Topic Highlight Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Feb 7, 2016; 22(5): 1745-1755
Published online Feb 7, 2016. doi: 10.3748/wjg.v22.i5.1745
Colorectal cancer tumour markers and biomarkers: Recent therapeutic advances
Gustaw Lech, Maciej Słodkowski, Ireneusz Wojciech Krasnodębski, Department of General, Gastroenterological and Oncological Surgery, Medical University of Warsaw, 02097 Warsaw, Poland
Robert Słotwiński, Department of Surgical Research and Transplantology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02106 Warsaw, Poland
Robert Słotwiński, Department of Immunology, Biochemistry and Nutrition, Medical University of Warsaw, 02007 Warsaw, Poland
Author contributions: Lech G wrote the manuscript; Słotwiński R, Słodkowski M and Krasnodębski IW reviewed the entire manuscript.
Conflict-of-interest statement: The authors declare no conflicts of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Gustaw Lech, MD, PhD, Department of General, Gastroenterological and Oncological Surgery, Medical University of Warsaw, 1a Banacha Street, 02097 Warsaw, Poland. gustaw.lech@wum.edu.pl
Telephone: +48-22-5992482 Fax: +48-22-5992057
Received: May 3, 2015
Peer-review started: May 8, 2015
First decision: September 9, 2015
Revised: October 5, 2015
Accepted: December 21, 2015
Article in press: December 21, 2015
Published online: February 7, 2016

Abstract

Colorectal cancer (CRC) is the second most commonly diagnosed cancer among females and third among males worldwide. It also contributes significantly to cancer-related deaths, despite the continuous progress in diagnostic and therapeutic methods. Biomarkers currently play an important role in the detection and treatment of patients with colorectal cancer. Risk stratification for screening might be augmented by finding new biomarkers which alone or as a complement of existing tests might recognize either the predisposition or early stage of the disease. Biomarkers have also the potential to change diagnostic and treatment algorithms by selecting the proper chemotherapeutic drugs across a broad spectrum of patients. There are attempts to personalise chemotherapy based on presence or absence of specific biomarkers. In this review, we update review published last year and describe our understanding of tumour markers and biomarkers role in CRC screening, diagnosis, treatment and follow-up. Goal of future research is to identify those biomarkers that could allow a non-invasive and cost-effective diagnosis, as well as to recognise the best prognostic panel and define the predictive biomarkers for available treatments.

Key Words: Colorectal cancer, Biomarker, Microsatellite instability, KRAS mutation, BRAF mutation, PIK3CA mutation, Chromosome 18q loss of heterozygosity, Anti-epidermal growth factor receptor therapy, Colorectal cancer biomarkers, Carcinoembryonic antigen

Core tip: This review summarizes data concerning clinical utility of biomarkers in colorectal cancer patients. Authors focus primarily on currently available diagnostic, prognostic and predictive biomarkers of the disease. Great attention is also paid to the advances achieved in personalized therapy of colorectal cancer.



INTRODUCTION

Worldwide, colorectal cancer (CRC) annually affects more than one million men and women and causes more than half a million deaths[1]. In Europe in 2010, CRC was the third most common malignant cancer in both men and women[2]. There are 250000 cases of colorectal cancer diagnosed on an annual basis in Europe only. Five-year survival was 54 percent among adult Europeans diagnosed with colorectal cancer between 1995 and 1999[3]. More recent available data report that the overall five-year relative survival can achieve 65 percent, but varies depending on stage of cancer disease[4].

The number of biomarkers used for tests continues to grow. The National Institute of Health defines a biomarker as a biological molecule found in blood, other body fluids, or tissues that is a sign of normal or abnormal process, or of a condition or disease[5]. A definition of biomarker mostly refers to DNA, RNA, microRNA (miRNA), epigenetic changes or antibodies. A term tumour marker, by some researchers considered as a synonym of biomarker, refers to substances (most typically proteins, glycolipids) representing biological structures, which can be attributed to the development of normal cells or carcinogenesis at different cell development stages e.g., tumour-associated antigens (TAAs) which are the largest group of clinically significant markers. As a result, the concentration of TAAs typically correlates with the number (or mass) of specific neoplastic cells.

In daily clinical practice, in the process of diagnosis and therapy, there are several parameters in use of long-established high sensitivity, specificity and positive predictive value. These parameters have been selected from among tens of molecules produced by cells in long-term laboratory tests, observational studies and clinical trials. The concentrations of tumour markers tested at the diagnostic stage are believed to assist in early cancer diagnosis and to be used in screening tests. Some of them are currently found to be more important during treatment and long-term follow-up. On the other hand, for some types of tumours, markers are also considered important in monitoring the progress of treatment, efficacy of neo-adjuvant therapy, surgery, adjuvant chemotherapy and radiation therapy and follow-up for possible recurrence. Long-term observational studies also point to the fact that, apart from determining antigen concentration, it can be also important to trace its progress and dynamics. In this review, we have updated a review published in 2014[6]. We examine molecular (genetic, epigenetic, protein) biomarkers associated with CRC and discuss their role in cancer screening, early detecting of disease recurrence and as prognostic and predictive factors.

BLOOD AND STOOL MARKERS FOR COLORECTAL CANCER SCREENING AND FOLLOW UP
Blood and stool genetic and epigenetic markers

Several authors have investigated molecular non-invasive screening tests for early detection of CRC. DNA, RNA and other molecules derived by tumour in stool, as well as their concentrations in blood have been studied extensively. Colorectal process of carcinogenesis is characterized by genetic and epigenetic alteration transforming normal cells into cancer cells. Most studies concerning molecular markers in stool have focused on the detection of tumour DNA. These investigations have concentrated on the detection of mutated KRAS, TP53, APC and markers for microsatellite instability (MSI)[7-9]. A faecal DNA test targeted at molecular biomarkers has been commercially available for twelve years, with reported sensitivity for cancer ranging from 25% up to 92% for the latest tests based on BEAMing technology, and 94%-98% specificity[10-12]. Apart from genetic alterations, the DNA promoter hypermethylation silencing the tumour suppressor genes has been widely investigated. Epigenetic changes, depending on the markers or their combinations evaluated, have been detected in CRC patients with 70%-96% sensitivity and 72%-96% specificity[9,13,14]. Many combinations of genetic and epigenetic markers have been studied, but until now, the results have not endorsed their use in clinical practice. Using blood instead of stool as a screening material could offer some obvious advantages. Several studies have evaluated potential plasma DNA genetic and epigenetic biomarkers in CRC detection. The overall sensitivity ranges from 30% to 87%, with specificity of up to 96%. The use of RNA biomarkers in stool has not been investigated as extensively as was the case for DNA biomarkers, mainly because stool environment is responsible for mRNA degradation, although improving laboratory retrieval methods seems to solve this problem. Koga et al[15] analysed mRNA expression of MMP7, PTGS2, TP53 and MYBL2 in colonocytes isolated from stool by quantitative real-time RT-PCR, to find out that these markers can identify CRC patients with 58% sensitivity and 88% specificity. Sensitivity was found to depend on tumour size and tumour location, but not cancer stage[15]. Most recently, the so called transcriptomic studies have investigated the expression of miRNAs - short, non-coding 18-22 nucleotide RNA molecules in stools of CRC patients. The most extensively studied miR21, miR106a, miR135, miR17-92 were found to be overexpressed in CRC patients compared with healthy individuals[16,17]. As was the case with RNA markers in stool, many studies have been evaluating mRNA of different tumour genes in whole blood, plasma or circulating tumour cells to identify new CRC screening markers. Most of them investigated mRNA molecules of CK19, CK20, or Carcinoembryonic antigen (CEA). The overall sensitivity of these markers was up to 72%, specifically when combinations of these markers were used[18,19]. The specificity was very high with healthy control samples or much lower when compared to other cancer or inflammatory bowel diseases samples[20]. Recent studies have indicated that circulating miRNAs may be involved in the process of oncogenesis. The use of miRNA as a biomarker is now being evaluated. A large number of miRNA molecules have been assessed, with a focus on miR145, miR143, miR135, miR17-92. More specifically, Huang et al[21] has found that plasma miR29a and miR92a demonstrated a significant diagnostic value for advanced neoplasia with 83% and 84% sensitivity and specificity, respectively, in discriminating CRC patients. These studies need to be validated in randomised trials to define their value in CRC screening.

