+ All Categories
Home > Documents > Biomarkers for gastric cancer: prognostic, predictive or targets of therapy?

Biomarkers for gastric cancer: prognostic, predictive or targets of therapy?

Date post: 23-Dec-2016
Category:
Upload: fatima
View: 213 times
Download: 1 times
Share this document with a friend
12
INVITED REVIEW Biomarkers for gastric cancer: prognostic, predictive or targets of therapy? Cecília Durães & Gabriela M. Almeida & Raquel Seruca & Carla Oliveira & Fátima Carneiro Received: 1 October 2013 /Revised: 12 November 2013 /Accepted: 23 December 2013 # Springer-Verlag Berlin Heidelberg 2014 Abstract Gastric cancer is an aggressive disease often diag- nosed at an advanced stage. Despite improvements in surgical and adjuvant treatment approaches, gastric cancer remains a global public health problem with a 5-year overall survival of less than 25 %. This is a heterogeneous disease, both in terms of biology and genetics, and many prognostic biomarkers have been pointed out in the literature; nevertheless, their application remains debatable. In this review, we opted to give relevance to those biomarkers that have been the subject of studies with significant statistical power, which have been replicated and have been/are in targeted therapy clinical trials and, which as a consequence, have their prognostic and/or predictive value established. Some gastric cancer biomarkers that may help in defining the course of treatment are also discussed. Accepted practical guidelines, wet-lab protocols for the detection of these biomarkers, as well as ongoing and completed clinical trials have been compiled. In summary, clinical approaches based on the combination of correct stag- ing with targeted and conventional systemic therapies may improve gastric cancer patientsoutcome, but are only in their infancy. Some major challenges in identifying reliable prognostic/predictive biomarkers are individual genetic vari- ation and tumour heterogeneity that often influence response to therapy and drug resistance. Prognostic and predictive biomarkers may nevertheless be extremely valuable to cor- rectly stratify gastric cancer patients for treatment and, ulti- mately, improve survival. Keywords Gastric cancer . Biomarkers . Prognostic . Predictive . Targeted therapy . Patient stratification Introduction Gastric cancer (GC), although declining in incidence in the last decades, is still the fourth most common malignancy and the second leading cause of cancer-related death worldwide [1, 2]. This disease affects close to one million people per year, particularly in Eastern Asian countries and Western Europe [2]. GC is a heterogeneous disease, both in terms of biology and genetics. Histologically, the main variants are the intesti- nal type, with clearly defined glandular structures and the diffuse type consisting of individually infiltrating neoplastic cells [3, 4]. About 90 % of the GCs are sporadic, whereas the remaining 10 % show familial clustering. The latter includes 13 % of hereditary forms of intestinal and diffuse GC (HDGC-hereditary diffuse gastric cancer) [5]. Germline mu- tations and large deletions of CDH1 (E-cadherin gene) are the underlying genetic defect in 45 % of families with clinical diagnosis of HDGC [6]. An endoscopic ultrasound performed concomitantly with esophagoduodenoscopy has high diagnostic accuracy for dif- ferent GC stages. A preoperative laparoscopy can also be performed to improve staging diagnosis [7]. An accurate preoperative staging of nodal involvement (N stage) is cur- rently a diagnostic challenge in GC, for which new sentinel lymph node mapping technologies are being tested [8, 9]. Multi-detector computer tomography is the gold standard for the detection of distant metastases [7]. Despite improvements in surgical and adjuvant treatment approaches, GC remains a global public health problem. Sur- gical resection offers the best chance for curative therapy, with 5-year survival of ca. 5070 %, if an early diagnosis is made. However, most newly diagnosed patients present with C. Durães : G. M. Almeida : R. Seruca : C. Oliveira : F. Carneiro Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal R. Seruca : C. Oliveira (*) : F. Carneiro (*) Pathology and Oncology Department, Faculty of Medicine of the University of Porto, Porto, Portugal e-mail: [email protected] e-mail: [email protected] Virchows Arch DOI 10.1007/s00428-013-1533-y
Transcript
Page 1: Biomarkers for gastric cancer: prognostic, predictive or targets of therapy?

INVITED REVIEW

Biomarkers for gastric cancer: prognostic, predictiveor targets of therapy?

Cecília Durães & Gabriela M. Almeida & Raquel Seruca &

Carla Oliveira & Fátima Carneiro

Received: 1 October 2013 /Revised: 12 November 2013 /Accepted: 23 December 2013# Springer-Verlag Berlin Heidelberg 2014

Abstract Gastric cancer is an aggressive disease often diag-nosed at an advanced stage. Despite improvements in surgicaland adjuvant treatment approaches, gastric cancer remains aglobal public health problem with a 5-year overall survival ofless than 25 %. This is a heterogeneous disease, both in termsof biology and genetics, and many prognostic biomarkershave been pointed out in the literature; nevertheless, theirapplication remains debatable. In this review, we opted to giverelevance to those biomarkers that have been the subject ofstudies with significant statistical power, which have beenreplicated and have been/are in targeted therapy clinical trialsand, which as a consequence, have their prognostic and/orpredictive value established. Some gastric cancer biomarkersthat may help in defining the course of treatment are alsodiscussed. Accepted practical guidelines, wet-lab protocolsfor the detection of these biomarkers, as well as ongoing andcompleted clinical trials have been compiled. In summary,clinical approaches based on the combination of correct stag-ing with targeted and conventional systemic therapies mayimprove gastric cancer patients’ outcome, but are only in theirinfancy. Some major challenges in identifying reliableprognostic/predictive biomarkers are individual genetic vari-ation and tumour heterogeneity that often influence responseto therapy and drug resistance. Prognostic and predictivebiomarkers may nevertheless be extremely valuable to cor-rectly stratify gastric cancer patients for treatment and, ulti-mately, improve survival.

Keywords Gastric cancer . Biomarkers . Prognostic .

Predictive . Targeted therapy . Patient stratification

Introduction

Gastric cancer (GC), although declining in incidence in thelast decades, is still the fourth most common malignancy andthe second leading cause of cancer-related death worldwide[1, 2]. This disease affects close to onemillion people per year,particularly in Eastern Asian countries and Western Europe[2]. GC is a heterogeneous disease, both in terms of biologyand genetics. Histologically, the main variants are the intesti-nal type, with clearly defined glandular structures and thediffuse type consisting of individually infiltrating neoplasticcells [3, 4]. About 90 % of the GCs are sporadic, whereas theremaining 10 % show familial clustering. The latter includes1–3 % of hereditary forms of intestinal and diffuse GC(HDGC-hereditary diffuse gastric cancer) [5]. Germline mu-tations and large deletions of CDH1 (E-cadherin gene) are theunderlying genetic defect in 45 % of families with clinicaldiagnosis of HDGC [6].

An endoscopic ultrasound performed concomitantly withesophagoduodenoscopy has high diagnostic accuracy for dif-ferent GC stages. A preoperative laparoscopy can also beperformed to improve staging diagnosis [7]. An accuratepreoperative staging of nodal involvement (N stage) is cur-rently a diagnostic challenge in GC, for which new sentinellymph node mapping technologies are being tested [8, 9].Multi-detector computer tomography is the gold standard forthe detection of distant metastases [7].

Despite improvements in surgical and adjuvant treatmentapproaches, GC remains a global public health problem. Sur-gical resection offers the best chance for curative therapy, with5-year survival of ca. 50–70 %, if an early diagnosis is made.However, most newly diagnosed patients present with

C. Durães :G. M. Almeida : R. Seruca : C. Oliveira : F. CarneiroInstitute of Molecular Pathology and Immunology of the Universityof Porto (IPATIMUP), Porto, Portugal

R. Seruca :C. Oliveira (*) : F. Carneiro (*)Pathology and Oncology Department, Faculty of Medicineof the University of Porto, Porto, Portugale-mail: [email protected]: [email protected]

Virchows ArchDOI 10.1007/s00428-013-1533-y

Page 2: Biomarkers for gastric cancer: prognostic, predictive or targets of therapy?

advanced, and unresectable, disease and 5-year survival dropsto 4–10 %. For these patients, chemotherapy is the maintreatment option; nevertheless, the median overall survival(OS) for advanced disease remains below 1 year, comparedwith best supportive care (3–5 months) [10, 11].

The aim of this review is to provide up-to-date informationabout prognostic and predictive biomarkers for GC patients. Acancer prognostic biomarker provides information on thelikely course of the disease. In contrast, a predictive biomarkeris defined as a marker which can be used to identify subpop-ulations of patients who are most likely to respond (or not) to atargeted therapy [12]. The search for cancer biomarkers iscarried out in order to identify tumour cells at early stagesand predict treatment response, ultimately leading to afavourable therapeutic outcome.

The literature concerning GC prognostic biomarkers israther vast. In this review, we opted to give relevance to thosebiomarkers that have been the subject of studies with signif-icant statistical power, which have been replicated and havebeen/are used in targeted therapy clinical trials and, which as aconsequence, have their prognostic and/or predictive valueestablished. Some GC biomarkers are neither drug targetsnor predictive markers. Nevertheless, they may be useful todefine the course of treatment, such as surgery, administrationof systemic therapy and/or radiotherapy and will, therefore, bediscussed (Table 1).

