Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Biochimica et Biophysica Acta 1854 (2015) 687–695

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta


journal homepage: www.elsevier.com/locate/bbapap

Review

Current advances in esophageal cancer proteomics☆


Norihisa Uemura a,1, Tadashi Kondo b,⁎,1
a
Department of Gastroenterological Surgery, Aichi Cancer Center Hospital, 1-1 Kanokoden, chikusa-ku, Nagoya, Aichi 464-8681, Japan
b
Division of Pharmacoproteomics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan

a r t i c l e i n f o a b s t r a c t

Article history: We review the current status of proteomics for esophageal cancer (EC) from a clinician's viewpoint. The ultimate
Received 4 June 2014 goal of cancer proteomics is the improvement of clinical outcome. The proteome as a functional translation of the
Received in revised form 4 September 2014 genome is a straightforward representation of genomic mechanisms that trigger carcinogenesis. Cancer proteo-
Accepted 9 September 2014
mics has identified the mechanisms of carcinogenesis and tumor progression, detected biomarker candidates for
Available online 16 September 2014
early diagnosis, and provided novel therapeutic targets for personalized treatments. Our review focuses on three
Keywords:
major topics in EC proteomics: diagnostics, treatment, and molecular mechanisms. We discuss the major
Esophageal cancer histological differences between EC types, i.e., esophageal squamous cell carcinoma and adenocarcinoma, and eval-
Proteomics uate the clinical significance of published proteomics studies, including promising diagnostic biomarkers and novel
Biomarker therapeutic targets, which should be further validated prior to launching clinical trials. Multi-disciplinary collabo-
Neoadjuvant therapy rations between basic scientists, clinicians, and pathologists should be established for inter-institutional validation.
Molecular mechanism In conclusion, EC proteomics has provided significant results, which after thorough validation, should lead to the
development of novel clinical tools and improvement of the clinical outcome for esophageal cancer patients.
This article is part of a Special Issue entitled: Medical Proteomics.
© 2014 Elsevier B.V. All rights reserved.

1. Introduction the patients with advanced disease, neoadjuvant therapy has been
proved extremely valuable and is established as a standard treatment
Esophageal cancer (EC) is the fifth for men and the eighth for [5–11]. However, a significant proportion of treated patients (60–70%)
women, most common cause of cancer-related death worldwide [1]. do not respond well to neoadjuvant regimens and develop severe
Despite the use of modern surgical techniques in combination with adverse effects [10,12]. Thus, risk stratification for neoadjuvant therapy
radiotherapy and chemotherapy, early recurrence is common in patients is required to improve treatment quality for patients with EC. Moreover,
with advanced disease and the overall 5-year survival rate remains below elucidation of molecular mechanisms underlying the resistance to treat-
40% [2]. In contrast, the 5-year survival rate for patients with early stage ment may lead to novel therapeutic strategies and/or agents.
lesions after endoscopic or surgical treatment exceeds 90% [3]. Survival Cancer proteomics has been conducted with the goal to identify
substantially decreases with the increase of tumor invasion and the pres- diagnostic biomarkers and improve clinical outcome. The biomarkers
ence of regional lymph node metastases and distant metastases [2]. These should be used for early cancer detection and effective prediction of
clinical facts clearly indicate that early detection is the key to EC cure. cancer clinical behavior, as well as identification of novel molecular
However, fewer than 30% of EC patients appeared in the hospital at targets involved in tumorigenesis and disease progression [13]. Cancer
early stage [4], and early screening modalities are critically needed. For proteomics is a promising approach because proteome is a functional
translation of the genomic aberrations that initiate and promote the dis-
Abbreviations: 2-DE, Two dimensional gel electrophoresis; 2D-DIGE, Two-dimensional
ease. Many lines of evidence indicate the discrepancy between protein
difference gel electrophoresis; BRE, Brain and reproductive organ-expressed; COX, and mRNA expression, and recent studies demonstrated that protein
Cytochrome c oxidase subunits; CRT, Chemoradiotherapy; EAC, Esophageal adenocarcino- levels are mainly determined at the translation rather than transcriptional
ma; EC, Esophageal cancer; ESCC, Esophageal squamous cell carcinoma; HER2, Human epi- step [14–16]. Posttranslational modifications, protein interactions,
dermal growth factor receptor 2; Hsp, Heat shock protein; KLH, Keyhole limpet
activity, and localization are also unique data that can be obtained
hemocyanin; LC-MS/MS, Liquid chromatography-tandem mass spectrometry; MALDI-TOF
MS, Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry; SELDI- only by proteomics [17]. Since aberrant protein expression is an impor-
TOF MS, Surface-enhanced laser desorption/ionization time-of-flight mass spectrometry tant feature of cancerous phenotype, the analysis of cancer proteome
☆ This article is part of a Special Issue entitled: Medical Proteomics. can result in the identification of biomarker candidates associated with
⁎ Corresponding author at: Division of Pharmacoproteomics, National Cancer Center clinical characteristics and can further understanding of the molecular
Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan. Tel.: +81 3 3542
2511; fax: +81 3 3547 5298.
mechanisms underlying the initiation and progression of EC [18].
E-mail addresses: nuemura@aichi-cc.jp (N. Uemura), takondo@ncc.go.jp (T. Kondo). In the last decade, the advancement in proteomics has led to a
1
Uemura N and Kondo T contributed equally to this study. comprehensive understanding of the EC proteome. For example, in

http://dx.doi.org/10.1016/j.bbapap.2014.09.011
1570-9639/© 2014 Elsevier B.V. All rights reserved.
688 N. Uemura, T. Kondo / Biochimica et Biophysica Acta 1854 (2015) 687–695

two-dimensional gel electrophoresis (2-DE), the separation in the first also clinically distinct. In a study of 8562 patients who underwent
dimension was improved by the use of immobiline dry strip gels [19], surgical resection, Merkow et al. found that ESCC histology was the
which enabled the isolation and identification of low-expressed pro- only factor predictive of pathologic complete response to neoadjuvant
teins by mass spectrometry. Two-dimensional difference gel electro- treatment [31], consistent with previous observations showing that his-
phoresis (2D-DIGE) provided compensation of gel-to-gel variations by tological type might affect response to neoadjuvant chemotherapy and
the use of an internal standard sample, while the application of highly subsequent prognosis for EC patients [32]. Thus, histologically different
sensitive fluorescent dyes enabled expression profiling of small protein ESCC and EAC should be discriminated in EC proteomics.
amounts from laser microdissected tissues [20–22]. The number of Second, we focus on the clinical significance of proteomic studies.
detectable protein spots has increased from hundreds to thousands We believe that the ultimate goal of cancer proteomics should be
through advances in analytical detection of low-molecular-weight pro- to bring solutions to clinical problems and provide a better grasp of
teins [23]. In addition to gel-based techniques, mass spectrometry and situations encountered in clinical practice. Accordingly, we reviewed
highly sensitive immunodetection methods have also been successfully previous studies of EC proteome from a clinician's viewpoint.
applied to EC proteomics [24,25] (Fig. 1).
In the early 2000s, proteomics studies in EC focused on the compar- 2. Diagnosis
ison between tumor and non-tumor tissues, and a considerable number
of proteins with differential expression in esophageal tumors was 2.1. Early EC detection
detected and functionally examined [26–28]. Although these studies
provided potential keys to the understanding of molecular background The most sensitive tool for the early detection of EC is
underlying EC development, considerable diversity of biological mech- chromoendoscopy with Lugol's dye [33]. During the 1990s, multiple
anisms involved in cancer initiation and progression, as well as hetero- reports demonstrated that endoscopy provided an easy, inexpensive,
geneity of esophageal tumors, made it difficult to link the detected and sensitive means of detecting early and late squamous cell neoplasia.
proteins to specific clinical and pathological data. Then, clinically However, because of an unfavorable balance of risks, benefits, costs, and
oriented proteomics studies, which integrated protein profiling with psychological burden, the implementation of a national endoscopic
clinical and pathological observations of primary tumors from different screening program has not been justified [34]. Currently, there is a
patients, were launched as the next step toward clinical application. need for new, less invasive, and less expensive screening tools, because
Our review focuses on three major topics in EC proteomics: diagnos- most gastroenterology facilities would not be able to support the endo-
tics, treatment, and molecular mechanisms. We will also consider histo- scopic screening program [35]. A simple blood test for early EC detection
logical types of EC, namely esophageal squamous cell carcinoma (ESCC) is superior to endoscopy in terms of cost, testability, and invasiveness. In
and adenocarcinoma (EAC). It has been shown that the predominant current practice, the greatest clinical benefit would be potentially
histological type of EC is region-specific. Although ESCC prevails in offered by novel serum biomarkers with high sensitivity and specificity
most of the world, the incidence of EAC now exceeds that of ESCC in for EC. With this notion, proteomics studies have been conducted to
Australia, the USA, and some Western European countries (e.g., the develop EC biomarkers that would allow early cancer detection in
UK, Finland, France, and the Netherlands) [29,30]. ESCC and EAC are blood instead of tissues.