Blood and stool protein markers

Protein markers for screening and early detection of CRC can be divided into tumour TAAs, antibodies against TAAs, and other CRC-relevant proteins. CEA was discovered almost 50 years ago, in 1965, and it still remains the only tumour marker of recognised efficacy in monitoring CRC patients’ therapy[22]. CEA was first considered specific for CRC, but elevated CEA levels were later detected in other neoplasms too, e.g. gastric and pancreatic cancers, and in inflammatory conditions. Elevated CEA concentrations are only rarely identified in CRC stage I. Moreover, CEA does not differentiate benign versus malignant polyps. According to The European Group on Tumor Markers, European Society of Medical Oncology and American Society of Clinical Oncology guidelines[2,23,24], CEA is not recommended for use in screening tests. Recently, some studies have investigated the advantages of mRNA molecules encoding CEA for the detection of CRC, but the results were not superior to those of CEA[19].

In some studies, high CEA concentrations in patients with CRC stage II and III were found to be potentially indicative of more aggressive types of cancer[25,26]. Earlier, the Colorectal Working Group of American Joint Committee on Cancer proposed to include CEA baseline concentration to the traditional TNM classification as the so-called C-stage. C-stage was proposed to be divided into Cx, C0 (CEA < 5 ng/mL) and C1 (CEA > 5 ng/mL) substages[27]. The meaning of CEA as an independent prognostic factor was also confirmed in a recent retrospective analysis of 17910 patients with CRC, with a mean 27-mo follow-up, with longer survival periods for patients with IIA C0 and IIIA C0 vs I C1, IIIA C0 vs IIA C1, and IIIB C0 vs IIB-C C1, respectively[28]. No study, however, has shown that CEA concentration level can be used to select those patients with stage II CRC who would benefit from adjuvant chemotherapy. From a prognostic point of view, it appears reasonable to determine CEA levels before surgery in patients with disseminated CRC. The roles of CEA in determining life expectancy was confirmed in several studies on patients with liver metastases[29,30]. Recent study proved that combined use of CEA and serum amyloid A (SAA) is able to identify patients with favourable and poor prognosis. In addition to tumour baseline parameters, routine analysis of CEA together with SAA provides improved prognosis value on cancer specific survival and disease-free survival in resected rectal cancers[31]. CEA half-life is known to last approximately 7 d. After R0 resection surgery, CEA levels should return to normal within 4 to 6 wk. Sustained elevated CEA levels can be indicative of infiltration or metastases. Slow increase in CEA concentrations after surgery is a typical sign of local recurrence, whereas dynamically increasing levels can be symptomatic of metastases, most probably in the liver[24,32]. Testing CEA levels is considered most cost-effective in detecting post-surgery recurrences[24]. Please note that CEA levels tested every 3 mo for the first 3 years and thereafter every 6 mo for subsequent 2-3 years is a golden follow-up standard after CRC therapy recommended by a number of scientific associations[2,23,33]. It appears particularly important in asymptomatic patients, in whom chemotherapy can be used, with a much longer life expectancy as compared to treatment administered after the onset of symptoms of recurrence. CEA is a marker of choice in monitoring disseminated disease during systemic therapy. Constant increase in CEA levels is typically associated with a progression of the disease, even though radiological tests may prove otherwise[23,24]. However, chemotherapy can also result in temporary increase in CEA concentration, which must be also taken into account. Therefore, it is not recommended to test CEA levels within 2 wk of chemotherapy, whereas in patients on oxaliplatin, tests can be carried out after 4 to 6 wk.

Cancer antigen 19-9 (CA 19-9) is a glycoprotein whose relevance in CRC diagnosis still remains an issue. The majority of researchers arrived at the conclusion that CA 19-9 sensitivity is much inferior to that of CEA, and that elevated CA 19-9 levels is a poor prognostic factor[2,23,34-36]. Other carbohydrate antigens: CA 195, CA 50 have been also investigated, but with comparatively disappointing results. CA 72-4 is a biomarker with poor sensitivity ranging from 9% to 31% and better specificity ranging from 89% to 95% in patients screened for CRC. The diagnostic information in recurrent CRC provided by CA 72-4 has borderline significance, by far worse than CEA. All authors conclude that CA 72-4 sensitivity is rather low and specificity incomplete in screening and following up in patients with CRC. On the other hand, an algorithm based on combination of CEA, CA 19-9, CA 72-4, CA 242, CYFRA21-1 improves the diagnostic accuracy compared with these biomarkers alone[34-39]. Among other protein markers examined for screening purposes, two have been extensively investigated: the tumour specific M2 isoform of pyruvate kinase (M2-PK) in stool and tissue inhibitor of matrix metalloproteinase 1 (TIMP1). M2-PK measured in stool showed relatively high sensitivity for CRC up to 91%, and much lower for adenomas[40,41]. Plasma level of TIMP1 is reported to be elevated in CRC patients and prospective studies have been carried to assess its utility as biomarker. The results of the study included more than 4500 patients screened by endoscopy for CRC demonstrated that TIMP1 is not significantly superior to CEA marker in cancer screening and is not suitable for the detection of premalignant lesions[42]. Tissue polypeptide-specific antigen (TPS) and tissue polypeptide antigen (TPA) which detects the fragments of cytokeratines 8, 18 and 19 due to lack of sensitivity and specificity can not to be recommended in CRC screening. The majority of investigators have found that increased levels of TPA and TPS are observed in metastatic stage of CRC. A further studies has suggested that combination of TPA and CEA rises the sensitivity of these biomarkers in identifying the patients with CRC recurrence[34,37,43,44]. Other biomarkers, such as: thymidine phosphorylase (TP), DNA ploidy were determined to be insignificant in detecting, staging and following-up of patients with CRC[23].

MOLECULAR PROGNOSTIC AND PREDICTIVE BIOMARKERS

With the recent progress in understanding the molecular mechanisms of cancer development, dissemination, resistance to chemotherapy, and radiation therapy, it is now easier to select the most proper strategy for managing CRC. Clinical prospective and retrospective studies open the door for biomarkers use in clinical practice to assist in selecting the best drugs, both standard, such as 5-fluorouracil, oxaliplatin or irinotecan, and new generation targeted drugs: cetuximab, panitumumab, or bevacizumab. Biomarker identification is particularly important for patients with CRC stage II. In this group of patients, the risk of recurrence is only 20 percent. It is also desirable to use adjuvant therapy in this type of patients. There are attempts to select this group of patients based on genetic tests, or to personalise chemotherapy based on specific biomarkers. The following markers discovered throughout the recent years continue to be closely examined: MSI, chromosome 18q loss of heterozygosity (18qLOH), p53, KRAS, BRAF, NRAS, PIK3CA mutations, PTEN expression, UGT1A1 gene polymorphism, and ezrin protein (Table 1).

