Recovering circulating extracellular or cell-free RNA from bodily fluids

10
Recovering circulating extracellular or cell-free RNA from bodily fluids Georgios Tzimagiorgis a, *, Evangelia Z. Michailidou b , Aristidis Kritis c , Anastasios K. Markopoulos b , Sofia Kouidou a a Laboratory of Biological Chemistry, Medical School, Aristotle University of Thessaloniki, 540 06 Thessaloniki, Greece b Department of Oral Medicine and Maxillofacial Pathology, Aristotle University, Thessaloniki, Greece c Laboratory of Physiology, Medical School, Aristotle University, Thessaloniki, Greece 1. Introduction Despite the advances in cancer therapeutic approaches, during the last decades, the morbidity and mortality rates in cancer still remain high [1]. The earliest possible diagnosis and treatment is still the best approach to improve survival rates to the disease [2]. The National Cancer Institute estimates that premature deaths, which may have been avoided through screening, range from 3% to 35% [3]. Screening for cancer is usually attempted whenever worrying symptoms arise, having as a result the diagnosis of cancer as a late stage disease [4]. The current methods for diagnosis of the disease are usually invasive (e.g. PAP smear, colonoscopy, etc.) and expensive whereas the existing biological markers are not definitive and lack high sensitivity and specificity [5]. Many researchers therefore work on developing new, sensitive and inexpensive methods for cancer diagnostics [6]. Detection of extracellular or cell-free nucleic acids (DNA or RNA) in plasma, serum and other bodily fluids using the PCR or RT-PCR methods have been suggested as non invasive and cost effective methods for cancer detection [7]. The first report on nucleic acid detection in plasma, was back in 1972 [8] and since then numerous are the reports on detection of cancer specific mutations in circulating DNA or RNA in plasma [9], as well as in other bodily fluids [10–12]. More recently, mRNA biomarkers in serum, plasma and other bodily fluids have been analyzed performing reverse transcription- PCR (RT-PCR)-based detection strategies in patients with cancer [13]. Parallel to the increasing number of such biomarkers in bodily fluids is the growing availability of technologies that enable mass screening programs [14]. Detection of mRNA markers compared to conventional biochemical methods, seems to increase specificity, sensitivity and timeliness of the cancer screening method. 2. Origin of circulating extracellular or cell-free nucleic acids Extracellular or cell-free nucleic acids (DNA or mRNA) have been isolated from various bodily fluids and used as biological markers for various diseases among which is cancer [15–18]. Several terms are used for these extracellular nucleic acids such as: (1) Circulating nucleic acids, mainly used for DNA and RNA, circulating in plasma or serum (2) Extracellular, or (3) Cell-free nucleic acids, when isolated from other body fluids such as: saliva, urine, cerebrospinal fluid (CSF), bronchoalveolar lavage fluid (BALF) amniotic fluid and other. Several different mechanisms have been implicated as a potential source for the circulating extracellular or cell-free nucleic acids. One possible mechanism is cell necrosis resulting Cancer Epidemiology 35 (2011) 580–589 A R T I C L E I N F O Article history: Received 16 April 2010 Received in revised form 28 February 2011 Accepted 28 February 2011 Available online 22 April 2011 Keywords: Extracellular or cell-free RNA Cancer detection Biological fluids RNA stabilizing reagents RNA isolation mRNA miRNA A B S T R A C T The presence of extracellular circulating or cell-free RNA in biological fluids is becoming a promising diagnostic tool for non invasive and cost effective cancer detection. Extracellular RNA or miRNA as biological marker could be used either for the early detection and diagnosis of the disease or as a marker of recurrence patterns and surveillance. In this review article, we refer to the origin of the circulating extracellular RNA, we summarise the data on the biological fluids (serum/plasma, saliva, urine, cerebrospinal fluid and bronchial lavage fluid) of patients suffering from various types of malignancies reported to contain a substantial amount of circulating extracellular (or cell-free) RNAs and we discuss the appropriate reagents and methodologies needed to be employed in order to obtain RNA material of high quality and integrity for the majority of the experimental methods used in RNA expression analysis. Furthermore, we discuss the advantages and disadvantages of the RT-PCR or microarray methodology which are the methods more often employed in procedures of extracellular RNA analysis. ß 2011 Elsevier Ltd. All rights reserved. * Corresponding author. Tel.: +30 2310999122; fax: +30 2310999004. E-mail address: [email protected] (G. Tzimagiorgis). Contents lists available at ScienceDirect Cancer Epidemiology The International Journal of Cancer Epidemiology, Detection, and Prevention jou r nal h o mep age: w ww.c an cer ep idem io log y.n et 1877-7821/$ see front matter ß 2011 Elsevier Ltd. All rights reserved. doi:10.1016/j.canep.2011.02.016

Transcript of Recovering circulating extracellular or cell-free RNA from bodily fluids

Recovering circulating extracellular or cell-free RNA from bodily fluids

Georgios Tzimagiorgis a,*, Evangelia Z. Michailidou b, Aristidis Kritis c, Anastasios K. Markopoulos b,Sofia Kouidou a

a Laboratory of Biological Chemistry, Medical School, Aristotle University of Thessaloniki, 540 06 Thessaloniki, Greeceb Department of Oral Medicine and Maxillofacial Pathology, Aristotle University, Thessaloniki, Greecec Laboratory of Physiology, Medical School, Aristotle University, Thessaloniki, Greece

1. Introduction

Despite the advances in cancer therapeutic approaches, duringthe last decades, the morbidity and mortality rates in cancer stillremain high [1]. The earliest possible diagnosis and treatment isstill the best approach to improve survival rates to the disease [2].The National Cancer Institute estimates that premature deaths,which may have been avoided through screening, range from 3% to35% [3]. Screening for cancer is usually attempted wheneverworrying symptoms arise, having as a result the diagnosis of canceras a late stage disease [4]. The current methods for diagnosis of thedisease are usually invasive (e.g. PAP smear, colonoscopy, etc.) andexpensive whereas the existing biological markers are notdefinitive and lack high sensitivity and specificity [5]. Manyresearchers therefore work on developing new, sensitive andinexpensive methods for cancer diagnostics [6]. Detection ofextracellular or cell-free nucleic acids (DNA or RNA) in plasma,serum and other bodily fluids using the PCR or RT-PCR methodshave been suggested as non invasive and cost effective methods forcancer detection [7]. The first report on nucleic acid detection inplasma, was back in 1972 [8] and since then numerous are the

reports on detection of cancer specific mutations in circulatingDNA or RNA in plasma [9], as well as in other bodily fluids [10–12].

More recently, mRNA biomarkers in serum, plasma and otherbodily fluids have been analyzed performing reverse transcription-PCR (RT-PCR)-based detection strategies in patients with cancer[13]. Parallel to the increasing number of such biomarkers in bodilyfluids is the growing availability of technologies that enable massscreening programs [14].

Detection of mRNA markers compared to conventionalbiochemical methods, seems to increase specificity, sensitivityand timeliness of the cancer screening method.

2. Origin of circulating extracellular or cell-free nucleic acids

Extracellular or cell-free nucleic acids (DNA or mRNA) havebeen isolated from various bodily fluids and used as biologicalmarkers for various diseases among which is cancer [15–18].Several terms are used for these extracellular nucleic acids such as:(1) Circulating nucleic acids, mainly used for DNA and RNA,circulating in plasma or serum (2) Extracellular, or (3) Cell-freenucleic acids, when isolated from other body fluids such as: saliva,urine, cerebrospinal fluid (CSF), bronchoalveolar lavage fluid(BALF) amniotic fluid and other.

Several different mechanisms have been implicated as apotential source for the circulating extracellular or cell-freenucleic acids. One possible mechanism is cell necrosis resulting

Cancer Epidemiology 35 (2011) 580–589

A R T I C L E I N F O

Article history:

Received 16 April 2010

Received in revised form 28 February 2011

Accepted 28 February 2011

Available online 22 April 2011

Keywords:

Extracellular or cell-free RNA

Cancer detection

Biological fluids

RNA stabilizing reagents

RNA isolation

mRNA

miRNA

A B S T R A C T

The presence of extracellular circulating or cell-free RNA in biological fluids is becoming a promising

diagnostic tool for non invasive and cost effective cancer detection. Extracellular RNA or miRNA as

biological marker could be used either for the early detection and diagnosis of the disease or as a marker

of recurrence patterns and surveillance. In this review article, we refer to the origin of the circulating

extracellular RNA, we summarise the data on the biological fluids (serum/plasma, saliva, urine,

cerebrospinal fluid and bronchial lavage fluid) of patients suffering from various types of malignancies

reported to contain a substantial amount of circulating extracellular (or cell-free) RNAs and we discuss

the appropriate reagents and methodologies needed to be employed in order to obtain RNA material of

high quality and integrity for the majority of the experimental methods used in RNA expression analysis.

Furthermore, we discuss the advantages and disadvantages of the RT-PCR or microarray methodology

which are the methods more often employed in procedures of extracellular RNA analysis.

� 2011 Elsevier Ltd. All rights reserved.

* Corresponding author. Tel.: +30 2310999122; fax: +30 2310999004.

E-mail address: [email protected] (G. Tzimagiorgis).

Contents lists available at ScienceDirect

Cancer EpidemiologyThe International Journal of Cancer Epidemiology, Detection, and Prevention

jou r nal h o mep age: w ww.c an cer ep idem io log y.n et

1877-7821/$ – see front matter � 2011 Elsevier Ltd. All rights reserved.

doi:10.1016/j.canep.2011.02.016

in the presence of high amounts of DNA (and probably RNA) inthe plasma of cancer patients with large or advanced tumors[19,20]. Apoptosis has also been considered as another possiblesource of circulating cell-free nucleic acids. Circulating DNAanalyzed by electrophoresis often exhibits a typical ladder offragmentation similar to that shown by apoptotic cells [21–23].Apoptotic bodies come from aged or damaged cells. Halicka et al.demonstrated that during apoptosis the nucleic acids, DNA andRNA, are packaged separately into two types of apoptotic bodies,one type contains only RNA and no detectable levels of DNA andthe other type contains only DNA and no detectable RNA [24].Furthermore, it was shown that extracellular, mRNA coding forthe enzyme tyrosinase which is associated with apoptoticbodies, is protected, from degradation in human serum [25].These finding suggests that extracellular RNA, circulating withinapoptotic bodies, is less susceptible to ribonucleases’ activities[5,25].

Spontaneous, active release of DNA and RNA by tumors inmicrovesicles is another possibility. Both normal but also cancercells release microvesicles which are spherical membrane frag-ments [26] and come from the surface of the cell or the endosomalmembranes. Microvesicles are released from viable cells and areusually smaller in size than the apoptotic bodies. Microvesicles arereported to be a normal constituent of human plasma [27]transferring proteins and RNA and contain no fragmented DNA[5,27–41]. In addition, lysis of cancer cells, trauma [5,42] andtherapeutic procedures [43–46] might also contribute to circulat-ing nucleic acids.

3. Biological fluids containing circulating extracellular (or cell-free) RNAs

3.1. Plasma/serum

The first reports on increased circulating nucleic acids – bothDNA and RNA – in the serum of cancer patients were back in 1977[47]. Circulating DNA and RNA in the serum of cancer patients wereisolated and characterized as tumor-derived and tumor-specific inthe late 1980s [23,19]. At first the cancer specific mRNA existing inserum was thought to come from circulating cancer cells [48]. Theamount of circulating nucleic acids however cannot be justified bythe number of cancer cells existing in serum [9], leading to thehypothesis that tumor specific circulating or cell-free nucleic acidscome from the necrosis [19,20] or apoptosis [24,25] of cancer cellsor they are even actively released by them in microvesicles[26,27,29].