Prognostic and predictive biomarkers for gastric cancer

The TNM staging (tumour, lymph nodes and metastasis) atdiagnosis has been the most important tool used to assessprognosis and predict the need for systemic treatment inresectable GC [13]. However, as this approach has shownlimited success, molecular biomarkers with either prognosticor predictive value have been comprehensively investigated inthe last decade and are the main focus of this review.

TNM staging

The TNM system describes the size and spread of the tumour,whether cancer cells have spread to lymph nodes and whetherthe cancer has spread to a different part of the body. The mainclassifications currently used are the Western Hemispherestaging system developed by the American Joint Committeeon Cancer (AJCC) and the Union for International CancerControl (UICC) [14], and the Japanese staging system, whichis more elaborated and based on anatomic involvement, par-ticularly the lymph node stations [15].

Despite the widespread use of TNM staging to assess GCpatients’ prognosis, and particularly the number of lymphnodes as a poor prognostic factor, prognosis often variesbetween patients at the same tumour stage. Therefore, in

addition to TNM staging, auxiliary methods have been inves-tigated to refine the accuracy of prognostic stratification. Astudy from 2004 established that performance status ≥2, thepresence of metastases (liver and peritoneal) and alkalinephosphatase ≥100 U/L were independent poor prognosticfactors in GC [16]. A prognostic index was constructed withthese factors by classifying patients as having good (no riskfactor), moderate (one or two factors) or poor prognosis (threeor four risk factors) [16] and was later validated in the REAL-2 trial [17].

Although TNM staging per se is insufficient to predict GCprognosis, accurate staging is mandatory to appropriatelytriage GC into potentially curative (resectable) versusnon-resectable categories and implement multimodalitytherapy [18].

Molecular biomarkers: prognostic, predictive and therapytargets

The main genetic alterations that can drive carcinogenesis aremutations (including small insertions/deletions), amplifica-tions and rearrangements. Gene amplification often leads toprotein overexpression.With the exception of dominant drivermutations in solid tumours such as BRAFV600E in melanoma,activating EGFR mutations in lung cancer and HER2 ampli-fication in breast and GC, other potentially driver geneticalterations are relatively rare [19].

Gene amplification is a frequent mode of genetic alterationin GC. Common methods of detecting gene amplificationinclude fluorescence in situ hybridisation (FISH), chromogen-ic ISH (CISH), silver ISH (SISH), real-time quantitative PCR(RT-qPCR) and single-nucleotide polymorphism (SNP)genotyping assay using next-generation sequencing (NGS).The most commonly accepted definition of amplification isgene/chromosome enumeration probe (CEP) ≥2. Protein over-expression is commonly detected by immunohistochemistry(IHC). Such a variety of available methods presents advan-tages towards a wider comprehension of biomarkers; howev-er, it can also bemisleading due to discrepancies obtainedwithdifferent techniques.

Human epidermal growth factor 2 (HER2)

HER2 (encoded by ERBB2, the v-erb-b2 avian erythroblasticleukaemia viral oncogene homolog 2) is one of the fourmembers of the human epidermal growth factor receptorfamily (EGFR or HER1, HER2, HER3 and HER4) in thereceptor tyrosine kinase (RTK) superfamily. Unlike otherHER family members, HER2 does not contain a ligand bind-ing site and signals through heterodimerisation with otherHER family members, primarily EGFR (Fig. 1) [20].

ERBB2 amplification and HER2 overexpression have beenmost widely studied in breast cancer, constituting established

Virchows Arch

Page 3: Biomarkers for gastric cancer: prognostic, predictive or targets of therapy?

Tab

le1

Interpretatio

nof

prognosticandpredictiv

evalueof

biom

arkersforadvanced

GC

Biomarkergroup/pathway

Biomarker

Detectio

nmethod

Biomarkerstatus

Prognostic

value

Clin

icaltrial

status

Targeted

therapya

Predictivevalue

Impact

Ref.

Growth

factor

receptors

EGFR

FISH

,SISH,C

ISH,

RT-qP

CR

EGFRam

plification

Yes

PhaseIII

Cetuxim

abNo

Decreased

PFSandOS

[40]

PhaseIII

Panitumum

abNo

Decreased

OS

[41]

IHC,R

T-qP

CR

Overexpression

PhaseIII

Nim

otuzum

abb

Pending

PhaseII/III

Lapatinibb

Pending

FGFR

2FISH

,RT-qP

CR

FGFR2am

plification

Yes

PhaseII

AZD4547

Pending

VEGFR

2FISH

,CISH

VEGFR

2am

plification

Yes

PhaseIII

Ram

ucirum

abYes

Modestincreased

OS

[51]

IHC

Overexpression

HER2

FISH

,SISH,C

ISH,

RT-qP

CR

ERBB2am

plification

NC

FDA-approved

Trastuzum

abb

Yes

IncreasedOS

[24]

PhaseII/III

T-DM1b

Pending

IHC,R

T-qP

CR

Overexpression

PhaseIIa

Pertuzum

abb

Pending

PhaseIII

Lapatinibb

Pending

MET(H

GFR

)FISH

,SISH,C

ISH,

RT-qP

CR

METam

plification

Yes

PhaseII

Rilo

tumum

abPending

IHC,R

T-qP

CR

Overexpression

PhaseIII

Onartuzum

abPending

Growth

factor

ligands

VEGFA

IHC

Overexpression

Yes

PhaseIII

Bevacizum

abNo

IncreasedOSbutn

otsignificant

[49]

PI3K

/AKT

PI3K

ASequencing

PIK3C

Amutations

Yes

PhaseIb

BYL719

Pending

FISH

,RT-qP

CR

PIK3C

Aam

plification

IHC,R

T-qP

CR

Overexpression

mTOR

FISH

MTO

Ram

plification

Yes

PhaseIII

Everolim

usNo

IncreasedOSbutn

otsignificant

[63]

IHC

Overexpression

MSI

BAT-25,B

AT-26,

NR-21,NR-24,

MONO-27

Sequencing

Nucleotideinsertionor

deletio

nYes

––

–[89]

KRAS/MAPK

KRAS

Sequencing

KRASmutations

Yes

––

–[77]

Celladhesion

E-cadherin

Sequencing

CDH1structuralalteratio

nsYes

––

–[81,82]

CDH1mutations

T-DM1trastuzumab

emtansine,OSoverallsurvival,IH

Cim

munohistochem

istry,RT-qP

CRreal-tim

equantitativePCR,FISHfluorescence

insitu

hybridization,

SISH

silver

insitu

hybridisation,

CISH

chromogenicin

situ

hybridisation,MSI

microsatelliteinstability,N

Cnotconsistentamongseries

aMosto

fthesetargeted

therapieshave

been

givenin

combinatio

nwith

conventio

nalchemotherapy

bGCpatientswereselected

basedon

thetargeted

moleculestatus

Virchows Arch

Page 4: Biomarkers for gastric cancer: prognostic, predictive or targets of therapy?

biomarkers of poor prognosis. The currently accepted proto-col to detect HER2 aberrations encloses the use of IHC withspecific antibodies and in situ hybridisation techniques (FISH,CISH and SISH) (Fig. 2). Those cancers scoring 3+ by IHCare considered HER2 positive; those scoring 2+ undergofurther confirmation by FISH, CISH or SISH; and thosescoring <2+ by IHC are considered HER2-negative cases.

Using similar scoring systems, amplification and/or overex-pression of ERBB2/HER2 has also been observed in colorectal,lung, gastric and ovarian cancers [21]. In GC, ERBB2 amplifi-cation or HER2 overexpression has been reported in 7–34 % ofthe tumours [21–23], particularly in the gastroesophageal junc-tion carcinomas (proximal) and in intestinal type GC [22–24].

The prognostic value of HER2 positivity in advanced GCis, however, a controversial issue. There are reports indicatingthat ERBB2 amplification is associated with poor prognosisand aggressive disease [21, 23, 25, 26], whereas other reportsshow no difference in prognosis when compared with HER2-negative tumours [27–29]. A recent study showed that HER2

status is not even an independent prognostic biomarker inearly gastroesophageal adenocarcinoma [30].

