Molecular Classification of Gastric Cancer

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Expert Review of Molecular Diagnostics

ISSN: 1473-7159 (Print) 1744-8352 (Online) Journal homepage: http://www.tandfonline.com/loi/iero20

Molecular classification of gastric cancer

Christoph Röcken

To cite this article: Christoph Röcken (2017) Molecular classification of gastric cancer, Expert
Review of Molecular Diagnostics, 17:3, 293-301, DOI: 10.1080/14737159.2017.1286985

To link to this article: http://dx.doi.org/10.1080/14737159.2017.1286985

Accepted author version posted online: 24


Jan 2017.
Published online: 06 Feb 2017.

Submit your article to this journal

Article views: 183

View related articles

View Crossmark data

Citing articles: 2 View citing articles

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=iero20

Download by: [La Trobe University] Date: 01 October 2017, At: 13:01
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS, 2017
VOL. 17, NO. 3, 293–301
http://dx.doi.org/10.1080/14737159.2017.1286985

REVIEW

Molecular classification of gastric cancer


Christoph Röcken
Department of Pathology, Christian-Albrechts-University, Kiel, Germany

ABSTRACT ARTICLE HISTORY


Introduction: Gastric cancer is among the most common cancers worldwide. Despite declining inci- Received 27 November 2016
dences, the prognosis remains dismal in Western countries and is better in Asian countries with national Accepted 23 January 2017
cancer screening programs. Complete endoscopic or surgical resection of the primary tumor with or KEYWORDS
without lymphadenectomy offers the only chance of cure in the early stage of the disease. Survival of Gastric cancer; molecular
more locally advanced gastric cancers was improved by the introduction of perioperative, adjuvant and classification; histology;
palliative chemotherapy. However, the identification and usage of novel predictive and diagnostic Epstein-Barr virus;
targets is urgently needed. microsatellite instability;
Areas covered: Recent comprehensive molecular profiling of gastric cancer proposed four molecular HER2
subtypes, i.e. Epstein-Barr virus-associated, microsatellite instable, chromosomal instable and genomi-
Downloaded by [La Trobe University] at 13:01 01 October 2017

cally stable carcinomas. The new molecular classification will spur clinical trials exploring novel targeted
therapeutics. This review summarizes recent advancements of the molecular classification, and based
on that, putative pitfalls for the development of tissue-based companion diagnostics, i.e. prevalence of
actionable targets and therapeutic efficacy, tumor heterogeneity and tumor evolution, impact of
ethnicity on gastric cancer biology, and standards of care in the East and West.
Expert commentary: The overall low prevalence of actionable targets and tumor heterogeneity are the
two main obstacles of precision medicine for gastric cancer.

1. Introduction sporadic GC: a chronic atrophic gastritis leads to intestinal


metaplasia, dysplasia, and finally the occurrence of GC [2]. In
Gastric cancer (GC) is the fifth most common cancer world-
his initial model, a diet rich in salt, and medication were
wide with an estimated 952,000 new cases (7% of total cancer
considered as driving forces of chronic gastritis. Later on,
incidence) and 723,000 deaths in 2012 [1]. The incidence and
after Barry J. Marshall and J. Robin Warren had identified H.
prevalence of GC vary greatly geographically. The highest
pylori as a cause of chronic bacterial (type B) gastritis, Correa
rates are observed in East Asia, Central Europe, and Eastern
modified his model and added H. pylori as causative agent of a
Europe accounting collectively for 87% of all new GC cases in
chronic inflammation [3]. Since then ample epidemiological,
the world. Significant lower rates are found in Africa and North
animal experimental, and therapeutic evidence support the
America [1]. Worldwide, the standardized incidence rates of
contention that chronic H. pylori-induced gastritis leads to
noncardia GC have been declining steadily over the last 30–
GC formation. In 1994, H. pylori was classified as group 1
50 years. To the contrary, cancer of the proximal stomach/
human carcinogen by the International Agency on Cancer.
cardia and esophago-gastric junction has been stable or even
Chronic colonization of the stomach mucosa by H. pylori may
increased in its incidence. Patient prognosis remains dismal in
lead to different phenotypes of chronic gastritis. The antrum-
countries without GC screening programs. GC is a disease of
predominant form is associated with ulcers of the stomach
the elderly often reaching the peak incidence in the seventh
and duodenal mucosa without an increased risk of cancer
and eighth decade of life. Five-year survival rate then ranges
development. A corpus-predominant gastritis carries a sub-
between 30–35% for men and women. Particularly, in low-
stantial risk for the development of GC through mucosal
incidence regions the disease is often diagnosed at an
atrophy, hypacidia, and intestinal and pseudopyloric metapla-
advanced stage limiting the therapeutic options. Prognosis
sia [4]. The indolent form of H. pylori-associated gastritis pre-
was improved in countries with high incidence rates and
sents neither with ulcers nor with an increased risk of GC
national cancer screening programs, e.g. Japan and Korea,
formation. Several bacterial virulence factors for colonization,
leading to diagnosis of GC in earlier disease stages.
persistence, ulcer, and cancer risk have been identified in
recent years, e.g. CagA, DupA, IceA1, NapA, oipA, and VacA
2. Etiology [5]. These are modulated by host factors, e.g. polymorphisms
in genes coding for pro- and anti-inflammatory cytokines, as
GC may occur sporadically, familial, or as a hereditary disease.
well as environmental and socioeconomic cues. The individual
In 1988, Correa described a model for the carcinogenesis of