Table 1 Recommendations for use of tumour markers and biomarkers in colorectal cancer by groups of experts.
BiomarkerApplicationsASCO[23,98,99]ESMO[2,33]NCCN[100,101]
CEAScreeningNoNone publishedNone published
Prognostic factorYesYesYes
Follow upYesYesYes
CA 19-9AllNoNoNone published
CA 72-4AllNone publishedNone publishedNone published
CA 242AllNone publishedNone publishedNone published
CA 195AllNone publishedNone publishedNone published
CYFRA 21-1AllNone publishedNone publishedNone published
MSIPrognostic factorNoYesYes
18qLOHPrognostic factorYesYes (potentially)None published
p53 genePrognostic factorNoYes (potentially)None published
KRASPrognostic factorNone publishedYes (potentially)None published
Predictive factorYesYesYes
BRAFPrognostic factorNone publishedYesYes
Predictive factorYesYes (potentially)Yes (potentially)
PIK3CAPredictive factorNone publishedYes (potentially)None published
PTENPredictive factorYes (potentially)Yes (potentially)None published
UGT1A1Predictive factorYesYes (only in case of severe toxicity of irinotecan)No
VEGFAllNone publishedNone publishedNone published
TPA, TPSAllNone publishedNone publishedNone published
EzrinAllNone publishedNone publishedNone published
DNA ploidyAllNoNone publishedNone published
TSPrognostic factorNoYes (potentially)None published
Prognostic factorYes (potentially)Yes (potentially)None published
TPAllNoNone publishedNone published
DPDPrognostic factorNoYes (only in case of severe toxicity of 5-FU)None published
MSI

MSI denotes changes in coding and non-coding sequences of microsatellite chromosomes, i.e. repeated DNA sequences. These sequences are particularly exposed to errors in the mutation repair system that consist in the loss or multiplication of nucleotide sequence repetitions, which results in shortening or extension of microsatellite regions in neoplastic cells. Mutations arising out of these processes are eliminated by mismatch repair genes (MMR) such as MSH2, MSH6, PMS2 and MLH1, which makes some researchers believe that MSI can be caused by mutations in these genes[45]. Microsatellite instability can be classified into microsatellite instability-high (MSI-H), and microsatellite instability-low (MSI-L), depending on the percentage of loci that correlate to MSI characteristics. Tumour cells that lack MSI features are designated as MSS.

In retrospective studies and meta-analyses in patients with CRC stage II and III, MSI-H was shown to be a predictive factor that improved overall survival (OS), irrespective of the progression (stage) of cancer. A lower incidence of lymph node metastases and distant metastases as compared to MSI-L or MSS cancer cells was also observed[46-49]. MSI status is currently recommended in the WHO classification of mucinous-type CRC - MSI-H indicates good prognosis, MSI-L or MSS - poor outcome. However, MSI should be considered more of a prognostic rather than predictive factor. This conclusion is based on equivocal results of studies evaluating the efficacy of 5-FU-based chemotherapy in groups of patients with MSI-H and MSI-L or MSS. Ribic et al[48] examined tumour specimens collected from 570 patients with CRC stage II and III and correlated the test results with chemotherapy outcomes in these patients to reveal a tendency for shorter overall survival in patients with MSI-H on adjuvant therapy. Significant improvement was observed in patients with MSS tumours. A recent pooled analysis of randomized clinical studies revealed significant decrease in the overall five-year survival rate for patients with CRC stage II and MSI-H on 5-FU-based chemotherapy. 5-FU-based chemotherapy was found to improve therapeutic outcomes only in patients with CRC stage III and MSI-L or MSS[50]. Some studies indicated potentially negative effects of 5-FU-based chemotherapy in patients with MSI-H. A longer survival rate as compared to patients on 5-FU-based adjuvant chemotherapy was observed in a reference group of patients undergoing surgical treatment. Resistance of MSI-H tumours to 5-FU was also confirmed in in vitro studies[51]. A completely different conclusion can be drawn from earlier studies of Elsaleh et al[52,53], which confirmed the efficacy of 5-FU in patients with CRC stage III and MSI-H. Recent study also proved that prognostic value of MMR mutation was similar in the presence or absence of fluorouracil and folinic acid chemotherapy[54]. Beragnolli et al[55] revealed that a higher rate of overall 5-year progression-free survival was observed in patients with CRC stage III and MSI-H on 5-FU and irinotecan vs 5-FU-based chemotherapy. To recap, the results of MSI studies and clinical experience in patients with CRC stage II indicate that the degree of microsatellite instability may be of significance as a prognostic factor. Also, adjuvant 5-FU-based chemotherapy was proved to provide no benefits (or potentially cause adverse reactions) in patients with MSI-H. Further research is needed to investigate whether the MSI status can predict benefit (in high-risk patients) from irinotecan-based treatment or oxaliplatinum-based therapy.

Chromosome 18q loss of heterozygosity

A number of studies were dedicated to another prognostic factor in patients with CRC stage II and III - chromosome 18q loss of heterozygosity in the coding place of, inter alia, SMAD 4 proteins specific to CRC. In these studies, the overall 5-year survival was poorer for patients with CRC stage III and 18qLOH as compared to non-18qLOH patients[56]. A meta-analysis of data from 27 studies and 2189 patients by Popat et al[57] confirmed that poorer survival was correlated with 18q chromosome deletion. Two years later, the same research team questioned these findings after re-examining the same data[58]. Likewise, no correlation was identified between the presence of 18qLOH and 5-year survival in patients with non-MSI-H phenotype[59]. The role of 18qLOH in predicting response to standard chemotherapy has not been yet fully confirmed. Watanabe et al[60] demonstrated better response to 5-FU-based chemotherapy in patients with CRC stage III and MSS and with the absence of 18q chromosome deletion vs. patients in whom 18q chromosome deletion was present. The recently published results of the same research team can be a proof that in patients with CRC stage II and III and MSS-H (> 33%), the level of LOH of four chromosomes, including 18, is correlated with significantly poorer survival rate as compared to patients with MSS and LOH-L or MSI-H phenotype[61].

Based on the available data, 18q chromosome deletion cannot be the sole basis for any therapeutic decisions, however, it is being more closely examined under ECOG 5202 study, featuring molecular markers identified so far in selecting the most proper adjuvant post-surgery treatment, by prospectively analysing the role of MSI and 18qLOH in prognosis and therapeutic decisions in patients with CRC stage II. Patients with good prognosis (with MSI-H and w/o 18qLOH) were followed-up, and patients with poor prognosis (with MSI-L or MSS and 18qLOH) were randomized to one of two groups on chemotherapy (FOLFOX alone or FOLFOX and bevacizumab). The results of E5202 are expected in the next few years. No conclusion can be drawn from this study about the possible inefficacy of chemotherapy in patients with MSI-H, however, the study will include a multifactor analysis of biomarkers that can assist in taking therapeutic decisions in other groups of patients[62].

P53 mutation

Mutation in the tumour suppressor gene p53 (chromosome region 17p13) occur in 50%-70% of all CRC and is associated with worse outcomes, including disease free survival and overall survival[63]. Results obtained from a study that included more than 3500 CRC patients confirm the prognostic value of p53 mutation, which seems to be determined by the primary tumour site. Patients with p53 mutation and tumour of proximal colon had better OS when treated with adjuvant chemotherapy compared to those treated by surgery alone[64].

Biomarkers suitable in anti-epidermal growth factor receptor therapy

A number of currently tested markers have been discovered in the course of studies on epidermal growth factor receptor (EGFR) signalling pathways. KRAS gene mutation on short arm of chromosome 12 at codon 12 (80% of patients) or, to a lesser extent, codon 13 is believed to be of use as a biomarker in patients on cetuximab or panitumumab[65]. These mutations are one of the most common in proliferative diseases (37% and 13%, respectively), and their significance in CRC carcinogenesis has been examined in much detail[66]. As these mutations are present in EGFR signalling pathway, they can be a predictive factor for therapy with anti-EGFR antibodies. In studies performed so far, KRAS mutation was found to be correlated with non-responsiveness to cetuximab and panitumumab[67,68]. CRYSTAL and OPUS data indicate that the effectiveness of FOLFOX or FOLFIRI alone is no inferior to that of cetuximab in patients with KRAS in combination with chemotherapy according to FOLFIRI and FOLFOX regimen, respectively. However, in non-KRAS patients, cetuximab improves the therapeutic outcome[69,70]. The same conclusions can be drawn from the results of other large clinical studies: COIN, NORDIC VII or PRIME[71-73]. Yet, the effects of KRAS mutation at codon 12 or 13 on tumour biology were found to differ. In two studies, the survival rate was higher in patients with an uncommon G13D mutation at codon 13 on cetuximab vs patients with other mutations, and similar to patients with no KRAS mutations identified[65,74]. It is presently believed that anti-EGFR antigens should not be used in patients with tumours indicative of G12V mutation of KRAS at codon 12. For bevacizumab, KRAS mutation was found to be of no use as a predictive factor[75].