Despite the controversy regarding HER2 prognostic value,inhibition of HER2 has been tested as a targeted therapyoption in several cancer types. Trastuzumab, a monoclonalantibody that targets HER2, inhibits HER2-mediated signal-ling and prevents cleavage of its extracellular domain [31]. InHER2-positive metastatic breast cancer, trastuzumab im-proves patient outcome and is now the standard of care [32,33]. The Trastuzumab for Gastric Cancer (ToGA,ClinicalTrials.gov Identifier: NCT01041404) phase III inter-national study assessed the efficacy of a combination oftrastuzumab with conventional chemotherapy (cisplatin plus5-fluorouracil or capecitabine) as a treatment for GC patients.This trial has demonstrated that advanced GC patients, strat-ified by HER2 amplification/overexpression, had longer me-dian OS when treated with trastuzumab/chemotherapy versuschemotherapy alone (13.8 versus 11.1 months) [24]. More-over, there was improved predictive value of trastuzumab

Fig. 1 Frontline therapy trials for advanced GC involve antibodiesagainst overexpressed and/or amplified receptor tyrosine kinases (RTKs)or their ligands, and specific small-molecule inhibitors. The targetedRTKs for advanced GC therapy are HER2, EGFR, MET, FGFR2 andVEGFR2. The activation of these RTKs signals through PI3K (among

other pathways) leading to the activation of mTOR, both novel bio-markers under investigation for targeted GC therapy. All these moleculesregulate signalling cascades important for neoplasia, including differen-tiation, proliferation, angiogenesis and survival

Virchows Arch

Page 5: Biomarkers for gastric cancer: prognostic, predictive or targets of therapy?

based on HER2 positivity thresholds. The median OS of the“higher HER2 threshold” categories (Very High: IHC3+/FISH+; High: IHC3+ and IHC2+/FISH+) was better than theremainder (IHC0+/FISH+, any IHC1+ and IHC2+/FISH−).The median OS within the whole trastuzumab arm was13.8 months, whereas in the “Very High HER2” and “HighHER2” groups, it was 17.9 and 16 months, respectively.Trastuzumab in combination with cisplatin and afluoropyrimidine is, therefore, currently approved by the Foodand Drug Administration (FDA, USA) and European Medi-cines Agency as first-line treatment of patients with HER2-overexpressing metastatic GC, who have not received priortreatment for metastatic disease.

Several promising GC clinical trials targeting HER2, usingcombinations of different drugs, are simultaneously undergoing.The LoGIC trial (ClinicalTrials.gov identifier: NCT00680901) isinvestigating the effect of lapatinib, a dual EGFR and HER2tyrosine kinase inhibitor. The effect of pertuzumab, amonoclonalantibody that binds to a different part of HER2 than trastuzumab,is also being studied (ClinicalTrials.gov identifier:NCT01461057). There is also an ongoing randomised trial in-vestigating the predictive value of trastuzumab emtansine (T-DM1), an antibody–drug conjugate incorporating HER2-targeted antitumour properties of trastuzumab with the cytotoxicactivity of the microtubule-inhibitory agent DM1(ClinicalTrials.gov identifier: NCT01641939).

Epidermal growth factor receptor (EGFR, HER1)

EGFR (encoded by EGFR) is another member of the HERfamily which is activated by the binding of specific ligands,including EGF and TGFα (Fig. 1). Kiyose et al. [34],employing FISH assays, reported the incidence of EGFRamplification to be 4.9 % in 365 GC samples. Another study,using SNP assays, reported that 7.7 % of GC harboured EGFRamplification [35]. A larger investigation (511 CG samples)examined EGFR overexpression by IHC and FISH and re-ported that 27.4 % of the samples were positive for EGFRprotein expression by IHC (IHC2+ and 3+) while only 2.3 %

were amplified when detected by FISH [36]. EGFR overex-pression has been shown to be a prognostic indicator of worseoutcome in GC [37, 38]. EGFR IHC or FISH positivity hasbeen associated with the presence of lymph node metastasis,higher stage and poor survival, however, after multivariateanalysis, only EGFR IHC positivity remained a poor prog-nostic factor [36].

EGFR has also been used as a target for therapy in GC.Monoclonal antibodies against EGFR (cetuximab orpanitumumab), combined with conventional chemotherapy,have been explored in two randomised GC clinical trials [39].Contrary to the ToGA study [24], the international phase IIIstudy EXPAND (ClinicalTrials.gov Identifier: NCT01611506),which used a combination of cetuximab and conventionalchemotherapy, did not provide benefit in OS of GC patients[40]. In fact, the cetuximab group had an inferior progression-free survival (PFS) (4.4 months) and inferior OS (9.4 months),compared with cisplatin/capecitabine alone (5.6 and10.7 months, respectively). Similarly, in the REAL3 trial(ClinicalTrials.gov Identifier: NCT00824785), addition ofpanitumumab to epirubicin/oxaliplatin/capecitabine resulted ina significantly lower OS (8.8 months) compared withepirubicin/oxaliplatin/capecitabine alone (11.3 months) [41].

As the results from the EXPAND and REAL3 studiessuggest, the addition of anti-EGFR antibodies to chemother-apy does not deliver additional benefit for patients with ad-vanced GC. In both studies, the prevalence of EGFR amplifi-cation or EGFR IHC positivity was not reported. In thissetting, data from single-group studies suggest that EGFRexpression, EGFR copy number and expression of otherEGFR ligands (epiregulin and amphiregulin) or downstreamcomponents of the EGFR-signalling pathway might be candi-date biomarkers for anti-EGFR-antibody efficacy [42–44]. Aphase III trial (ENRICH, ClinicalTrials.gov identifier:NCT01813253) is investigating the addition of the anti-EGFR antibody nimotuzumab to irinotecan as second-linetreatment for IHC 2+/3+ GC patients. The results from thisstudy might elucidate if selecting EGFR IHC 2+/3+ GCpatients for anti-EGFR therapy will improve survival. Indeed,

Fig. 2 HER2 expression and ERBB2amplification in one GC: a IHC. In most cells, the staining is not limited to basolateral membranous reactivity but tothe entire membrane (original magnification×400). b IHC. Higher magnification highlighting completeness of membrane staining (original magnifi-cation×600). c SISH. Uniform ERBB2 gene amplification

Virchows Arch

Page 6: Biomarkers for gastric cancer: prognostic, predictive or targets of therapy?

the prognostic and predictive role of EGFR expression hasalready been demonstrated in the FLEX trial for advancednon-small-cell lung cancer (NSCLC) [45]. This study showedthat high EGFR expression is a tumour biomarker that canpredict survival benefit from the addition of cetuximab tochemotherapy, therefore indicating that assessment of EGFRexpression could offer a personalised treatment approach.

Small-molecule inhibitors of EGFR like erlotinib andlapatinib are also in GC clinical trials; however, the numberof patients included was small and/or the series unselected forEGFR overexpression and/or amplification. A phase II clini-cal trial (ClinicalTrials.gov identifier: NCT01123473) inves-tigating lapatinib in combination with chemotherapy selectedpatients for EGFR or HER2 positivity; however, the resultsare still pending.

Angiogenesis pathway biomarkers

Angiogenesis, the growth of new blood vessels, is an impor-tant aspect of tumourigenesis. It is primarily modulated by thevascular endothelial growth factor A (VEGFA) and its recep-tors VEGFRs, in particular types 1 and 2 (Fig. 1). In GC,expression of VEGFA and VEGFR was reported in 40 and36 % of cases, respectively [38]. VEGFA expression in GCand serum has been associated with poor prognosis, lymphnode involvement and metastasis, both in resectable and ad-vanced GC [46]. Targeted inhibition of VEGFA and VEGFRsis possible and has been a commonly employed strategy inoncology to decrease tumour vascular supply and metastasis,leading to tumour shrinkage. The use of monoclonal antibod-ies against VEGFA, such as bevacizumab, has successfullyshowed to improve OS in patients with advanced colorectalcancer [47] and NSCLC [48]. In the AVAGAST GC trial(ClinicalTrials.gov Identifier: NCT00548548), the additionof bevacizumab to cisplatin and capecitabine resulted inhigher overall response rate (46.0 versus 37.4 %) and higherPFS (6.7 versus 5.3 months) [49]. However, OS was notsignificantly improved (12.1 versus 10.1 months). TheAVAGAST trial did not report the level of VEGFR1 orVEGFR2 expression which is neither prognostic for OS norpredictive to bevacizumab response [50]. A recentrandomised trial (REGARD, ClinicalTrials.gov Identifier:NCT00917384), using ramucirumab, a monoclonal antibodyagainst VEGFR2, instead of the VEGFA, successfullyprolonged OS (5.2 versus 3.8 months on placebo) [51].Ramucirumab is the first biological treatment given as a singledrug that has survival benefits in patients with advanced GCor gastroesophageal cancer progressing after first-line chemo-therapy [51]. Although these findings validate VEGFR2 sig-nalling as an important therapeutic target in advanced GC, theOS improvement with ramucirumab was small, and it will beimportant to investigate if there are any biomarkers underlyingits clinical activity, such as overexpression of VEGFR2.

Emerging biomarkers for target therapy in gastric cancer

The poor long-term outcomes associated with current chemo-therapy and scarce targeted therapy treatment for advancedGC suggest a need for other targeted agents that may confer abetter survival benefit. The most relevant novel biomarkers inGC currently being investigated, some of which are already inclinical trials, include the fibroblast growth factor receptor(FGFR), the hepatocyte growth factor receptor (HGFR,MET) and the mammalian target of rapamycin (mTOR).