CONTACT Christoph Röcken christoph.roecken@uksh.de Department of Pathology, Christian-Albrechts-University, Arnold-Heller-Str. 3, Haus 14, D-24105
Kiel, Germany
© 2017 Informa UK Limited, trading as Taylor & Francis Group
294 C. RÖCKEN

disease manifestation and cancer risk is a result of a complex categorizes four subtypes, i.e. microsatellite instable (MSI),
interaction between host and microbiotic traits [6]. Epstein-Barr-virus (EBV)-positive, chromosomal instable, and
The familial forms of GC may share risk factors, such as H. genomically stable GCs [17,18]. These studies for the first
pylori-infection, nutritional habits, and gene polymorphisms, time build a bridge between distinct molecular subtypes of
e.g. in pro- and anti-inflammatory cytokine genes. GC and some phenotypes. It may form the basis for the
Approximately 5–10% of the patients with GC carry germ development of novel therapeutic strategies [17,18]. The chro-
line mutations and are truly hereditary forms [7,8]. A total of mosomal instable type often shows an intestinal phenotype
30–40% of the cases with a diffuse-type hereditary GC harbor according to Laurén, frequently harbors mutations in the P53-
mutations or copy number changes in the CDH1 gene, which tumor suppressor gene as well as activating mutations in
codes for E-cadherin. More than 50 different mutations of the genes coding for receptor tyrosine kinases. The genomically
CDH1 gene have been described hitherto. The mutations are stable GC often is of a diffuse type, with mutations occurring
found across the entire gene. α-E-catenin (CTNNA1) was also in CDH1 and RHOA. They also show alterations in cell adhesion
linked to diffuse type hereditary GC and germline mutations, molecules. The EBV-associated variant and microsatellite
including nonsense and frameshift mutations, were found in instable GCs (MSI-GCs) usually present with an unusual phe-
three families [9,10]. Other genes possibly associated with notype [18,19].
hereditary GC are insulin receptor (INSR), F-Box-protein 24 Based on the molecular classification of GC one might raise
(FBXO24), mitogen-activated protein kinase kinase kinase 6 the question as which genotype is associated with which
(MAP3K6), protease serine 1 (PRSS1), and DOT1-like histone phenotype (=morpho-molecular classification of GC).
Downloaded by [La Trobe University] at 13:01 01 October 2017

H3K79 methyltransferase (DOT1L), and require verification


[10,11]. Recently, Worthley et al. described the first kindred
4.1. Microsatellite instable gastric cancer (MSI-GC)
with an intestinal type hereditary GC syndrome associated
with proximal polyposis [12]. The underlying mutation was MSI-GCs are hypermutated tumors carrying a CpG-island
found in the APC Promoter 1B [13]. GC can also be part of methylator phenotype (CIMP). They usually demonstrate silen-
other hereditary cancer syndromes such as familial adenoma- cing of MLH1, which encodes a DNA-mismatch repair protein.
tous polyposis coli (APC), Lynch syndrome (hMLH1, hMLH2), In the literature, the prevalence of MSI-GC ranges from 0% to
Cowden syndrome (PTEN), juvenile polyposis, Li-Fraumeni syn- 44.5% [20]. MSI-GCs are significantly more prevalent in elderly
drome (TP53), MUTYH-associated adenomatous polyposis patients, the distal stomach and are associated with a signifi-
(MUTYH), Peutz-Jeghers syndrome (STK11), and hereditary cantly lower number of lymph node metastases and a signifi-
breast and ovarian cancer (BRCA1/2) [14]. cantly better overall and tumor-specific survival. MSI-GC lack
BRAF-mutations commonly found in sporadic MSI colorectal
cancer [18]. In fact, BRAF-mutations are virtually absent in
3. Histology of GC
GC [21].
The World Health Organization (WHO) published in 2010 a In our own GC cohort we found 34 (7.5%) patients with
new classification system for malignant tumors of the gastro- MSI-GC [20]. A total of 21 (6.2%) patients with MSI-GC showed
intestinal tract [15]. Several modifications were introduced for unusual histological features. The tumors consisted predomi-
GC. The adenocarcinomas were subclassified into tubular, nantly of highly pleomorphic tumor cells with large vesicular
papillary, mucinous, poorly cohesive (including the signet nuclei in a trabecular, nested, microalveolar, or solid growth
ring cell variant), and mixed type. The carcinoma with lym- pattern. Tumor cell size was variable, sometimes exhibiting
phoid stroma (medullary carcinoma) and the hepatoid adeno- tumor cells that had lymphocytoid or blastoid appearance.
carcinoma were added. While no recommendation was given An abundant tumor-associated inflammatory stroma consist-
for ancient classification schemes [15], classification according ing of either polymorphs and/or lymphocytes was frequently
to Laurén is still widely used in many clinical trials and GC observed in these cases, usually with little or no desmoplastic
research publications [16]. According to Laurén, GC is classi- stroma. Thirteen patients demonstrated several of these his-
fied into intestinal, diffuse, mixed, and unclassified [16]. The tological features, whereas eight had only few discriminating
vast majority (>90%) of GCs are adenocarcinomas. Recent features mainly consisting of pushing margins and/or a lym-
comprehensive molecular characterization studies were car- phocytes or polymorphs. In three cases the inflammatory
ried out with gastric adenocarcinomas and utilized the component was the only unusual feature [20]. Interestingly,
Laurén classification [17]. Thus, the morpho-molecular classifi- the tumors of 13 (3.8%) patients showed no distinctive histo-
cation of GC matches only rudimentary with the current WHO logical features and were indistinguishable from any of the
classification [18]. microsatellite stable GCs. We found no pure poorly cohesive,
signet ring cell carcinomas in the MSH-GC group even though
signet ring cells can be part of a mixed pattern in MSI-GC.
4. Molecular subtypes of GC
The accurate classification of MSI-GC may become clinically
The genetic complexity of GC has been recently shown in an relevant for two reasons: (1) MSI-GCs may not require any stan-
integrative genomic analysis including array-based somatic dard adjuvant (radio-)chemotherapy in a curative setting [20]. (2)
copy number analysis, whole-exome sequencing, array-based MSI-GCs express the immune checkpoint molecules PD-L1 and
DNA methylation profiling, messenger RNA sequencing, PD-1 and may be considered suitable for the treatment with
microRNA sequencing, and reverse-phase protein array analy- immune checkpoint inhibitors in the palliative setting [18,22].
sis [18]. A molecular classification was proposed, which The classification and identification of MSI-GCs can be done
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS 295