The same applies to BRAF mutations found in 8%-13% of patients with CRC, which are mutually exclusive with KRAS mutations. The most frequently observed BRAF mutation is V600E mutation. BRAF mutations make the tumour to a large extent resistant to anti-EGFR monoclonal antibodies, and significantly worsen prognosis, especially in patients with MSI-L and MSS[66,70,76-78]. Based on the available data, National Comprehensive Cancer Network (NCCN) suggests considering BRAF mutation testing when KRAS is mutation negative. Interestingly, good prognosis was reported even in those MSI-H CRC patients who had coincident BRAF mutations[78]. In one of studies, the OS period was shown to be slightly longer in patients on cetuximab even if the BRAF mutation was present[69]. Very limited response to vemurafenib, recently approved for metastatic melanoma patients harboring BRAF (V660E) mutation, was demonstrated in CRC patients. Researchers reported that by adding cetuximab strongly synergistic reaction with BRAF inhibitors was observed[79]. NRAS is another member of RAS proto-oncogenes which was found to be rarely mutated, while BRAF is mutually exclusive with KRAS mutations. Since NRAS mutation can predict resistance to EGFR therapy, NCCN suggests considering NRAS mutation testing when KRAS is mutation negative. To date, NRAS mutation does not appear to be associated with the prognosis[80].

Phosphatidylinositide-3-kinases (PI3K) are kinases that promote cellular proliferation. Mutations in PIK3CA gene encoding p110α catalytic subunit of PI3K have been identified in different human solid tumours, including CRC. PIK3CA gene is mutated in 10%-20% of CRC tumours. PIK3CA gene encodes the kinase that regulates, alongside with KRAS, downstream signalling pathways of EGFR. Moreover, PI3K-initiated signalling is inhibited by phosphatase and tensin homologue deleted on chromosome 10 (PTEN). Recent studies have revealed an increase in colon cancer-specific mortality in patients with PIK3CA-mutated tumours, as compared with patients with PIK3CA wild-type tumours[81,82]. However only the coexistence of PIK3CA exon 9 and 20 mutations but not PIK3CA mutation in either exon 9 or 20 alone has been reported to be associated with the worse prognosis[82]. Among patients with KRAS wild-type tumours, the presence of PIK3CA mutation correlated with a significant increase in CRC specific mortality. In contrast, PIK3CA mutation did not significantly affect mortality among patients with KRAS-mutated tumours. Thus, the effect of PIK3CA mutation may be potentially limited to patients with KRAS wild-type tumours[81]. Following the fact that only patients with KRAS-wild type CRC may respond to anti-EGFR antibodies, several studies have investigated the role of PIK3CA mutations on CRC cells response to cetuximab or panitumumab. The data collected so far indicate that CRC with PIK3CA mutations are significantly resistant to anti-EGFR antibodies. When only KRAS wild-type tumours are analyzed, the correlation is even stronger[83-85]. Changes in PIK3 signalling and loss of PTEN expression have been generally linked with the lack of response to EGFR-targeted therapy[86,87]. Recent studies have found that inhibition of cyclooxygenase-2 by regular use of aspirin after CRC diagnosis was associated with longer cancer specific survival time among patients with mutated as opposed to wild-type PIK3CA. The authors conclude that PIK3CA mutations may serve as a predictive biomarker for adjuvant aspirin therapy[88]. Further studies involving KRAS mutated CRC patients are necessary to establish the role of aspirin in PI3K pathway.

Biomarker of the potential toxicity of irinotecan

Irinotecan is a chemotherapeutic agent that inhibits topoisomerase I, thereby inhibiting replication and stimulating cell apoptosis. Advanced neutropenia and intensive diarrhoea caused by damaged intestinal epithelium are the most common adverse effects of irinotecan, which significantly limit its use. UGT1A1 gene polymorphism is a very useful biomarker of the potential toxicity of irinotecan. It appears that the use of genetic tests is reasonable before treatment initiation with irinotecan to avoid severe adverse effects - mainly neutropenia in women. Genotyping for UGT1A1 can be carried out to select a group of sensitive patients with UGT1A1*28 allele, of whom lower initial doses would be recommended. Hopefully, it will also allow to administer a higher accumulated dose of the drug, divided into smaller portions, to limit its toxicity[89,90]. However, according to a recent meta-analysis, genotyping for UGT1A1 has no predictive value in terms of responsiveness to various doses of irinotecan among patients with CRC[91]. On the other is recommended by ESMO for patients with several toxicity reaction in whom irinotecan in high doses should be used[33]. Furthermore, homozygosity for the UGT1A1*28 has been linked with improved efficacy of FOLFIRI[92].

Potential biomarkers of vascular endothelial growth factor - targeted therapy

Since the vascular endothelial growth factor (VEGF) - targeted therapy has been integrated into CRC treatment protocols, some anti-angiogenic drugs have been introduced (bevacizumab, regorafenib, aflibercept). However, a patient selection strategy to identify those patients who benefit most from this therapy has yet to be developed. To date, a predictive biomarker for bevacizumab - the most commonly administered anti-angiogenic drug in CRC therapy - has not yet been identified. Several studies on the identification of predictive biomarkers of bevacizumab have been performed. Jürgensmeier et al[93] evaluated retrospectively, using samples from randomised trial HORIZON III, the prognostic/predictive value of VEGF and soluble VEGF receptor-2. High baseline values of VEGF were associated with worse progression free survival (PFS) and overall survival. These data have revealed that baseline VEGF levels were not predictive of PFS or OS outcome in bevacizumab-treated patients[93]. Other studies have demonstrated that plasma VEGF-A may serve as a prognostic marker, but is unable to predict response to VEGF-targeted therapy in advanced CRC[94,95]. At the same time, KRAS mutation was found to be of no use as a predictive factor for bevacizumab[75].

Ezrin

Ezrin protein, a part of ezrin/radixin/moesin family may play an important role in tumour invasion process. Recent studies has found that overexpression of ezrin protein correlates with CRC aggressiveness, its metastatic potential and worse prognosis. High ezrin expression was also identified as marker of early local recurrence of rectal cancer[96,97]. Although further investigation is needed, ezrin may represent a relevant biomarker and target for personalized anti-metastatic therapies.

CONCLUSION

The recent studies result in a better understanding of colorectal cancer and assist in the development of new treatment regimens, especially in advanced CRC stages. The new predictive factors, molecular imaging, or even commercial genome tests increasingly facilitate tumour genome testing and assist in selecting targeted therapies. Adjuvant targeted therapy with anti-EGFR antibodies is required in advanced CRC patients and absence of KRAS, BRAF, NRAS and PIK3CA genes mutation. Tests for MSI or MSS tumour phenotype and the presence or absence of 18q chromosome deletion is very much desirable in standard therapy based on 5-FU. Genotyping of UGT1A1 alleles is reasonable before treatment initiation with irinotecan to avoid severe adverse effects. Further studies are necessary to identify predictive biomarker of bevacizumab. Targeted therapy against membrane receptors appears to be the future of CRC therapy. Some promising studies are now carried out in this area, dedicated to, inter alia, other EGFR ligands, insulin-like growth factor receptor 1, platelet-derived growth factor receptors and c-MET inhibitors. The aim of future research is to identify those biomarkers that can provide a non-invasive and cost-effective diagnosis, as well as to recognise the best prognostic panel of biomarkers and define the predictive biomarkers for available treatments.