Fig. 1. Workflow of proteomics application to biomarker development in EC. Proteins in surgical specimens are extracted and subjected to gel-based and antibody-based proteomics. The
results are verified by immunohistochemistry, and applied to clinical applications [23,24].
N. Uemura, T. Kondo / Biochimica et Biophysica Acta 1854 (2015) 687–695 689

2.1.1. ESCC utility should be validated in different populations of EC patients. First,


Fan et al. analyzed serum biomarkers associated with ESCC using the the number of cases in the previous studies was limited, and it is ques-
ClinProt profiling technology based on mass spectrometry [36]. Tubulin tionable whether high biomarker performance observed in the exam-
beta chain, filamin A alpha isoform 1, and cytochrome b-c1 complex ined cohort could be reproduced in the hospitals without further
subunit 1 were identified as ESCC diagnostic serum biomarkers applica- validation. Second, the reported biomarkers have moderate diagnostic
ble also to stage I disease (Table 1). Zhang et al. compared protein accuracy, and in screening of a low-prevalence disease, such as EC,
expression profiles in pre-surgery and post-surgery sera of 17 ESCC pa- many persons can have false-positive test results. As these patients
tients by 2-DE and matrix-assisted laser desorption/ionization time-of- will be monitored by expensive and invasive examinations such as
flight mass spectrometry (MALDI-TOF MS) [26]. One of the differentially imaging and biopsy, in order to avoid this, the accuracy of biomarker
expressed proteins, clusterin, was found to be downregulated in both performance should be additionally validated in a low-prevalence
cancer tissue and pre-surgery serum, which was validated by RT-PCR population. The adverse consequences of high false-positive rate in EC
and Western blotting, suggesting that clusterin is a candidate serum screening should be thoroughly considered before applying the results
biomarker for ESCC. Liu et al. used 2-DE to examine tumor proteins of biomarker studies to early disease detection.
that elicit humoral response in ESCC patients [37] and detected autoan-
tibodies to oncogene CDC25B phosphatase in 36.29% of ESCC patients,
2.2. Prediction of lymph node metastasis
8.67% of other cancer patients, and none of the healthy controls. Given
that anti-CDC25B antibodies were found in patients with both early
In EC, lymph node metastasis originates from the superficial cancer
and late ESCC stages, these antibodies might have clinical utility in
and spreads quickly from the neck to the abdomen [41]. The status of
ESCC screening and diagnosis. Fujita et al. examined novel tumor anti-
lymph node metastasis is one of the most critical parameters that affects
gens in an ESCC cell line and related autoantibodies in sera of ESCC
prognosis and determines treatment strategy. Computed tomography,
patients by proteomics [38]. The levels of an autoantibody against heat
magnetic resonance imaging, ultrasonography, endoscopic ultrasonog-
shock protein Hsp70 appeared to be significantly higher in patients
raphy, positron emission tomography, and other methods have been
with ESCC than in healthy individuals or patients with gastric or colon
employed for the diagnostics of nodal metastases. Despite the use of
cancer, suggesting that anti-Hsp70 autoantibody may have clinical
such modern diagnostic modalities, the diagnosis of lymph node metas-
utility as a diagnostic marker for ESCC.
tasis has insufficient accuracy. Stiles et al. reported that most patients
with cT2-T3N0M0 cancer, a resectable advanced cancer without clinical
2.1.2. EAC lymph node metastases, had pathological lymph node metastases and
Mechref et al. performed glycosylation profiling of serum samples despite induction therapy, more than 50% had persistent nodal disease
from patients with EAC, high-grade dysplasia, Barrett's esophagus, and [42]. The accurate clinical N staging of EC is difficult, and the improve-
healthy individuals and showed that the changes in the relative intensi- ment in diagnostic accuracy could play a crucial role in selecting ade-
ties of three glycan structures predicted EAC with 94% sensitivity quate treatment strategy. The identification of predictive biomarkers
and more than 60% specificity [39], suggesting that comparative for lymph node metastasis would have important clinical implications
EAC glycomics can be used for the identification of EAC candidate and could resolve particular concerns regarding diagnostics of lymph
biomarkers. node metastasis.
The survival rate of EC patients remains low largely because of the
absence of effective screening markers for early detection. Because
2.2.1. ESCC
patients with early EC stages benefit from endoscopic treatment or min-
Hatakeyama et al. examined the ESCC proteome in 72 laser micro-
imally invasive esophagectomy as curative resection [40], it is hoped
dissection samples using two-dimensional difference gel electrophore-
that novel serum biomarkers for early EC detection, as well as less
sis (2D-DIGE) and liquid chromatography-tandem mass spectrometry
invasive and cheaper screening tools, can be developed through multi-
(LC-MS/MS) [43]. Protein spots with intensities statistically different
center validation studies.
between patients without nodal metastasis and patients with more
than five lymph node metastases included cancer-associated factors
2.1.3. Perspectives of biomarkers for early detection such as alpha-actinin 4, hnRNP K, periplakin, squamous cell carcinoma
Cancer proteomics allows identification of early disease biomarkers antigen 1, NudC, and Hsp60; the latter was increased in tumor tissues
with high sensitivity and specificity; however, the biomarker clinical almost in parallel with the number of lymph node metastases.

Table 1
Summary of biomarker candidates discovered by proteomics in EC.

Purpose Histology Protein name Reference

Early detection ESCC tubulin beta chain, filamin A alpha isoform 1, and cytochrome b-c1 complex subunit 1 36
clusterin 26
autoantibodies to oncogene CDC25B phosphatase 37
autoantibody against heat shock protein Hsp70 38
EAC glycan structures 39
Prediction of lymph node metastasis ESCC alpha-actinin 4, hnRNP K, periplakin, squamous cell carcinoma antigen 1, NudC, and Hsp60 43
alpha-actinin 4 44
annexin A2, Cdc42 45
EAC Hsp27 family proteins, HER2 46
Prdiction of response to treatments ESCC thioredoxin domain-containing protein 4 precursor and cystathionine gamma-lyase 53
EAC COX protein 54
apolipoprotein A-1, serum amyloid A, and transthyretin 55
Hsp27 56
ESCC and EAC C4a and C3a 57
Prediction of prognosis ESCC transglutaminase 3 23
calreticulin and a 78-kDa glucose-regulated protein 59
EAC Hsp27 and HER2 46
ESCC and EAC apolipoprotein A-I, serum amyloid A, and transthyretin 60
690 N. Uemura, T. Kondo / Biochimica et Biophysica Acta 1854 (2015) 687–695