Circulating RNA has been isolated from the plasma or serum ofpatients suffering from various types of malignancies such asbreast cancer [49–56], lung cancer [57–63], prostate cancer [64–78], thyroid cancer [79–98], hepatocellular carcinoma [5,72,75,99–112], melanoma [113–116], gastric cancer [117], renal cell cancer[118], oesophageal cancer [119,120], rectal carcinoma [121],gynecologic malignancies [122], pancreatic cancer [61], coloncancer [61], bladder cancer [16] and solid tumors [123], and hasbeen used as a biological marker either for the early detection anddiagnosis of the disease [64,100], or as a marker of recurrencepatterns [81,121], survival predictor [51], follow up and surveil-lance marker [79,82].

In particular, in breast cancer, measurement of serummetastasin mRNA has been proposed as a screening tool,predicting poor survival and distant metastases [51], whereasmeasurement of circulating 5T4 mRNA as having the potential toidentify patients who could benefit from a 5T4-targetedtherapy [53]. Moreover, the measurement of circulating Dkk-1 levels in breast cancer patients could predict bonemetastases [56].

In lung cancer, serum hTERT mRNA together with EGFR mRNAwas proposed as a good biomarker for diagnosis and clinical stageassessment [58].

In prostate cancer, serum PCA3 mRNA detection [64], HIF-1alpha mRNA quantitative RT-PCR [65], and serum E2F3 mRNAmeasurement [67], are proposed for the diagnosis of the disease.Furthermore, circulating PSA mRNA was proposed either as apreoperative prognostic marker for organ-confined or locallyadvanced prostate cancer [71], or as a tool to detect potentialsurgical failures pre-operatively [77] and monitor patients withmetastatic prostate cancer [69].

In thyroid cancer, peripheral thyrotropin hormone receptor(TSHR) mRNA has been proposed for cancer detection in patientswith thyroid nodules [87], or in indeterminate fine-needleaspiration [83] or for predicting recurrence [79]. Moreover someauthors suggest measurement of serum thyroglobulin mRNA indetecting thyroid cancer [87] or monitoring metastatic disease[89], whereas others do not support this idea [90,91].

In hepatocellular carcinoma, Dong et al. suggested thatmeasuring circulating plasma hepatic TGF-beta1 might proveuseful in the diagnosis and prognosis of HBV-induced hepatocel-lular carcinoma [100]. Wu et al. proposed hepatoma-specificalpha-fetoprotein mRNA (HS-AFP mRNA) and circulating alpha-fetoprotein (AFP)-mRNA [103] and Dong et al., insulin-like growthfactor II (IGF-II) mRNA [107] as markers to diagnose hepatocellularcarcinoma, monitor metastasis and relapse.

In esophageal squamous cell carcinoma, preoperative squa-mous cell carcinoma–antigen messenger RNA (SCC–Ag mRNA)levels in the peripheral blood are proposed as predictive factor forrecurrence [120].

The detection of circulating fetal RNA (placenta derived) in thematernal circulation has been tested in prenatal diagnostics as anon-invasive procedure compared to amniocentesis, chorionicvillus sampling and cordocentesis which are for the moment theroutine methods. The first to show that fetal RNA is present in theplasma of pregnant women was Poon in 2000 [124]. Ng et al. in2003 [125] showed that placenta-derived RNA coding for humanplacental lactogen and the b-subunit of human chorionicgonadotrophin and corticotrophin-releasing hormone (CRH)[126] are detectable using Real-time PCR in the maternal plasma.Detection of placental circulating RNA in the maternal plasma hasalso been proposed for the early diagnosis of several pregnancycomplications such as preeclampsia [127] and chromosomalaneuploidies [128]. These markers have been shown to beindependent of gender and polymorphisms [129]. Moreover,Oudejans et al. in 2003 [130], detected placenta-derived mRNAspecific for chromosome 21, whereas Wataganara et al. in 2004[131] detected fetal-derived gamma-globin mRNA in the maternalplasma. Further more in 2004, Tsui et al. [132] using expressionmicroarrays identified new mRNA species detectable in maternalplasma that can be used in the future as markers in prenataldiagnostics. Establishing these markers in the clinical diagnosticroutine is the next step to be taken.

Cell-free DNA was demonstrated to increase in the circulationof patients after acute trauma giving its measurement a possibleprognostic ability [133]. On this basis, there was an attempt tomeasure glyceraldehyde-3-phosphate dehydrogenase mRNA inthe plasma of trauma patients which was found significantlyincreased to that of healthy individuals when plasma was filteredthrough a 0.22 mm pore size filter [134]. Orlandi et al. attempted tomeasure circulating mRNA for nephrin in healthy volunteers andrenal transplantation patients [135]. They found that mRNA fornephrin reduces significantly with age and transplantationprobably due to the reduced renal mass. Circulating mRNA fornephrin was found however higher in females something whichmust be taken into account when accessing the results of cell-free

G. Tzimagiorgis et al. / Cancer Epidemiology 35 (2011) 580–589 581

mRNA for nephrin in plasma. Circulating cell-free RNA was alsodetected in the plasma of patients suffering from diabeticretinopathy in diabetes mellitus [136]. Two are the majorcomplications in diabetes mellitus, retinopathy and neuropathy.Circulating rhodopsin mRNA [137,138] and neuron-specificenolase mRNA [139] were found to significantly follow theseverity of the retinopathy.

3.2. Saliva

Saliva is the bodily fluid secreted in the oral cavity as theproduct of a set of 3 major salivary glands (the parotid,submandibular, and sublingual gland) and the numerous minorsalivary glands lying beneath the oral mucosa. It contains variousproteins, microorganisms, blood cells, desquamated epithelialcells, cell free-DNA and RNA and other components [140], forexample a breast cancer marker protein, c-erb-B-2, is reported tobe found in stratified saliva fractions [141,142]. RNA in the salivamay come from: (1) the secretions of the salivary glands (by theacinar cells or by the circulation [143], (2) passive diffusion fromserum to saliva across cell membranes [141,142], (3) desquamatedoral epithelial cells, (4) apoptotic oral epithelial cells [5,25], (5)blood cells or directly as cell-free RNA from blood released in salivathrough the crevicular fluid or microwounds, (6) cell death ortrauma and (7) even active release from epithelial or cancerouscells [26,27]. For years however RNA was thought to quicklydegrade in saliva due to the numerous RNases present in the saliva[144,145]. It was therefore a surprise that RNA molecules werediscovered intact in the saliva [18,146–162]. Detection of salivaryRNA has been used extensively in Forensic Medicine for body fluididentification [146–153], for the early diagnosis of oral squamouscell carcinoma [155,156], as genomic biomarker for primarySjogren’s syndrome [157], as a biomarker for the identification ofsleep drive in flies and humans [158]. Despite the opposite reports[159] salivary RNA generally appears to show stability andconsistency and has the potential to be used as a biomarker fororal cancer and possibly other diseases [143,154,160–162].

3.3. Urine

Numerous are the reports on the detection of various mRNAs inurine [163] for the diagnosis or surveillance of prostate [164,165]and bladder cancer [16,176–195], since urine is the bodily fluiddraining these tumors. Muthukumar et al. proposed the measure-ment of messenger RNA for FOXP3 in the urine of renal-allograftrecipients as a non invasive method for predicting acute rejectionof renal transplants [196]. Lately however, two reports on mRNAmarkers in urine concerning colorectal cancer [197] and metastaticmelanoma [198] have been published and raise the question ofhow these cancer specific mRNAs were present in urine.

PCA 3 (prostate cancer antigen 3) is the main cancer specificmRNA marker reported to have been detected in urine and hasbeen suggested for the diagnosis [164–171] and active surveillance[172,173] of prostate cancer. Molecular urine tests measuring theprostate cancer antigen 3 (RNA detection) are even commerciallyavailable [174,175]. Detection of PSA mRNA [165], TMPRSS2-ERGfusion [170] transcripts and Golgi protein GOLM1 [173] in urine asbiomarkers for prostate cancer have been also sporadicallyreported.

The detection of survivin, a member of the inhibitor of apoptosisfamily [176–181], cytokeratin 20 [177,182–185] and humantelomerase (hTERT) [186–191] mRNAs in urine have been reportedand used for the diagnosis and prognosis [16,177,182,194] ofbladder cancer or as a detection, characterisation and recurrencemarker [175,176,178–181,183–185,187–193,195] for the samedisease. The use of mucin 7 [177], hTERT [192], ETS2/urokinase

plasminogen activator [193], CD4 [183] and UHRF1 [194] mRNAshas been also reported as molecular markers for the diagnosis ofbladder cancer.

3.4. Cerebrospinal fluid (CSF)

In a study by de Bont the mRNA levels of insulin-like growthfactor-binding proteins, IGFBP-2 and IGFBP-3 were found signifi-cantly increased in paediatric medulloblastoma patients and theIGFBP-5 and IGF-II mRNA levels in paediatric ependymomapatients [199].

3.5. Bronchial lavage fluid

Free mRNA in bronchial lavage fluid has been suggested as anew diagnostic tool in lung cancer detection by Schmidt et al. in astudy published in 2005 [60].

3.6. miRNA

All the above mentioned studies refer to total and exogenousRNA or mRNA. Lately however there are some reports in theliterature concerning miRNA (both endogenous and exogenous) inplasma, serum, saliva and urine [200] that can be used asbiomarkers for the detection, monitoring and prognosis of cancer[201–221] and other diseases [217,222–226] or for body fluididentification [227]. Fully processed miRNAs are small RNAmolecules, approximately 18–24 nucleotides in length[220,228], which bind to mRNA and seem to regulate itstranscription [216]. miRNA can be isolated using the samemethods which are mentioned later in this document for theisolation of mRNA.

3.7. RNA stabilising reagents

RNA in biological samples and especially cell-free RNA in bodilyfluids is extremely unstable, degrading quickly after the samplecollection [229]. Several RNases are present in water, buffers, handsmear and various bodily fluids (tears, saliva and perspiration),which quickly degrade RNA in biological samples, cell cultures andexperiments. Therefore, the use of a method for stabilizing mRNAis important in order to ensure the isolation of a high qualitymRNA, preserving the gene expression profile.

Various methods have been used for the stabilisation of mRNA.These in general stabilise mRNA and help with handling thesamples at room temperature as well as in long term storage at�80 8C (archiving).

The first RNase inhibitor was isolated from placenta back in1979 [230]. Since then, several types of RNase inhibitors areavailable from many companies enabling RNA purification,transcription, translation, RT-PCR reactions, cDNA synthesis, etc.The various RNase inhibitors offered vary in the temperatures andpH at which they are active, the mechanism of their action, thetypes of RNases they block such as RNase A, B, C, T1 and S1 nucleaseand the need for dithiothreitol (DDT) addition for activity. Usuallythese RNase inhibitors do not block Taq polymerase, reversetranscriptase, RNA polymerase and other enzymes important forthe experiments. Some inhibitors are to be used for RNA isolation(RNAlater by Ambion, Qiagen, Superase-in by Ambion) (Table 1),and others are most effective as prophylactics during enzymaticreactions (Ribonuclease Inhibitor (Recombinant) by Affymetrix,RNasin1 Plus RNase Inhibitor and Recombinant RNasin1 Ribonu-clease Inhibitor by Promega, RNasin, Electrophoresis by SBSGenetech, RNaseOUTTM Recombinant Ribonuclease Inhibitor).