with antibodies directed against MLH1, PMS2, MSH2, and MSH6 diagnosis of EBVaGC may become clinically relevant in the near
as well as usage of the mononucleotide markers BAT-25, BAT-26, future.
NR-21, NR-24, and NR-27 [20]. However, there is currently no
international consensus on the diagnosis of MSI-GCs and our
4.3. Chromosomal instable GCs
group adapted the procedure utilized for colorectal cancer [20].
Tumor heterogeneity is an issue in MSI-GCs. We identified The chromosomal instable GCs often show an intestinal
a single case which showed a biphasic MSH2-expression phenotype according to Laurén, frequently harbor mutations
pattern. Staining of the whole tissue slides unexpectedly in the P53-tumor suppressor gene and activating mutations
demonstrated a loss of MSH2 in the major part (between in genes coding for receptor tyrosine kinases [18].
77% and 95%) of the tumor whereas a small part (min. 5%, Approximately 37% of all GCs harbor amplifications of
max. 23%) expressed MSH2. Subsequent molecular biologi- genes coding for receptor tyrosine kinases such as epider-
cal studies of the microdissected tumor areas confirmed mal growth factor receptor (EGFR), FGFR2, HER2, and MET
microsatellite stability in the MSH2-positive and MSI in the [19,29]. EGFR-amplified GCs present with a male preponder-
MSH2-negative area. Furthermore, analyses of the lymph ance and affect more commonly the distal stomach [28].
node metastases confirmed that only the microsatellite HER2-amplified GCs are equally distributed among men
stable tumor subclone had metastasized. Interindividual and women, and tend to be more prevalent in the proximal
and intratumoral heterogeneity of MSI-GCs may significantly stomach. HER2-amplification does not correlate with patient
impact on clinical decision-making. Histological phenotype prognosis [30,31]. MET-amplified GCs are found more com-
Downloaded by [La Trobe University] at 13:01 01 October 2017

may raise suspicion in the majority of cases. However, one monly in men and proximal GCs. They have significantly
third of the tumors fail to exhibit any specific phenotype more lymph node and distant metastases, and are charac-
and MSI-testing may need a more general approach. The terized by a very poor prognosis with a median overall
vast majority of MSI-GC demonstrates loss of MLH1-expres- survival of less than 6 months [32,33].
sion and immunostaining for MLH1 may be considered as FGFR2 is another receptor tyrosine kinase, which is ampli-
the most cost-effective approach to screen for MSI [23]. fied in approximately 7–10% of GCs [19,33]. Up to now, no
However, it has to be kept in mind that other members of association was found between FGFR2 amplification and
the DNA-repair machinery can be lost and that in patients patient gender, anatomical site, histology, or TNM classifica-
with lymph node metastases, testing the primary tumor and tion [19,33]. However, there has been some evidence that
lymph nodes metastases might help to unravel intratumoral FGFR2-amplified GCs may exert poor survival outcome [19].
heterogeneity. Amplification of FGFR2 is not restricted to chromosomal
instable GCs and can also be observed in genomically stable
GCs. This might explain why no association was found
4.2. Epstein-bar-virus-associated gastric cancer
between tumor type and FGFR2 status [33].
(EBVaGC)
There is currently no uniform approach for the classification
EBVaGC is another molecular subtype of GC [17,18]. EBVaGC of chromosomal instable GCs. Immunostaining of p53 and
also show a CIMP-phenotype, frequently harbor PIK3CA- and detection of receptor tyrosine kinase expression and amplifi-
ARID1A-mutations and dysregulation of immune cell signal- cation have been proposed recently as surrogate markers
ing. EBVaGC are characterized by male predominance, occur- [23,28]. However, in view of the observations made with
rence primarily in the proximal and postgastrectomy FGFR2, other markers might be needed.
remnant stomach, multiplicity, and association with lympho-
cytic infiltration [24]. The number of lymph node metastases
4.4. Genomically stable GCs
was negatively associated with EBV status [25], and in Asian
populations EBVaGc may exert a better prognosis compared Genomically stable GCs are supposed to have primarily a
with EBV-negative GCs [26]. EBVaGC and MSI-GC are diffuse histological phenotype according to Laurén, harbor
mutually exclusive [20]. The phenotype of EBVaGC is variable CDH1- and RHOA mutations as well as rearrangements
with tubular and intestinal differentiation. It may exert an between CLDN18 and ARHGAP26 or ARHGAP6 [18,34].
undifferentiated phenotype (lymphoepithelioma-like or Claudin 18 is a component of the tight junctions and its
medullary). Any GC with an unusual phenotype should be down regulation was associated with impaired survival [35].
forwarded to EBV-testing using EBER-in situ-hybridization, Elevated expression of cell adhesion pathways, including the
which gives a nuclear signal. Immunostaining is less sensi- B1/B3 integrins, syndecan-1-mediated signaling, and angio-
tive, because GC is commonly of latency type 1 and hence genesis-related pathways are also associated with the geno-
immunonegative for EBNA2, LMP1, and ZEBRA [24,27]. Given mically stable genotype. However, our own validation
the wide morphological spectrum of EBVaGC, testing should studies demonstrated that RHOA mutations also occur in
be done at leisure [28]. In addition, all EBVaGCs harbor intestinal type GCs [36] and the integrins αvβ3 and αvβ5
promotor hypermethylation of CDKN2A (p16INK4A) and immu- are expressed significantly more often in the intestinal phe-
nostaining of p16 may serve as a putative surrogate marker notype [37]. The expression of αvβ5 on stroma cells was
for EBVaGC [18]. found to be an independent prognostic factor of intestinal-
Similar to MSI-GC, EBVaGCs significantly more commonly type GC [37]. Thus, while comprehensive molecular charac-
express PD-L1 and PD-1 and might thus be sensitive to therapy terization of GC has generated highly informative data, vali-
with immune checkpoint inhibitors [22]. Thus recognition and dation studies are needed to make the findings more robust.
296 C. RÖCKEN

The morpho-molecular classification of GC remains interindividual variability of mutational load [40]. Only P53-
cumbersome. mutations are highly prevalent in GC. The vast majority of
mutations is in the range of <10%, which supports the con-
tention that GC is a complex disease. A low prevalence of
5. Translational aspects actionable targets complicates clinical trials. Pharmaceutical
Molecular classification raises hopes not only for an improved companies consider issues of financing and re-imbursement.
morpho-molecular classification of GC but also for novel treat- Principle investigators need to consider patient numbers for
ment options (see Table 1). This applies for the prognostica- statistical analyses. Basket trials with upfront molecular testing
tion of patient outcome and for the biomarker-based can help to overcome these limitations. However, they may
prediction of therapeutic response (companion diagnostics). need to focus on GC rather than being part of diverse other
With regard to prognosis, there has been some evidence in solid organ tumors under investigation, and each actionable
colorectal cancers that stage II MSI colorectal cancers may not target requires verification for each individual tumor type. The
benefit from therapy with 5-fluoruracil [38,39]. This may also EXPAND study, which tested capecitabine and cisplatin with or
apply to GC. It is a disease of the elderly, often presenting with without cetuximab for patients with previously untreated
co-morbidities, and avoiding perioperative/neoadjuvant or advanced GC, nicely illustrated the obstacles: adding cetuxi-
adjuvant oncological treatment might be an option, worth to mab to capecitabine–cisplatin as first-line treatment did not
consider prospectively in MSI-GC. result in any benefit [41]. Cetuximab is a fully humanized
With regard to the other three molecular subtypes, i.e. antibody directed against the EGFR and provides significant
Downloaded by [La Trobe University] at 13:01 01 October 2017