Fibroblast growth factor receptor (FGFR)

FGFR family members (FGFR1, FGFR2, FGFR3 andFGFR4) belong to the RTK superfamily (Fig. 1). In a recentgenomic survey of GC using high-resolution SNP arrays,FGFR2 copy number gain was found in 9.3 % of tumoursand was more common than EGFR (7.7 %), HER2 (7.2 %) orMET (4.3 %) copy number gains [35]. Although FGFR2FISHwas also performed in the study, it was not reported if all thesamples with FGFR2 copy number gain were also trulyFGFR2-amplified. Another large screening study, includingCaucasian and Korean GC samples [52], found that 5.9 % ofthe GC presented FGFR2 amplification (cut-off: FGFR2/CEP10 >2). The FGFR2 amplification was more common inCaucasians (7.4 %) than in Koreans (4.2 %) and was associ-ated with lymph node metastasis in both cohorts and thediffuse type of GC in Koreans. Moreover, FGFR2 amplifica-tion was found to be a prognostic biomarker of shorter OS inboth cohorts. Jung et al. also showed the presence of FGFR2amplification in Korean GC samples (4.5 %) and its associa-tion with shorter OS [53].

FGFR2 has therefore attracted significant attention as apotential candidate for targeted therapy in GC. A number ofFGFR2 inhibitors are in clinical development, having demon-strated efficacy in cell lines and in human xenograft modelsin vivo [35, 54]. A randomised phase II trial comparing theselective FGFR inhibitor AZD4547 to paclitaxel as second-line treatment of advanced GC, harbouring FGFR2 polysomyor amplification (SHINE, ClinicalTrials.gov identifier:NCT01457846), is ongoing, and it will inform on the valueof this marker as a therapeutic target.

Hepatocyte growth factor receptor (HGFR, MET)

MET (encoded byMET) belongs to the HGFR family (Fig. 1).MET amplification and/or overexpression of its protein prod-uct has long been implicated in the pathogenesis of GC, withmany reports based on gene copy number, RNA expressionand/or protein expression, supporting its role as a poor prog-nosis factor [37, 55–57]. Nevertheless, the prevalence ofMETamplification in GC varies widely in the literature from 0 %[58] to 21 % [55]. This discrepancy is greatly attributed to the

Virchows Arch

Page 7: Biomarkers for gastric cancer: prognostic, predictive or targets of therapy?

methodology employed to detect gene amplification/copynumber gain and/or protein expression. MET copy numbergain detected by RT-qPCR is sometimes interpreted as ampli-fication, similarly to the common definition of gene amplifi-cation by FISH (gene/CEP ≥2), and this may bemisleading. Astudy employing NGS SNP assays reported MET amplifica-tion in 4.1 % of GC cases, while using the FISH criterion, thevalue was lower [35]. Another study did not findMETampli-fication in 38 GC using the FISH criterion [58]. A larger studyincluding 216 GC samples showed by RT-qPCR that 10 % ofGC displayed five or more MET copies. However, a FISHancillary analysis of 32 random samples did not show trueMET amplification [37]. Depending on the method used andthe stringiness of the FISH criteria, different MET amplifica-tion prevalences have been further reported [34, 55–57].Nonetheless, in all these studies, the GC patients withpolysomic and/or amplified MET showed poorer DFS andOS in comparison with the non-polysomic MET.

Recently, a randomised phase II metastatic gastroesopha-geal cancer t r ia l (Cl in ica lTr ia ls .gov Ident i f ie r :NCT00719550) using chemotherapy with and withoutrilotumumab, an antibody against the MET ligand HGF, con-firmed a significantly worse prognosis for MET-positive (IHC2+ in ≥50 % of cells) (median OS of 5.7 months) comparedwith MET-negative tumours within the placebo group (medi-an OS of 8.9 months) [18].

There is also an ongoing randomised phase III trial(MetGastric, ClinicalTrials.gov identifier: NCT01662869) in-vestigating whether the addition of onartuzumab, an antibodyagainst MET, to chemotherapy in MET-positive (IHC 2+/3+)and HER2-negative GC patients will significantly improveOS, when compared with chemotherapy alone.

PI3K/mTOR signalling pathway

The phosphatidylinositol-3-kinase (PI3K)/mTOR pathwayrepresents one common final convergence signalling pathwayoriginated by the activation of several RTKs (Fig. 1), such asthe RTKs herein reviewed. Activation of the PI3K/mTORpathway in GC has been demonstrated in preclinical studies[59, 60], and its deregulation has been associated with in-creased lymph node metastasis and decreased survival of GCpatients [61, 62].

Although alteration of PI3K/mTOR related genes and pro-teins has not been formally and specifically associated withGC prognosis, this pathway has been seen as a target fortherapeutic intervention. In particular, the role of everolimus,an mTOR inhibitor, has been investigated in advanced meta-static GC in two randomised trials. The recently publishedphase III randomised study (GRANITE-1, ClinicalTrials.govIdentifier: NCT00879333) [63] compared the use of everoli-mus or placebo plus best supportive care in second-line treat-ment of advanced GC. This strategy did not significantly

improve the median OS for the everolimus treatment arm(5.4 months on everolimus versus 4.3 months on placebo).The median PFS was only modest with estimates of1.7 months in the everolimus arm and 1.4 months in theplacebo arm. These results were interpreted as possibly indi-cating a subgroup treatment effect and to address that a retro-spective identification of specific biomarkers for patient strat-ification is ongoing in the GRANITE-1 study. Another bio-marker retrospective analysis in a different phase II study(ClinicalTrials.gov Identifier: NCT00729482) of everolimusin GC [64] did not yield significant results; thus, furtherinvestigation is warranted to select GC patients that maybenefit from this particular targeted therapy.

Oncogenic mutations in PIK3CA (gene encoding the alphap110 catalytic subunit of PI3K) have been observed in GC,constitutively activating the PI3KA/mTOR pathway. The firststudies in GC reported a PIK3CAmutation frequency of 25 %[65] and 10 % [66]. Other recent studies report PIK3CAmutation frequencies varying from 5.1 to 16 % [67–69].One of these studies reported PIK3CA amplification in 67 %of GC [68], and association with poor prognosis, indicatingthat this is a major mechanism leading to the activation thePI3K/mTOR pathway in GC.

Small molecules inhibiting PI3KA have still to prove theirefficacy. In view of this, a recently initiated trial(ClinicalTrials.gov identifier: NCT01613950) is intended toinvestigate the safety and preliminary efficacy of the PI3KAinhibitor BYL719, in combination with AUY922 (heat shockprotein HSP90A inhibitor), in patients with advanced GC.Only patients with tumours carrying either a molecular alter-ation of PIK3CA, or amplification of ERBB2, are eligible forthis study, therefore allowing assessment of biomarker sub-groups. Additionally, the dual PI3KA and mTOR inhibitorBEZ235 has been shown to decrease cell viability and induceapoptosis in a GC cell line harbouring PIK3CA and KRASmutations [70]. PI3K/mTOR dual inhibitors have not yetentered early-stage clinical trials for advanced GC.

Non-targetable prognostic biomarkers in gastric cancer

In addition to the most frequently used molecular biomarkersin GC, for which specific targeting in combination with con-ventional chemotherapy has been the subject of several clin-ical trials, there are also a series of alterations that, althoughrare and possibly non-targetable, may potentially be used inpatient stratification and towards improvedGC patient therapyselection/personalised therapy.

Mismatch repair (MMR) deficiency leads to a tumourphenotype known as microsatellite instability (MSI), in whichcells accumulate genetic errors rather than correcting thoseerrors. MSI has been reported in 18 to 28% of GC, mainly dueto hypermethylation of the MLH1 promoter [71]. Patients

Virchows Arch

Page 8: Biomarkers for gastric cancer: prognostic, predictive or targets of therapy?

bearing GC tumours with MSI generally develop GC later inlife and have favourable prognosis when compared with tu-mours with MMR proficient tumours [72, 73].

KRAS activating mutations (frequently found in codons 12,13 and 61) are present in ca. 4 % of GC [74, 75]. Although notcurrently used as a prognostic biomarker, KRAS mutationsoccur mainly in GC with MSI [72, 74, 76]. A recent reportclaims that the presence of KRASmutations is associated withworse prognosis in patients with proximal GC [77]. Morestudies are needed to clarify whether the presence of KRASmutations may underlie worse prognosis among GCwithMSIphenotype.

Moreover, KRASmutations may have predictive value forresponsiveness to EGFR-tyrosine kinase inhibitors in GC,similarly to what has been shown in colorectal cancer [78,79]. Therefore, KRAS activating mutations possibly constitutea marker for patient stratification. KRAS is, however, consid-ered to be a non-targetable biomarker since attempts to targetthis protein directly with small-molecule inhibitors have, sofar, proved unsuccessful [80].

E-cadherin (encoded by CDH1) dysfunction is one of themost important factors and frequent aberration (ca. 90%) in GCinitiation and progression, so far described. However, only thepresence of E-cadherin structural alterations (in 10 % of GCcases) represents a poor prognostic factor [81, 82]. Thesealterations are, presently, non-targetable as this would requirerestoring E-cadherin expression by gene therapy. Nevertheless,E-cadherin is a potential predictive marker of response totherapy, since its impairment decreases tumour cell sensitivityto conventional and targeted therapies [83–85]. Retrospectivestudies evaluating E-cadherin status in GC samples integratedin specific clinical trials (and correlating them with patientsurvival) are required in order to fully assess this possibility.