Footnotes

P- Reviewer: Bajenova O S- Editor: Gong ZM L- Editor: A E- Editor: Wang CH

References
1.  Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin. 2013;63:11-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9215]  [Cited by in F6Publishing: 9719]  [Article Influence: 883.5]  [Reference Citation Analysis (3)]
2.  Labianca R, Nordlinger B, Beretta GD, Brouquet A, Cervantes A. Primary colon cancer: ESMO Clinical Practice Guidelines for diagnosis, adjuvant treatment and follow-up. Ann Oncol. 2010;21 Suppl 5:v70-v77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 217]  [Cited by in F6Publishing: 235]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
3.  Berrino F, De Angelis R, Sant M, Rosso S, Bielska-Lasota M, Coebergh JW, Santaquilani M. Survival for eight major cancers and all cancers combined for European adults diagnosed in 1995-99: results of the EUROCARE-4 study. Lancet Oncol. 2007;8:773-783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 588]  [Cited by in F6Publishing: 583]  [Article Influence: 34.3]  [Reference Citation Analysis (0)]
4.  National Cancer Institute SEER Stat Fact Sheets: Colon and rectum. 2011.  Available from: http://seer.cancer.gov/statfacts/html/colorect.html.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Langan RC, Mullinax JE, Raiji MT, Upham T, Summers T, Stojadinovic A, Avital I. Colorectal cancer biomarkers and the potential role of cancer stem cells. J Cancer. 2013;4:241-250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 60]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
6.  Lech G, Slotwinski R, Krasnodebski IW. The role of tumor markers and biomarkers in colorectal cancer. Neoplasma. 2014;61:1-8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 33]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
7.  Osborn NK, Ahlquist DA. Stool screening for colorectal cancer: molecular approaches. Gastroenterology. 2005;128:192-206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 169]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
8.  Traverso G, Shuber A, Levin B, Johnson C, Olsson L, Schoetz DJ, Hamilton SR, Boynton K, Kinzler KW, Vogelstein B. Detection of APC mutations in fecal DNA from patients with colorectal tumors. N Engl J Med. 2002;346:311-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 248]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
9.  Bosch LJ, Carvalho B, Fijneman RJ, Jimenez CR, Pinedo HM, van Engeland M, Meijer GA. Molecular tests for colorectal cancer screening. Clin Colorectal Cancer. 2011;10:8-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 80]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
10.  Ahlquist DA, Sargent DJ, Loprinzi CL, Levin TR, Rex DK, Ahnen DJ, Knigge K, Lance MP, Burgart LJ, Hamilton SR. Stool DNA and occult blood testing for screen detection of colorectal neoplasia. Ann Intern Med. 2008;149:441-50, W81.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Calistri D, Rengucci C, Molinari C, Ricci E, Cavargini E, Scarpi E, Milandri GL, Fabbri C, Ravaioli A, Russo A. Quantitative fluorescence determination of long-fragment DNA in stool as a marker for the early detection of colorectal cancer. Cell Oncol. 2009;31:11-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 10]  [Reference Citation Analysis (0)]
12.  Diehl F, Schmidt K, Durkee KH, Moore KJ, Goodman SN, Shuber AP, Kinzler KW, Vogelstein B. Analysis of mutations in DNA isolated from plasma and stool of colorectal cancer patients. Gastroenterology. 2008;135:489-498.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 168]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
13.  Leung WK, To KF, Man EP, Chan MW, Hui AJ, Ng SS, Lau JY, Sung JJ. Detection of hypermethylated DNA or cyclooxygenase-2 messenger RNA in fecal samples of patients with colorectal cancer or polyps. Am J Gastroenterol. 2007;102:1070-1076.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 84]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
14.  Nagasaka T, Tanaka N, Cullings HM, Sun DS, Sasamoto H, Uchida T, Koi M, Nishida N, Naomoto Y, Boland CR. Analysis of fecal DNA methylation to detect gastrointestinal neoplasia. J Natl Cancer Inst. 2009;101:1244-1258.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 107]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
15.  Koga Y, Yasunaga M, Moriya Y, Akasu T, Fujita S, Yamamoto S, Kozu T, Baba H, Matsumura Y. Detection of colorectal cancer cells from feces using quantitative real-time RT-PCR for colorectal cancer diagnosis. Cancer Sci. 2008;99:1977-1983.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 20]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
16.  Koga Y, Yasunaga M, Takahashi A, Kuroda J, Moriya Y, Akasu T, Fujita S, Yamamoto S, Baba H, Matsumura Y. MicroRNA expression profiling of exfoliated colonocytes isolated from feces for colorectal cancer screening. Cancer Prev Res (Phila). 2010;3:1435-1442.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 162]  [Article Influence: 11.6]  [Reference Citation Analysis (1)]
17.  Link A, Balaguer F, Shen Y, Nagasaka T, Lozano JJ, Boland CR, Goel A. Fecal MicroRNAs as novel biomarkers for colon cancer screening. Cancer Epidemiol Biomarkers Prev. 2010;19:1766-1774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 242]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
18.  Wang JY, Wu CH, Lu CY, Hsieh JS, Wu DC, Huang SY, Lin SR. Molecular detection of circulating tumor cells in the peripheral blood of patients with colorectal cancer using RT-PCR: significance of the prediction of postoperative metastasis. World J Surg. 2006;30:1007-1013.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 92]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
19.  Shen C, Hu L, Xia L, Li Y. Quantitative real-time RT-PCR detection for survivin, CK20 and CEA in peripheral blood of colorectal cancer patients. Jpn J Clin Oncol. 2008;38:770-776.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 45]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
20.  Dandachi N, Balic M, Stanzer S, Halm M, Resel M, Hinterleitner TA, Samonigg H, Bauernhofer T. Critical evaluation of real-time reverse transcriptase-polymerase chain reaction for the quantitative detection of cytokeratin 20 mRNA in colorectal cancer patients. J Mol Diagn. 2005;7:631-637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 32]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
21.  Huang Z, Huang D, Ni S, Peng Z, Sheng W, Du X. Plasma microRNAs are promising novel biomarkers for early detection of colorectal cancer. Int J Cancer. 2010;127:118-126.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Gold P, Freedman SO. Demonstration of tumor-specific antigens in human colonic carcinomata by immunological tolerance and absorption techniques. J Exp Med. 1965;121:439-462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1572]  [Cited by in F6Publishing: 1505]  [Article Influence: 53.8]  [Reference Citation Analysis (0)]
23.  Locker GY, Hamilton S, Harris J, Jessup JM, Kemeny N, Macdonald JS, Somerfield MR, Hayes DF, Bast RC. ASCO 2006 update of recommendations for the use of tumor markers in gastrointestinal cancer. J Clin Oncol. 2006;24:5313-5327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1057]  [Cited by in F6Publishing: 1042]  [Article Influence: 57.9]  [Reference Citation Analysis (0)]
24.  Duffy MJ, Lamerz R, Haglund C, Nicolini A, Kalousová M, Holubec L, Sturgeon C. Tumor markers in colorectal cancer, gastric cancer and gastrointestinal stromal cancers: European group on tumor markers 2014 guidelines update. Int J Cancer. 2014;134:2513-2522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 224]  [Article Influence: 20.4]  [Reference Citation Analysis (1)]
25.  Chen CC, Yang SH, Lin JK, Lin TC, Chen WS, Jiang JK, Wang HS, Chang SC. Is it reasonable to add preoperative serum level of CEA and CA19-9 to staging for colorectal cancer? J Surg Res. 2005;124:169-174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 82]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
26.  Weissenberger C, Von Plehn G, Otto F, Barke A, Momm F, Geissler M. Adjuvant radiochemotherapy of stage II and III rectal adenocarcinoma: role of CEA and CA 19-9. Anticancer Res. 2005;25:1787-1793.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Compton C, Fenoglio-Preiser CM, Pettigrew N, Fielding LP. American Joint Committee on Cancer Prognostic Factors Consensus Conference: Colorectal Working Group. Cancer. 2000;88:1739-1757.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