EBVaGC, chromosomal instable, and genomically stable GCs, survival benefit to patients with advanced colorectal cancer
currently there is not sufficient evidence that they might be with wild-type KRAS status [42]. As in comparison with color-
suitable for risk stratification [18]. However, single genetic ectal cancer, KRAS mutations are rare in GC while they do
alterations have been associated with poor patient outcome, express EGFR. Therefore, it was somewhat reasonable to test
e.g. MET-amplification, and might outperform grouping efficacy and safety of cetuximab in KRAS-wildtype GCs.
according to one of the three other aforementioned molecular However, cetuximab failed to show any benefit and the rela-
subtypes. tive success in colorectal cancer could not be recapitulated in
With regard to predictive biomarkers, recent in-depth GC [41]. These data provide evidence that the presence of an
molecular analyses of GC unraveled many actionable targets actionable target per se does not guarantee therapeutic effi-
and raise hope that these offer new chances for targeted cacy in a given tumor/patient.
therapy of GC [17,18] (see Table 1). However, a few issues
merit consideration. 5.2. Tumor heterogeneity and tumor evolution
Our own recent validation studies showed that the different
5.1. Low prevalence of actionable targets and molecular subtypes overlap. GC may harbor microsatellite
therapeutic efficacy stable and instable areas adjacent to each other [20]. RHOA
mutations are found in intestinal type GCs [36]. In addition,
The subgrouping into four molecular subtypes cannot mask
the amplification of HER2 and MET may occur in the same
the problem of an overall low prevalence of the individual
primary tumor albeit in different areas [32]. These observa-
genetic alterations. GC belongs to the group of cancers with a
tions highlight a specific issue of GC, i.e. intratumoral hetero-
high frequency of somatic mutations as well as a substantial
geneity. Approximately 30% of all GCs are heterogeneous
and may compromise morpho-molecular classification of GC
Table 1. Comprehensive molecular characterization of gastric cancer classified and hence selection of an appropriate targeted therapy.
four molecular subtypes, which harbor several interesting actionable targets and Tumor heterogeneity is becoming one of the main obstacles
treatment options.
of cancer treatment in the era of precision medicine.
Molecular subtype Target Treatment/drug
Sequencing of multiple samples of primary clear cell renal
MSI No adjuvant chemotherapy?
MSI/EBV-associated PD-L1/PD-1 Nivolumab, Pembrolizumab, cell carcinoma and its metastatic sites revealed the complex-
Atezolizumab, ity of intraindividual cancer genetics, and allowed reconstruc-
Durvalumab, tion of its evolutionary history [43,44]. Genetically distinct
Avelumab,
Ipilimumab subclones were found in the primary tumor and distant
EBV-associated PIK3CA/AKT/mTOR PX-866, NVP-BYL719, metastases. Cancer is now viewed as a highly dynamic evolu-
MLN1117, NVP-BEZ236, tionary disease [45], with continuing mutations favoring the
Temsirolimus, Everolimus,
Ridaforolimus emergence of new (sub)clones with distinct biological prop-
Chromosomal instable EGFR Cetuximab, Panitumumab erties already in precursor lesions [46]. Subclones may show
HER2 Trastuzumab, Pertuzumab different patterns of interaction; they may compete, overtake
MET Onartuzumab, Rilotumumab,
Crizotinib, Foretinib, other clones, parasitize, or peacefully coexist. A ‘trunk and
Tivantinib branch’ model has been proposed for tumor evolution [47].
VEGFA Bevacizumab Common events, found in every subclone of the tumor
VEGFR2 Ramucirumab
Genomically stable Integrins Cilengetide region, are represented in the trunk. Heterogeneous somatic
EpCam Catumaxumab events occurring in a limited number of (or even a single)
MSI: microsatellite instable; EBV: Epstein-Barr-virus subclone(s) represent the branches of the tree [47]. While
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS 297

targeting alterations in the trunk might be most promising as


they occur in all tumor cells, many actionable targets exclu-
sively occur in branches. This subclonal heterogeneity of
actionable targets poses problems in diagnosis (tissue sam-
pling error) and therapy (emergence of resistance).
Treatment and eradication of a subclone may provide a
growth advantage for a competing non-responsive subclone,
which finally may lead to patient death. This is probably why
the efficacy of several targeted therapies was limited in time
and also somewhat unpredictable [47]. Intratumoral molecu-
lar heterogeneity appears to be inherent to tumor evolution
and has also consequences for sampling procedures. We
have studied the impact of tissue sampling on Her2/neu
testing in GC. Her2/neu has been introduced as a predictive
biomarker for treatment of GC with trastuzumab [48].
Amplification of genes encoding receptor-tyrosine kinases
usually occurs in genomically instable GCs [18], which is
probably why in GC Her2/neu expression might be intrinsi-
Downloaded by [La Trobe University] at 13:01 01 October 2017