Future perspectives

Two major issues contribute towards the general poor prognosisofGCpatients: diagnosis at late stages and the lack of appropriatetherapies. Early diagnosis may be solved with adequate publichealth screening programs, such as those developed in Japan andKorea [86], and improvement in imaging technologies, such asendoscopy coupled with high-resolution microscopy [87].

The combination of correct GC staging with targeted ther-apy and conventional chemotherapy has made worthy prog-ress in the treatment of patients; nevertheless, the most recenttherapy regimens have had little impact on prognosis im-provement and OS. The development of new therapies willonly be possible if the molecular landscaping of GC is knownfor appropriate patient stratification, as has occurred in coloncancer [88]. The use of targeted therapies in combination withconventional chemotherapy offers limited success in GC pa-tients’ survival, as determined in the ToGA trial, in which

trastuzumab administration only improved OS of HER2-positive GC patients by 3 months [24]. Nevertheless, therapyagainst HER2 is the only targeted therapy that is currentlyaccepted as standard of care in GC. This was possible due tothe demonstration that only GCs with very high expression ofHER2 would benefit from this targeted therapy. In all otherclinical trials herein cited and/or published, with the exceptionof the ongoing ENRICH and lapatinib trials using EGFRantibody/inhibitor, GC patients have not been stratified fortreatment according to a similar premise, and this may be oneof the most obvious reasons for trial failure in GC. It isessential to make progress in GC patient molecular stratifica-tion to improve the clinical trial output, to perform retrospec-tive analyses to understand if particular subsets of GC patientsdid in fact respond to targeted treatment and to identify thelandscape of markers that may constitute targets fortherapy in each GC case.

The major challenges in identifying reliable prognosticbiomarkers are inter-individual variability of response totargeted therapy and drug resistance. At present, althoughthe role of many genetic alterations discovered in GC seemsunclear, they represent a promising tool for stratifying patientsaccording to tumour biological behaviour and likelihood ofresponse to systemic therapy. Additionally, independent vali-dation of the most promising prognostic and predictive bio-markers is required before they can be routinely employed inclinical practice. It is also important to undertake retrospectivestudies in which tumour samples from patients that haveundergone GC therapy are mined for biomarkers, or combi-nation of biomarkers, that can be predictive of favourable/unfavourable response towards a certain chemotherapeuticregimen or define the use of multiple therapy regimens in GC.

Acknowledgments The salary support to CD and GMA from POPH-QREN/Type 4.1, European Social Fund and Portuguese Ministry ofScience and Technology (MCTES), SFRH/BPD/62974/2009 andSFRH/BPD/87257/2012, respectively, is acknowledged. IPATIMUP isan Associate Laboratory of the Portuguese Ministry of Science, Technol-ogy and Higher Education and is partially supported by FCT, the Portu-guese Foundation for Science and Technology.

Conflict of interest The authors declare that they have no conflict ofinterest.

References

1. Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM (2010)Estimates of worldwide burden of cancer in 2008: GLOBOCAN2008. Int J Cancer 127(12):2893–2917. doi:10.1002/ijc.25516

2. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D (2011)Global cancer statistics. Ca-a Cancer Journal for Clinicians 61(2):69–90. doi:10.3322/Caac.20107

3. Houghton J, Wang TC (2005) Helicobacter pylori and gastric cancer:a new paradigm for inflammation-associated epithelial cancers.Gastroenterology 128(6):1567–1578

Virchows Arch

Page 9: Biomarkers for gastric cancer: prognostic, predictive or targets of therapy?

4. Yamashita K, Sakuramoto S, Katada N, Futawatari N, Moriya H,Hirai K, Kikuchi S, Watanabe M (2009) Diffuse type advancedgastric cancer showing dismal prognosis is characterized by deeperinvasion and emerging peritoneal cancer cell: the latest comparativestudy to intestinal advanced gastric cancer. Hepatogastroenterology56(89):276–281

5. Oliveira C, Seruca R, Carneiro F (2009) Hereditary gastric cancer.Best Pract Res Clin Gastroenterol 23(2):147–157. doi:10.1016/j.bpg.2009.02.003

6. Oliveira C, Senz J, Kaurah P, Pinheiro H, Sanges R, Haegert A, CorsoG, Schouten J, Fitzgerald R, Vogelsang H, Keller G, Dwerryhouse S,Grimmer D, Chin SF, Yang HK, Jackson CE, Seruca R, Roviello F,Stupka E, Caldas C, Huntsman D (2009) Germline CDH1 deletionsin hereditary diffuse gastric cancer families. Hum Mol Genet 18(9):1545–1555. doi:10.1093/hmg/ddp046

7. Mihaljevic AL, Friess H, Schuhmacher C (2013) Clinical trials ingastric cancer and the future. J Surg Oncol 107(3):289–297. doi:10.1002/Jso.23120

8. Takeuchi H, Kitagawa Y (2013) New sentinel node mapping tech-nologies for early gastric cancer. Ann Surg Oncol 20(2):522–532.doi:10.1245/s10434-012-2602-1

9. Kitagawa Y, Takeuchi H, Takagi Y, Natsugoe S, Terashima M,Murakami N, Fujimura T, Tsujimoto H, Hayashi H, Yoshimizu N,Takagane A, Mohri Y, Nabeshima K, Uenosono Y, Kinami S,Sakamoto J, Morita S, Aikou T, Miwa K, Kitajima M (2013)Sentinel node mapping for gastric cancer: a prospective multicentertrial in Japan. J Clin Oncol. doi:10.1200/JCO.2013.50.3789

10. Cervantes A, Roda D, Tarazona N, Rosello S, Perez-Fidalgo JA(2013) Current questions for the treatment of advanced gastric cancer.Cancer Treat Rev 39(1):60–67. doi:10.1016/j.ctrv.2012.09.007

11. GASTRIC Group, Oba K, Paoletti X, Bang YJ, Bleiberg H,Burzykowski T, Fuse N, Michiels S, Morita S, Ohashi Y, PignonJP, Rougier P, Sakamoto J, Sargent D, Sasako M, Shitara K,Tsuburaya A, Van Cutsem E, Buyse M (2013) Role of chemotherapyfor advanced/recurrent gastric cancer: an individual-patient-datameta-analysis. Eur J Cancer 49(7):1565–1577. doi:10.1016/j.ejca.2012.12.016

12. Italiano A (2011) Prognostic or predictive? It’s time to get back todefinitions! J Clin Oncol 29(35):4718. doi:10.1200/JCO.2011.38.3729, author reply 4718-4719

13. Pietrantonio F, Braud F, Da Prat V, Perrone F, Pierotti MA, GariboldiM, Fanetti G, Biondani P, Pellegrinelli A, Bossi I, Di Bartolomeo M(2013) A review on biomarkers for prediction of treatment outcomein gastric cancer. Anticancer Res 33(4):1257–1266

14. Edge S, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A (eds)(2010) AJCC cancer staging manual, 7th edn. New York, Springer

15. Japanese Research Society for Gastric Cancer (1993) The general rulesfor the gastric cancer study in surgery and pathology, 12 edn. JapaneseResearch Society for Gastric Cancer, Kanahara Shuppan, Tokyo

16. Chau I, Norman AR, Cunningham D, Waters JS, Oates J, Ross PJ(2004) Multivariate prognostic factor analysis in locally advancedand metastatic esophago-gastric cancer—pooled analysis from threemulticenter, randomized, controlled trials using individual patientdata. J Clin Oncol 22(12):2395–2403. doi:10.1200/JCO.2004.08.154

17. Chau I, Ashley S, Cunningham D (2009) Validation of the RoyalMarsden hospital prognostic index in advanced esophagogastric can-cer using individual patient data from the REAL 2 study. J Clin Oncol27(19):e3–e4. doi:10.1200/JCO.2009.22.0863

18. Sehdev A, Catenacci DV (2013) Gastroesophageal cancer: focus onepidemiology, classification, and staging. DiscovMed 16(87):103–111

19. Lee J, Ou SH (2013) Towards the goal of personalized medicine ingastric cancer—time to move beyond HER2 inhibition. Part I:targeting receptor tyrosine kinase gene amplification. Discov Med15(85):333–341

20. Ou SH (2012) Second-generation irreversible epidermal growth fac-tor receptor (EGFR) tyrosine kinase inhibitors (TKIs): a better

mousetrap? A review of the clinical evidence. Crit Rev OncolHematol 83(3):407–421. doi:10.1016/j.critrevonc.2011.11.010

21. Gravalos C, Jimeno A (2008) HER2 in gastric cancer: a new prog-nostic factor and a novel therapeutic target. Ann Oncol 19(9):1523–1529. doi:10.1093/annonc/mdn169

22. HofmannM, StossO, Shi D, Buttner R, van deVijverM,KimW,OchiaiA, Ruschoff J, Henkel T (2008) Assessment of a HER2 scoring systemfor gastric cancer: results from a validation study. Histopathology 52(7):797–805. doi:10.1111/j.1365-2559.2008.03028.x

