control
Breast cancerCloning and sequencing of cfDNASouth Africavan der Vaart and Pretorius [42]
miRNA
Serum65 LC patients
29 pulmonary tuberculosis patients
29 pneumonia
37 healthy controls
Lung cancerExpression levels of miR-21, miR-155, miR-182, and miR-197 assessed using qPCREgyptAbd-El-Fattah, et al. [43]
Serum60 HCV-related HCC patients
60 HCV-related liver cirrhosis patients
60 healthy controls
Liver cancerExpression levels of miRNAs determined using qPCREgyptAli, et al. [44]
Serum60 HCC patients
30 healthy controls
Liver cancerExpression levels of microRNAs 191, 203 and 335 determined using qPCREgyptEzzat, et al. [45]
Plasma45 LC patients
40 healthy controls
Lung cancerThe expression level of miR-21 and miR-23a was detected by qPCREgyptHetta, et al. [46]
Serum60 ovarian cancer patients
30 healthy controls
Ovarian cancerSerum miR-21 levels were measured by TaqMan-qPCREgyptMahmoud, et al. [47]
Serum35 CRC patients
51 patients with benign lesions
101 healthy controls
Colorectal cancerThe expression of miR-210, miR-21 and miR-126 was performed using qPCREgyptSabry, et al. [48]
Serum106 BC patients
49 benign breast lesion patients
40 healthy controls
Breast CancerThe expression level of miR-335 was detected by qPCREgyptSwellam, et al. [49]
Serum137 BC patients
60 benign breast lesion patients
38 healthy controls
Breast cancermiRNAs expression levels were determined using reaction qPCREgyptSwellam, et al. [50]
Serum30 HCC patients
20 healthy controls
Liver cancerlncRNA GAS5 and miR-34a expression level measured using qPCREgyptToraih, et al. [51]
Blood9 CHC patients
6 liver cirrhosis patients
9 HCC patients
8 healthy controls
Liver cancermiRNAs expression levels were determined using reaction qPCREgyptZekri, et al. [52]
Plasma60 HCC patients
60 CHC patients
60 healthy controls
Liver cancermiRNA expression levels assessed using qPCREgyptDemerdash, et al. [53]
Serum224 HCC patients
250 CHC patients
84 healthy controls
Liver cancermiRNAs (hsa-miR-1269, hsa-miR-125b, hsa-miR-138, hsa-miR-214-5p, hsa-miR-494, hsa-miR-375 and hsa-miR-145) were assessed using qPCREgyptElemeery, et al. [54]
Plasma65 AML patients
50 healthy controls
Acute myeloid leukemiaExpression of miR-92a, miR-143 and miR-342 was measured using qPCREgyptElhamamsy, et al. [55]
Serum64 CRC patients
27 healthy controls
Colorectal cancerExpression levels of miR-92a, miR-375, and miR-760 assessed using qPCREgyptElshafei, et al. [56]
Serum23 HCC patients
25 post-HCV liver cirrhosis patients
30 HCV patients
10 healthy controls
Liver cancermiRNA expression levels using qPCREgyptKhairy, et al. [57]
Plasma70 bladder cancer patients
62 healthy controls
Bladder cancerExpression levels of miR-92a, miR-100 and miR-143 measured using qPCREgyptMotawi, et al. [58]
Serum60 HCC patients
40 CHC patients
30 healthy controls
Liver cancerExpression levels of miRNA-122 and miRNA-222 assessed using qPCREgyptMotawi, et al. [59]
Peripheral blood mononuclear cells85 ALL patients
25 healthy controls
Acute lymphoblastic leukemiaExpression levels of miR-92a, miR-100 and miR-143 were measured using qPCREgyptSwellam and El-Khazragy [60]
Serum30 CRC patients
18 IBD patients
18 colonic polyps’ patients
24 colonic symptoms patients
100 CRC patients (validation)
Colorectal cancermiRNAs expression levels were determined using reaction qPCREgyptZekri, et al. [61]
Blood30 HCC patients
20 HCV patients
20 healthy controls
Liver cancermiRNA expression levels assessed using qPCREgyptAlnoanmany, et al. [62]
Urine188 Bladder cancer patients
88 Benign bladder lesions
92 healthy controls
Bladder cancermiR-210, miR-10b, miR-29c, miR-221, and miR-23a expression levels assessed using qPCREgyptEissa, et al. [63]
Serum40 HCC patients
40 HCV patients
20 Healthy controls
Liver cancermiRNA expression levels using qPCREgyptEl-Abd, et al. [64]
Serum120 BC patients
50 healthy controls
Breast cancerExpression levels of miRNAs (miR10b, miR34a, miR155, miR195 and miR16) determined using qPCREgyptHagrass, et al. [65]