Fu et al. employed proteomics to analyze protein extracts of 12 ESCC that of patients treated by surgery alone [50]. Thus, to avoid potential
specimens from patients with distinctly different tumor stages [44]. morbidity due to ineffective treatment and prevent further disease
Using 2D-GE and MALDI-TOF MS, they identified 18 proteins differen- progression, it is essential to perform accurate risk stratification of EC
tially expressed in tumors compared to matched surrounding normal patients. The clinical significance of response predictions for patients
tissues; among them, the expression of alpha-actinin 4 was shown to with unresectable EC can be interpreted in a similar fashion.
be significantly associated with lymph node metastasis by immunohis- Nonresponding patients with unresectable EC should receive a different
tochemistry analysis using two-tissue microarray blocks that contained treatment at early stages. In this context, the identification of predictive
442 primary tumor samples. Because two independent studies consis- markers is important for the risk stratifications and individualization of
tently identified alpha-actinin 4, it may be a promising biomarker for multimodality treatments for patients with advanced EC [51].
nodal metastasis.
Other proteins related to lymph node metastasis were identified by 2.3.1. ESCC
Feng et al., who reported that the expression of annexin A2 in ESCCs was Hayashida et al. used SELDI-MS to analyze proteomic profiles in
significantly downregulated and that of Cdc42 upregulated compared 27 serum samples from untreated ESCC patients and predict the efficacy
with corresponding normal esophagus mucosa; it was also shown that of preoperative chemoradiotherapy [52]. The authors found that out of
their levels significantly correlated with nodal metastasis [45]. The 859 protein peaks, a set of four (at 7,420, 9,112, 17,123, and 12,867 m/z)
authors proposed that the aberrant expression of annexin A2 and could be used to distinguish responders from nonresponders with
Cdc42 played a role in the carcinogenesis and metastasis of ESCC. a sensitivity of 100% and 93.3% in the training and validation sets,
respectively.
2.2.2. EAC Wen et al. investigated the mechanisms of drug resistance in ESCC
Berg et al. analyzed expression profiles of 17 cancer-related signal- by comparing expression profiles of a multidrug-resistant ESCC cell
ing proteins in a series of 87 formalin fixed and paraffin-embedded line EC 109/CDDP with its parental drug-sensitive cell line using
EAC tissue samples using a reverse phase protein array [46]. They 2D-GE and MALDI-TOF MS [53]. The identified 44 differentially
found a molecular subtype of EAC characterized by low levels of the expressed proteins were involved in endoplasmic reticulum stress
Hsp27 family proteins and high expression of the human epidermal response, metabolic processes, DNA replication and repair, nucleotide
growth factor receptor 2 (HER2) family members. Patients with this binding, calcium binding, and cytoskeleton formation; among them,
EAC subtype were more likely to have lymph node and distant metasta- thioredoxin domain-containing protein 4 precursor and cystathionine
ses and short overall survival. Compared with all other EACs, this EAC gamma-lyase were validated by western blot and RT-PCR.
subtype included significantly more cases positive for pN (lymph node
status) and cM (occurrence of distant metastasis): 67% and 15%, 2.3.2. EAC
respectively. Another new EAC subtype had the opposite pattern: low In a study of 23 EAC pre-therapeutic biopsy samples, Aichler et al.
expression of HER2 proteins and high expression of Hsp27 family analyzed proteomic changes associated with response to chemotherapy
members, few pN-positive (21%), and no cM-positive (0%) cases. by MALDI imaging mass spectrometry and LC-MS/MS, and showed that
clinical response to cisplatin was associated with defects in cancer
2.2.3. Perspectives of biomarkers for lymph node metastasis cell mitochondrial respiratory chain caused by the loss of specific
Cancer proteomics provided promising predictive biomarkers for cytochrome c oxidase (COX) subunits [54]. These results were further
lymph node metastasis. However, the benefit of these biomarkers re- validated in an independent study showing that reduced COX protein
mains unproven in clinical settings. First, the significance of correlation expression prior to treatment correlated with chemotherapeutic
between the biomarkers and nodal metastasis should be validated in sensitivity and favorable clinical outcomes, whereas unchanged COX
larger cohorts in multiple institutions. Second, therapeutic strategies expression indicated chemoresistance.
for the treatment of biomarker-positive but otherwise negative Plasma protein profiling in mice with OE19 EAC xenografts
cases should be established by randomized trials. Nodal metastases un- that were treated with clinically relevant chemotherapeutic agents
detectable by imaging tests are very small, and the presence of such epirubicin, cisplatin, or 5-fluorouracil resulted in the identification of
micrometastases normally has no impact on the prognosis for ESCC pa- apolipoprotein A-1, serum amyloid A, and transthyretin as candidate
tients [47]. For the patients with image-negative nodal micrometastases biomarkers of response to chemotherapy [55]. However, although the
who are positive in biomarker and pathology tests, the prognostic value results were confirmed in clinical samples, the number of patients
of other clinical examinations must be considered when planning for with evident pathological response was relatively small; the biomarker
pre-operative therapy. pattern could not be compared with other indicators of clinical response
and required further validation.
2.3. Therapy response prediction To identify predictive biomarkers, Langer et al. analyzed pre-
therapeutic protein expression profiles in tumor biopsy specimens of
Patients at the early EC stage can be cured by endoscopic or surgical 34 EAC patients treated with neoadjuvant chemotherapy [56]. The
treatment. However, the patients seen at the hospital usually have a authors found that the levels of cellular stress response-associated
developed disease, and a number of multimodal approaches to the Hsp27 and Hsp60, glucose-regulated proteins 94 and 78, and a number
treatment of late EC have been developed. Neoadjuvant chemotherapy, of cytoskeletal proteins differed significantly between responders
alone or in combination with radiotherapy (CRT), became a standard and nonresponders. Immunohistochemistry and gene expression
approach to locally advanced EC [48]. Although favorable response to analysis confirmed a significant association between low Hsp27 expres-
multimodal treatment results in better clinical outcome, randomized sion and resistance to neoadjuvant chemotherapy, suggesting that
trials of different neoadjuvant regimens for locally advanced cancers chemosensitivity may be related to stress-activated inflammatory
have shown modest increase in survival [5–11]. Only those patients mechanisms.
who achieved complete histopathologic response seem to significantly
benefit from neoadjuvant therapy, whereas most of the treated patients 2.3.3. ESCC and EAC
(60–70%) did not respond well and experienced severe adverse effects Maher et al. examined proteomic profiles of ESCC and EAC patients
[10,12]; treatment responses may vary even among patients with the using surface-enhanced laser desorption/ionization time-of-flight
same clinical stage of the disease. The problem is that nonresponsive mass spectrometry (SELDI-TOF MS) and ELISA [57]. By comparing
patients may lose the option of surgical resection after ineffective pre-treatment serum samples from 16 poor responders and 15 good re-
chemotherapy [49], and their prognosis has been found inferior to sponders, it was found that higher serum levels of complement factors
N. Uemura, T. Kondo / Biochimica et Biophysica Acta 1854 (2015) 687–695 691