The most widely used RNase inhibitors are the placentalribonuclease inhibitors (RIPs) (Ribonuclease Inhibitor (Human

G. Tzimagiorgis et al. / Cancer Epidemiology 35 (2011) 580–589582

Placenta) – Affymetrix/USB, Ribonuclease inhibitor from humanplacenta – AppliChem, Placenta Ribonuclease Inhibitor – BioChain,etc.), which however are effective only against RNase A typeenzymes (RNAse A, B, C, human placental RNase) and not RNases 1,T1, T2, H, U1, U2 or CL3.

The type of RNase inhibitor used is dependent on theapplication. For example, Wong discusses upon RNAlater as asuitable RNase inhibitor for saliva that ‘‘RNAlater increases cellularmembrane permeability and its high salt content (ammoniumsulfate) changes intracellular salt concentration, leading to leakageof cellular components, including genomic DNA. RNAlatercompared with a number of RNA stabilizers was found to be theleast suitable, even worse than without any stabilizer’’ [231].

4. Methods for RNA isolation and analysis

4.1. Guanidinium-based protocols

The most commonly used techniques for rapid purification ofRNA are the methods employed guanidinium–acid–phenol extrac-tions [232–234]. RNA lysis buffers containing the chaotropicagents, guanidinium thiocyanate or guanidinium HCl, lead to theisolation of a very high quality and integrity of RNA. These reagentsare among the strongest protein denaturants and immediatelyinactivate RNases in the lysate. In all cases, protein denaturation(including disruption of RNases) is enhanced by the addition of theb-mercaptoethanol (b-ME). b-ME acts as a reducing agentbreaking intramolecular protein bisulfide bonds. The partitioningof RNA, DNA and protein in the resulting lysate is accomplished byacid–phenol extraction The RNA is then precipitated withisopropanol. Although the addition of exogenous RNase inhibitorsto eliminate both exogenous and endogenous nucleases is notrequired, the use of an RNase inhibitor in the RNA solutionenhances the stability of the stored material, and is highlyrecommended (see above).

4.2. Silica-columns based protocols

Recently, the development of new techniques employing silicacolumns for the isolation of nucleic acids provided new tools in theisolation of high quality extracellular RNA (Qiagen, Manual of viralRNA mini kit, Qiagen) [235]. This method is based on the formationof complexes between nucleic acids and the detergent (silica gelmatrix) in the presence of chaotropic salts. These complexes can beprecipitated by centrifugation to form a pellet. The resuspendedpellet, containing the desired RNA, is then treated with proteinaseK solution. The silica-based methods have several benefits, are lesslaborious and time-consuming, need less manipulation, involve nopellet handling, and therefore result to a reproducible high RNAyield [236]. All these advantages make the silica methods moreapplicable for routine molecular diagnostic methodology. In allcases, an internal control RNA can be added in the beginning of anyRNA isolation procedure in order to evaluate the method itself andmake a decision on which method is more suitable in yourresearch.

4.3. Isolation of DNA-free RNA from the samples

In all RNA experiments and particular in reverse transcription-polymerase chain reaction) (RT-PCR), it is crucial to eliminate anyresidual amount of DNA. Due to the sensitivity of PCR reaction, thepresence of genomic DNA contamination in an RNA preparationmay lead to ambiguous or misleading results. This problem isexaggerated when the analysis is performed using extracellularRNA, due to the small amounts of RNA obtained. To avoid thisproblem, it is highly recommended that RNA preparations betreated with DNase I (RNase-free) prior to cDNA synthesis[236,237], and that control reactions be performed in whichreverse transcriptase is omitted [238]. Furthermore, it is alsorecommended that all PCR primer pairs should be designed in amanner that the sequence for each primer may be located inadjacent exons of the gene of interest. This will allow thedistinction on the basis of size of the PCR products resultingeither from the amplification of the cDNA or from contaminatinggenomic DNA if there relatively small differences in the size [239].

4.4. Evaluation of the RNA quality obtained from the different samples

Although, there are a number of methods employed to obtainhigh quality RNA from a variety of sources, the isolation of highquality and integrity, extracellular RNA, is a very difficult task.Sample handling and variations in RNA isolation procedures mightaffect quality of the isolated and therefore could introduce errorsinto the analysis process. Therefore, and in order to minimize theseerrors, is necessary to standardize the input amount of RNA (orcDNA) and to simultaneously measure the levels of expression ofan endogenous RNA, which is expressed more or less in a constantmanner between the different samples or to employ an externalstandard [240,241].

The cellular genes used to normalize for the variability ofclinical samples or the integrity of the isolated nucleic acid in RNAprocedures, are genes involved in the regulation of basic andubiquitous cellular functions, and they are members of thehousekeeping gene family. They most widely used of them codefor components of the cytoskeleton (b-actin), major histocompat-ibility complex (b-2-microglobulin), glycolytic pathway (glycer-aldehyde-3-phosphate dehydrogenase, GAPDH andphosphoglycerokinase 1), metabolic salvage of nucleotides (hypo-xanthine ribosyltransferase, HPRT1), or synthesis of ribosomesubunits (rRNA) [240,241].

First of all, in the case of extracellular or circulating RNA, it iscrucial to evaluate the integrity of the isolated mRNA. This can besimply assessed by electrophoresis. In the cases, when a smallquantity of RNA is available this can be performed by PCRamplification using the selected control genes. We can postulatethat at least two or three of these genes can be selected in each casein order to obtain the best gene(s) standards. The choice can besimply done based on the biological sample used and the level ofexpression of the gene(s) of interest. In general, it was demon-strated that when only a small amount of RNA is available and thegene of interest exhibits a high level of expression, the high-expression GAPDH is the most suitable as an endogenous control

Table 1RNase inhibitors that are commonly used to stabilise RNA in the sample.

Agilent technologies Ambion Bioline Sigma-Aldrich Qiagen

RNase block RNAlater1 RiboSafe RNase inhibitor ProtectRNATM RNase inhibitor RNAlater

RNAlater1-ICE RNAprotect cell

SUPERaseIn RNAprotect saliva

Paxgene blood RNA

G. Tzimagiorgis et al. / Cancer Epidemiology 35 (2011) 580–589 583

gene with b-actin as a second choice [242]. Furthermore, de Koket al. argues that when the gene of interest has intermediate or lowexpression the best choice for the endogenous control gene is theHPRT1 gene, a low-copy housekeeping gene, which in combinationwith GAPDH, gives the best results in order to estimate RNAisolation efficiency, RNA quality, and RT-efficiency [243]. Otherstudies revealed that b-2 microglobulin and 18S rRNA are the mostsuitable internal control genes in quantitative serum-stimulationstudies, while b-actin and GAPDH are not so efficient [244]. On theother hand, some other studies proposed that the expression of theHPRT gene more accurately reflects the mean expression ofmultiple commonly used housekeeping genes [244]. Taking intoaccount all these considerations, we conclude that the internalcontrol gene(s) needs to be properly evaluated in order to selectthe most appropriate ones in a particular study.

4.5. RT-PCR or microarrays: advantages and disadvantages of

methods that can be used in extracellular RNA analysis

Analysis of gene expression, in general, can be done by severaldifferent methods including Northern blotting, RNase protectionassays, serial analysis of gene expression (SAGE), RT-PCR (andlately qRT-PCR), as well as with the microarray technology[245,246]. Among these techniques, although Northern blotanalysis and RNase protection assays are the ideal methods forthe measurement of gene expression/mRNA levels both require theuse of intact RNA, which rarely is the case in isolated extracellularRNA. On the other hand, the SAGE analysis is a highly complicatedand laborious technique, which also requires high quality RNA.Thus, the methods of choice to detect RNA species e.g. in plasma orserum is the techniques of RT-PCR or microarray analysis.

Microarray experiments allow for comparison of gene expres-sion profiles between two mRNA samples. There are twomicroarray platforms in common use cDNA microarrays, whichutilize cloned probe molecules corresponding to characterizedexpressed sequences, and oligonucleotide microarrays, made ofsynthetic probe sequences based on database information [247].Microarrays are quite commonly used and are usually consistentwith data obtained from northern blots. The major advantage thatmicroarrays have over all the other methods used in RNAexpression analysis is that thousands of genes can be visualizedat a time with a microarray, while all other techniques are usuallylooking at one or a small number of genes [247].

Real-time PCR is a type of quantitative PCR that can be used tocompare normal (control) samples to disease’s samples, providingclues as to the gene expression changes that occur in pathogenesis.The advent of real-time quantitative PCR (qRT-PCR) has eliminatedthe variability traditionally associated with quantitative PCR, thusallowing the routine and reliable quantitation of PCR products. Dueto its sensitivity, qRT-PCR, is the method of choice in order toquantify mRNA levels from even very small numbers of cells andlately even single cells or small amounts of biological fluids.Several advantages of this method over the microarrays are worthmentioning e.g. the low cost, the need of less expensive equipment,the relatively easy sample analysis. As with microarray analysis, inqRT-PCR, there is no need to work with intact RNA. Thus, althoughboth methods can be easily employed, the simplicity and thesensitivity of the qRT-PCR remain the major advantages.

5. Advantages and disadvantages of analyzing circulatingextracellular RNA instead of intracellular RNA

The analysis of extracellular circulating RNA is a powerful toolin order to evaluate a number of different pathological conditions,since it has proven to provide valuable information on theexpression pattern of several candidate genes [143,147]. Further-

more, the analysis is not limited to the RNA material released fromcirculating ‘blood’ cells but is also able to detect all circulating RNAoriginated from different ‘‘damaged’’ or ‘‘lysed’’ tissue cells, oftenexhibiting a malignant phenotype [5,19,20,42]. Moreover, theanalysis of cell free circulating RNA can provide information for thepresence of any abnormality without the need of material obtainedfrom biopsies, in the same way plasma/serum protein and enzymeanalysis is used in the diagnosis of several diseases andpathophysiological conditions. Furthermore, the analysis ofcirculating extracellular RNA can be employed easily in massscreening of high risk population [100,155,156] and in monitoringof metastases in cancer patients [51,69,89]. Additionally, theanalysis of extracellular RNA is the method of choice in order toavoid tedious and time consuming methods for the recruitment ofa number of circulating malignant cells in the blood in order toisolate and characterize the intracellular RNA content. Along thesame line, biopsies, the major source of intracellular RNA for geneexpression analysis studies, have as a prerequisite the presence of adisease diagnosis.

Despite the above-mentioned advantages, there are severaldisadvantages in working with extracellular RNA. The majority ofthese disadvantages arise from the low quality/quantity of theisolated RNA. Low RNA quality is a major obstacle in the analysis ofextracellular RNA. The isolated RNA if often degraded or fragmentedin small parts. As a consequence, different regions of the sametranscript might be represented differentially in the isolated RNA. Toovercome this problem there is a need to design carefully the PCRprimers in order to obtain the desired result. Often, there is a need todesign primer pairs detecting different regions of the transcriptunder investigation. In addition, and in order to have a roughestimation of the RNA quantity, it is necessary to use simultaneouslyas internal control, several genes (b-actin, b2m, GAPDH, 18S rRNA,etc.) [240,241]. Usually, the presence of amplification of at least twoof them in an RT-PCR reaction is required in order to select thesamples to be included in the study. Low RNA quantity, althoughmight be a potential problem, can be easily solved using real time RT-PCR protocols which are providing increased sensitivity. On thecontrary, the analysis of intracellular RNA, which is the alternativemethod, although it is free from all the above mentioneddisadvantages, request in all cases an invasive procedure (biopsy,fine needle aspiration, etc.) and in several circumstances to have adisease diagnosis.