28.  Thirunavukarasu P, Sukumar S, Sathaiah M, Mahan M, Pragatheeshwar KD, Pingpank JF, Zeh H, Bartels CJ, Lee KK, Bartlett DL. C-stage in colon cancer: implications of carcinoembryonic antigen biomarker in staging, prognosis, and management. J Natl Cancer Inst. 2011;103:689-697.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 123]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
29.  Ueno H, Mochizuki H, Hatsuse K, Hase K, Yamamoto T. Indicators for treatment strategies of colorectal liver metastases. Ann Surg. 2000;231:59-66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 118]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
30.  Mala T, Bøhler G, Mathisen Ø, Bergan A, Søreide O. Hepatic resection for colorectal metastases: can preoperative scoring predict patient outcome? World J Surg. 2002;26:1348-1353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 78]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
31.  Giessen C, Nagel D, Glas M, Spelsberg F, Lau-Werner U, Modest DP, Michl M, Heinemann V, Stieber P, Schulz C. Evaluation of preoperative serum markers for individual patient prognosis in stage I-III rectal cancer. Tumour Biol. 2014;35:10237-10248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 33]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
32.  Goldstein MJ, Mitchell EP. Carcinoembryonic antigen in the staging and follow-up of patients with colorectal cancer. Cancer Invest. 2005;23:338-351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 265]  [Cited by in F6Publishing: 264]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
33.  Schmoll HJ, Van Cutsem E, Stein A, Valentini V, Glimelius B, Haustermans K, Nordlinger B, van de Velde CJ, Balmana J, Regula J. ESMO Consensus Guidelines for management of patients with colon and rectal cancer. a personalized approach to clinical decision making. Ann Oncol. 2012;23:2479-2516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1035]  [Cited by in F6Publishing: 1034]  [Article Influence: 86.2]  [Reference Citation Analysis (1)]
34.  Nicolini A, Ferrari P, Duffy MJ, Antonelli A, Rossi G, Metelli MR, Fulceri F, Anselmi L, Conte M, Berti P. Intensive risk-adjusted follow-up with the CEA, TPA, CA19.9, and CA72.4 tumor marker panel and abdominal ultrasonography to diagnose operable colorectal cancer recurrences: effect on survival. Arch Surg. 2010;145:1177-1183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 34]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
35.  Carpelan-Holmström M, Louhimo J, Stenman UH, Alfthan H, Järvinen H, Haglund C. CEA, CA 242, CA 19-9, CA 72-4 and hCGbeta in the diagnosis of recurrent colorectal cancer. Tumour Biol. 2004;25:228-234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 40]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
36.  Lumachi F, Marino F, Orlando R, Chiara GB, Basso SM. Simultaneous multianalyte immunoassay measurement of five serum tumor markers in the detection of colorectal cancer. Anticancer Res. 2012;32:985-988.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Levy M, Visokai V, Lipska L, Topolcan O. Tumor markers in staging and prognosis of colorectal carcinoma. Neoplasma. 2008;55:138-142.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Marrelli D, Caruso S, Neri A, Pedrazzani C, Capuano L, Mazzei MA, Roviello F. Clinical utility of serum tumor markers in the diagnosis of malignant intestinal occlusion. A prospective observational study. Int J Biol Markers. 2011;26:58-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
39.  Zhong W, Yu Z, Zhan J, Yu T, Lin Y, Xia ZS, Yuan YH, Chen QK. Association of serum levels of CEA, CA199, CA125, CYFRA21-1 and CA72-4 and disease characteristics in colorectal cancer. Pathol Oncol Res. 2015;21:83-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 45]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
40.  Koss K, Maxton D, Jankowski JA. Faecal dimeric M2 pyruvate kinase in colorectal cancer and polyps correlates with tumour staging and surgical intervention. Colorectal Dis. 2008;10:244-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 45]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
41.  Mulder SA, van Leerdam ME, van Vuuren AJ, Francke J, van Toorenenbergen AW, Kuipers EJ, Ouwendijk RJ. Tumor pyruvate kinase isoenzyme type M2 and immunochemical fecal occult blood test: performance in screening for colorectal cancer. Eur J Gastroenterol Hepatol. 2007;19:878-882.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Nielsen HJ, Brünner N, Jorgensen LN, Olsen J, Rahr HB, Thygesen K, Hoyer U, Laurberg S, Stieber P, Blankenstein MA. Plasma TIMP-1 and CEA in detection of primary colorectal cancer: a prospective, population based study of 4509 high-risk individuals. Scand J Gastroenterol. 2011;46:60-69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 63]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
43.  Byström P, Berglund Å, Nygren P, Wernroth L, Johansson B, Larsson A, Glimelius B. Evaluation of predictive markers for patients with advanced colorectal cancer. Acta Oncol. 2012;51:849-859.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 34]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
44.  Holdenrieder S, Stieber P, Liska V, Treska V, Topolcan O, Dreslerova J, Matejka VM, Finek J, Holubec L. Cytokeratin serum biomarkers in patients with colorectal cancer. Anticancer Res. 2012;32:1971-1976.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Baba Y, Nosho K, Shima K, Irahara N, Kure S, Toyoda S, Kirkner GJ, Goel A, Fuchs CS, Ogino S. Aurora-A expression is independently associated with chromosomal instability in colorectal cancer. Neoplasia. 2009;11:418-425.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Popat S, Hubner R, Houlston RS. Systematic review of microsatellite instability and colorectal cancer prognosis. J Clin Oncol. 2005;23:609-618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1267]  [Cited by in F6Publishing: 1270]  [Article Influence: 66.8]  [Reference Citation Analysis (1)]
47.  Samowitz WS, Curtin K, Ma KN, Schaffer D, Coleman LW, Leppert M, Slattery ML. Microsatellite instability in sporadic colon cancer is associated with an improved prognosis at the population level. Cancer Epidemiol Biomarkers Prev. 2001;10:917-923.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Ribic CM, Sargent DJ, Moore MJ, Thibodeau SN, French AJ, Goldberg RM, Hamilton SR, Laurent-Puig P, Gryfe R, Shepherd LE. Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer. N Engl J Med. 2003;349:247-257.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1585]  [Cited by in F6Publishing: 1563]  [Article Influence: 74.4]  [Reference Citation Analysis (0)]
49.  Kim GP, Colangelo LH, Wieand HS, Paik S, Kirsch IR, Wolmark N, Allegra CJ. Prognostic and predictive roles of high-degree microsatellite instability in colon cancer: a National Cancer Institute-National Surgical Adjuvant Breast and Bowel Project Collaborative Study. J Clin Oncol. 2007;25:767-772.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 252]  [Cited by in F6Publishing: 264]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
50.  Sinicrope FA, Sargent DJ. Clinical implications of microsatellite instability in sporadic colon cancers. Curr Opin Oncol. 2009;21:369-373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 68]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
51.  Meyers M, Wagner MW, Hwang HS, Kinsella TJ, Boothman DA. Role of the hMLH1 DNA mismatch repair protein in fluoropyrimidine-mediated cell death and cell cycle responses. Cancer Res. 2001;61:5193-5201.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Elsaleh H, Iacopetta B. Microsatellite instability is a predictive marker for survival benefit from adjuvant chemotherapy in a population-based series of stage III colorectal carcinoma. Clin Colorectal Cancer. 2001;1:104-109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 58]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
53.  Elsaleh H, Joseph D, Grieu F, Zeps N, Spry N, Iacopetta B. Association of tumour site and sex with survival benefit from adjuvant chemotherapy in colorectal cancer. Lancet. 2000;355:1745-1750.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 406]  [Cited by in F6Publishing: 438]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