C4a and C3a were significantly associated with favorable CRT response. and transthyretin are promising candidate prognostic biomarkers that
The leave-one-out cross-validation analysis confirmed that C4a and C3a should be validated in EC clinical studies.
could predict the response to neoadjuvant CRT with a sensitivity and
specificity of 78.6% and 83.3%, respectively. 2.4.4. Perspectives of prognostic biomarkers
Cancer proteomics provided promising prognostic biomarkers for
2.3.4. Perspectives of treatment response predictive biomarkers EC. In clinical practice, post-operative EC prognosis has been made
Cancer proteomics has provided promising predictive biomarkers based on multiple clinicopathological parameters [61,62]; however,
for multimodal therapies. Therapeutic resistance is multi-factorial and novel biomarkers will be beneficial in the cases when thus made predic-
is associated with several cell signaling mechanisms, including mito- tion was not confirmed. Clinicopathological characteristics can directly
chondrial redox [53], respiration [54], and cellular stress response path- reflect EC malignancy, but more accurate prognosis could be obtained
ways [56]. Proteomics can be a powerful tool in identifying protein by the combination of traditional clinical parameters with novel
biomarkers associated with resistance to anticancer therapy [18]. molecular biomarkers identified by proteomics. By conducting sub-
As EC is a clinically diverse disease and treatment modalities are not group analysis based on the state of lymph node metastasis, a significant
fully standardized among hospitals, extensive multi-institutional, clinicopathological prognostic factor, Uemura et al. succeeded in discov-
large-scale studies are required to validate the clinical utility of predic- ering a reliable prognostic biomarker [23]. Similarly, Jiang et al. identi-
tive biomarker candidates. It is also urgent to develop novel therapeutic fied a prognostic biomarker by correlation with EC clinical stage [58].
approaches for patients with biomarker-predicted resistance to Patients' stratification according to clinical background would allow
standardized treatment regimens. Thorough investigation of the mech- compensation for the heterogeneity of molecular mechanisms involved
anisms underlying functional association of biomarker proteins with in EC and provide a potential to increase the sensitivity and specificity of
clinical parameters should provide a key to the development of novel EC prognostic biomarkers.
effective clinical applications to treat CRT-resistant EC.
3. Treatment: Novel targets and chemotherapeutic drugs
2.4. Prognostic prediction
The prediction of responses to CRT may help reduce the number of
Prognostic prediction in EC has important clinical implications be-
unnecessary treated patients. However, without effective therapies to
cause it critically affects decisions regarding the treatment strategy
be offered to nonresponders, the prediction of CRT outcome would be
and follow-up schedule. Patients with good prognosis do not require
tantamount to abandoning nonresponders to their fate. Significant
intensive adjuvant therapy or routine follow-up, whereas those with
improvement in overall EC outcome has to be parallel to that for
poor prognosis must receive strong adjuvant treatment and be
CRT-resistant population; accordingly, there is an urgent need for new
frequently examined. Thus, an accurate risk stratification of EC patients
effective therapeutic approaches for patients unresponsive to conven-
is of paramount importance to prevent further disease progression and
tional treatment. The identification of reliable prognostic biomarkers
reduce morbidity due to ineffective/unnecessary clinical intervention.
through proteomic approaches may identify novel drug targets and
lead to the development of new more effective therapeutic strategies
2.4.1. ESCC
for CRT-resistant EC patients [63].
Uemura et al. compared proteomic profiles of ESCC patients with
favorable and poor prognosis using 2D-DIGE and mass spectrometry
[23] and identified 18 differentially expressed proteins. Of them, 3.1. ESCC
transglutaminase 3 expression was validated in 76 primary tumor sam-
ples by immunohistochemistry and found to be inversely correlated Using 2-DE, Fu et al. compared proteomic profiles of ESCC tumors
with shorter patient survival. at different stages and adjacent normal esophageal tissues [44] and
In search of ESCC prognostic serum biomarkers using SELDI-TOF MS, identified two proteins, alpha-actinin 4 and a 67-kDa laminin receptor
Jiang et al. found no difference between protein profiles patients with associated with ESCC progression (Table 2). The authors suggested
more than 2-year and less than 2-year survival [58]. Furthermore, only that these candidate biomarkers might be useful for prognostic evalua-
1 protein peak differed significantly between stage III disease patients tion, molecular classification, and therapeutic targeting of ESCCs.
stratified by this survival criterion.
However, Du et al., using 2-DE and MALDI-TOF or LC-ESI MS, 3.2. EAC
found 22 proteins differentially expressed in ESCCs compared to
tumor-adjacent normal esophageal tissues [59] and showed that high In the analysis of proteomic changes associated with EAC response
expression of calreticulin and a 78-kDa glucose-regulated protein to chemotherapy, Aichler et al. discovered that favorable response of
were correlated with poor prognosis. cancer cells to cisplatin was related to the defects in mitochondrial
respiratory complexes caused by the loss of COX subunits [54]. The
2.4.2. EAC authors noted that the association between cancer cell respiration and
Berg et al. reported that a particular EAC molecular subtype charac- chemosensitivity was consistent with anticancer therapeutics that
terized by low levels of the Hsp27 proteins and high HER2 expression target the mitochondrial electron transport chain.
correlated with nodal and distant metastases, and short overall survival Vona-Davis et al. used 2-DE to examine the response of Barrett's
[46]. cancer cells to a novel therapeutic agent keyhole limpet hemocyanin
(KLH) [64], an immunostimulatory oxygen-carrying metalloprotein
2.4.3. ESCC and EAC from a marine mollusk Megathura crenulata, which inhibits the growth
Kelly et al. used SELDI-TOF MS to examine EC prognostic biomarkers of Barrett's EC cell lines through both apoptotic and nonapoptotic mech-
in plasma of 20 patients with EAC, 1 with ESCC, 2 with poorly differen- anisms [65,66]. Proteomic profiling showed that KLH downregulated
tiated carcinomas, and 1 with severe dysplasia [60]. Apolipoprotein A-I, proteins associated with glycolysis and protein synthesis, while upreg-
serum amyloid A, and transthyretin in pre-treatment plasma samples ulating those related to oxidative stress (the Hsp70 proteins and
were found to be associated with disease-free survival and overall sur- UDP-glucose 6-dydrogenase), indicating the molecular mechanisms of
vival. The authors also reported the correlation of these three proteins KLH anticancer effects. These results suggest that KLH used as an adju-
with response to chemotherapy in a mouse xenograft model [55]. vant or topical therapy is a promising drug in the treatment of Barrett's
These data suggest that plasma apolipoprotein A-I, serum amyloid A, adenocarcinoma.
692 N. Uemura, T. Kondo / Biochimica et Biophysica Acta 1854 (2015) 687–695

Table 2
Summary of key proteins on treatment and molecular mechanisms discovered by proteomics in EC.

Purpose Histology Protein name Reference

Novel melecular target ESCC alpha-actinin 4 and a 67-kDa laminin receptor 44


EAC COX protein 54
migration-stimulating factor 67
Novel chemotheraupeutic drug EAC keyhole limpet hemocyanin 64
Elucidation of molecular mechanism underlying carcinogenesis EC cell line brain and reproductive organ-expressed protein 80
EAC Rho GDP dissociation inhibitor 2, alpha-enolase, lamin A/C, and 81
nucleoside-diphosphate kinase A
cathepsin D and aldo-ketoreductases 1C2 and 1B10 82
Elucidation of molecular mechanism underlying histological differentiation ESCC stratifin, prohibitin, and squamous cell carcinoma antigen 1 83
annexin A2 84
cathepsin D 85

Hu et al. screened a mouse-derived antibody library generated 4.1. Carcinogenesis


against EC endothelial cells to identify proteins that participate in
tumor angiogenesis [67]. Through selection of monoclonal antibodies To explore carcinogenic mechanisms through protein profiling,
that reacted with EC cell-surface antigens and influenced tumor cell Chen et al. examined soluble proteins differentially expressed in EC
behavior, they identified the antibody against migration-stimulating and non-cancerous cells using 2-DE and MALDI-TOF MS [80]. Among
factor (MSF), which significantly suppressed tumor growth through 20 differentially expressed protein spots, brain and reproductive
inhibition of human tumor-related angiogenesis. These results indicate organ-expressed (BRE) protein was selected and functionally analyzed
that MSF may be a target for the anti-angiogenic treatment of esophageal using small interference RNA (Table 2). BRE silencing upregulated
cancer. tumor-suppressor p53 and downregulated prohibitin, cyclin A, and
CDK2, suggesting that BRE plays an important role in mediating
antiapoptotic and proliferative responses in EC cells.
3.3. Perspectives of novel therapeutics identified by EC proteomics
The insight into potential mechanisms underlying the progression
from Barrett's metaplasia to EAC was gained by Zhao et al. [81], who
Protein profiling resulted in the identification of novel therapeutic
found that Rho GDP dissociation inhibitor 2, alpha-enolase, lamin A/C,
targets, which can lead to a significant improvement of EC prognosis, es-
and nucleoside-diphosphate kinase A were upregulated at both mRNA
pecially for nonresponders diagnosed by predictive protein biomarkers.
and protein levels in EAC compared to Barrett's metaplasia. The results
In other solid tumors, several targeted therapeutic agents, such as
suggest that the identified proteins play a role in EAC development
bevacizumab and cetuximab in colorectal cancer [68], lapatinib and
and may be candidate biomarkers of the progression from Barrett's
trastuzumabin in breast cancer [69,70], and imatinib and sunitinib in
metaplasia to EAC.
gastrointestinal stromal tumor [71,72] have been widely used in clinical
Breton et al. used 2-DE and MALDI-TOF MS to examine proteomic
practice. In EC, the addition of anti-HER2 antibody trastuzumab to the
profiles of 10 esophageal cell lines representing distinct stages in EAC
platinum and fluoropyrimidine doublet regimen is adopted as the stan-
development [82]. Among 33 differentially expressed proteins, the in-
dard care for HER2-positive tumors of the esophagogastric junction [73].
creased levels of cathepsin D and aldo-ketoreductases 1C2 and 1B10
Therapies targeting epidermal growth factor receptor and vascular
in metaplastic and dysplastic cell lines were confirmed by Western blot-
endothelial growth factor remain under active investigation but have
ting and qRT-PCR. The expression patterns of these proteins analyzed
yet to definitively demonstrate clinically significant benefits for EC
from esophageal epithelium from patients with non-erosive and erosive
patients [74]. Multiple molecular targets remain of interest in EC, in-
gastroesophageal reflux disease, Barrett's esophagus, and EAC, suggest
cluding mammalian target of rapamycin, c-MET, insulin-like growth
the contribution to EAC development through effects on apoptosis,
factor, and cytotoxic T-lymphocyte antigen 4 [74]. Clinical application
transport of bile acids, and retinoid metabolism. Further mechanistic
of targeted therapies in EC is less advanced than in other solid tumors,
and clinical investigations are required to fully elucidate the role of
and further proteome studies are required to identify more promising
these molecules in EAC.
molecules.