6. Conclusions

Circulating extracellular or cell-free RNA obtained from avariety of bodily fluids can became a valuable tool in earlydiagnosis of several diseases including cancer. The major obstaclein order to be applicable for routine molecular diagnosticmethodology is to ensure the high quality and the integrity ofthe isolated RNA. In order to develop a reproducible methodologyof diagnostic value care should be taken in the use of the properRNA stabilizer in the clinical sample, in choosing of the appropriateRNA isolation procedure, ensuring the absence of any DNAcontamination, and finally, in the use of the most suitableendogenous housekeeping control genes, factors which arediscussed in detail in this article.

Conflict of interest statement

There is no conflict of interest.

References

[1] American Cancer Society. Cancer facts and figures 2009. Atlanta, GA: Ameri-can Cancer Society, 2009.

G. Tzimagiorgis et al. / Cancer Epidemiology 35 (2011) 580–589584

[2] Welch HG, Schwartz LM, Woloshin S. Are increasing 5-year survival ratesevidence of success against cancer? JAMA 2000;283(22):2975–8.

[3] National Cancer Institute. www.cancer.gov.[4] Ellison LF, Wilkins K. Cancer prevalence in the Canadian population. Health

Rep 2009;20(1):7–19.[5] El-Hefnawy T, Raja S, Kelly L, Bigbee WL, Kirkwood JM, Luketich JD, et al.

Characterization of amplifiable, circulating RNA in plasma and its potential asa tool for cancer diagnostics. Clin Chem 2004;50(3):564–73.

[6] Goessl C. Diagnostic potential of circulating nucleic acids for oncology. ExpertRev Mol Diagn 2003;3(4):431–42 [Review].

[7] Fleischhacker M, Schmidt B. Circulating nucleic acids (CNAs) and cancer – asurvey. Biochim Biophys Acta 2007;1775(1):181–232.

[8] Kamm RC, Smith AG. Nucleic acid concentrations in normal human plasma.Clin Chem 1972;18(6):519–22.

[9] Goebel G, Zitt M, Zitt M, Muller HM. Circulating nucleic acids in plasma orserum (CNAPS) as prognostic and predictive markers in patients with solidneoplasias. Dis Markers 2005;21(3):105–20.

[10] Haas C, Klesser B, Maake C, Bar W, Kratzer A. mRNA profiling for body fluididentification by reverse transcription endpoint PCR and realtime PCR. Fo-rensic Sci Int Genet 2009;3(2):80–8.

[11] O’Driscoll L. Extracellular nucleic acids and their potential as diagnostic,prognostic and predictive biomarkers. Anticancer Res 2007;27(3A):1257–65.

[12] Chan AK, Chiu RW, Lo YM. Cell-free nucleic acids in plasma, serum and urine:a new tool in molecular diagnosis. Ann Clin Biochem 2003;40(Pt 2):122–30.

[13] Sidransky D. Emerging molecular markers of cancer. Nat Rev Cancer2002;2(3):210–9.

[14] Kramer BS. The science of early detection. Urol Oncol 2004;22(4):344–7.[15] Zhou H, Xu W, Qian H, Yin Q, Zhu W, Yan Y. Circulating RNA as a novel tumor

marker: an in vitro study of the origins and characteristics of extracellularRNA. Cancer Lett 2008;259(1):50–60.

[16] Lodde M, Fradet Y. The detection of genetic markers of bladder cancer in urineand serum. Curr Opin Urol 2008;18(5):499–503.

[17] Taback B, Hoon DS. Circulating nucleic acids and proteomics of plasma/serum: clinical utility. Ann N Y Acad Sci 2004;1022:1–8.

[18] Zhang L, Farrell JJ, Zhou H, Elashoff D, Akin D, Park NH, et al. Salivarytranscriptomic biomarkers for detection of resectable pancreatic cancer.Gastroenterology 2010;138(3):949–57.

[19] Stroun M, Anker P, Maurice P, Lyautey J, Lederrey C, Beljanski M, et al.Neoplastic characteristics of the DNA found in the plasma of cancer patients.Oncology 1989;46(5):318–22.

[20] Li C, Hsu H, Wu T, Tsao KC, Sun CF, Wu JT. Cell-free DNA is released fromtumor cells upon cell death: a study of tissue cultures of tumor cell lines. J ClinLab Anal 2003;17(4):103–7.

[21] Giacona M, Ruben G, Iczkowski K, Roos T, Porter D, Sorenson G. Cell-free DNAin human blood plasma: length measurements in patients with pancreaticcancer and healthy controls. Pancreas 1998;17(1):89–97.

[22] Fournie G, Courtin J, Laval F, Chale JJ, Pourrat JP, Pujazon MC, et al. PlasmaDNA as a marker of cancerous cell death. Investigations in patients sufferingfrom lung cancer and in nude mice bearing human tumours. Cancer Lett1995;91(2):221–7.

[23] Stroun M, Anker P, Lyautey J, Lederey C, Maurice PA. Isolation and character-ization of DNA from the plasma of cancer patients. Eur J Cancer Clin Oncol1987;23(6):707–12.

[24] Halicka HD, Bedner E, Darzynkiewicz Z. Segregation of RNA and separatepackaging of DNA and RNA in apoptotic bodies during apoptosis. Exp Cell Res2000;260(2):248–56.

[25] Hasselmann D, Rappl G, Tilgen W, Reinhold U. Extracellular tyrosinase mRNAwithin apoptotic bodies is protected from degradation in human serum. ClinChem 2001;47(8):1488–9.

[26] Ratajczak J, Wysoczynski M, Hayek F, Janowska-Wieczorek A, Ratajczak MZ.Membrane-derived microvesicles: important and under appreciated media-tors of cell-to-cell communication. Leukemia 2006;20(9):1487–95.

[27] Yuan A, Farber EL, Rapoport AL, Tejada D, Deniskin R, Akhmedov NB, et al.Transfer of microRNAs by embryonic stem cell microvesicles. PLoS One2009;4(3):e4722.

[28] Aharon A, Brenner B. Microparticles, thrombosis and cancer. Best Pract ResClin Haematol 2009;22(1):61–9. Review.

[29] Skog J, Wurdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, et al.Glioblastoma microvesicles transport RNA and proteins that promote tumourgrowth and provide diagnostic biomarkers. Nat Cell Biol 2008;10(12):1470–6.

[30] Hunter MP, Ismail N, Zhang X, Aguda BD, Lee EJ, Yu L, et al. Detection ofmicroRNA expression in human peripheral blood microvesicles. PLoS One2008;3(11):e3694.

[31] Giusti I, D’Ascenzo S, Millimaggi D, Taraboletti G, Carta G, Franceschini N,et al. Cathepsin B mediates the pH-dependent proinvasive activity of tumor-shed microvesicles. Neoplasia 2008;10(5):481–8.

[32] Garcıa JM, Garcıa V, Pena C, Domınguez G, Silva J, Diaz R, et al. Extracellularplasma RNA from colon cancer patients is confined in a vesicle-like structureand is mRNA-enriched. RNA 2008;14(7):1424–32.

[33] Baj-Krzyworzeka M, Szatanek R, Weglarczyk K, Baran J, Zembala M. Tumour-derived microvesicles modulate biological activity of human monocytes.Immunol Lett 2007;113(2):76–82.

[34] Ratajczak J, Miekus K, Kucia M, Zhang J, Reca R, Dvorak P, et al. Embryonicstem cell-derived microvesicles reprogram hematopoietic progenitors: evi-

dence for horizontal transfer of mRNA and protein delivery. Leukemia2006;20(5):847–56.

[35] Baj-Krzyworzeka M, Szatanek R, Weglarczyk K, Baran J, Urbanowicz B,Branski P, et al. Tumour-derived microvesicles carry several surface deter-minants and mRNA of tumour cells and transfer some of thesedeterminants to monocytes. Cancer Immunol Immunother 2006;55(7):808–18.

[36] Janowska-Wieczorek A, Wysoczynski M, Kijowski J, Marquez-Curtis L,Machalinski B, Ratajczak J, et al. Microvesicles derived from activated plate-lets induce metastasis and angiogenesis in lung cancer. Int J Cancer2005;113(5):752–60.

[37] Stroun M, Anker P. Nucleic acids spontaneously released by living frogauricles. Biochem J 1972;128(3):100P–1P.

[38] Anker P, Stroun M, Maurice PA. Spontaneous release of DNA by human bloodlymphocytes as shown in an in vitro system. Cancer Res 1975;35(9):2375–82.

[39] Anker P, Stroun M, Maurice PA. Spontaneous extracellular synthesis of DNAreleased by human blood lymphocytes. Cancer Res 1976;36(8):2832–9.

[40] Stroun M, Anker P, Maurice P, Gahan PB. Circulating nucleic acids in higherorganisms. Int Rev Cytol 1977;51:1–48.

[41] Anker P, Stroun M, Maurice P. Characteristics of nucleic acids excreted bynon-stimulated normal human lymphocytes. Schweiz Med Wochenschr1977;107(41):1457.

[42] Laktionov PP, Tamkovich SN, Rykova EY, Bryzgunova OE, Starikov AV, Kuz-netsova NP, et al. Extracellular circulating nucleic acids in human plasma inhealth and disease. Nucleosides Nucleotides Nucleic Acids 2004;23(6–7):879–83.

[43] Ross JS, Slodkowska EA, Symmans WF, Pusztai L, Ravdin PM, Hortobagyi GN.The HER-2 receptor and breast cancer: ten years of targeted anti-HER-2therapy and personalized medicine. Oncologist 2009;14(4):320–68.

[44] Ross JS. Breast cancer biomarkers and HER2 testing after 10 years of anti-HER2 therapy. Drug News Perspect 2009;22(2):93–106.

[45] Davis G, Davis J. Detection of circulating DNA by counterimmunoelectro-phoresis (CIE). Arthritis Rheum 1973;16(1):52–8.

[46] Hughes G, Cohen S, Lightfoot R, Meltzer J, Christian C. The release of DNA intoserum and synovial fluid. Arthritis Rheum 1971;14(2):259–66.

[47] Leon SA, Shapiro B, Sklaroff DM, Yaros MJ. Free DNA in the serum of cancerpatients and the effect of therapy. Cancer Res 1977;37(3):646–50.

[48] Johnson PW, Burchill SA, Selby PJ. The molecular detection of circulatingtumour cells. Br J Cancer 1995;72(2):268–76.

[49] Savino M, Parrella P, Copetti M, Barbano R, Murgo R, Fazio VM, et al.Comparison between real-time quantitative PCR detection of HER2 mRNAcopy number in peripheral blood and ELISA of serum HER2 protein fordetermining HER2 status in breast cancer patients. Cell Oncol2009;31(3):203–11.

[50] Marrakchi R, Ouerhani S, Benammar S, Rouissi K, Bouhaha R, Bougatef K, et al.Detection of cytokeratin 19 mRNA and CYFRA 21-1 (cytokeratin 19 frag-ments) in blood of Tunisian women with breast cancer. Int J Biol Markers2008;23(4):238–43.

[51] El-Abd E, El-Tahan R, Fahmy L, Zaki S, Faid W, Sobhi A, et al. Serum metastasinmRNA is an important survival predictor in breast cancer. Br J Biomed Sci2008;65(2):90–4.

[52] Perhavec A, Cerkovnik P, Novakovic S, Zgajnar J. The hTERT mRNA in plasmasamples of early breast cancer patients, non-cancer patients and healthyindividuals. Neoplasma 2008;55(6):549–54.

[53] Kopreski MS, Benko FA, Gocke CD. Circulating RNA as a tumor marker:detection of 5T4 mRNA in breast and lung cancer patient serum. Ann N YAcad Sci 2001;945:172–8.

[54] Chen XQ, Bonnefoi H, Pelte MF, Lyautey J, Lederrey C, Movarekhi S, et al.Telomerase RNA as a detection marker in the serum of breast cancer patients.Clin Cancer Res 2000;6(10):3823–6.