cally heterogeneous (Figure 1). We assessed Her2/neu status


(according to the GC scoring system [49]) using a tissue
micro array approach, in which five tissue cores per paraffin
‘donor’ block served as surrogate for a biopsy procedure, and
compared the results with those obtained on whole tissue
sections cut from the same paraffin block. On the TMA cores
we obtained a ‘false-negative’ rate of 24% and a ‘false-posi-
tive’ rate of 3% [31]. Silva et al. [33] made similar observa-
tions when they explored gene copy number status of
receptor tyrosine kinase and downstream signaling genes in
primary GC and matched lymph node metastases. A total of
68% of their 103 patients with results from primary GC and
matched lymph node metastasis showed a conversion of the
copy number status: they found a significant intraindividual
difference for KRAS, MYC, CCNE, HER2, and FGFR2 [33].
Heterogeneous expression of predictive biomarkers poses a
major challenge in clinical trials but this issue is often neglected.
The phenomenon is often first explored in retrospective bio-
marker studies, once a treatment modality has been formally
approved by a regulatory body. As an example, many studies
on Her2/neu expression were carried out after approval of
trastuzumab for the treatment of GC (for a review see [50]).
These issues should be addressed prior to implementation of a
trial and whether (expression of) the molecular target of the
treatment is homogeneous (‘trunk-alteration’) or heteroge-
Figure 1. Morpho-molecular classification of gastric cancer. A molecular
neous (‘branch-alteration’) should be explored in advance and classification was proposed, which categorizes four subtypes, i.e. microsatel-
translated into suitable biopsy procedures [51]. However, many lite instable (a, b), Epstein-Barr-virus-positive (c, d), chromosomal instable (d-
studies address treatment in later stages of cancer with pallia- g), and genomically stable gastric cancers (h). This microsatellite instable
gastric cancer has a solid growth pattern (a) and a loss of MLH1-expression
tive intent and analyses rely on biopsy samples only, as patients (b). MLH1-immunopositive tumor-infiltrating lymphocytes serve as internal
may not be eligible for tumor resection. However, an essential positive control (b). Epstein-Barr-virus-positive cancers (c) often yield a
requirement for any trial is the exploration of tumor heteroge- lymphocytic infiltration (lymphoepithelioma-like) and nuclear staining after
EBER-in situ hybridization (d). Chromosomal instable gastric cancers often
neity before the trial takes off, to avoid sampling errors and have an intestinal phenotype according to Laurén (e) and activating muta-
favor representative tissue biopsies [51]. In this respect it might tions in genes coding for receptor tyrosine kinases, such as Her2/neu (f).
not be surprising that results of recent studies communicated However, Her2/neu-overexpression (f) and HER2-gene amplification (g) can
be heterogeneous, with positive tumor areas (arrow heads) and negative
during the annual meeting of the American Society of Clinical areas (arrows) within the same primary tumor. Genomically stable gastric
Oncologists failed to show a benefit of MET inhibitors in GC. In cancers are often of a diffuse phenotype according to Laurén (h).
both studies, drug efficacy was tested along with a potential Hematoxylin and eosin (a, c, e, h); anti-MLH1-immunostaining (b); EBER-in
situ hybridization (d); Her2/neu-immunostaining (d); HER2-in situ hybridiza-
companion diagnostic [52,53]. In fact, apart from HER2, no other tion (g). Original magnifications 400-fold.
298 C. RÖCKEN

companion diagnostic reached clinical application in GC, so far. of the recurrent tumor in a palliative setting. Second, ther-
Ramucirumab, which targets VEGF-receptor 2, does not require apy affects tumor biology. Companion diagnostics explored
companion diagnostics. in untreated tumors may necessitate validation in neoadju-
Based on the observations made for Her2/neu, an interdis- vantly treated tumors due to subclone eradiation and sub-
ciplinary group of German experts has taken on the challenges clone selection. This may become particularly relevant for
of Her2/neu testing in GC and recommended that at least five the use of immune checkpoint inhibitors and their cognate
tumor-bearing biopsies should be obtained from different companion diagnostics.
sites of the primary tumor [54]. Similarly, the College of
American Pathologists, the American Society of Clinical
6. Expert commentary
Pathology and the American Society of Clinical Oncology
recently published recommendations for HER2 testing [55]. The recent comprehensive molecular characterization has
Testing of multiple biopsy fragments (from a primary or meta- advanced our understanding of the complexity and diversity
static site), or from the resected primary tumor, is preferred. of GC. Trastuzumab (Her2/neu) [48] and ramucirumab (VEGF-
For biopsy specimens, current recommendations state that, receptor 2) have been approved as targeted therapy of GC.
when possible, a minimum of five biopsy specimens, and Administration of trastuzumab only applies to a small sub-
optimally six to eight, should be obtained to account for group of patients with overexpression of Her2/neu and/or
intratumoral heterogeneity and to provide sufficient tumor amplification of HER2 [40]. No other companion diagnostic
specimens for diagnosis and biomarker testing [55]. has reached clinics. Neither EGFR nor MET had been validated
Downloaded by [La Trobe University] at 13:01 01 October 2017

as suitable targets for precision medicine of GC. However, as


shown in Table 1, there are a substantial number of drugs
5.3. Impact of ethnicity on GC biology available, which are directed against a variety of dysregulated
molecules of the four molecular subtypes. The overall low
Several studies carried out on the U.S. patient populations
prevalence of many actionable targets and tumor heteroge-
have shown that ethnicity impacts on patient survival prob-
neity of GC are probably the two main obstacles. Sampling
ably reflecting a difference in GC tumor biology [56–63]. The
errors leading to false negative and false positive test results is
recent comprehensive molecular characterization also
a specific risk in GC. Patients with advanced GC may be
explored the putative impact of ethnicity. The authors did
considered ineligible for gastrectomy and biopsy samples
not observe any systematic differences in distribution of sub-
might be the only tissue specimen available for genetic testing
types between patients of East Asian and Western origin and
and companion diagnostics. The histological phenotype may
they also were unable to identify a strong biologic difference
not be representative in a biopsy specimen as well as the
between tumors of East Asian origin compared with other
diverse genetic alterations described earlier. Recently, expert
tumors [18]. However, patient numbers were low and further
panels suggested that at least five tumor-bearing biopsies
analysis with larger sample cohorts will be required to better
should be studied in GC in order to enable adequate HER2-
delineate differences that may exist between GCs originating
testing. This may also apply to other putative biomarkers of
from different regions of the world and in patients of different
GC. Thus, future studies also need to consider the risk of
ethnic backgrounds [18]. This may become particularly impor-
misinterpretation, and lack of response as well as tumor pro-
tant, when treatment with checkpoint inhibitors is to be
gression may require repeat tissue sampling in order to clas-
explored. GCs in Asian and non-Asian patient populations
sify GC correctly. In our own cohort the evidence of tumor
exhibit distinct tumor immunity signatures related to T-cell
heterogeneity in GC was overwhelming (even including the
function [64]. This may have an effect on the development
Ki67-status [68]) and we discontinued the use of tissue micro
of companion diagnostics and therapeutic efficacy [22].
arrays for biomarker analyses.