4.2. Histological differentiation


4. Molecular mechanisms
Using 2-DE and MALDI-TOF MS, Qi et al. examined protein changes
Elucidation of disease molecular mechanisms is directly linked to the
associated with the degree of ESCC differentiation [83] and found that
development of novel diagnostic biomarkers and therapeutic targets
the expression of stratifin, prohibitin, and squamous cell carcinoma
that would bring significant benefits to cancer patients. In the molecular
antigen 1 directly correlated with ESCC differentiation. The same group
mechanistic analysis, proteomics provides tools essential to the discov-
also reported that the expression of annexin A2 was step-wise downreg-
ery of signaling paradigms that trigger tumor initiation and promote
ulated during malignant transformation of epithelial cells [84]: in poorly
cancer progression. The transformation from normal esophageal epithe-
differentiated ESCCs, annexin A2 was either not detected (46%) or had
lium to invasive EC is a multistep process caused by accumulation of
low expression levels (36%). Furthermore, it has been shown that overex-
multiple genetic alterations [75], mainly involving the Wnt, cell cycle,
pression of cathepsin D inversely correlated with ESCC differentiation
and Notch pathways [76]. Despite many genetic studies, the exact mo-
[85]. The significant changes in the expression of these proteins may indi-
lecular mechanisms underlying carcinogenesis and tumor progression
cate that many pathways are involved in ESCC dedifferentiation.
in EC remain unclear [77–79], and comprehensive protein expression
profiling provided by cancer proteomics should contribute to our un-
derstanding of EC biology. Proteomics has identified numerous intrigu- 4.3. Perspectives of proteomics approach to EC molecular mechanisms
ing biomarkers for early cancer diagnostics and prediction of nodal
metastasis, treatment response, and prognosis, which are critically in- Cancer proteomics has resulted in the identification of proteins
volved in each process and require further mechanistic characterization. involved in cancer initiation and progression; further investigation of
N. Uemura, T. Kondo / Biochimica et Biophysica Acta 1854 (2015) 687–695 693

the identified molecules may provide deeper understanding of junction: a meta-analysis of randomized clinical trials, Ann. Surg. Oncol. 10 (2003)
754–761.
EC-related mechanisms and contribute to the development of novel [7] R.A. Malthaner, R.K. Wong, R.B. Rumble, L. Zuraw, C. Members of the Gastrointestinal
therapeutic strategies. However, the current state of cancer proteomics Cancer Disease Site Group of Cancer Care Ontario's Program in Evidence-based,
indicates fundamental problems and the need for improvement. Neoadjuvant or adjuvant therapy for resectable esophageal cancer: a systematic
review and meta-analysis, BMC Med. 2 (2004) 35.
Although proteomics provides a unique opportunity of generating com- [8] F. Fiorica, D. Di Bona, F. Schepis, A. Licata, L. Shahied, A. Venturi, A.M. Falchi, A. Craxi,
prehensive information regarding cellular proteome, it has not been C. Camma, Preoperative chemoradiotherapy for oesophageal cancer: a systematic
fully realized in cancer research. At present, cancer proteomics focuses review and meta-analysis, Gut 53 (2004) 925–930.
[9] S.E. Greer, P.P. Goodney, J.E. Sutton, J.D. Birkmeyer, Neoadjuvant chemoradiotherapy
on a limited portion of proteome, thus excluding some potentially for esophageal carcinoma: a meta-analysis, Surgery 137 (2005) 172–177.
important molecules, and consequently, it has not yet provided [10] V. Gebski, B. Burmeister, B.M. Smithers, K. Foo, J. Zalcberg, J. Simes, G. Australasian
pathway-wide and molecular family-wide analyses. Gastro-Intestinal Trials, Survival benefits from neoadjuvant chemoradiotherapy or
chemotherapy in oesophageal carcinoma: a meta-analysis, Lancet Oncol. 8 (2007)
Furthermore, in EC, proteomics has been mostly applied to analyze
226–234.
expression levels, leaving out important proteome features such as [11] X.H. Xu, X.H. Peng, P. Yu, X.Y. Xu, E.H. Cai, P. Guo, K. Li, Neoadjuvant chemotherapy
posttranslational modifications, molecular interactions, activity, and for resectable esophageal carcinoma: a meta-analysis of randomized clinical trials,
localization, which should be extensively examined in association Asian Pac. J. Cancer Prev. 13 (2012) 103–110.
[12] N.P. Nguyen, S.P. Krafft, V. Vinh-Hung, P. Vos, F. Almeida, S. Jang, M. Ceizyk, A. Desai,
with EC clinical and pathological features. R. Davis, R. Hamilton, H. Modarresifar, D. Abraham, L. Smith-Raymond, Feasibility
Finally, although cancer proteomics provides high-throughput as- of tomotherapy to reduce normal lung and cardiac toxicity for distal esophageal
sessment of protein expression, subsequent functional analysis remains cancer compared to three-dimensional radiotherapy, Radiother. Oncol. 101 (2011)
438–442.
labor-intense and low-scale. As a consequence, only a small number of [13] M. Tyers, M. Mann, From genomics to proteomics, Nature 422 (2003) 193–197.
proteins are further investigated, and potentially valuable molecules [14] G. Chen, T.G. Gharib, C.C. Huang, J.M. Taylor, D.E. Misek, S.L. Kardia, T.J. Giordano,
may be filtered through functional assessment. All these issues should M.D. Iannettoni, M.B. Orringer, S.M. Hanash, D.G. Beer, Discordant protein and
mRNA expression in lung adenocarcinomas, Mol. Cell. Proteomics 1 (2002)
be resolved by continuous technological advances. 304–313.
[15] S. Varambally, J. Yu, B. Laxman, D.R. Rhodes, R. Mehra, S.A. Tomlins, R.B. Shah,
5. Conclusions U. Chandran, F.A. Monzon, M.J. Becich, J.T. Wei, K.J. Pienta, D. Ghosh, M.A. Rubin,
A.M. Chinnaiyan, Integrative genomic and proteomic analysis of prostate cancer
reveals signatures of metastatic progression, Cancer Cell 8 (2005) 393–406.
Cancer proteomics has provided a number of promising diagnostic [16] S.P. Gygi, Y. Rochon, B.R. Franza, R. Aebersold, Correlation between protein and
biomarkers and novel therapeutic targets, as well as intriguing proteins mRNA abundance in yeast, Mol. Cell. Biol. 19 (1999) 1720–1730.
[17] R.E. Banks, M.J. Dunn, D.F. Hochstrasser, J.C. Sanchez, W. Blackstock, D.J. Pappin,
underlying the molecular mechanisms of carcinogenesis and progres-
P.J. Selby, Proteomics: new perspectives, new biomedical opportunities, Lancet
sion of EC. Although individual proteins seem to be promising, the num- 356 (2000) 1749–1756.
ber of clinical samples examined by proteomics has been limited, and [18] L. Smith, M.J. Lind, K.J. Welham, L. Cawkwell, G. Cancer Biology Proteomics, Cancer
multi-institutional validation studies are required to launch clinical proteomics and its application to discovery of therapy response markers in human
cancer, Cancer 107 (2006) 232–241.
trials of biomarkers and therapeutic targets. In parallel with confirming [19] F. Sabounchi-Schutt, J. Astrom, I. Olsson, A. Eklund, J. Grunewald, B. Bjellqvist, An
proteomic data, biomarker-based therapeutic strategies integrating the immobiline DryStrip application method enabling high-capacity two-dimensional
diverse clinical features of ES should be developed. Current studies have gel electrophoresis, Electrophoresis 21 (2000) 3649–3656.
[20] M. Unlu, M.E. Morgan, J.S. Minden, Difference gel electrophoresis: a single gel method
shed a light on limited aspects of the EC proteome, and comprehensive for detecting changes in protein extracts, Electrophoresis 18 (1997) 2071–2077.
analysis will be possible with further technological progress. For exam- [21] R. Tonge, J. Shaw, B. Middleton, R. Rowlinson, S. Rayner, J. Young, F. Pognan,
ple, novel antibody production engineering systems that allow generate E. Hawkins, I. Currie, M. Davison, Validation and development of fluorescence two-
dimensional differential gel electrophoresis proteomics technology, Proteomics 1
appropriate antibody for immunohistochemistry will facilitate the (2001) 377–396.
discovery of biomarker candidates as well as the validation study. In [22] T. Kondo, M. Seike, Y. Mori, K. Fujii, T. Yamada, S. Hirohashi, Application of sensitive
addition to improving the performance of proteomic modalities, inter- fluorescent dyes in linkage of laser microdissection and two-dimensional gel
electrophoresis as a cancer proteomic study tool, Proteomics 3 (2003) 1758–1766.
disciplinary collaboration is essential to obtain research data that will [23] N. Uemura, Y. Nakanishi, H. Kato, S. Saito, M. Nagino, S. Hirohashi, T. Kondo,
benefit patients with EC. Transglutaminase 3 as a prognostic biomarker in esophageal cancer revealed by
proteomics, Int. J. Cancer 124 (2009) 2106–2115.
[24] N. Uemura, Y. Nakanishi, H. Kato, M. Nagino, S. Hirohashi, T. Kondo, Antibody-based
Conflicts of interest
proteomics for esophageal cancer: identification of proteins in the nuclear factor-
kappaB pathway and mitotic checkpoint, Cancer Sci. 100 (2009) 1612–1622.
The authors report no conflicts of interest. [25] Y.J. Qi, W.X. Chao, J.F. Chiu, An overview of esophageal squamous cell carcinoma
proteomics, J. Proteome 75 (2012) 3129–3137.
[26] L.Y. Zhang, W.T. Ying, Y.S. Mao, H.Z. He, Y. Liu, H.X. Wang, F. Liu, K. Wang, D.C. Zhang,
Role of the funding source Y. Wang, M. Wu, X.H. Qian, X.H. Zhao, Loss of clusterin both in serum and tissue
correlates with the tumorigenesis of esophageal squamous cell carcinoma via
No funding was received for this review. proteomics approaches, World J. Gastroenterol. 9 (2003) 650–654.
[27] G. Zhou, H. Li, D. DeCamp, S. Chen, H. Shu, Y. Gong, M. Flaig, J.W. Gillespie, N. Hu,
P.R. Taylor, M.R. Emmert-Buck, L.A. Liotta, E.F. Petricoin III, Y. Zhao, 2D differential
References in-gel electrophoresis for the identification of esophageal scans cell cancer-specific
protein markers, Mol. Cell. Proteomics 1 (2002) 117–124.
[1] A. Jemal, F. Bray, M.M. Center, J. Ferlay, E. Ward, D. Forman, Global cancer statistics, [28] X. Chen, N. Li, S. Wang, J. Hong, M. Fang, J. Yousselfson, P. Yang, R.A. Newman,
CA Cancer J. Clin. 61 (2011) 69–90. R.A. Lubet, C.S. Yang, Aberrant arachidonic acid metabolism in esophageal
[2] T.W. Rice, V.W. Rusch, C. Apperson-Hansen, M.S. Allen, L.Q. Chen, J.G. Hunter, adenocarcinogenesis, and the effects of sulindac, nordihydroguaiaretic acid, and
K.A. Kesler, S. Law, T.E. Lerut, C.E. Reed, J.A. Salo, W.J. Scott, S.G. Swisher, T.J. Watson, alpha-difluoromethylornithine on tumorigenesis in a rat surgical model, Carcinogen-
E.H. Blackstone, Worldwide esophageal cancer collaboration, Dis. Esophagus 22 esis 23 (2002) 2095–2102.
(2009) 1–8. [29] C. Lepage, B. Rachet, V. Jooste, J. Faivre, M.P. Coleman, Continuing rapid increase
[3] Y. Shimizu, H. Tsukagoshi, M. Fujita, M. Hosokawa, M. Kato, M. Asaka, Long-term in esophageal adenocarcinoma in England and Wales, Am. J. Gastroenterol. 103
outcome after endoscopic mucosal resection in patients with esophageal squamous (2008) 2694–2699.
cell carcinoma invading the muscularis mucosae or deeper, Gastrointest. Endosc. 56 [30] H. Pohl, H.G. Welch, The role of overdiagnosis and reclassification in the marked
(2002) 387–390. increase of esophageal adenocarcinoma incidence, J. Natl. Cancer Inst. 97 (2005)
[4] T.W. Rice, G. Zuccaro Jr., D.J. Adelstein, L.A. Rybicki, E.H. Blackstone, J.R. Goldblum, 142–146.
Esophageal carcinoma: depth of tumor invasion is predictive of regional lymph [31] R.P. Merkow, K.Y. Bilimoria, M.D. McCarter, W.B. Chow, C.Y. Ko, D.J. Bentrem, Use
node status, Ann. Thorac. Surg. 65 (1998) 787–792. of multimodality neoadjuvant therapy for esophageal cancer in the United States:
[5] J.D. Urschel, H. Vasan, A meta-analysis of randomized controlled trials that assessment of 987 hospitals, Ann. Surg. Oncol. 19 (2012) 357–364.
compared neoadjuvant chemoradiation and surgery to surgery alone for resectable [32] E. Bollschweiler, R. Metzger, U. Drebber, S. Baldus, D. Vallbohmer, M. Kocher,
esophageal cancer, Am. J. Surg. 185 (2003) 538–543. A.H. Holscher, Histological type of esophageal cancer might affect response to
[6] I.G. Kaklamanos, G.R. Walker, K. Ferry, D. Franceschi, A.S. Livingstone, Neoadjuvant neo-adjuvant radiochemotherapy and subsequent prognosis, Ann. Oncol. 20
treatment for resectable cancer of the esophagus and the gastroesophageal (2009) 231–238.
694 N. Uemura, T. Kondo / Biochimica et Biophysica Acta 1854 (2015) 687–695