23. Tanner M, Hollmen M, Junttila TT, Kapanen AI, Tommola S, SoiniY, Helin H, Salo J, Joensuu H, Sihvo E, Elenius K, Isola J (2005)Amplification of HER-2 in gastric carcinoma: association withTopoisomerase IIalpha gene amplification, intestinal type, poor prog-nosis and sensitivity to trastuzumab. Ann Oncol 16(2):273–278. doi:10.1093/annonc/mdi064

24. BangYJ, VanCutsemE, FeyereislovaA, ChungHC, Shen L, Sawaki A,Lordick F, Ohtsu A, Omuro Y, Satoh T, Aprile G, Kulikov E, Hill J,Lehle M, Ruschoff J, Kang YK, To GATI (2010) Trastuzumab incombination with chemotherapy versus chemotherapy alone for treat-ment of HER2-positive advanced gastric or gastro-oesophageal junctioncancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet376(9742):687–697. doi:10.1016/S0140-6736(10)61121-X

25. Bar-Sela G, Hershkovitz D, Haim N, Kaidar-Person O, Shulman K,Ben-Izhak O (2013) The incidence and prognostic value of HER2overexpression and cyclin D1 expression in patients with gastric orgastroesophageal junction adenocarcinoma in Israel. Oncol Lett 5(2):559–563. doi:10.3892/ol.2012.1031

26. Barros-Silva JD, Leitao D, Afonso L, Vieira J, Dinis-Ribeiro M,Fragoso M, Bento MJ, Santos L, Ferreira P, Rego S, Brandao C,Carneiro F, Lopes C, Schmitt F, Teixeira MR (2009) Association ofERBB2 gene status with histopathological parameters and disease-specific survival in gastric carcinoma patients. Br J Cancer 100(3):487–493. doi:10.1038/sj.bjc.6604885

27. Aizawa M, Nagatsuma AK, Kitada K, Kuwata T, Fujii S, KinoshitaT, Ochiai A (2013) Evaluation of HER2-based biology in 1,006 casesof gastric cancer in a Japanese population. Gastric Cancer. doi:10.1007/s10120-013-0239-9

28. Gordon MA, Gundacker HM, Benedetti J, Macdonald JS, BarandaJC, Levin WJ, Blanke CD, Elatre W, Weng P, Zhou JY, Lenz HJ,Press MF (2013) Assessment of HER2 gene amplification in adeno-carcinomas of the stomach or gastroesophageal junction in the INT-0116/SWOG9008 clinical trial. Ann Oncol 24(7):1754–1761. doi:10.1093/annonc/mdt106

29. Grabsch H, Sivakumar S, Gray S, Gabbert HE, Muller W (2010)HER2 expression in gastric cancer: rare, heterogeneous and of noprognostic value—conclusions from 924 cases of two independentseries. Cell Oncol 32(1–2):57–65. doi:10.3233/CLO-2009-0497

30. Okines AF, Thompson LC, Cunningham D, Wotherspoon A, Reis-Filho JS, Langley RE, Waddell TS, Noor D, Eltahir Z, Wong R,Stenning S (2013) Effect of HER2 on prognosis and benefit fromperi-operative chemotherapy in early oesophago-gastric adenocarci-noma in the MAGIC trial. Ann Oncol 24(5):1253–1261. doi:10.1093/annonc/mds622

31. Hudis CA (2007) Trastuzumab—mechanism of action and use inclinical practice. N Engl J Med 357(1):39–51. doi:10.1056/NEJMra043186

32. Smith I, Procter M, Gelber RD, Guillaume S, Feyereislova A,Dowsett M, Goldhirsch A, Untch M, Mariani G, Baselga J,Kaufmann M, Cameron D, Bell R, Bergh J, Coleman R, WardleyA, Harbeck N, Lopez RI, Mallmann P, Gelmon K, Wilcken N, WistE, Sanchez Rovira P, Piccart-Gebhart MJ, team Hs (2007) 2-yearfollow-up of trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer: a randomised controlled trial. Lancet369(9555):29–36. doi:10.1016/S0140-6736(07)60028-2

33. Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, BajamondeA, Fleming T, EiermannW,Wolter J, PegramM, Baselga J, Norton L

Virchows Arch

Page 10: Biomarkers for gastric cancer: prognostic, predictive or targets of therapy?

(2001) Use of chemotherapy plus a monoclonal antibody againstHER2 for metastatic breast cancer that overexpresses HER2. NEngl JMed 344(11):783–792. doi:10.1056/NEJM200103153441101

34. Kiyose S, Nagura K, Tao H, Igarashi H, Yamada H, Goto M, MaedaM, KurabeN, SuzukiM, TsuboiM, Kahyo T, Shinmura K, Hattori N,Sugimura H (2012) Detection of kinase amplifications in gastriccancer archives using fluorescence in situ hybridization. Pathol Int62(7):477–484. doi:10.1111/j.1440-1827.2012.02832.x

35. Deng N, Goh LK, Wang H, Das K, Tao J, Tan IB, Zhang S,Lee M, Wu J, Lim KH, Lei Z, Goh G, Lim QY, Tan AL, SinPoh DY, Riahi S, Bell S, Shi MM, Linnartz R, Zhu F, YeohKG, Toh HC, Yong WP, Cheong HC, Rha SY, Boussioutas A,Grabsch H, Rozen S, Tan P (2012) A comprehensive surveyof genomic alterations in gastric cancer reveals systematicpatterns of molecular exclusivity and co-occurrence amongdistinct therapeutic targets. Gut 61(5):673–684. doi:10.1136/gutjnl-2011-301839

36. Kim MA, Lee HS, Lee HE, Jeon YK, Yang HK, Kim WH (2008)EGFR in gastric carcinomas: prognostic significance of protein over-expression and high gene copy number. Histopathology 52(6):738–746. doi:10.1111/j.1365-2559.2008.03021.x

37. Graziano F, Galluccio N, Lorenzini P, Ruzzo A, Canestrari E,D’Emidio S, Catalano V, Sisti V, Ligorio C, Andreoni F, Rulli E,Di Oto E, Fiorentini G, Zingaretti C, De Nictolis M, Cappuzzo F,Magnani M (2011) Genetic activation of the MET pathway andprognosis of patients with high-risk, radically resected gastric cancer.J Clin Oncol 29(36):4789–4795. doi:10.1200/JCO.2011.36.7706

38. Lieto E, Ferraraccio F, Orditura M, Castellano P, Mura AL, Pinto M,Zamboli A, De Vita F, Galizia G (2008) Expression of vascularendothelial growth factor (VEGF) and epidermal growth factor re-ceptor (EGFR) is an independent prognostic indicator of worseoutcome in gastric cancer patients. Ann Surg Oncol 15(1):69–79.doi:10.1245/s10434-007-9596-0

39. Chan JA, Blaszkowsky LS, Enzinger PC, Ryan DP, Abrams TA, ZhuAX, Temel JS, Schrag D, Bhargava P, Meyerhardt JA, Wolpin BM,Fidias P, Zheng H, Florio S, Regan E, Fuchs CS (2011) Amulticenterphase II trial of single-agent cetuximab in advanced esophageal andgastric adenocarcinoma. Ann Oncol 22(6):1367–1373. doi:10.1093/annonc/mdq604

40. Lordick F, Kang YK, Chung HC, Salman P, Oh SC, Bodoky G,Kurteva G, Volovat C, Moiseyenko VM, Gorbunova V, Park JO,Sawaki A, Celik I, Gotte H, Melezinkova H, Moehler M,Arbeitsgemeinschaft Internistische O, Investigators E (2013)Capecitabine and cisplatin with or without cetuximab for patientswith previously untreated advanced gastric cancer (EXPAND): arandomised, open-label phase 3 trial. Lancet Oncol 14(6):490–499.doi:10.1016/S1470-2045(13)70102-5

41. Okines AF, Ashley SE, Cunningham D, Oates J, Turner A, Webb J,Saffery C, Chua YJ, Chau I (2010) Epirubicin, oxaliplatin, andcapecitabine with or without panitumumab for advancedesophagogastric cancer: dose-finding study for the prospective mul-ticenter, randomized, phase II/III REAL-3 trial. J Clin Oncol 28(25):3945–3950. doi:10.1200/JCO.2010.29.2847

42. Luber B, Deplazes J, Keller G, Walch A, Rauser S, Eichmann M,Langer R, Hofler H, Hegewisch-Becker S, Folprecht G, Woll E,Decker T, Endlicher E, Lorenzen S, Fend F, Peschel C, Lordick F(2011) Biomarker analysis of cetuximab plus oxaliplatin/leucovorin/5-fluorouracil in first-line metastatic gastric and oesophago-gastricjunct ion cancer : resul ts from a phase II t r ia l of theArbeitsgemeinschaft Internistische Onkologie (AIO). BMC Cancer11:509. doi:10.1186/1471-2407-11-509

43. Han SW, Oh DY, Im SA, Park SR, Lee KW, Song HS, Lee NS, LeeKH, Choi IS, LeeMH, KimMA,KimWH,BangYJ, KimTY (2009)Phase II study and biomarker analysis of cetuximab combined withmodified FOLFOX6 in advanced gastric cancer. Br J Cancer 100(2):298–304. doi:10.1038/sj.bjc.6604861