5.4. Standards of care for the treatment of GC 7. Five-year view


Standards of care vary in different regions of the world, In the following years, further efforts will be spent to improve
which may also effect molecular classification and compa- (perioperative, adjuvant, and palliative) oncological treatment
nion diagnostics of GC. Perioperative and adjuvant chemo of GC. These will include checkpoint inhibitors and they
(radio-)therapy of GC has become standard of care in should explore the necessity of chemotherapy in MSI-GC.
Western countries [65,66], while in Eastern Asia, adjuvant Validation studies hopefully unravel the impact of ethnicity
chemotherapy is standard of care [67]. In general, adjuvant on the molecular classification of GC and should also spend
chemotherapy offers the chance for an in-depth exploitation major attention on tumor heterogeneity. Ultimately, the
of the resected primary tumor and its corresponding lymph upcoming years should show whether the four-tire molecular
node metastases prior to (radio-)chemotherapy. Intratumoral classification stands the test of time or needs revision/
heterogeneity can be explored systematically on a patient- amendment.
by-patient basis. Neoadjuvant/perioperative chemotherapy
carries the risk to compromise molecular classification in
Key issues
two ways. At first, therapy-induced regression may prohibit
accurate molecular classification by reducing overall tumor ● Gastric cancer is among the most common cancers world-
burden. Companion diagnostic may need biopsy sampling wide with a dismal prognosis. It may occur sporadically,
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS 299

familial or as a hereditary disease. Nutritional habits and 4. Uemura N, Okamoto S, Yamamoto S, et al. Helicobacter pylori
colonization of the stomach by H. pylori are major causes of infection and the development of gastric cancer. N Engl J Med.
2001;345(11):784–789.
gastric cancer.
5. Malfertheiner P, Link A, Selgrad M. Helicobacter pylori: perspectives
● Histological phenotypes of gastric cancer according to the and time trends. Nat Rev Gastroenterol Hepatol. 2014;11(10):628–
World Health Organization-criteria are the tubular, papillary, 638.
mucinous, poorly cohesive, mixed type, medullary and 6. Amieva M, Peek RM Jr. Pathobiology of Helicobacter pylori-induced
hepatoid carcinoma. According to Laurén, gastric cancer is gastric cancer. Gastroenterology. 2016;150(1):64–78.
7. Oliveira C, Pinheiro H, Figueiredo J, et al. Familial gastric cancer:
classified into intestinal, diffuse, mixed and unclassified.
genetic susceptibility, pathology, and implications for manage-
● A recent molecular classification proposed four subtypes, ment. Lancet Oncol. 2015;16(2):e60–e70.
i.e. microsatellite instable, Epstein-Barr-virus-positive, chro- 8. Yamada H, Sakamato H, Sugimura H. Sexy small copy numbers in
mosomal instable, and genomically stable gastric cancers. hereditary gastric carcinogenesis. J Gastroint Dig Syst. 2014;4:4.
● Microsatellite instable and Epstein-Barr-virus associated 9. Majewski IJ, Kluijt I, Cats A, et al. An α-E-catenin (CTNNA1) mutation
in hereditary diffuse gastric cancer. J Pathol. 2013;229(4):621–629.
gastric cancers significantly more commonly express PD-
10. Petrovchich I, Ford JM. Genetic predisposition to gastric cancer.
L1 and PD-1 and might thus be sensitive to therapy with Semin Oncol. 2016;43(5):554–559.
immune checkpoint inhibitors. 11. Donner I, Kiviluoto T, Ristimäki A, et al. Exome sequencing reveals
● Chromosomal instable gastric cancers frequently harbor three novel candidate predisposition genes for diffuse gastric can-
P53-mutations and activating mutations in genes coding cer. Fam Cancer. 2015;14(2):241–246.
12. Worthley DL, Phillips KD, Wayte N, et al. Gastric adenocarcinoma
for receptor tyrosine kinases, e.g. EGFR, FGFR2, HER2,
and proximal polyposis of the stomach (GAPPS): a new autosomal
Downloaded by [La Trobe University] at 13:01 01 October 2017

and MET. dominant syndrome. Gut. 2012;61(5):774–779.