[33] A.B. Lopes, R.B. Fagundes, Esophageal squamous cell carcinoma—precursor lesions [57] S.G. Maher, D.T. McDowell, B.C. Collins, C. Muldoon, W.M. Gallagher, J.V. Reynolds,
and early diagnosis, World J. Gastrointest. Endosc. 4 (2012) 9–16. Serum proteomic profiling reveals that pretreatment complement protein levels
[34] L.B. Gerson, P.W. Groeneveld, G. Triadafilopoulos, Cost-effectiveness model of are predictive of esophageal cancer patient response to neoadjuvant chemoradia-
endoscopic screening and surveillance in patients with gastroesophageal reflux tion, Ann. Surg. 254 (2011) 809–816 (discussion 816–807).
disease, Clin. Gastroenterol. Hepatol. 2 (2004) 868–879. [58] H. Jiang, X.H. Wang, X.M. Yu, Z.G. Zheng, Detection and prognostic analysis of serum
[35] E.L. Bird-Lieberman, R.C. Fitzgerald, Early diagnosis of oesophageal cancer, Br. J. Cancer protein expression in esophageal squamous cell cancer, Asian Pac. J. Cancer Prev. 13
101 (2009) 1–6. (2012) 1579–1582.
[36] N.J. Fan, C.F. Gao, X.L. Wang, Tubulin beta chain, filamin A alpha isoform 1, and [59] X.L. Du, H. Hu, D.C. Lin, S.H. Xia, X.M. Shen, Y. Zhang, M.L. Luo, Y.B. Feng, Y. Cai, X. Xu,
cytochrome b-c1 complex subunit 1 as serological diagnostic biomarkers of esoph- Y.L. Han, Q.M. Zhan, M.R. Wang, Proteomic profiling of proteins dysregulted in
ageal squamous cell carcinoma: a proteomics study, OMICS J. Integr. Biol. 17 (2013) Chinese esophageal squamous cell carcinoma, J. Mol. Med. 85 (2007) 863–875.
215–223. [60] P. Kelly, F. Paulin, D. Lamont, L. Baker, S. Clearly, D. Exon, A. Thompson, Pre-treatment
[37] W.L. Liu, G. Zhang, J.Y. Wang, J.Y. Cao, X.Z. Guo, L.H. Xu, M.Z. Li, L.B. Song, W.L. Huang, plasma proteomic markers associated with survival in oesophageal cancer, Br. J. Cancer
M.S. Zeng, Proteomics-based identification of autoantibody against CDC25B as a 106 (2012) 955–961.
novel serum marker in esophageal squamous cell carcinoma, Biochem. Biophys. [61] H. Igaki, H. Kato, Y. Tachimori, Y. Nakanishi, T. Shimoda, Surgery for clinical T3
Res. Commun. 375 (2008) 440–445. carcinomas of the upper thoracic oesophagus and the need for new strategies, Br.
[38] Y. Fujita, T. Nakanishi, Y. Miyamoto, M. Hiramatsu, H. Mabuchi, A. Miyamoto, J. Surg. 92 (2005) 1235–1240.
A. Shimizu, T. Takubo, N. Tanigawa, Proteomics-based identification of autoantibody [62] H. Igaki, Y. Tachimori, H. Kato, Improved survival for patients with upper and/or
against heat shock protein 70 as a diagnostic marker in esophageal squamous cell middle mediastinal lymph node metastasis of squamous cell carcinoma of the
carcinoma, Cancer Lett. 263 (2008) 280–290. lower thoracic esophagus treated with 3-field dissection, Ann. Surg. 239 (2004)
[39] Y. Mechref, A. Hussein, S. Bekesova, V. Pungpapong, M. Zhang, L.E. Dobrolecki, 483–490.
R.J. Hickey, Z.T. Hammoud, M.V. Novotny, Quantitative serum glycomics of esophageal [63] M. Kavallaris, G.M. Marshall, Proteomics and disease: opportunities and challenges,
adenocarcinoma and other esophageal disease onsets, J. Proteome Res. 8 (2009) Med. J. Aust. 182 (2005) 575–579.
2656–2666. [64] L. Vona-Davis, T. Vincent, S. Zulfiqar, B. Jackson, D. Riggs, D.W. McFadden, Proteomic
[40] S.S. Biere, M.I. van Berge Henegouwen, K.W. Maas, L. Bonavina, C. Rosman, J.R. Garcia, analysis of SEG-1 human Barrett's-associated esophageal adenocarcinoma cells
S.S. Gisbertz, J.H. Klinkenbijl, M.W. Hollmann, E.S. de Lange, H.J. Bonjer, D.L. van der treated with keyhole limpet hemocyanin, J. Gastrointest. Surg. Off. J. Soc. Surg.
Peet, M.A. Cuesta, Minimally invasive versus open oesophagectomy for patients with Aliment. Tract. 8 (2004) 1018–1023.
oesophageal cancer: a multicentre, open-label, randomised controlled trial, Lancet [65] D.W. McFadden, D.R. Riggs, B.J. Jackson, L. Vona-Davis, Keyhole limpet hemocyanin,
379 (2012) 1887–1892. a novel immune stimulant with promising anticancer activity in Barrett's esophageal
[41] Y. Tachimori, Y. Nagai, N. Kanamori, N. Hokamura, H. Igaki, Pattern of lymph adenocarcinoma, Am. J. Surg. 186 (2003) 552–555.
node metastases of esophageal squamous cell carcinoma based on the anatomical [66] J.R. Harris, J. Markl, Keyhole limpet hemocyanin: molecular structure of a potent
lymphatic drainage system, Dis. Esophagus 24 (2011) 33–38. marine immunoactivator, Rev. Eur. Urol. 37 (Suppl. 3) (2000) 24–33.
[42] B.M. Stiles, F. Mirza, A. Coppolino, J.L. Port, P.C. Lee, S. Paul, N.K. Altorki, Clinical [67] H. Hu, Y. Ran, Y. Zhang, Z. Zhou, S.J. Harris, L. Yu, L. Sun, J. Pan, J. Liu, J. Lou, Z. Yang,
T2-T3N0M0 esophageal cancer: the risk of node positive disease, Ann. Thorac. Antibody library-based tumor endothelial cells surface proteomic functional screen
Surg. 92 (2011) 491–496 (discussion 496–498). reveals migration-stimulating factor as an anti-angiogenic target, Mol. Cell. Proteomics
[43] H. Hatakeyama, T. Kondo, K. Fujii, Y. Nakanishi, H. Kato, S. Fukuda, S. Hirohashi, 8 (2009) 816–826.
Protein clusters associated with carcinogenesis, histological differentiation and [68] J. Tol, M. Koopman, A. Cats, C.J. Rodenburg, G.J. Creemers, J.G. Schrama, F.L. Erdkamp,
nodal metastasis in esophageal cancer, Proteomics 6 (2006) 6300–6316. A.H. Vos, C.J. van Groeningen, H.A. Sinnige, D.J. Richel, E.E. Voest, J.R. Dijkstra,
[44] L. Fu, Y.R. Qin, D. Xie, H.Y. Chow, S.M. Ngai, D.L. Kwong, Y. Li, X.Y. Guan, Identification M.E. Vink-Borger, N.F. Antonini, L. Mol, J.H. van Krieken, O. Dalesio, C.J. Punt,
of alpha-actinin 4 and 67 kDa laminin receptor as stage-specific markers in esoph- Chemotherapy, bevacizumab, and cetuximab in metastatic colorectal cancer, N. Engl.
ageal cancer via proteomic approaches, Cancer 110 (2007) 2672–2681. J. Med. 360 (2009) 563–572.
[45] J.G. Feng, Q. Liu, X. Qin, Y.H. Geng, S.T. Zheng, T. Liu, I. Sheyhidin, X.M. Lu, Clinico- [69] D.J. Slamon, B. Leyland-Jones, S. Shak, H. Fuchs, V. Paton, A. Bajamonde, T. Fleming,
pathological pattern and Annexin A2 and Cdc42 status in patients presenting with W. Eiermann, J. Wolter, M. Pegram, J. Baselga, L. Norton, Use of chemotherapy
differentiation and lymphnode metastasis of esophageal squamous cell carcinomas, plus a monoclonal antibody against HER2 for metastatic breast cancer that
Mol. Biol. Rep. 39 (2012) 1267–1274. overexpresses HER2, N. Engl. J. Med. 344 (2001) 783–792.