54.  Hutchins G, Southward K, Handley K, Magill L, Beaumont C, Stahlschmidt J, Richman S, Chambers P, Seymour M, Kerr D. Value of mismatch repair, KRAS, and BRAF mutations in predicting recurrence and benefits from chemotherapy in colorectal cancer. J Clin Oncol. 2011;29:1261-1270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 473]  [Cited by in F6Publishing: 494]  [Article Influence: 38.0]  [Reference Citation Analysis (0)]
55.  Bertagnolli MM, Niedzwiecki D, Compton CC, Hahn HP, Hall M, Damas B, Jewell SD, Mayer RJ, Goldberg RM, Saltz LB. Microsatellite instability predicts improved response to adjuvant therapy with irinotecan, fluorouracil, and leucovorin in stage III colon cancer: Cancer and Leukemia Group B Protocol 89803. J Clin Oncol. 2009;27:1814-1821.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 279]  [Cited by in F6Publishing: 305]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
56.  Sarli L, Bottarelli L, Bader G, Iusco D, Pizzi S, Costi R, D’Adda T, Bertolani M, Roncoroni L, Bordi C. Association between recurrence of sporadic colorectal cancer, high level of microsatellite instability, and loss of heterozygosity at chromosome 18q. Dis Colon Rectum. 2004;47:1467-1482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 37]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
57.  Popat S, Houlston RS. A systematic review and meta-analysis of the relationship between chromosome 18q genotype, DCC status and colorectal cancer prognosis. Eur J Cancer. 2005;41:2060-2070.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 139]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
58.  Popat S, Zhao D, Chen Z, Pan H, Shao Y, Chandler I, Houlston RS. Relationship between chromosome 18q status and colorectal cancer prognosis: a prospective, blinded analysis of 280 patients. Anticancer Res. 2007;27:627-633.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Ogino S, Nosho K, Irahara N, Shima K, Baba Y, Kirkner GJ, Meyerhardt JA, Fuchs CS. Prognostic significance and molecular associations of 18q loss of heterozygosity: a cohort study of microsatellite stable colorectal cancers. J Clin Oncol. 2009;27:4591-4598.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 95]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
60.  Watanabe T, Wu TT, Catalano PJ, Ueki T, Satriano R, Haller DG, Benson AB, Hamilton SR. Molecular predictors of survival after adjuvant chemotherapy for colon cancer. N Engl J Med. 2001;344:1196-1206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 648]  [Cited by in F6Publishing: 631]  [Article Influence: 27.4]  [Reference Citation Analysis (0)]
61.  Watanabe T, Kobunai T, Yamamoto Y, Matsuda K, Ishihara S, Nozawa K, Yamada H, Hayama T, Inoue E, Tamura J. Chromosomal instability (CIN) phenotype, CIN high or CIN low, predicts survival for colorectal cancer. J Clin Oncol. 2012;30:2256-2264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 76]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
62.   Available from: http://clinicaltrials.gov/ct2/show/study/NCT00217737?Term=ecog 5202&rank=1.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Tejpar S, Bertagnolli M, Bosman F, Lenz HJ, Garraway L, Waldman F, Warren R, Bild A, Collins-Brennan D, Hahn H. Prognostic and predictive biomarkers in resected colon cancer: current status and future perspectives for integrating genomics into biomarker discovery. Oncologist. 2010;15:390-404.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 129]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
64.  Russo A, Bazan V, Iacopetta B, Kerr D, Soussi T, Gebbia N; TP53-CRC Collaborative Study Group. The TP53 colorectal cancer international collaborative study on the prognostic and predictive significance of p53 mutation: influence of tumor site, type of mutation, and adjuvant treatment. J Clin Oncol. 2005;23:7518-7528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 277]  [Cited by in F6Publishing: 272]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
65.  De Roock W, Jonker DJ, Di Nicolantonio F, Sartore-Bianchi A, Tu D, Siena S, Lamba S, Arena S, Frattini M, Piessevaux H. Association of KRAS p.G13D mutation with outcome in patients with chemotherapy-refractory metastatic colorectal cancer treated with cetuximab. JAMA. 2010;304:1812-1820.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 570]  [Cited by in F6Publishing: 561]  [Article Influence: 40.1]  [Reference Citation Analysis (0)]
66.  Markowitz SD, Bertagnolli MM. Molecular origins of cancer: Molecular basis of colorectal cancer. N Engl J Med. 2009;361:2449-2460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1274]  [Cited by in F6Publishing: 1304]  [Article Influence: 86.9]  [Reference Citation Analysis (2)]
67.  Di Fiore F, Blanchard F, Charbonnier F, Le Pessot F, Lamy A, Galais MP, Bastit L, Killian A, Sesboüé R, Tuech JJ. Clinical relevance of KRAS mutation detection in metastatic colorectal cancer treated by Cetuximab plus chemotherapy. Br J Cancer. 2007;96:1166-1169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 576]  [Cited by in F6Publishing: 616]  [Article Influence: 36.2]  [Reference Citation Analysis (0)]
68.  Lièvre A, Bachet JB, Boige V, Cayre A, Le Corre D, Buc E, Ychou M, Bouché O, Landi B, Louvet C. KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab. J Clin Oncol. 2008;26:374-379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1117]  [Cited by in F6Publishing: 1107]  [Article Influence: 69.2]  [Reference Citation Analysis (0)]
69.  Van Cutsem E, Köhne CH, Láng I, Folprecht G, Nowacki MP, Cascinu S, Shchepotin I, Maurel J, Cunningham D, Tejpar S. Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treatment for metastatic colorectal cancer: updated analysis of overall survival according to tumor KRAS and BRAF mutation status. J Clin Oncol. 2011;29:2011-2019.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1314]  [Cited by in F6Publishing: 1452]  [Article Influence: 111.7]  [Reference Citation Analysis (0)]
70.  Bokemeyer C, Van Cutsem E, Rougier P, Ciardiello F, Heeger S, Schlichting M, Celik I, Köhne CH. Addition of cetuximab to chemotherapy as first-line treatment for KRAS wild-type metastatic colorectal cancer: pooled analysis of the CRYSTAL and OPUS randomised clinical trials. Eur J Cancer. 2012;48:1466-1475.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 393]  [Cited by in F6Publishing: 417]  [Article Influence: 34.8]  [Reference Citation Analysis (0)]
71.  Douillard JY, Siena S, Cassidy J, Tabernero J, Burkes R, Barugel M, Humblet Y, Bodoky G, Cunningham D, Jassem J. Randomized, phase III trial of panitumumab with infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first-line treatment in patients with previously untreated metastatic colorectal cancer: the PRIME study. J Clin Oncol. 2010;28:4697-4705.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1296]  [Cited by in F6Publishing: 1336]  [Article Influence: 95.4]  [Reference Citation Analysis (0)]
72.  Maughan TS, Adams RA, Smith CG, Meade AM, Seymour MT, Wilson RH, Idziaszczyk S, Harris R, Fisher D, Kenny SL. Addition of cetuximab to oxaliplatin-based first-line combination chemotherapy for treatment of advanced colorectal cancer: results of the randomised phase 3 MRC COIN trial. Lancet. 2011;377:2103-2114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 702]  [Cited by in F6Publishing: 731]  [Article Influence: 56.2]  [Reference Citation Analysis (2)]
73.  Tveit KM, Guren T, Glimelius B, Pfeiffer P, Sorbye H, Pyrhonen S, Sigurdsson F, Kure E, Ikdahl T, Skovlund E. Phase III trial of cetuximab with continuous or intermittent fluorouracil, leucovorin, and oxaliplatin (Nordic FLOX) versus FLOX alone in first-line treatment of metastatic colorectal cancer: the NORDIC-VII study. J Clin Oncol. 2012;30:1755-1762.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 398]  [Cited by in F6Publishing: 407]  [Article Influence: 33.9]  [Reference Citation Analysis (0)]
74.  Tejpar S, Celik I, Schlichting M, Sartorius U, Bokemeyer C, Van Cutsem E. Association of KRAS G13D tumor mutations with outcome in patients with metastatic colorectal cancer treated with first-line chemotherapy with or without cetuximab. J Clin Oncol. 2012;30:3570-3577.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 250]  [Cited by in F6Publishing: 272]  [Article Influence: 22.7]  [Reference Citation Analysis (0)]
75.  Price TJ, Hardingham JE, Lee CK, Weickhardt A, Townsend AR, Wrin JW, Chua A, Shivasami A, Cummins MM, Murone C. Impact of KRAS and BRAF Gene Mutation Status on Outcomes From the Phase III AGITG MAX Trial of Capecitabine Alone or in Combination With Bevacizumab and Mitomycin in Advanced Colorectal Cancer. J Clin Oncol. 2011;29:2675-2682.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 166]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