[55] O’Driscoll L, Kenny E, Mehta JP, Doolan P, Joyce H, Gammell P, et al. Feasibilityand relevance of global expression profiling of gene transcripts in serum frombreast cancer patients using whole genome microarrays and quantitative RT-PCR. Cancer Genomics Proteomics 2008;5(2):94–104.

[56] Voorzanger-Rousselot N, Goehrig D, Journe F, Doriath V, Body JJ, Clezardin P,et al. Increased Dickkopf-1 expression in breast cancer bone metastases. Br JCancer 2007;97(7):964–70.

[57] Sugai S, Satoh Y, Komatsu M, Okumura S, Nakagawa K, Ishikawa Y, et al.Recurrence pattern and rapid intraoperative detection of carcinoembryonicantigen (CEA) mRNA in pleural lavage in patients with non-small cell lungcancer (NSCLC). Rinsho Byori 2008;56(10):851–7.

[58] Miura N, Nakamura H, Sato R, Tsukamoto T, Harada T, Takahashi S, et al.Clinical usefulness of serum telomerase reverse transcriptase (hTERT) mRNAand epidermal growth factor receptor (EGFR) mRNA as a novel tumor markerfor lung cancer. Cancer Sci 2006;97(12):1366–73.

[59] Bremnes RM, Sirera R, Camps C. Circulating tumour-derived DNA and RNAmarkers in blood: a tool for early detection, diagnostics, and follow-up? LungCancer 2005;49(1):1–12.

[60] Schmidt B, Engel E, Carstensen T, Weickmann S, John M, Witt C, et al.Quantification of free RNA in serum and bronchial lavage: a new diagnostictool in lung cancer detection? Lung Cancer 2005;48(1):145–7.

[61] Clarke LE, Leitzel K, Smith J, Ali SM, Lipton A. Epidermal growth factorreceptor mRNA in peripheral blood of patients with pancreatic, lung, andcolon carcinomas detected by RT-PCR. Int J Oncol 2003;22(2):425–30.

G. Tzimagiorgis et al. / Cancer Epidemiology 35 (2011) 580–589 585

[62] Fleischhacker M, Beinert T, Ermitsch M, Seferi D, Possinger K, Engelmann C,et al. Detection of amplifiable messenger RNA in the serum of patients withlung cancer. Ann N Y Acad Sci 2001;945:179–88.

[63] Johnson PJ, Lo YM. Plasma nucleic acids in the diagnosis and management ofmalignant disease. Clin Chem 2002;48(8):1186–93.

[64] Neves AF, Araujo TG, Biase WK, Meola J, Alcantara TM, Freitas DG, et al.Combined analysis of multiple mRNA markers by RT-PCR assay for prostatecancer diagnosis. Clin Biochem 2008;41(14–15):1191–8.

[65] Pipinikas CP, Carter ND, Corbishley CM, Fenske CD. HIF-1alpha mRNA geneexpression levels in improved diagnosis of early stages of prostate cancer.Biomarkers 2008;13(7):680–91.

[66] Bai VU, Kaseb A, Tejwani S, Divine GW, Barrack ER, Menon M, et al. Identifi-cation of prostate cancer mRNA markers by averaged differential expressionand their detection in biopsies, blood, and urine. Proc Natl Acad Sci U S A2007;104(7):2343–8.

[67] Pipinikas CP, Nair SB, Kirby RS, Carter ND, Fenske CD. Measurement of bloodE2F3 mRNA in prostate cancer by quantitative RT-PCR: a preliminary study.Biomarkers 2007;12(5):541–57.

[68] Zambon CF, Basso D, Prayer-Galetti T, Navaglia F, Fasolo M, Fogar P, et al.Quantitative PSA mRNA determination in blood: a biochemical tool forscoring localized prostate cancer. Clin Biochem 2006;39(4):333–8.

[69] Patel K, Whelan PJ, Prescott S, Brownhill SC, Johnston CF, Selby PJ, et al. Theuse of real-time reverse transcription-PCR for prostate-specific antigenmRNA to discriminate between blood samples from healthy volunteersand from patients with metastatic prostate cancer. Clin Cancer Res2004;10(22):7511–9.

[70] Siddiqua A, Chendil D, Rowland R, Meigooni AS, Kudrimoti M, Mohiuddin M,et al. Increased expression of PSA mRNA during brachytherapy in peripheralblood of patients with prostate cancer. Urology 2002;60(2):270–5.

[71] Straub B, Muller M, Krause H, Schrader M, Goessl C, Heicappell R, et al.Detection of prostate-specific antigen RNA before and after radical retropubicprostatectomy and transurethral resection of the prostate using ‘‘Light-Cycler’’-based quantitative real-time polymerase chain reaction. Urology2001;58(5):815–20.

[72] Pirisi M, Fabris C, Luisi S, Santuz M, Toniutto P, Vitulli D, et al. Evaluation ofcirculating activin-A as a serum marker of hepatocellular carcinoma. CancerDetect Prev 2000;24(2):150–5.

[73] Ghossein RA, Osman I, Bhattacharya S, Ferrara J, Fazzari M, Cordon-Cardo C,et al. Detection of prostatic specific membrane antigen messenger RNA usingimmunobead reverse transcriptase polymerase chain reaction. Diagn MolPathol 1999;8(2):59–65.

[74] Ogawa O, Iinuma M, Sato K, Sasaki R, Shimoda N, Satoh S, et al. Circulatingprostate-specific antigen mRNA during radical prostatectomy in patientswith localized prostate cancer: with special reference to neoadjuvant hor-monal therapy. Urol Res 1999;27(4):291–6.

[75] Ishikawa T, Kashiwagi H, Iwakami Y, Hirai M, Kawamura T, Aiyoshi Y, et al.Expression of alpha-fetoprotein and prostate-specific antigen genes in sev-eral tissues and detection of mRNAs in normal circulating blood by reversetranscriptase-polymerase chain reaction. Jpn J Clin Oncol 1998;28(12):723–8.

[76] Castaldo G, Cecere G, di Fusco V, Prezioso D, d’Armiento M, Salvatore F.Prostate-specific antigen (protein and mRNA) analysis in the differentialdiagnosis and staging of prostate cancer. Clin Chim Acta 1997;265(1):65–76.

[77] Olsson CA, De Vries GM, Benson MC, Raffo A, Buttyan R, Cama C, et al. The useof RT-PCR for prostate-specific antigen assay to predict potential surgicalfailures before radical prostatectomy: molecular staging of prostate cancer.Br J Urol 1996;77(3):411–7.

[78] Papadopoulou E, Davilas E, Sotiriou V, Georgakopoulos E, Georgakopoulou S,Koliopanos A, et al. Cell-free DNA and RNA in plasma as a new molecularmarker for prostate and breast cancer. Ann N Y Acad Sci 2006;1075:235–43.

[79] Milas M, Barbosa GF, Mitchell J, Berber E, Siperstein A, Gupta M. Effectivenessof peripheral thyrotropin receptor mRNA in follow-up of differentiatedthyroid cancer. Ann Surg Oncol 2009;16(2):473–80.

[80] Coelho SM, Buescu A, Corbo R, Carvalho DP, Vaisman M. Recurrence ofpapillary thyroid cancer suspected by high anti-thyroglobulin antibody levelsand detection of peripheral blood thyroglobulin mRNA. Arq Bras EndocrinolMetabol 2008;52(8):1321–5.

[81] Lombardi CP, Bossola M, Princi P, Boscherini M, La Torre G, Raffaelli M, et al.Circulating thyroglobulin mRNA does not predict early and midterm recur-rences in patients undergoing thyroidectomy for cancer. Am J Surg2008;196(3):326–32.

[82] Barbosa GF, Milas M. Peripheral thyrotropin receptor mRNA as a novelmarker for differentiated thyroid cancer diagnosis and surveillance. ExpertRev Anticancer Ther 2008;8(9):1415–24.

[83] Gupta M, Chia SY. Circulating thyroid cancer markers. Curr Opin EndocrinolDiabetes Obes 2007;14(5):383–8.

[84] Chia SY, Milas M, Reddy SK, Siperstein A, Skugor M, Brainard J, et al. Thyroid-stimulating hormone receptor messenger ribonucleic acid measurement inblood as a marker for circulating thyroid cancer cells and its role in thepreoperative diagnosis of thyroid cancer. J Clin Endocrinol Metab2007;92(2):468–75.

[85] Harish K. Thyroglobulin: current status in differentiated thyroid carcinoma.Endocr Regul 2006;40(2):53–67.

[86] Ishikawa T, Miwa M, Uchida K. Quantitation of thyroid peroxidase mRNA inperipheral blood for early detection of thyroid papillary carcinoma. Thyroid2006;16(5):435–42.

[87] Chinnappa P, Taguba L, Arciaga R, Faiman C, Siperstein A, Mehta AE, et al.Detection of thyrotropin-receptor messenger ribonucleic acid (mRNA) andthyroglobulin mRNA transcripts in peripheral blood of patients with thyroiddisease: sensitive and specific markers for thyroid cancer. J Clin EndocrinolMetab 2004;89(8):3705–9.

[88] Novakovic S, Hocevar M, Zgajnar J, Besic N, Stegel V. Detection of telomeraseRNA in the plasma of patients with breast cancer, malignant melanoma orthyroid cancer. Oncol Rep 2004;11(1):245–52.

[89] Grammatopoulos D, Elliott Y, Smith SC, Brown I, Grieve RJ, Hillhouse EW, et al.Measurement of thyroglobulin mRNA in peripheral blood as an adjunctivetest for monitoring thyroid cancer. Mol Pathol 2003;56(3):162–6.

[90] Denizot A, Delfino C, Dutour-Meyer A, Fina F, Ouafik L. Evaluation of quanti-tative measurement of thyroglobulin mRNA in the follow-up of differentiatedthyroid cancer. Thyroid 2003;13(9):867–72.

[91] Eszlinger M, Neumann S, Otto L, Paschke R. Thyroglobulin mRNA quantifica-tion in the peripheral blood is not a reliable marker for the follow-up ofpatients with differentiated thyroid cancer. Eur J Endocrinol2002;147(5):575–82.

[92] Fugazzola L, Mihalich A, Persani L, Cerutti N, Reina M, Bonomi M, et al. Highlysensitive serum thyroglobulin and circulating thyroglobulin mRNA evalua-tions in the management of patients with differentiated thyroid cancer inapparent remission. J Clin Endocrinol Metab 2002;87(7):3201–8.

[93] Bellantone R, Lombardi CP, Bossola M, Ferrante A, Princi P, Boscherini M, et al.Validity of thyroglobulin mRNA assay in peripheral blood of postoperativethyroid carcinoma patients in predicting tumor recurrences varies accordingto the histologic type: results of a prospective study. Cancer2001;92(9):2273–9.

[94] Bojunga J, Dragan C, Schumm-Draeger PM, Usadel KH, Kusterer K. Circulatingcalcitonin and carcinoembryonic antigen m-RNA detected by RT-PCR astumour markers in medullary thyroid carcinoma. Br J Cancer2001;85(10):1546–50.

[95] Biscolla RP, Cerutti JM, Maciel RM. Detection of recurrent thyroid cancer bysensitive nested reverse transcription–polymerase chain reaction of thyro-globulin and sodium/iodide symporter messenger ribonucleic acid tran-scripts in peripheral blood. J Clin Endocrinol Metab 2000;85(10):3623–7.

[96] Bojunga J, Roddiger S, Stanisch M, Kusterer K, Kurek R, Renneberg H, et al.Molecular detection of thyroglobulin mRNA transcripts in peripheral blood ofpatients with thyroid disease by RT-PCR. Br J Cancer 2000;82(10):1650–5.