[46] D. Berg, C. Wolff, R. Langer, T. Schuster, M. Feith, J. Slotta-Huspenina, K. Malinowsky, [70] J. Baselga, I. Bradbury, H. Eidtmann, S. Di Cosimo, E. de Azambuja, C. Aura, H. Gomez,
K.F. Becker, Discovery of new molecular subtypes in oesophageal adenocarcinoma, P. Dinh, K. Fauria, V. Van Dooren, G. Aktan, A. Goldhirsch, T.W. Chang, Z. Horvath,
PLoS One 6 (2011) e23985. M. Coccia-Portugal, J. Domont, L.M. Tseng, G. Kunz, J.H. Sohn, V. Semiglazov, G. Lerzo,
[47] F. Sato, Y. Shimada, Z. Li, G. Watanabe, M. Maeda, M. Imamura, Lymph node M. Palacova, V. Probachai, L. Pusztai, M. Untch, R.D. Gelber, M. Piccart-Gebhart, A.S.T.
micrometastasis and prognosis in patients with oesophageal squamous cell carcinoma, Neo, Lapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO):
Br. J. Surg. 88 (2001) 426–432. a randomised, open-label, multicentre, phase 3 trial, Lancet 379 (2012) 633–640.
[48] K. Almhanna, R. Shridhar, K.L. Meredith, Neoadjuvant or adjuvant therapy for resect- [71] G.D. Demetri, M. von Mehren, C.D. Blanke, A.D. Van den Abbeele, B. Eisenberg,
able esophageal cancer: is there a standard of care? Cancer Control 20 (2013) 89–96. P.J. Roberts, M.C. Heinrich, D.A. Tuveson, S. Singer, M. Janicek, J.A. Fletcher,
[49] N.S. Blencowe, A.G. McNair, C.R. Davis, S.T. Brookes, J.M. Blazeby, Standards of out- S.G. Silverman, S.L. Silberman, R. Capdeville, B. Kiese, B. Peng, S. Dimitrijevic,
come reporting in surgical oncology: a case study in esophageal cancer, Ann. Surg. B.J. Druker, C. Corless, C.D. Fletcher, H. Joensuu, Efficacy and safety of imatinib mesylate
Oncol. 19 (2012) 4012–4018. in advanced gastrointestinal stromal tumors, N. Engl. J. Med. 347 (2002) 472–480.
[50] G.W. Dittrick, J.M. Weber, R. Shridhar, S. Hoffe, M. Melis, K. Almhanna, J. Barthel, [72] G.D. Demetri, A.T. van Oosterom, C.R. Garrett, M.E. Blackstein, M.H. Shah, J. Verweij,
J. McLoughlin, R.C. Karl, K.L. Meredith, Pathologic nonresponders after neoadjuvant G. McArthur, I.R. Judson, M.C. Heinrich, J.A. Morgan, J. Desai, C.D. Fletcher, S. George,
chemoradiation for esophageal cancer demonstrate no survival benefit compared C.L. Bello, X. Huang, C.M. Baum, P.G. Casali, Efficacy and safety of sunitinib in patients
with patients treated with primary esophagectomy, Ann. Surg. Oncol. 19 (2012) with advanced gastrointestinal stromal tumour after failure of imatinib: a
1678–1684. randomised controlled trial, Lancet 368 (2006) 1329–1338.
[51] D. Vallbohmer, A.H. Holscher, S. DeMeester, T. DeMeester, J. Salo, J. Peters, T. Lerut, [73] Y.J. Bang, E. Van Cutsem, A. Feyereislova, H.C. Chung, L. Shen, A. Sawaki, F. Lordick,
S.G. Swisher, W. Schroder, E. Bollschweiler, W. Hofstetter, A multicenter study of A. Ohtsu, Y. Omuro, T. Satoh, G. Aprile, E. Kulikov, J. Hill, M. Lehle, J. Ruschoff, Y.K. Kang,
survival after neoadjuvant radiotherapy/chemotherapy and esophagectomy for G.A.T.I. To, Trastuzumab in combination with chemotherapy versus chemotherapy
ypT0N0M0R0 esophageal cancer, Ann. Surg. 252 (2010) 744–749. alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction
[52] Y. Hayashida, K. Honda, Y. Osaka, T. Hara, T. Umaki, A. Tsuchida, T. Aoki, S. Hirohashi, cancer (ToGA): a phase 3, open-label, randomised controlled trial, Lancet 376 (2010)
T. Yamada, Possible prediction of chemoradiosensitivity of esophageal cancer by 687–697.
serum protein profiling, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 11 (2005) [74] P.M. Boland, B. Burtness, Esophageal carcinoma: are modern targeted therapies
8042–8047. shaking the rock? Curr. Opin. Oncol. 25 (2013) 417–424.
[53] J. Wen, B. Zheng, Y. Hu, X. Zhang, H. Yang, Y. Li, C.Y. Zhang, K.J. Luo, X. Zang, Y.F. Li, [75] P.H. Riegman, K.J. Vissers, J.C. Alers, E. Geelen, W.C. Hop, H.W. Tilanus, H. van
X.Y. Guan, J.H. Fu, Comparative proteomic analysis of the esophageal squamous Dekken, Genomic alterations in malignant transformation of Barrett's esophagus,
carcinoma cell line EC109 and its multi-drug resistant subline EC109/CDDP, Int. Cancer Res. 61 (2001) 3164–3170.
J. Oncol. 36 (2010) 265–274. [76] Y. Song, L. Li, Y. Ou, Z. Gao, E. Li, X. Li, W. Zhang, J. Wang, L. Xu, Y. Zhou, X. Ma, L. Liu,
[54] M. Aichler, M. Elsner, N. Ludyga, A. Feuchtinger, V. Zangen, S.K. Maier, B. Balluff, Z. Zhao, X. Huang, J. Fan, L. Dong, G. Chen, L. Ma, J. Yang, L. Chen, M. He, M. Li,
C. Schone, L. Hierber, H. Braselmann, S. Meding, S. Rauser, H. Zischka, M. Aubele, X. Zhuang, K. Huang, K. Qiu, G. Yin, G. Guo, Q. Feng, P. Chen, Z. Wu, J. Wu, L. Ma,
M. Schmitt, M. Feith, S.M. Hauck, M. Ueffing, R. Langer, B. Kuster, H. Zitzelsberger, J. Zhao, L. Luo, M. Fu, B. Xu, B. Chen, Y. Li, T. Tong, M. Wang, Z. Liu, D. Lin, X. Zhang,
H. Hofler, A.K. Walch, Clinical response to chemotherapy in oesophageal adenocarci- H. Yang, J. Wang, Q. Zhan, Identification of genomic alterations in oesophageal
noma patients is linked to defects in mitochondria, J. Pathol. 230 (2013) 410–419. squamous cell cancer, Nature 509 (2014) 91–95.
[55] P. Kelly, V. Appleyard, K. Murray, F. Paulin, D. Lamont, L. Baker, S. Suttie, D. Exon, [77] E.P. Xing, G.Y. Yang, L.D. Wang, S.T. Shi, C.S. Yang, Loss of heterozygosity of the Rb gene
A. Thompson, Detection of oesophageal cancer biomarkers by plasma proteomic correlates with pRb protein expression and associates with p53 alteration in human
profiling of human cell line xenografts in response to chemotherapy, Br. J. Cancer esophageal cancer, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 5 (1999) 1231–1240.
103 (2010) 232–238. [78] L.D. Wang, Y.R. Qin, Z.M. Fan, D. Kwong, X.Y. Guan, G.S. Tsao, J. Sham, J.L. Li, X.S. Feng,
[56] R. Langer, K. Ott, K. Specht, K. Becker, F. Lordick, M. Burian, K. Herrmann, Comparative genomic hybridization: comparison between esophageal squamous
A. Schrattenholz, M.A. Cahill, M. Schwaiger, H. Hofler, H.J. Wester, Protein expression cell carcinoma and gastric cardia adenocarcinoma from a high-incidence area for
profiling in esophageal adenocarcinoma patients indicates association of heat-shock both cancers in Henan, northern China, Dis. Esophagus 19 (2006) 459–467.
protein 27 expression and chemotherapy response, Clin. Cancer Res. Off. J. Am. [79] K.F. Kwong, Molecular biology of esophageal cancer in the genomics era, Surg. Clin.
Assoc. Cancer Res. 14 (2008) 8279–8287. North Am. 85 (2005) 539–553.
N. Uemura, T. Kondo / Biochimica et Biophysica Acta 1854 (2015) 687–695 695