76.  Di Nicolantonio F, Martini M, Molinari F, Sartore-Bianchi A, Arena S, Saletti P, De Dosso S, Mazzucchelli L, Frattini M, Siena S. Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. J Clin Oncol. 2008;26:5705-5712.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1211]  [Cited by in F6Publishing: 1218]  [Article Influence: 76.1]  [Reference Citation Analysis (0)]
77.  Roth AD, Tejpar S, Delorenzi M, Yan P, Fiocca R, Klingbiel D, Dietrich D, Biesmans B, Bodoky G, Barone C. Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: results of the translational study on the PETACC-3, EORTC 40993, SAKK 60-00 trial. J Clin Oncol. 2010;28:466-474.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 801]  [Cited by in F6Publishing: 880]  [Article Influence: 58.7]  [Reference Citation Analysis (0)]
78.  Siena S, Sartore-Bianchi A, Di Nicolantonio F, Balfour J, Bardelli A. Biomarkers predicting clinical outcome of epidermal growth factor receptor-targeted therapy in metastatic colorectal cancer. J Natl Cancer Inst. 2009;101:1308-1324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 386]  [Cited by in F6Publishing: 417]  [Article Influence: 27.8]  [Reference Citation Analysis (0)]
79.  Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D, Beijersbergen RL, Bardelli A, Bernards R. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature. 2012;483:100-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1390]  [Cited by in F6Publishing: 1456]  [Article Influence: 121.3]  [Reference Citation Analysis (0)]
80.  Irahara N, Baba Y, Nosho K, Shima K, Yan L, Dias-Santagata D, Iafrate AJ, Fuchs CS, Haigis KM, Ogino S. NRAS mutations are rare in colorectal cancer. Diagn Mol Pathol. 2010;19:157-163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 142]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
81.  Ogino S, Nosho K, Kirkner GJ, Shima K, Irahara N, Kure S, Chan AT, Engelman JA, Kraft P, Cantley LC. PIK3CA mutation is associated with poor prognosis among patients with curatively resected colon cancer. J Clin Oncol. 2009;27:1477-1484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 249]  [Cited by in F6Publishing: 264]  [Article Influence: 17.6]  [Reference Citation Analysis (1)]
82.  Liao X, Morikawa T, Lochhead P, Imamura Y, Kuchiba A, Yamauchi M, Nosho K, Qian ZR, Nishihara R, Meyerhardt JA. Prognostic role of PIK3CA mutation in colorectal cancer: cohort study and literature review. Clin Cancer Res. 2012;18:2257-2268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 188]  [Cited by in F6Publishing: 214]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
83.  Jhawer M, Goel S, Wilson AJ, Montagna C, Ling YH, Byun DS, Nasser S, Arango D, Shin J, Klampfer L. PIK3CA mutation/PTEN expression status predicts response of colon cancer cells to the epidermal growth factor receptor inhibitor cetuximab. Cancer Res. 2008;68:1953-1961.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 354]  [Cited by in F6Publishing: 378]  [Article Influence: 23.6]  [Reference Citation Analysis (0)]
84.  Sartore-Bianchi A, Martini M, Molinari F, Veronese S, Nichelatti M, Artale S, Di Nicolantonio F, Saletti P, De Dosso S, Mazzucchelli L. PIK3CA mutations in colorectal cancer are associated with clinical resistance to EGFR-targeted monoclonal antibodies. Cancer Res. 2009;69:1851-1857.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 560]  [Cited by in F6Publishing: 577]  [Article Influence: 38.5]  [Reference Citation Analysis (0)]
85.  De Roock W, Claes B, Bernasconi D, De Schutter J, Biesmans B, Fountzilas G, Kalogeras KT, Kotoula V, Papamichael D, Laurent-Puig P. Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol. 2010;11:753-762.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1491]  [Cited by in F6Publishing: 1585]  [Article Influence: 113.2]  [Reference Citation Analysis (1)]
86.  Sood A, McClain D, Maitra R, Basu-Mallick A, Seetharam R, Kaubisch A, Rajdev L, Mariadason JM, Tanaka K, Goel S. PTEN gene expression and mutations in the PIK3CA gene as predictors of clinical benefit to anti-epidermal growth factor receptor antibody therapy in patients with KRAS wild-type metastatic colorectal cancer. Clin Colorectal Cancer. 2012;11:143-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 81]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
87.  Grady WM, Pritchard CC. Molecular alterations and biomarkers in colorectal cancer. Toxicol Pathol. 2014;42:124-139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 64]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
88.  Liao X, Lochhead P, Nishihara R, Morikawa T, Kuchiba A, Yamauchi M, Imamura Y, Qian ZR, Baba Y, Shima K. Aspirin use, tumor PIK3CA mutation, and colorectal-cancer survival. N Engl J Med. 2012;367:1596-1606.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
89.  Shulman K, Cohen I, Barnett-Griness O, Kuten A, Gruber SB, Lejbkowicz F, Rennert G. Clinical implications of UGT1A1*28 genotype testing in colorectal cancer patients. Cancer. 2011;117:3156-3162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 63]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
90.  Rouits E, Charasson V, Pétain A, Boisdron-Celle M, Delord JP, Fonck M, Laurand A, Poirier AL, Morel A, Chatelut E. Pharmacokinetic and pharmacogenetic determinants of the activity and toxicity of irinotecan in metastatic colorectal cancer patients. Br J Cancer. 2008;99:1239-1245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 47]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
91.  Dias MM, McKinnon RA, Sorich MJ. Impact of the UGT1A1*28 allele on response to irinotecan: a systematic review and meta-analysis. Pharmacogenomics. 2012;13:889-899.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 46]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
92.  Cecchin E, Innocenti F, D’Andrea M, Corona G, De Mattia E, Biason P, Buonadonna A, Toffoli G. Predictive role of the UGT1A1, UGT1A7, and UGT1A9 genetic variants and their haplotypes on the outcome of metastatic colorectal cancer patients treated with fluorouracil, leucovorin, and irinotecan. J Clin Oncol. 2009;27:2457-2465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 161]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
93.  Jürgensmeier JM, Schmoll HJ, Robertson JD, Brooks L, Taboada M, Morgan SR, Wilson D, Hoff PM. Prognostic and predictive value of VEGF, sVEGFR-2 and CEA in mCRC studies comparing cediranib, bevacizumab and chemotherapy. Br J Cancer. 2013;108:1316-1323.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 50]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
94.  Jubb AM, Harris AL. Biomarkers to predict the clinical efficacy of bevacizumab in cancer. Lancet Oncol. 2010;11:1172-1183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 237]  [Cited by in F6Publishing: 231]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
95.  Luo HY, Xu RH. Predictive and prognostic biomarkers with therapeutic targets in advanced colorectal cancer. World J Gastroenterol. 2014;20:3858-3874.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
96.  Patara M, Santos EM, Coudry Rde A, Soares FA, Ferreira FO, Rossi BM. Ezrin expression as a prognostic marker in colorectal adenocarcinoma. Pathol Oncol Res. 2011;17:827-833.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
97.  Jörgren F, Nilbert M, Rambech E, Bendahl PO, Lindmark G. Ezrin expression in rectal cancer predicts time to development of local recurrence. Int J Colorectal Dis. 2012;27:893-899.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
98.  Lenz HJ Established biomarkers for colon cancer. In: American Society of Clinical Oncology 2009; ASCO, 2009: 215-219.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Ellis LM Promising future biomarkers for colorectal cancer. In: American Society of Clinical Oncology 2009; ASCO, 2009: 212-214.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Engstrom PF, Arnoletti JP, Benson AB, Chen YJ, Choti MA, Cooper HS, Covey A, Dilawari RA, Early DS, Enzinger PC. NCCN Clinical Practice Guidelines in Oncology: colon cancer. J Natl Compr Canc Netw. 2009;7:778-831.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Rose J, Augestad KM, Cooper GS. Colorectal cancer surveillance: what’s new and what’s next. World J Gastroenterol. 2014;20:1887-1897.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]