[97] Ringel MD, Balducci-Silano PL, Anderson JS, Spencer CA, Silverman J, SparlingYH, et al. Quantitative reverse transcription-polymerase chain reaction ofcirculating thyroglobulin messenger ribonucleic acid for monitoring patientswith thyroid carcinoma. J Clin Endocrinol Metab 1999;84(11):4037–42.

[98] Ringel MD, Ladenson PW, Levine MA. Molecular diagnosis of residual andrecurrent thyroid cancer by amplification of thyroglobulin messenger ribo-nucleic acid in peripheral blood. J Clin Endocrinol Metab 1998;83:4435–42.

[99] Kijima H, Sawada T, Tomosugi N, Kubota K. Expression of hepcidin mRNA isuniformly suppressed in hepatocellular carcinoma. BMC Cancer 2008;8:167.

[100] Dong ZZ, Yao DF, Yao M, Qiu LW, Zong L, Wu W, et al. Clinical impact ofplasma TGF-beta1 and circulating TGF-beta1 mRNA in diagnosis of hepato-cellular carcinoma. Hepatobiliary Pancreat Dis Int 2008;7(3):288–95.

[101] Miura N, Maruyama S, Oyama K, Horie Y, Kohno M, Noma E, et al. Develop-ment of a novel assay to quantify serum human telomerase reverse tran-scriptase messenger RNA and its significance as a tumor marker forhepatocellular carcinoma. Oncology 2007;72(Suppl 1):45–51.

[102] Lu Y, Wu LQ, Lu ZH, Wang XJ, Yang JY. Expression of SSX-1 and NY-ESO-1mRNA in tumor tissues and its corresponding peripheral blood expression inpatients with hepatocellular carcinoma. Chin Med J (Engl)2007;120(12):1042–6.

[103] Wu W, Yao DF, Yuan YM, Fan JW, Lu XF, Li XH, et al. Combined serumhepatoma-specific alpha-fetoprotein and circulating alpha-fetoprotein-mRNA in diagnosis of hepatocellular carcinoma. Hepatobiliary PancreatDis Int 2006;5(4):538–44.

[104] Yao DF, Wu W, Yao M, Qiu LW, Wu XH, Su XQ, et al. Dynamic alteration oftelomerase expression and its diagnostic significance in liver or peripheralblood for hepatocellular carcinoma. World J Gastroenterol2006;12(31):4966–72.

[105] Wu LQ, Lu Y, Wang XF, Lv ZH, Zhang B, Yang JY. Expression of cancer-testisantigen (CTA) in tumor tissues and peripheral blood of Chinese patients withhepatocellular carcinoma. Life Sci 2006;79(8):744–8.

[106] Wu W, Yao DF, Qiu LW, Wu XH, Yao M, Su XQ, et al. Abnormal expression ofhepatomas and circulating telomerase and its clinical values. HepatobiliaryPancreat Dis Int 2005;4(4):544–9.

[107] Dong ZZ, Yao DF, Yao DB, Wu XH, Wu W, Qiu LW, et al. Expression andalteration of insulin-like growth factor II-messenger RNA in hepatomatissues and peripheral blood of patients with hepatocellular carcinoma.World J Gastroenterol 2005;11(30):4655–60.

[108] Yao F, Guo JM, Xu CF, Lou YL, Xiao BX, Zhou WH, et al. Detecting AFP mRNA inperipheral blood of the patients with hepatocellular carcinoma, liver cirrho-sis and hepatitis. Clin Chim Acta 2005;361(1–2):119–27.

[109] Ng EK, Tsui NB, Lam NY, Chiu RW, Yu SC, Wong SC, et al. Presence of filterableand nonfilterable mRNA in the plasma of cancer patients and healthyindividuals. Clin Chem 2002;48(8):1212–7.

[110] Mou DC, Cai SL, Peng JR, Wang Y, Chen HS, Pang XW, et al. Evaluation ofMAGE-1 and MAGE-3 as tumour-specific markers to detect blood dissemi-nation of hepatocellular carcinoma cells. Br J Cancer 2002;86(1):110–6.

G. Tzimagiorgis et al. / Cancer Epidemiology 35 (2011) 580–589586

[111] Barbu V, Bonnand AM, Hillaire S, Coste T, Chazouilleres O, Gugenheim J, et al.Circulating albumin messenger RNA in hepatocellular carcinoma: results of amulticenter prospective study. Hepatology 1997;26(5):1171–5.

[112] Matsumura M, Niwa Y, Kato N, Komatsu Y, Shiina S, Kawabe T, et al. Detectionof alpha-fetoprotein mRNA, an indicator of hematogenous spreading hepa-tocellular carcinoma, in the circulation: a possible predictor of metastatichepatocellular carcinoma. Hepatology 1994;20(6):1418–25.

[113] Osella-Abate S, Quaglino P, Savoia P, Leporati C, Comessatti A, Bernengo MG.VEGF-165 serum levels and tyrosinase expression in melanoma patients:correlation with the clinical course. Melanoma Res 2002;12(4):325–34.

[114] Rappl G, Hasselmann DO, Rossler M, Ugurel S, Tilgen W, Reinhold U. Detec-tion of tumor-associated circulating mRNA in patients with disseminatedmalignant melanoma. Ann N Y Acad Sci 2001;945:189–91.

[115] Quereux G, Denis M, Khammari A, Lustenberger P, Dreno B. Prognostic valueof tyrosinase reverse-transcriptase polymerase chain reaction in metastaticmelanoma. Dermatology 2001;203(3):221–5.

[116] Hasselmann DO, Rappl G, Rossler M, Ugurel S, Tilgen W, Reinhold U. Detec-tion of tumor-associated circulating mRNA in serum, plasma and blood cellsfrom patients with disseminated malignant melanoma. Oncol Rep2001;8(1):115–8.

[117] Ishigami S, Sakamoto A, Uenosono Y, Nakajo A, Okumura H, Matsumoto M,et al. Carcinoembryonic antigen messenger RNA expression in blood canpredict relapse in gastric cancer. J Surg Res 2008;148(2):205–9.

[118] Feng G, Li G, Gentil-Perret A, Tostain J, Genin C. Elevated serum-circulatingRNA in patients with conventional renal cell cancer. Anticancer Res2008;28(1A):321–6.

[119] Yang YF, Li H, Xu XQ, Diao YT, Fang XQ, Wang Y, et al. An expression ofsquamous cell carcinoma antigen 2 in peripheral blood within the differentstages of esophageal carcinogenesis. Dis Esophagus 2008;21(5):395–401.

[120] Honma H, Kanda T, Ito H, Wakai T, Nakagawa S, Ohashi M, et al. Squamouscell carcinoma-antigen messenger RNA level in peripheral blood predictsrecurrence after resection in patients with esophageal squamous cell carci-noma. Surgery 2006;139(5):678–85.

[121] Kocakova I, Svoboda M, Kubosova K, Chrenko V, Roubalova E, Krejci E, et al.Preoperative radiotherapy and concomitant capecitabine treatment inducethymidylate synthase and thymidine phosphorylase mRNAs in rectal carci-noma. Neoplasma 2007;54(5):447–53.

[122] Miura N, Kanamori Y, Takahashi M, Sato R, Tsukamoto T, Takahashi S, et al. Adiagnostic evaluation of serum human telomerase reverse transcriptasemRNA as a novel tumor marker for gynecologic malignancies. Oncol Rep2007;17(3):541–8.

[123] Valenti MT, Dalle Carbonare L, Donatelli L, Bertoldo F, Giovanazzi B, Caliari F,et al. STEAP mRNA detection in serum of patients with solid tumours. CancerLett 2009;273(1):122–6.

[124] Poon LL, Leung TN, Lau TK, Lo YM. Presence of fetal RNA in maternal plasma.Clin Chem 2000;46:1832–4.

[125] Ng EKO, Tsui NBY, Lau TK, Leung TN, Chiu RWK, Panesar NS, et al. mRNA ofplacental origin is readily detectable in maternal plasma. Proc Natl Acad Sci US A 2003;100:4748–53.

[126] Ng EKO, Leung TN, Tsui NBY, Lau TK, Panesar NS, Chiu RW, et al. Theconcentration of circulating corticotropin-releasing hormone mRNA in ma-ternal plasma is increased in preeclampsia. Clin Chem 2003;49:727–31.

[127] Hahn S, Rusterholz C, Hosli I, Lapaire O. Cell-free nucleic acids as potentialmarkers for preeclampsia. Placenta 2011;32S1:S17–20.

[128] Ng EKO, El-Sheikhah A, Chiu RWK, Chan KCA, Hogg M, Bindra R, et al. Humanchorionic gonadotropin beta-subunit mRNA concentrations in maternalserum in aneuploid pregnancies. Clin Chem 2004;50:1055–7.

[129] Lo YM. Recent advances in fetal nucleic acids in maternal plasma. J Histo-chem Cytochem 2005;53:293–6.

[130] Oudejans CB, Go AT, Visser A, Mulders MA, Westerman BA, Blankenstein MA,et al. Detection of chromosome 21-encoded mRNA of placental origin inmaternal plasma. Clin Chem 2003;49:1445–9.

[131] Wataganara T, LeShane ES, Chen AY, Borgatta L, Peter I, Johnson KL, et al.Plasma gamma-globin gene expression suggests that fetal hematopoieticcells contribute to the pool of circulating cell-free fetal nucleic acids duringpregnancy. Clin Chem 2004;50:689–93.

[132] Tsui NB, Chim SS, Chiu RW, Lau TK, Ng EKO, Leung TN, et al. Systematic micro-array based identification of placental mRNA in maternal plasma: towards non-invasive prenatal gene expression profiling. J Med Genet 2004;41:461–7.

[133] Lo YMD, et al. Plasma DNA as a prognostic marker in trauma patients. ClinChem 2000;46:319–23.

[134] Rainer TH, Lam NY, Tsui NB, Ng EK, Chiu RW, Joynt GM, et al. Effects offiltration on glyceraldehyde-3-phosphate dehydrogenase mRNA in the plas-ma of trauma patients and healthy individuals. Clin Chem 2004;50:206–8.

[135] Orlandi E, Butt A, Goldsmith D, Swaminathan R. Factors affecting circulatingmRNA for nephrin. Clin Chem 2005;51:1982–3.

[136] Butt AN, Swaminathan R. Overview of circulating nucleic acids in plasma/serum. Ann N Y Acad Sci 2008;1137:236–42.

[137] Hamaoui K, Butt A, Powrie J, Swaminathan R. Concentration of circulatingrhodopsin mRNA in diabetic retinopathy. Clin Chem 2004;50:2152–5.

[138] Shalchi Z, Sandhu HS, Butt AN, Smith S, Powrie J, Swaminathan R. Retina-specific mRNA in the assessment of diabetic retinopathy. Ann N Y Acad Sci2008;1137:253–7.

[139] Sandhu HS, Butt AN, Powrie J, Swaminathan R. Measurement of circulatingneuron-specific enolase mRNA in diabetes mellitus. Ann N Y Acad Sci2008;1137:258–63.

[140] de Almeida Pdel V, Gregio A, Machado M, de Lima A, Azevedo L. Salivacomposition and functions: a comprehensive review. J Contemp Dent Pract2008;9(3):72–80.

[141] Streckfus C, Bigler L. The use of soluble, salivary c-erbB-2 for the detectionand post-operative follow-up of breast cancer in women: the results of a five-year translational research study. Adv Dent Res 2005;18(1):17–24.