44. Moehler M, Mueller A, Trarbach T, Lordick F, Seufferlein T,Kubicka S, Geissler M, Schwarz S, Galle PR, Kanzler S, GermanArbeitsgemeinschaft Internistische O (2011) Cetuximab withirinotecan, folinic acid and 5-fluorouracil as first-line treatment inadvanced gastroesophageal cancer: a prospective multi-centerbiomarker-oriented phase II study. Ann Oncol 22(6):1358–1366.doi:10.1093/annonc/mdq591

45. Pirker R, Pereira JR, von Pawel J, Krzakowski M, Ramlau R, Park K,de Marinis F, Eberhardt WE, Paz-Ares L, Storkel S, SchumacherKM, von Heydebreck A, Celik I, O’Byrne KJ (2012) EGFR expres-sion as a predictor of survival for first-line chemotherapy pluscetuximab in patients with advanced non-small-cell lung cancer:analysis of data from the phase 3 FLEX study. Lancet Oncol 13(1):33–42. doi:10.1016/S1470-2045(11)70318-7

46. Yoshikawa T, Tsuburaya A, Kobayashi O, Sairenji M, Motohashi H,Yanoma S, Noguchi Y (2000) Plasma concentrations of VEGF andbFGF in patients with gastric carcinoma. Cancer Lett 153(1–2):7–12

47. Hurwitz H, Fehrenbacher L, NovotnyW, Cartwright T, Hainsworth J,Heim W, Berlin J, Baron A, Griffing S, Holmgren E, Ferrara N, FyfeG, Rogers B, Ross R, Kabbinavar F (2004) Bevacizumab plusirinotecan, fluorouracil, and leucovorin for metastatic colorectal can-cer. N Engl JMed 350(23):2335–2342. doi:10.1056/NEJMoa032691

48. Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati A,Lilenbaum R, Johnson DH (2006) Paclitaxel-carboplatin alone orwith bevacizumab for non-small-cell lung cancer. N Engl J Med355(24):2542–2550. doi:10.1056/NEJMoa061884

49. Ohtsu A, ShahMA, Van Cutsem E, Rha SY, Sawaki A, Park SR, LimHY, Yamada Y, Wu J, Langer B, Starnawski M, Kang YK (2011)Bevacizumab in combination with chemotherapy as first-line therapyin advanced gastric cancer: a randomized, double-blind, placebo-controlled phase III study. J Clin Oncol 29(30):3968–3976. doi:10.1200/JCO.2011.36.2236

50. Van Cutsem E, de Haas S, Kang YK, Ohtsu A, Tebbutt NC,Ming XuJ, Peng Yong W, Langer B, Delmar P, Scherer SJ, Shah MA (2012)Bevacizumab in combination with chemotherapy as first-line therapyin advanced gastric cancer: a biomarker evaluation from theAVAGAST randomized phase III trial. J Clin Oncol 30(17):2119–2127. doi:10.1200/JCO.2011.39.9824

51. Fuchs CS, Tomasek J, Yong CJ, Dumitru F, Passalacqua R, GoswamiC, Safran H, Dos Santos LV, Aprile G, Ferry DR, Melichar B, TehfeM, Topuzov E, Zalcberg JR, Chau I, Campbell W, Sivanandan C,Pikiel J, Koshiji M, Hsu Y, Liepa AM, Gao L, Schwartz JD,Tabernero J, for the RTI (2013) Ramucirumab monotherapy forpreviously treated advanced gastric or gastro-oesophageal junctionadenocarcinoma (REGARD): an international, randomised,multicentre, placebo-controlled, phase 3 trial. Lancet. doi:10.1016/S0140-6736(13)61719-5

52. Kilgour E, Su X, Zhan P, Gavine P, Morgan S, Womack C, Jung E-J,Bang Y-J, Im S-A, Kim W, Grabsch H (2012) Prevalence andprognostic significance of FGF receptor 2 (FGFR2) gene amplifica-tion in Caucasian and Korean gastric cancer cohorts. J Clin Oncol30(15_suppl):4124

53. Jung EJ, Jung EJ, Min SY, Kim MA, Kim WH (2012) Fibroblastgrowth factor receptor 2 gene amplification status and its clinicopath-ologic significance in gastric carcinoma. Hum Pathol 43(10):1559–1566. doi:10.1016/j.humpath.2011.12.002

54. Xie L, Su X, Zhang L, Yin X, Tang L, Zhang X, Xu Y, Gao Z, Liu K,Zhou M, Gao B, Shen D, Zhang L, Ji J, Gavine PR, Zhang J, KilgourE, Zhang X, Ji Q (2013) FGFR2 gene amplification in gastric cancerpredicts sensitivity to the selective FGFR inhibitor AZD4547. ClinCancer Res 19(9):2572–2583. doi:10.1158/1078-0432.CCR-12-3898

55. Lee J, Seo JW, Jun HJ, Ki CS, Park SH, Park YS, LimHY, ChoiMG,Bae JM, Sohn TS, Noh JH, Kim S, Jang HL, Kim JY, KimKM, KangWK, Park JO (2011) Impact of METamplification on gastric cancer:possible roles as a novel prognostic marker and a potential therapeu-tic target. Oncol Rep 25(6):1517–1524. doi:10.3892/or.2011.1219

Virchows Arch

Page 11: Biomarkers for gastric cancer: prognostic, predictive or targets of therapy?

56. Lennerz JK, Kwak EL, Ackerman A, Michael M, Fox SB, BergethonK, Lauwers GY, Christensen JG, Wilner KD, Haber DA, Salgia R,Bang YJ, Clark JW, Solomon BJ, Iafrate AJ (2011) MET amplifica-tion identifies a small and aggressive subgroup of esophagogastricadenocarcinoma with evidence of responsiveness to crizotinib. J ClinOncol 29(36):4803–4810. doi:10.1200/JCO.2011.35.4928

57. Lee HE, Kim MA, Lee HS, Jung EJ, Yang HK, Lee BL, Bang YJ,Kim WH (2012) MET in gastric carcinomas: comparison betweenprotein expression and gene copy number and impact on clinicaloutcome. Br J Cancer 107(2):325–333. doi:10.1038/bjc.2012.237

58. Janjigian YY, Tang LH, Coit DG, Kelsen DP, Francone TD, WeiserMR, Jhanwar SC, ShahMA (2011) METexpression and amplificationin patients with localized gastric cancer. Cancer Epidemiol BiomarkersPrev 20(5):1021–1027. doi:10.1158/1055-9965.EPI-10-1080

59. Taguchi F, Kodera Y, Katanasaka Y, Yanagihara K, Tamura T,Koizumi F (2011) Efficacy of RAD001 (everolimus) against ad-vanced gastric cancer with peritoneal dissemination. Invest NewDrugs 29(6):1198–1205. doi:10.1007/s10637-010-9464-9

60. Lang SA, Gaumann A, Koehl GE, Seidel U, Bataille F, Klein D, EllisLM, Bolder U, Hofstaedter F, Schlitt HJ, Geissler EK, Stoeltzing O(2007) Mammalian target of rapamycin is activated in human gastriccancer and serves as a target for therapy in an experimental model. IntJ Cancer 120(8):1803–1810. doi:10.1002/ijc.22442

61. Xu DZ, Geng QR, Tian Y, Cai MY, Fang XJ, Zhan YQ, Zhou ZW, LiW, Chen YB, Sun XW, Guan YX, Li YF, Lin TY (2010) Activatedmammalian target of rapamycin is a potential therapeutic target ingastric cancer. BMC Cancer 10:536. doi:10.1186/1471-2407-10-536

62. Yu G,Wang J, Chen Y,Wang X, Pan J, Li G, Jia Z, Li Q, Yao JC, XieK (2009) Overexpression of phosphorylated mammalian target ofrapamycin predicts lymph node metastasis and prognosis of chinesepatients with gastric cancer. Clin Cancer Res 15(5):1821–1829. doi:10.1158/1078-0432.CCR-08-2138

63. Ohtsu A, Ajani JA, Bai YX, Bang YJ, Chung HC, Pan HM,Sahmoud T, Shen L, Yeh KH, Chin K, Muro K, Kim YH, Ferry D,Tebbutt NC, Al-Batran SE, Smith H, Costantini C, Rizvi S, LebwohlD, Van Cutsem E (2013) Everolimus for previously treated advancedgastric cancer: results of the randomized, double-blind, phase IIIGRANITE-1 Study. J Clin Oncol. doi:10.1200/JCO.2012.48.3552

64. Yoon DH, Ryu MH, Park YS, Lee HJ, Lee C, Ryoo BY, Lee JL,Chang HM, Kim TW, Kang YK (2012) Phase II study of everolimuswith biomarker exploration in patients with advanced gastric cancerrefractory to chemotherapy including fluoropyrimidine and platinum.Br J Cancer 106(6):1039–1044. doi:10.1038/bjc.2012.47

65. Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, Yan H,Gazdar A, Powell SM, Riggins GJ, Willson JK, Markowitz S,Kinzler KW, Vogelstein B, Velculescu VE (2004) High frequencyof mutations of the PIK3CA gene in human cancers. Science304(5670):554. doi:10.1126/science.1096502