[80] H.B. Chen, K. Pan, M.K. Tang, Y.L. Chui, L. Chen, Z.J. Su, Z.Y. Shen, E.M. Li, W. Xie, Barrett's esophagus and esophageal adenocarcinoma, J. Proteome Res. 7 (2008)
K.K. Lee, Comparative proteomic analysis reveals differentially expressed proteins 1953–1962.
regulated by a potential tumor promoter, BRE, in human esophageal carcinoma [83] Y. Qi, J.F. Chiu, L. Wang, D.L. Kwong, Q.Y. He, Comparative proteomic analysis of
cells, Biochem. Cell Biol. Biochim. Biol. Cell. 86 (2008) 302–311. esophageal squamous cell carcinoma, Proteomics 5 (2005) 2960–2971.
[81] J. Zhao, A.C. Chang, C. Li, K.A. Shedden, D.G. Thomas, D.E. Misek, A.P. Manoharan, [84] Y.J. Qi, Q.Y. He, Y.F. Ma, Y.W. Du, G.C. Liu, Y.J. Li, G.S. Tsao, S.M. Ngai, J.F. Chiu, Prote-
T.J. Giordano, D.G. Beer, D.M. Lubman, Comparative proteomics analysis of Barrett omic identification of malignant transformation-related proteins in esophageal
metaplasia and esophageal adenocarcinoma using two-dimensional liquid mass squamous cell carcinoma, J. Cell. Biochem. 104 (2008) 1625–1635.
mapping, Mol. Cell. Proteomics 6 (2007) 987–999. [85] Z. Liu, J.G. Feng, A. Tuersun, T. Liu, H. Liu, Q. Liu, S.T. Zheng, C.G. Huang, G.D. Lv,
[82] J. Breton, M.C. Gage, A.W. Hay, J.N. Keen, C.P. Wild, C. Donnellan, J.B. Findlay, I. Sheyhidin, X.M. Lu, Proteomic identification of differentially-expressed proteins in
L.J. Hardie, Proteomic screening of a cell line model of esophageal carcinogenesis esophageal cancer in three ethnic groups in Xinjiang, Mol. Biol. Rep. 38 (2011)
identifies cathepsin D and aldo-keto reductase 1C2 and 1B10 dysregulation in 3261–3269.

You might also like