● Genomically stable cancers are characterized by CDH1- and 13. Li J, Woods SL, Healey S, et al. Point mutations in exon 1B of APC
RHOA mutations as well as rearrangements between reveal gastric adenocarcinoma and proximal polyposis of the sto-
CLDN18 and ARHGAP26 or ARHGAP6. mach as a familial adenomatous polyposis variant. Am J Hum
Genet. 2016;98(5):830–842.
● Overall low prevalence of mutations in actionable targets,
14. van der Post RS, Vogelaar IP, Carneiro F, et al. Hereditary diffuse
intra- and interindividual tumor heterogeneity, as well as gastric cancer: updated clinical guidelines with an emphasis on
different standards of stomach cancer treatment in the East germline CDH1 mutation carriers. J Med Genet. 2015;52(6):361–
and West are obstacles for the development of companion 374.
diagnostics. 15. Lauwers GY, Carneiro F, Graham DY, et al. Tumours of the stomach.
In: Bosman FT, Carneiro F, Hruban RH, et al., editors. WHO classifi-
● Implementation of check point inhibitors in gastric cancer
cation of tumours of the digestive system. Lyon: International
care needs consideration of ethnicity: Asian and non-Asian Agency for Research on Cancer; 2010. p. 48–80.
patient populations exhibit distinct tumor immunity 16. Lauren P. The two histologic main types of gastric carcinoma:
signatures. diffuse and so-called intestinal-type carcinoma. Acta Pathol
Microbiol Scand. 1965;64:31–49.
17. Wang K, Yuen ST, Xu J, et al. Whole-genome sequencing and
Funding comprehensive molecular profiling identify new driver mutations
in gastric cancer. Nat Genet. 2014;46(6):573–582.
This paper was not funded. •• This and the following publication are seminal papers on the
genetics of gastric cancer using whole-genome sequencing
and comprehensive molecular profiling.
18. Cancer Genome Atlas Research . Comprehensive molecular char-
Declaration of interest acterization of gastric adenocarcinoma. Nature. 2014;513
(7517):202–209.
The author has no relevant affiliations or financial involvement with any •• This seminal publication describes four molecular subtypes of
organization or entity with a financial interest in or financial conflict with gastric cancer based on a comprehensive molecular character-
the subject matter or materials discussed in the manuscript. This includes ization of 295 gastric cancer patients.
employment, consultancies, honoraria, stock ownership or options, expert 19. Deng N, Goh LK, Wang H, et al. A comprehensive survey of geno-
testimony, grants or patents received or pending, or royalties. mic alterations in gastric cancer reveals systematic patterns of
molecular exclusivity and co-occurrence among distinct therapeu-
tic targets. Gut. 2012;61(5):673–684.
• This study applied single nucelotide poylmorphism arrays in a
References panel of 193 gastric cancer patients and 40 gastric cancer cell
lines and found 22 recurrent genomic alterations including
Papers of special note have been highlighted as either of interest (•) or of several actionable targets.
considerable interest (••) to readers. 20. Mathiak M, Warneke VS, Behrens HM, et al. Clinicopathologic char-
1. Carneiro F. Stomach cancer. In: Stewart BWWCP, editor. World acteristics of microsatellite instable gastric carcinomas revisited:
cancer report 2014. Lyon: IARC; 2014. p. 383–391. urgent need for standardization. Appl Immunohistochem Mol
2. Correa P. A human model of gastric carcinogenesis. Cancer Res. Morphol. 2017;25:12–24.
1988;48(13):3554–3560. 21. Warneke V, Behrens H-M, Haag J, et al. Prognostic and putative
• This publication describes a etiopathogentic model of gastric predictive biomarkers of gastric cancer for personalized medicine.
cancer development, commonly known as the Correa-model. Diagn Mol Pathol. 2013;22(3):127–137.
3. Correa P. Human gastric carcinogenesis: a multistep and multi- 22. Böger C, Behrens H-M, Mathiak M, et al. PD-L1 is an independent
factorial process–First American Cancer Society award lecture on prognostic predictor in gastric cancer of Western patients.
cancer epidemiology and prevention. Cancer Res. 1992;52 Oncotarget. 2016;7(17):24269–24283.
(24):6735–6740.
300 C. RÖCKEN

23. Gonzalez RS, Messing S, Tu X, et al. Immunohistochemistry as a 44. Gerlinger M, Horswell S, Larkin J, et al. Genomic architecture and
surrogate for molecular subtyping of gastric adenocarcinoma. Hum evolution of clear cell renal cell carcinomas defined by multiregion
Pathol. 2016;56:16–21. sequencing. Nat Genet. 2014;46(3):225–233.
24. Fukayama M, Ushiku T. Epstein-Barr virus-associated gastric carci- 45. Yates LR, Campbell PJ. Evolution of the cancer genome. Nat Rev
noma. Pathol Res Pract. 2011;207(9):529–537. Genet. 2012;13(11):795–806.
25. Tokunaga M, Land CE. Epstein-Barr virus involvement in gastric 46. Humphries A, Cereser B, Gay LJ, et al. Lineage tracing reveals
cancer: biomarker for lymph node metastasis. Cancer Epidemiol multipotent stem cells maintain human adenomas and the pattern
Biomarkers Prev. 1998;7(5):449–450. of clonal expansion in tumor evolution. Proc Natl Acad Sci USA.
26. Liu X, Liu J, Qiu H, et al. Prognostic significance of Epstein-Barr virus 2013;110(27):E2490–E2499.
infection in gastric cancer: a meta-analysis. BMC Cancer. 47. Swanton C. Intratumor heterogeneity: evolution through space and
2015;15:782. time. Cancer Res. 2012;72(19):4875–4882.
27. Tokunaga M, Land CE, Uemura Y, et al. Epstein-Barr virus in gastric 48. Bang Y-J, Van CE, Feyereislova A, et al. Trastuzumab in combination
carcinoma. Am J Pathol. 1993;143(5):1250–1254. with chemotherapy versus chemotherapy alone for treatment of
28. Park CK, Park JS, Kim HS, et al. Receptor tyrosine kinase amplified HER2-positive advanced gastric or gastro-oesophageal junction
gastric cancer: clinicopathologic characteristics and proposed cancer (ToGA): a phase 3, open-label, randomised controlled trial.
screening algorithm. Oncotarget. 2016. DOI:10.18632/ Lancet. 2010;376(9742):687–697.
oncotarget.12291 •• This seminal study introduced targeted therapy for gastric
29. Kiyose S, Nagura K, Tao H, et al. Detection of kinase amplifications cancer.
in gastric cancer archives using fluorescence in situ hybridization. 49. Rüschoff J, Dietel M, Baretton G, et al. HER2 diagnostics in gastric
Pathol Int. 2012;62(7):477–484. cancer-guideline validation and development of standardized
30. Shen G-S, Zhao J-D, Zhao J-H, et al. Association of HER2 status with immunohistochemical testing. Virchows Arch. 2010;457(3):299–307.
prognosis in gastric cancer patients undergoing R0 resection: a 50. Lordick F, Röcken C. The identification of predictive factors for
Downloaded by [La Trobe University] at 13:01 01 October 2017