66. Velho S, Oliveira C, Ferreira A, Ferreira AC, Suriano G, Schwartz SJr, Duval A, Carneiro F, Machado JC, Hamelin R, Seruca R (2005)The prevalence of PIK3CA mutations in gastric and colon cancer.Eur J Cancer 41(11):1649–1654. doi:10.1016/j.ejca.2005.04.022

67. Lee J, van Hummelen P, Go C, Palescandolo E, Jang J, Park HY,Kang SY, Park JO, KangWK,MacConaill L, Kim KM (2012) High-throughput mutation profiling identifies frequent somatic mutationsin advanced gastric adenocarcinoma. PLoS One 7(6):e38892. doi:10.1371/journal.pone.0038892

68. Shi J, Yao D, LiuW,Wang N, Lv H, Zhang G, Ji M, Xu L, He N, ShiB, Hou P (2012) Highly frequent PIK3CA amplification is associatedwith poor prognosis in gastric cancer. BMC Cancer 12:50. doi:10.1186/1471-2407-12-50

69. Barbi S, Cataldo I, De Manzoni G, Bersani S, Lamba S, Mattuzzi S,Bardelli A, Scarpa A (2010) The analysis of PIK3CA mutations ingastric carcinoma and metanalysis of literature suggest that exon-selectivity is a signature of cancer type. J Exp Clin Cancer Res 29:32.doi:10.1186/1756-9966-29-32

70. Mueller A, Bachmann E, Linnig M, Khillimberger K, SchimanskiCC, Galle PR, Moehler M (2012) Selective PI3K inhibition byBKM120 and BEZ235 alone or in combination with chemotherapyin wild-type and mutated human gastrointestinal cancer cell lines.Cancer Chemother Pharmacol 69(6):1601–1615. doi:10.1007/s00280-012-1869-z

71. Pinto M, Oliveira C, Machado JC, Cirnes L, Tavares J, Carneiro F,Hamelin R, Hofstra R, Seruca R, Sobrinho-Simoes M (2000) MSI-Lgastric carcinomas share the hMLH1 methylation status of MSI-Hcarcinomas but not their clinicopathological profile. Lab Invest80(12):1915–1923

72. Corso G, Velho S, Paredes J, Pedrazzani C, Martins D, Milanezi F,Pascale V, Vindigni C, Pinheiro H, Leite M, Marrelli D, Sousa S,Carneiro F, Oliveira C, Roviello F, Seruca R (2011) Oncogenicmutations in gastric cancer with microsatellite instability. Eur JCancer 47(3):443–451. doi:10.1016/j.ejca.2010.09.008

73. Oliveira C, Seruca R, Seixas M, Sobrinho-Simoes M (1998) Theclinicopathological features of gastric carcinomas with microsatelliteinstability may be mediated by mutations of different “target genes”:a study of the TGFbeta RII, IGFII R, and BAX genes. Am J Pathol153(4):1211–1219

74. Oliveira C, Pinto M, Duval A, Brennetot C, Domingo E, Espin E,Armengol M, Yamamoto H, Hamelin R, Seruca R, Schwartz S Jr(2003) BRAF mutations characterize colon but not gastric cancerwith mismatch repair deficiency. Oncogene 22(57):9192–9196. doi:10.1038/sj.onc.1207061

75. van Grieken NC, Aoyma T, Chambers PA, Bottomley D, Ward LC,Inam I, Buffart TE, Das K, Lim T, Pang B, Zhang SL, Tan IB,Carvalho B, Heideman DA, Miyagi Y, Kameda Y, Arai T, MeijerGA, Tsuburaya A, Tan P, Yoshikawa T, Grabsch HI (2013) KRASand BRAF mutations are rare and related to DNA mismatch repairdeficiency in gastric cancer from the East and theWest: results from alarge international multicentre study. Br J Cancer 108(7):1495–1501.doi:10.1038/bjc.2013.109

76. Brennetot C, Duval A, Hamelin R, Pinto M, Oliveira C, Seruca R,Schwartz S Jr (2003) Frequent Ki-ras mutations in gastric tumors ofthe MSI phenotype. Gastroenterology 125(4):1282

77. Warneke VS, Behrens HM, Haag J, Balschun K, Boger C, Becker T,Ebert MP, Lordick F, Rocken C (2013) Prognostic and putativepredictive biomarkers of gastric cancer for personalized medicine.Diagn Mol Pathol 22(3):127–137. doi :10.1097/PDM.0b013e318284188e

78. Amado RG, Wolf M, Peeters M, Van Cutsem E, Siena S, FreemanDJ, Juan T, Sikorski R, Suggs S, Radinsky R, Patterson SD, ChangDD (2008) Wild-type KRAS is required for panitumumab efficacy inpatients with metastatic colorectal cancer. J Clin Oncol 26(10):1626–1634. doi:10.1200/JCO.2007.14.7116

79. Karapetis CS, Khambata-Ford S, Jonker DJ, O’Callaghan CJ, Tu D,Tebbutt NC, Simes RJ, Chalchal H, Shapiro JD, Robitaille S, PriceTJ, Shepherd L, Au HJ, Langer C, Moore MJ, Zalcberg JR (2008) K-ras mutations and benefit from cetuximab in advanced colorectalcancer. N Engl J Med 359(17):1757–1765. doi:10.1056/NEJMoa0804385

80. Wittinghofer A, Scheffzek K, Ahmadian MR (1997) The interactionof Ras with GTPase-activating proteins. FEBS Lett 410(1):63–67

81. Carvalho J, van Grieken NC, Pereira PM, Sousa S, Tijssen M, BuffartTE, Diosdado B, Grabsch H, Santos MA, Meijer G, Seruca R,Carvalho B, Oliveira C (2012) Lack of microRNA-101 causes E-cadherin functional deregulation through EZH2 up-regulation in intes-tinal gastric cancer. J Pathol 228(1):31–44. doi:10.1002/path.4032

82. Corso G, Carvalho J, Marrelli D, Vindigni C, Carvalho B, Seruca R,Roviello F, Oliveira C (2013) Somatic mutations and deletions of theE-cadherin gene predict poor survival of patients with gastric cancer.J Clin Oncol 31(7):868–875. doi:10.1200/JCO.2012.44.4612

83. Ferreira P, Oliveira MJ, Beraldi E, Mateus AR, Nakajima T, GleaveM, Yokota J, Carneiro F, Huntsman D, Seruca R, Suriano G (2005)

Virchows Arch

Page 12: Biomarkers for gastric cancer: prognostic, predictive or targets of therapy?

Loss of functional E-cadherin renders cells more resistant to theapoptotic agent taxol in vitro. Exp Cell Res 310(1):99–104. doi:10.1016/j.yexcr.2005.07.010

84. Witta SE, Gemmill RM, Hirsch FR, Coldren CD, Hedman K, RavdelL, Helfrich B, Dziadziuszko R, Chan DC, Sugita M, Chan Z, BaronA, Franklin W, Drabkin HA, Girard L, Gazdar AF, Minna JD, BunnPA Jr (2006) Restoring E-cadherin expression increases sensitivity toepidermal growth factor receptor inhibitors in lung cancer cell lines.Cancer Res 66(2):944–950. doi:10.1158/0008-5472.CAN-05-1988

85. Xin HW, Yang JH, Nguyen DM (2013) Sensitivity to epidermalgrowth factor receptor tyrosine kinase inhibitor requires E-cadherinin esophageal cancer and malignant pleural mesothelioma.Anticancer Res 33(6):2401–2408

86. Leung WK, Wu MS, Kakugawa Y, Kim JJ, Yeoh KG, Goh KL, WuKC, Wu DC, Sollano J, Kachintorn U, Gotoda T, Lin JT, You WC,Ng EK, Sung JJ, Asia Pacific Working Group on Gastric C (2008)

Screening for gastric cancer in Asia: current evidence and practice.Lancet Oncol 9(3):279–287. doi:10.1016/S1470-2045(08)70072-X

87. Thekkek N, Muldoon T, Polydorides AD, Maru DM, Harpaz N,Harris MT, Hofstettor W, Hiotis SP, Kim SA, Ky AJ,Anandasabapathy S, Richards-Kortum R (2012) Vital-dye enhancedfluorescence imaging of GI mucosa: metaplasia, neoplasia,inflammation. Gastrointest Endosc 75(4):877–887. doi:10.1016/j.gie.2011.10.004

88. Catenacci DV, Kozloff M, Kindler HL, Polite B (2011) Personalizedcolon cancer care in 2010. SeminOncol 38(2):284–308. doi:10.1053/j.seminoncol.2011.01.001

89. Suraweera N, Duval A, Reperant M, Vaury C, Furlan D, Leroy K,Seruca R, Iacopetta B, Hamelin R (2002) Evaluation of tumor mi-crosatellite instability using five quasimonomorphic mononucleotiderepeats and pentaplex PCR. Gastroenterology 123(6):1804–1811.doi:10.1053/gast.2002.37070

Virchows Arch


Recommended