[142] Streckfus CF, Bigler L, Dellinger T, Kuhn M, Chouinard N, Dai X. The expressionof the c-erbB-2 receptor protein in glandular salivary secretions. J Oral PatholMed 2004;33(10):595–600.

[143] Park N, Li Y, Yu T, Brinkman B, Wong D. Characterization of RNA in saliva. ClinChem 2006;52(6):988–94.

[144] Bardon A, Shugar D. Properties of purified salivary ribonuclease, and salivaryribonuclease levels in children with cystic fibrosis and in heterozygouscarriers. Clin Chim Acta 1980;101(1):17–24.

[145] Eichel H, Conger N, Chernick W. Acid and alkaline ribonucleases of humanparotid, submaxillary, and whole saliva. Arch Biochem Biophys1964;107:197–208.

[146] Haas C, Klesser B, Maake C, Bar W, Kratzer A. mRNA profiling for body fluididentification by reverse transcription endpoint PCR and realtime PCR.Forensic Sci Int 2009;3(2):80–8.

[147] Zubakov D, Hanekamp E, Kokshoorn M, van Ijcken W, Kayser M. Stable RNAmarkers for identification of blood and saliva stains revealed from wholegenome expression analysis of time-wise degraded samples. Int J Legal Med2008;122(2):135–42.

[148] Nussbaumer C, Gharehbaghi-Schnell E, Korschineck I. Messenger RNA pro-filing: a novel method for body fluid identification by real-time PCR. ForensicSci Int 2006;157(2–3):181–6.

[149] Bauer M. RNA in forensic science. Forensic Sci Int Genet 2007;1(1):69–74.[150] Juusola J, Ballantyne J. Multiplex mRNA profiling for the identification of body

fluids. Forensic Sci Int 2005;152(1):1–12.[151] Juusola J, Ballantyne J. Messenger RNA profiling: a prototype method to

supplant conventional methods for body fluid identification. Forensic Sci Int2003;135(2):85–96.

[152] St John M, Li Y, Zhou X, Denny P, Ho CM, Montemagno C, et al. Interleukin 6and interleukin 8 as potential biomarkers for oral cavity and oropharyngealsquamous cell carcinoma. Arch Otolaryngol Head Neck Surg2004;130(8):929–35.

[153] Hu S, Yu T, Xie Y, Yang Y, Li Y, Zhou X, et al. Discovery of oral fluid biomarkersfor human oral cancer using mass spectrometry. Cancer Genomics Proteo-mics 2007;4(2):55–64.

[154] Li Y, Zhou X, St John MA, Wong DT. RNA profiling of cell-free saliva usingmicroarray technology. J Dent Res 2004;83(3):199–203.

[155] Li Y, St John MA, Zhou X, Kim Y, Sinha U, Jordan RC, et al. Salivary tran-scriptome diagnostics for oral cancer detection. Clin Cancer Res2004;10(24):8442–50.

[156] Zimmermann B, Wong D. Salivary mRNA targets for cancer diagnostics. OralOncol 2008;44(5):425–9.

[157] Hu S, Wang J, Meijer J, Ieong S, Xie Y, Yu T, et al. Salivary proteomic andgenomic biomarkers for primary Sjogren’s syndrome. Arthritis Rheum2007;56(11):3588–600.

[158] Seugnet L, Boero J, Gottschalk L, Duntley S, Shaw P. Identification of abiomarker for sleep drive in flies and humans. Proc Natl Acad Sci U S A2006;103(52):19913–8.

[159] Kumar S, Hurteau G, Spivack S. Validity of Messenger RNA expressionanalyses of human saliva. Clin Cancer Res 2006;12(17):5033–9.

[160] Park NJ, Zhou X, Yu T, Brinkman BM, Zimmermann BG, Palanisamy V, et al.Characterization of salivary RNA by cDNA library analysis. Arch Oral Biol2007;52(1):30–5.

[161] Park NJ, Li Y, Yu T, Brinkman BM, Wong DT. Characterization of RNA in saliva.Clin Chem 2006;52(6):988–94.

[162] Park NJ, Li Y, Yu T, Brinkman BM, Wong DT. RNA protect saliva: an optimalroom-temperature stabilization reagent for the salivary transcriptome. ClinChem 2006;52(12):2303–4.

[163] Taniguchi M, Miura K, Iwao H, Yamanaka S, Bryzgunova OE, Skvortsova TE,et al. Isolation and comparative study of cell-free nucleic acids from humanurine. Ann N Y Acad Sci 2006;1075:334–44.

[164] Wang R, Chinnaiyan AM, Dunn RL, Wojno KJ, Wei JT. Rational approach toimplementation of prostate cancer antigen 3 into clinical care. Cancer2009;115(17):3879–86.

[165] Mearini E, Antognelli C, Del Buono C, Cochetti G, Giannantoni A, Nardelli E,et al. The combination of urine DD3 (PCA3) mRNA and PSA mRNA asmolecular markers of prostate cancer. Biomarkers 2009;14(4):235–43.

[166] Hessels D, Schalken JA. The use of PCA3 in the diagnosis of prostate cancer.Nat Rev Urol 2009;6(5):255–61.

[167] Nilsson J, Skog J, Nordstrand A, Baranov V, Mincheva-Nilsson L, BreakefieldXO, et al. Prostate cancer-derived urine exosomes: a novel approach tobiomarkers for prostate cancer. Br J Cancer 2009;100(10):1603–7.

[168] Whitman EJ, Groskopf J, Ali A, Chen Y, Blase A, Furusato B, et al. PCA3 scorebefore radical prostatectomy predicts extracapsular extension and tumorvolume. J Urol 2008;180(5):1975–8.

[169] Deras IL, Aubin SM, Blase A, Day JR, Koo S, Partin AW, et al. PCA3: a molecularurine assay for predicting prostate biopsy outcome. J Urol2008;179(4):1587–92.

[170] Hessels D, Smit FP, Verhaegh GW, Witjes JA, Cornel EB, Schalken JA. Detectionof TMPRSS2-ERG fusion transcripts and prostate cancer antigen 3 in urinary

G. Tzimagiorgis et al. / Cancer Epidemiology 35 (2011) 580–589 587

sediments may improve diagnosis of prostate cancer. Clin Cancer Res2007;13(17):5103–8.

[171] de Kok JB, Verhaegh GW, Roelofs RW, Hessels D, Kiemeney LA, Aalders TW,et al. DD3 (PCA3), a very sensitive and specific marker to detect prostatetumors. Cancer Res 2002;62(9):2695–8.

[172] Nakanishi H, Groskopf J, Fritsche HA, Bhadkamkar V, Blase A, Kumar SV, et al.PCA3 molecular urine assay correlates with prostate cancer tumor volume:implication in selecting candidates for active surveillance. J Urol2008;179(5):1804–9.

[173] Varambally S, Laxman B, Mehra R, Cao Q, Dhanasekaran SM, Tomlins SA, et al.Golgi protein GOLM1 is a tissue and urine biomarker of prostate cancer.Neoplasia 2008;10(11):1285–94.

[174] Fradet Y. Biomarkers in prostate cancer diagnosis: beyond prostate specificantigen. Curr Opin Urol 2009;19(3):243–6.

[175] Fradet Y, Saad F, Aprikian A, Dessureault J, Elhilali M, Trudel C, et al. uPM3, anew molecular urine test for the detection of prostate cancer. Urology2004;64(2):311–5.

[176] Pina-Cabral L, Santos L, Mesquita B, Amaro T, Magalhaes S, Criado B. Detec-tion of survivin mRNA in urine of patients with superficial urothelial cellcarcinomas. Clin Transl Oncol 2007;9(11):731–6.

[177] Pu XY, Wang ZP, Chen YR, Wang XH, Wu YL, Wang HP. The value of combineduse of survivin, cytokeratin 20 and mucin 7 mRNA for bladder cancerdetection in voided urine. J Cancer Res Clin Oncol 2008;134(6):659–65.

[178] Kenney DM, Geschwindt RD, Kary MR, Linic JM, Sardesai NY, Li ZQ. Detectionof newly diagnosed bladder cancer, bladder cancer recurrence and bladdercancer in patients with hematuria using quantitative RT-PCR of urinarysurvivin. Tumour Biol 2007;28(2):57–62.

[179] Hou JQ, He J, Wen DG, Chen ZX, Zeng J. Survivin mRNA expression in urine as abiomarker for patients with transitional cell carcinoma of bladder. Chin Med J(Engl) 2006;119(13):1118–20.

[180] Weikert S, Christoph F, Schrader M, Krause H, Miller K, Muller M. Quantitativeanalysis of survivin mRNA expression in urine and tumor tissue of bladdercancer patients and its potential relevance for disease detection and prog-nosis. Int J Cancer 2005;116(1):100–4.

[181] Wang H, Xi X, Kong X, Huang G, Ge G. The expression and significance ofsurvivin mRNA in urinary bladder carcinomas. J Cancer Res Clin Oncol2004;130(8):487–90.

[182] Guo B, Luo C, Xun C, Xie J, Wu X, Pu J. Quantitative detection of cytokeratin 20mRNA in urine samples as diagnostic tools for bladder cancer by real-timePCR. Exp Oncol 2009;31(1):43–7.

[183] Siracusano S, Niccolini B, Knez R, Tiberio A, Benedetti E, Bonin S, et al. Thesimultaneous use of telomerase, cytokeratin 20 and CD4 for bladder cancerdetection in urine. Eur Urol 2005;47(3):327–33.

[184] Christoph F, Muller M, Schostak M, Soong R, Tabiti K, Miller K. Quantitativedetection of cytokeratin 20 mRNA expression in bladder carcinoma by real-time reverse transcriptase-polymerase chain reaction. Urology2004;64(1):157–61.

[185] Inoue T, Nakanishi H, Inada K, Hioki T, Tatematsu M, Sugimura Y. Real timereverse transcriptase polymerase chain reaction of urinary cytokeratin 20detects transitional cell carcinoma cells. J Urol 2001;166(6):2134–41.

[186] Weikert S, Krause H, Wolff I, Christoph F, Schrader M, Emrich T, et al.Quantitative evaluation of telomerase subunits in urine as biomarkers fornoninvasive detection of bladder cancer. Int J Cancer 2005;117(2):274–80.

[187] Bowles L, Bialkowska-Hobrzanska H, Bukala B, Nott L, Razvi H. A prospectiveevaluation of the diagnostic and potential prognostic utility of urinaryhuman telomerase reverse transcriptase mRNA in patients with bladdercancer. Can J Urol 2004;11(6):2438–44.

[188] Melissourgos N, Kastrinakis NG, Davilas I, Foukas P, Farmakis A, LykourinasM. Detection of human telomerase reverse transcriptase mRNA in urine ofpatients with bladder cancer: evaluation of an emerging tumor marker.Urology 2003;62(2):362–7.

[189] Bialkowska-Hobrzanska H, Bowles L, Bukala B, Joseph MG, Fletcher R, RazviH. Comparison of human telomerase reverse transcriptase messenger RNAand telomerase activity as urine markers for diagnosis of bladder carcinoma.Mol Diagn 2000;5(4):267–77.

[190] de Kok JB, van Balken MR, Ruers TJ, Swinkels DW, Klein Gunnewiek JM.Detection of telomerase activity in urine as a tool for noninvasive detection ofrecurrent bladder tumors is poor and cannot be improved by timing ofsampling. Clin Chem 2000;46(12):2014–5.

[191] Ito H, Kyo S, Kanaya T, Takakura M, Koshida K, Namiki M, et al. Detection ofhuman telomerase reverse transcriptase messenger RNA in voided urinesamples as a useful diagnostic tool for bladder cancer. Clin Cancer Res1998;4(11):2807–10.