large-scale multicenter study in China. World J Gastroenterol. perioperative chemotherapy in esophago-gastric cancer. Ann
2016;22(23):5406–5414. Oncol. 2013;24(5):1135–1138.
31. Warneke VS, Behrens H-M, Böger C, et al. Her2/neu testing in 51. Röcken C. Quality assurance in clinical trials-the role of pathology.
gastric cancer: evaluating the risk of sampling errors. Ann Oncol. Virchows Arch. 2016;468(1):83–92.
2013;24(3):725–733. 52. Cunningham D, Tebbutt NC, Davidenko I, et al. Phase 3, rando-
32. Metzger M-L, Behrens H-M, Böger C, et al. MET in gastric cancer - mized, double-blind, multicenter, placebo-controlled trial of rilotu-
discarding a 10% cutoff rule. Histopathology. 2016;68(2):241–253. mumab plus epirubicin, cisplatin, and capecitabine as first-line
33. Silva ANS, Coffa J, Menon V, et al. Frequent coamplification of therapy in patients with advanced MET-positive gastric or gastro-
receptor tyrosine kinase and downstream signaling genes in esophageal junction cancer: RILOMET-1 study. J Clin Oncol. 2015;33
Japanese primary gastric cancer and conversion in matched (Suppl.):abstr4000.
lymph node metastasis. Ann Surg. 2016;1. DOI:10.1097/ 53. Shah MA, Bang YJ, Lordick F, et al. METGastric: a phase III study of
SLA.0000000000002042 onartuzumab plus mFOLFOX6 in patients with metastatic HER2-
34. Kakiuchi M, Nishizawa T, Ueda H, et al. Recurrent gain-of-function negative (HER2-) and MET-positive (MET+) adenocarcinoma of the
mutations of RHOA in diffuse-type gastric carcinoma. Nat Genet. stomach or gastroesophageal junction (GEC). J Clin Oncol. 2015;33
2014;46(6):583–587. (Suppl.):abstr4012.
35. Oshima T, Shan J, Okugawa T, et al. Down-regulation of claudin-18 54. Baretton G, Dietel M, Gaiser T, et al. HER2 testing in gastric cancer:
is associated with the proliferative and invasive potential of gastric results of a meeting of German experts. Pathologe. 2016;37(4):361–
cancer at the invasive front. PLoS One. 2013;8(9):e74757. 366.
36. Röcken C, Behrens H-M, Böger C, et al. Clinicopathological charac- 55. Bartley AN, Washington MK, Ventura CB, et al. HER2 testing and
teristics of RHOA mutations in a Central European gastric cancer clinical decision making in gastroesophageal adenocarcinoma:
cohort. J Clin Pathol. 2016;69(1):70–75. guideline from the College of American Pathologists, American
37. Böger C, Warneke VS, Behrens HM, et al. Integrins αβ3 and αβ5 as Society for Clinical Pathology, and American Society of Clinical
prognostic, diagnostic, and therapeutic targets in gastric cancer. Oncology. Arch Pathol Lab Med. 2016;140:1345–1363.
Gastric Cancer. 2015;18(4):784–795. DOI:10.5858/arpa.2016-0331-CP
38. Sargent DJ, Marsoni S, Monges G, et al. Defective mismatch repair 56. Al-Refaie WB, Tseng JF, Gay G, et al. The impact of ethnicity on the
as a predictive marker for lack of efficacy of fluorouracil-based presentation and prognosis of patients with gastric adenocarci-
adjuvant therapy in colon cancer. J Clin Oncol. 2010;28(20):3219– noma. Results from the National Cancer Data Base. Cancer.
3226. 2008;113(3):461–469.
39. Sinicrope FA, Foster NR, Thibodeau SN, et al. DNA mismatch repair 57. Byfield SAD, Earle CC, Ayanian JZ, et al. Treatment and outcomes of
status and colon cancer recurrence and survival in clinical trials of gastric cancer among United States-born and foreign-born Asians
5-fluorouracil-based adjuvant therapy. J Natl Cancer Inst. 2011;103 and Pacific Islanders. Cancer. 2009;115(19):4595–4605.
(11):863–875. 58. Kim J, Sun C-L, Mailey B, et al. Race and ethnicity correlate with
40. Lawrence MS, Stojanov P, Polak P, et al. Mutational heterogeneity survival in patients with gastric adenocarcinoma. Ann Oncol.
in cancer and the search for new cancer-associated genes. Nature. 2010;21(1):152–160.
2013;499(7457):214–218. 59. Kunz PL, Gubens M, Fisher GA, et al. Long-term survivors of gastric
41. Lordick F, Kang Y-K, Chung H-C, et al. Capecitabine and cisplatin cancer: a California population-based study. J Clin Oncol. 2012;30
with or without cetuximab for patients with previously untreated (28):3507–3515.
advanced gastric cancer (EXPAND): a randomised, open-label 60. Nelson R, Ko EB, Arrington A, et al. Race and correlations between
phase 3 trial. Lancet Oncol. 2013;14(6):490–499. lymph node number and survival for patients with gastric cancer. J
42. De Roock W, Piessevaux H, De Schutter J, et al. KRAS wild-type state Gastrointest Surg. 2013;17(3):471–481.
predicts survival and is associated to early radiological response in 61. Röcken C, Behrens H-M. Validating the prognostic and discriminat-
metastatic colorectal cancer treated with cetuximab. Ann Oncol. ing value of the TNM-classification for gastric cancer - a critical
2008;19(3):508–515. appraisal. Eur J Cancer. 2015;51(5):577–586.
43. Gerlinger M, Rowan AJ, Horswell S, et al. Intratumor heterogeneity 62. Theuer CP, Kurosaki T, Ziogas A, et al. Asian patients with gastric
and branched evolution revealed by multiregion sequencing. N carcinoma in the United States exhibit unique clinical features and
Engl J Med. 2012;366(10):883–892.
EXPERT REVIEW OF MOLECULAR DIAGNOSTICS 301

superior overall and cancer specific survival rates. Cancer. 2000;89 • The MAGIC-trial introduced perioperative chemotherapy in
(9):1883–1892. Europe.
63. Theuer CP. Asian gastric cancer patients at a southern California 66. Macdonald JS, Smalley SR, Benedetti J, et al. Chemoradiotherapy
comprehensive cancer center are diagnosed with less advanced after surgery compared with surgery alone for adenocarcinoma of
disease and have superior stage-stratified survival. Am Surg. the stomach or gastroesophageal junction. N Engl J Med. 2001;345
2000;66(9):821–826. (10):725–730.
64. Lin SJ, Gagnon-Bartsch JA, Tan IB, et al. Signatures of tumour • This study introduced adjuvant chemoradiotherapy in the U.S.
immunity distinguish Asian and non-Asian gastric adenocarcino- 67. Japanese Gastric Cancer . Japanese gastric cancer treatment guide-
mas. Gut. 2015;64(11):1721–1731. lines 2014 (ver. 4). Gastric Cancer. 2017;20(1):1–19.
65. Cunningham D, Allum WH, Stenning SP, et al. Perioperative che- 68. Böger C, Behrens HM, Röcken C. Ki67 – An unsuitable marker of
motherapy versus surgery alone for resectable gastroesophageal gastric cancer prognosis unmasks intratumoral heterogeneity. J
cancer. N Engl J Med. 2006;355(1):11–20. Surg Oncol. 2015;113(1):46–54.
Downloaded by [La Trobe University] at 13:01 01 October 2017

You might also like