Serum112 HCV-related HCC patients
125 HCV-related CLD patients
42 healthy controls
Liver cancerExpression miRNA was measured using qPCREgyptMotawi, et al. [66]
Urine32 HCC patients with post-HCV infection
74 chronic HCV patients
12 healthy controls
Liver cancermiRNA whole-genome expression profiling and relative expression profiling for candidate miRNAs using qPCREgyptAbdalla and Haj-Ahmad [67]
Serum20 Inflammatory BC patients
20 non-inflammatory BC patients
20 healthy controls
Breast cancerTaqMan qPCR was performed to detect the circulating expression of miRNAsTunisiaHamdi, et al. [68]
mRNA
Serum40 HCC patients
10 healthy controls
Liver cancermRNA expression levels using qPCREgyptAbdelgawad, et al. [69]
Serum25 HCC patients
15 healthy controls
Liver cancermRNA expression levels using qPCREgyptIbrahim, et al. [70]
lncRNAs
Serum80 BC patients
80 healthy controls
Breast cancermRNA expression levels using qPCREgyptZidan, et al. [71]
Serum120 CRC patients
30 adenomatous polyps’ patients
96 healthy controls
Colorectal cancerSerum expression levels of lncRNAs and miRNA using qPCREgyptShaker, et al. [72]
Serum78 HCC patients
36 CHC patients
44 healthy controls
Liver cancermRNA expression levels using qPCREgyptEl-Tawdi, et al. [73]
Plasma32 gastric cancer patients
30 healthy controls
Gastric cancermRNA expression levels using qPCREgyptHashad, et al. [74]
Exosomes
Serum60 HCC patients
42 CHC patients
18 healthy controls
Liver cancerExpression of exosomal RNA using qPCREgyptAbd El Gwad, et al. [75]
Serum20 LC patientsLung cancerExpression of exosomal RNA using qPCREgyptKhalil, et al. [76]
Abbreviations: ALL—Acute lymphoblastic leukemia, AML—Acute myeloid leukemia, BC—Breast cancer, BPH—Benign prostatic hyperplasia, CHC—Chronic hepatitis C, CLD—Chronic liver disease, COPD—Chronic obstructive pulmonary disease, CRC—Colorectal cancer, CSC—Cancer stem cell, HCC—Hepatocellular carcinoma, HCV—Hepatitis C-Virus, IBD—Inflammatory bowel disease, LC—Lung cancer, mtDNA—mitochondrial DNA, NHL—Non-Hodgkin’s lymphoma, PCa—Prostate cancer, PC—Pancreatic cancer, qPCR—Quantitative real-time PCR.
Table 2. Comparison of the circulating biomarkers, CTC, cfDNA, circulating tumor RNA and Exosomes in cancer management.
Table 2. Comparison of the circulating biomarkers, CTC, cfDNA, circulating tumor RNA and Exosomes in cancer management.
Analysis CapabilityCTCcfDNACirculating Tumor RNAExosomes
Genomic mutationsYesYesYesYes
RNA profilingYesNoYesYes
Phenotypic studies of tumor cellYesNoNoNo
Proteomic analysisYesNoNoYes
Clinical Applications
Clinical trialsPhase IVPhase IVPhase IVPhase II
Clinical approved techniquesCellSearch
  • Cobas® EGFR Mutation Test v2 assay
  • Epi proColon test
  • AmoyDx Super-ARMS EGFR mutation test
  • Therascreen EGFR RGQ Plasma PCR kit
  • Therascreen PIK3CA RGQ PCR Kit
  • Sysmex Inostics OncoBEAM RAS CRC Kit
  • Idylla™ ctKRAS Mutation Test
  • Idylla™ ctNRAS-BRAF mutation test
Progensa™ PCA3No
Cost of clinical use (outside Africa)$350$170–470$220-

Share and Cite

MDPI and ACS Style

Temilola, D.O.; Wium, M.; Coulidiati, T.H.; Adeola, H.A.; Carbone, G.M.; Catapano, C.V.; Zerbini, L.F. The Prospect and Challenges to the Flow of Liquid Biopsy in Africa. Cells 2019, 8, 862. https://doi.org/10.3390/cells8080862

AMA Style

Temilola DO, Wium M, Coulidiati TH, Adeola HA, Carbone GM, Catapano CV, Zerbini LF. The Prospect and Challenges to the Flow of Liquid Biopsy in Africa. Cells. 2019; 8(8):862. https://doi.org/10.3390/cells8080862

Chicago/Turabian Style

Temilola, Dada Oluwaseyi, Martha Wium, Tangbadioa Herve Coulidiati, Henry Ademola Adeola, Giuseppina Maria Carbone, Carlo Vittorio Catapano, and Luiz Fernando Zerbini. 2019. "The Prospect and Challenges to the Flow of Liquid Biopsy in Africa" Cells 8, no. 8: 862. https://doi.org/10.3390/cells8080862

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