[192] Xie XY, Yang X, Zhang JH, Liu ZJ. Analysis of hTERT expression in exfoliatedcells from patients with bladder transitional cell carcinomas using SYBRgreen real-time fluorescence quantitative PCR. Ann Clin Biochem 2007;44(Pt6):523–8.

[193] Hanke M, Kausch I, Dahmen G, Jocham D, Warnecke JM. Detailed technicalanalysis of urine RNA-based tumor diagnostics reveals ETS2/urokinase plas-minogen activator to be a novel marker for bladder cancer. Clin Chem2007;53(12):2070–7.

[194] Unoki M, Kelly JD, Neal DE, Ponder BA, Nakamura Y, Hamamoto R. UHRF1 is anovel molecular marker for diagnosis and the prognosis of bladder cancer. BrJ Cancer 2009;101(1):98–105.

[195] Holyoake A, O’Sullivan P, Pollock R, Best T, Watanabe J, Kajita Y, et al.Development of a multiplex RNA urine test for the detection and stratifica-

tion of transitional cell carcinoma of the bladder. Clin Cancer Res2008;14(3):742–9.

[196] Muthukumar T, Dadhania D, Ding R, Snopkowski C, Naqvi R, Lee JB, et al.Messenger RNA for FOXP3 in the urine of renal-allograft recipients. N Engl JMed 2005;353:2342–51.

[197] Yoneda K, Iida H, Endo H, Hosono K, Akiyama T, Takahashi H, et al. Identifi-cation of Cystatin SN as a novel tumor marker for colorectal cancer. Int JOncol 2009;35(1):33–40.

[198] Savoia P, Osella-Abate S, Comessatti A, Nardo T, Marchio C, Pacchioni D, et al.Traditional urinary cytology and tyrosinase RT-PCR in metastatic melanomapatients: correlation with clinical status. J Clin Pathol 2008;61(2):179–83.

[199] de Bont JM, van Doorn J, Reddingius RE, Graat GH, Passier MM, den Boer ML,et al. Various components of the insulin-like growth factor system in tumortissue, cerebrospinal fluid and peripheral blood of pediatric medulloblastomaand ependymoma patients. Int J Cancer 2008;123(3):594–600.

[200] Weber JA, Baxter DH, Zhang S, Huang DY, Huang KH, Lee MJ, et al. ThemicroRNA spectrum in 12 body fluids. Clin Chem 2010;56(11):1733–41.

[201] Kosaka N, Iguchi H, Ochiya T. Circulating microRNA in body fluid: a newpotential biomarker for cancer diagnosis and prognosis. Cancer Sci2010;101(10):2087–92.

[202] Ferracin M, Veronese A, Negrini M, Micromarkers. miRNAs in cancer diag-nosis and prognosis. Expert Rev Mol Diagn 2010;10(3):297–308.

[203] Shah AA, Leidinger P, Blin N, Meese E. miRNA: small molecules as potentialnovel biomarkers in cancer. Curr Med Chem 2010;17(36):4427–32.

[204] Heneghan HM, Miller N, Kerin MJ. MiRNAs as biomarkers and therapeutictargets in cancer. Curr Opin Pharmacol 2010;10(5):543–50.

[205] Scholer N, Langer C, Dohner H, Buske C, Kuchenbauer F. Serum microRNAs asa novel class of biomarkers: a comprehensive review of the literature. ExpHematol 2010;38(12):1126–30.

[206] Rahbari R, Holloway AK, He M, Khanafshar E, Clark OH, Kebebew E. Identifi-cation of differentially expressed microRNA in parathyroid tumors. Ann SurgOncol 2010;18(4):1158–65.

[207] Zen K, Zhang CY. Circulating microRNAs: a novel class of biomarkers todiagnose and monitor human cancers. Med Res Rev 2010;Nov 9 (forthcom-ing).

[208] Wang J, Chen J, Chang P, LeBlanc A, Li D, Abbruzzesse JL, et al. MicroRNAs inplasma of pancreatic ductal adenocarcinoma patients as novel blood-basedbiomarkers of disease. Cancer Prev Res (Phila) 2009;2(9):807–13.

[209] Zhou SL, Wang LD. Circulating microRNAs: novel biomarkers for esophagealcancer. World J Gastroenterol 2010;16(19):2348–54.

[210] Schee K, Fodstad Ø, Flatmark K. MicroRNAs as biomarkers in colorectalcancer. Am J Pathol 2010;177(4):1592–9.

[211] Roth C, Rack B, Muller V, Janni W, Pantel K, Schwarzenbach H. CirculatingmicroRNAs as blood-based markers for patients with primary and metastaticbreast cancer. Breast Cancer Res 2010;12(6):R90.

[212] Gotte M. MicroRNAs in breast cancer pathogenesis. Minerva Ginecol2010;62(6):559–65.

[213] Wang F, Zheng Z, Guo J, Ding X. Correlation and quantitation of microRNAaberrant expression in tissues and sera from patients with breast tumor.Gynecol Oncol 2010;119(3):586–93.

[214] Slaby O, Svoboda M, Michalek J, Vyzula R. MicroRNAs in colorectal cancer:translation of molecular biology into clinical application. Mol Cancer2009;8:102.

[215] Huang Z, Huang D, Ni S, Peng Z, Sheng W, Du X. Plasma microRNAs arepromising novel biomarkers for early detection of colorectal cancer. Int JCancer 2010;127(1):118–26.

[216] Lodes MJ, Caraballo M, Suciu D, Munro S, Kumar A, Anderson B. Detection ofcancer with serum miRNAs on an oligonucleotide microarray. PLoS One2009;4(7):e6229.

[217] Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, et al. Characterization of microRNAsin serum: a novel class of biomarkers for diagnosis of cancer and otherdiseases. Cell Res 2008;18(10):997–1006.

[218] Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-AgadjanyanEL, et al. Circulating microRNAs as stable blood-based markers for cancerdetection. Proc Natl Acad Sci U S A 2008;105(30):10513–8.

[219] Hanke M, Hoefig K, Merz H, Feller AC, Kausch I, Jocham D, et al. A robustmethodology to study urine microRNA as tumor marker: microRNA-126 andmicroRNA-182 are related to urinary bladder cancer. Urol Oncol2010;28(6):655–61.

[220] Park NJ, Zhou H, Elashoff D, Henson BS, Kastratovic DA, Abemayor E, et al.Salivary microRNA: discovery, characterization, and clinical utility for oralcancer detection. Clin Cancer Res 2009;15(17):5473–7.

[221] Farazi TA, Spitzer JI, Morozov P, Tuschl T. miRNAs in human cancer. J Pathol2011;223(2):102–15.

[222] Michael A, Bajracharya SD, Yuen PS, Zhou H, Star RA, Illei GG, et al. Exosomesfrom human saliva as a source of microRNA biomarkers. Oral Dis2010;16(1):34–8.

[223] Cortez MA, Calin GA. MicroRNA identification in plasma and serum: a newtool to diagnose and monitor diseases. Expert Opin Biol Ther 2009;9(6):703–11.

[224] Wang K, Zhang S, Marzolf B, Troisch P, Brightman A, Hu Z, et al. CirculatingmicroRNAs, potential biomarkers for drug-induced liver injury. Proc NatlAcad Sci U S A 2009;106(11):4402–7.

[225] Cheng Y, Tan N, Yang J, Liu X, Cao X, He P, et al. A translational study ofcirculating cell-free microRNA-1 in acute myocardial infarction. Clin Sci(Lond) 2010;119(2):87–95.

G. Tzimagiorgis et al. / Cancer Epidemiology 35 (2011) 580–589588

[226] Wang G, Tam LS, Li EK, Kwan BC, Chow KM, Luk CC, et al. Serum and urinarycell-free MiR-146a and MiR-155 in patients with systemic Lupus erythema-tosus. J Rheumatol 2011;223(2):102–15.

[227] Hanson EK, Lubenow H, Ballantyne J. Identification of forensically relevantbody fluids using a panel of differentially expressed microRNAs. Anal Bio-chem 2009;387(2):303–14.

[228] Bentwich I, Avniel A, Karov Y, Aharonov R, Gilad S, Barad O, et al. Identifica-tion of hundreds of conserved and nonconserved human microRNAs. NatGenet 2005;37(7):766–70.

[229] Tsui NB, Ng EK, Lo YM. Stability of endogenous and added RNA in bloodspecimens, serum, and plasma. Clin Chem 2002;48(10):1647–53.

[230] Blackburn P. Ribonuclease inhibitor from human placenta: rapid purificationand assay. J Biol Chem 1979;254(24):12484–7.

[231] Wong DT. Salivary transcriptome. Clin Cancer Res 2007;13(4):1350–1 [au-thor reply 1351].

[232] Chirgwin J, Przybyla A, MacDonald R, Rutter W. Isolation of biologically activeribonucleic acid from sources enriched in ribonuclease. Biochemistry1979;18(24):5294–9.

[233] Chomczynski P, Sacchi N. Single-step method of RNA isolation by acidguanidinium thiocyanate–phenol–chloroform extraction. Anal Biochem1987;162(1):156–9.

[234] Chomczynski P. A reagent for the single-step simultaneous isolation of RNA,DNA and proteins from cell and tissue samples. Biotechniques 1993;15(3).532–4, 536–7.

[235] Perez S, Royo LJ, Astudillo A, Escudero D, Alvarez F, Rodrıguez A, et al.Identifying the most suitable endogenous control for determininggene expression in hearts from organ donors. BMC Mol Biol2007;8:114.

[236] Huang Z, Fasco M, Kaminsky L. Optimization of DNase I removal of contami-nating DNA from RNA for use in quantitative RNA-PCR. Biotechniques1996;20(6). 1012–4, 1016, 1018–20.

[237] Wiame I, Remy S, Swennen R, Sagi L. Irreversible heat inactivation of DNase Iwithout RNA degradation. Biotechniques 2000;29(2). 252–4, 256.

[238] Dirnhofer S, Berger C, Untergasser G, Geley S, Berger P. Human beta-actinretropseudogenes interfere with RT-PCR. Trends Genet 1995;11(10):380–1.

[239] Reue K. mRNA quantitation techniques: considerations for experimentaldesign and application. J Nutr 1998;128(11):2038–44.

[240] Lossos IS, Czerwinski DK, Wechser MA, Levy R. Optimization of quantitativereal-time RT-PCR parameters for the study of lymphoid malignancies. Leu-kemia 2003;17(4):789–95.

[241] www.ambion.com/techlib/tb/tb_151.html.[242] Liu D, Chen S, Liu H. Choice of endogenous control for gene expression in non

small cell lung cancer. Eur Respir J 2005;26(6):1002–8.[243] de Kok J, Roelofs R, Giesendorf B, Pennings JL, Waas ET, Feuth T, et al.

Normalization of gene expression measurements in tumor tissues: compari-son of 13 endogenous control genes. Lab Invest 2005;85(1):154–9.

[244] Schmittgen TD, Zakrajsek BA. Effect of experimental treatment on house-keeping gene expression: validation by real-time, quantitative RT-PCR. JBiochem Biophys Methods 2000;46(1–2):69–81.

[245] Taniguchi M, Miura K, Iwao H, Yamanaka S. Quantitative assessment of DNAmicroarrays – comparison with Northern blot analyses. Genomics2001;71(1):34–9.

[246] Baldwin D, Crane V, Rice D. A comparison of gel-based, nylon filter andmicroarray techniques to detect differential RNA expression in plants. Cur-rent Opinion in Plant Biol 1999;2(2):96–103.

[247] Gershon D. Microarray technology: an array of opportunities. Nature2002;416(6883):885–91.

G. Tzimagiorgis et al. / Cancer Epidemiology 35 (2011) 580–589 589