You are on page 1of 9

Pharmacological Research 113 (2016) 236–244

Contents lists available at ScienceDirect

Pharmacological Research
journal homepage: www.elsevier.com/locate/yphrs

Invited Review

Esophageal cancer: The latest on chemoprevention and state of the art


therapies
Gregoire F. Le Bras a , Muhammad H. Farooq a , Gary W. Falk b ,
Claudia D. Andl (Ph.D.,) (Associate Professor) a,∗
a
Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
b
Division of Gastroenterology, Department of Internal Medicine, University of Pennsylvania, Philadelphia, PA, United States

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

Article history: Esophageal cancer is currently the 8th most common cancer worldwide and the 6th leading cause of
Received 19 April 2016 cancer-related mortality. Despite remarkable advances, the mortality for those suffering from esophageal
Received in revised form 11 August 2016 cancer remains high, with 5-year survival rates of less than 20%. In part, because most patients present
Accepted 16 August 2016
with late-stage disease, long-term survival even after resection and therapy is disappointingly low. As we
Available online 24 August 2016
will discuss in this review, multiple characteristics specific to the disease stage and patient must be con-
sidered when choosing a treatment plan. This article will summarize current standard therapies, potential
Keyword:
application of chemoprevention drugs and the promise and partial failure of personalized medicine, as
Esophageal cancer
Cancer prevention
well as novel treatments addressing this disease.
Treatment © 2016 Elsevier Ltd. All rights reserved.
Chemotherapy
Immunotherapy

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
2. Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
2.1. ESCC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
2.2. EAC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
3. Clinical diagnosis and presentation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
4. Prevention . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
4.1. Proton pump inhibitors (PPI) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
4.2. Aspirin and NSAIDs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 238
4.3. Statins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 238
4.4. Antibacterial drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 238
5. Tumor staging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 238
6. Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 238
6.1. Multimodal treatment for ESCC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 239
6.2. Multi-modal treatments for EAC and tumors of the gastro-esophageal junction (GEJ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 239
6.3. Positron emission tomography (PET): a role in evaluating prognosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 239
6.4. Chemotherapy advances: improvements of the classics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 239
6.5. Targeted therapies: challenges . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 241
6.6. Immunotherapy: failed hope and uncertainties . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 242
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 242
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 242

∗ Corresponding author at: University of Central Florida, Burnett School of


Biomedical Sciences, College of Medicine, 4110 Libra Drive, Building 20, BMS 223,
Orlando, FL 32816, United States.
E-mail address: Claudia.andl@ucf.edu (C.D. Andl).

http://dx.doi.org/10.1016/j.phrs.2016.08.021
1043-6618/© 2016 Elsevier Ltd. All rights reserved.
G.F. Le Bras et al. / Pharmacological Research 113 (2016) 236–244 237

1. Introduction with non-Hispanic white people, with white Hispanics having an


intermediate risk [20].
Esophageal cancer is currently the 8th most common cancer
worldwide and the 6th leading cause of cancer-related mortality
[1–3]. This year, an estimated 17,000 adults (13,500 men and 3500 3. Clinical diagnosis and presentation
women) in the United States will be diagnosed with esophageal
cancer. It is expected that almost 16,000 deaths from this disease The clinical presentation of patients with esophageal cancer
will occur this year. In the period from 2005 to 2011 the 5-years can be attributed to the direct effects of tumor growth on local
survival was 17.9% according to the SEER Cancer Statistics review. and regional structures. Both, ESCC and EAC show similar man-
As we will discuss in this review, multiple characteristics spe- ifestations, such as difficulties swallowing (dysphagia) being the
cific to the disease stage and patient are considered when choosing most common symptom. Dysphagia initially occurs upon inges-
a treatment plan. With incidence rates rising and the 5-year survival tion of dense solid food, and progresses gradually to interference
being stagnant at below 20%, we must develop better diagnostics with the consumption of soft foods and ultimately even liquids.
and therapies. In part, because most patients present with late- Pain is a common symptom even in the absence of dysphagia, so is
stage disease, long-term survival even after resection and therapy weight loss, which correlates with the occurrence of tumor-related
is disappointingly low. For advanced cancer, multimodal therapy anorexia [21].
such as chemoradiation or combination chemotherapy are the cur- Imaging modalities such as endoscopy, endoscopic ultrasonog-
rent standards. Yet, targeted therapies, e.g. against the programmed raphy, narrow band imaging or computed tomography and others
cell death protein 1 signaling pathway (PD1-PDL1), a so-called are used in the diagnosis and staging of ESCC. Endoscopic screen-
immune checkpoint, show promise. ing as well as brush cytologic testing has been performed in China
Upon review of clinical presentation and diagnosis, we will focus for patients with mild, moderate and severe dysplasia who have in
on standard therapies and the promise of personalized medicine increased risk of developing squamous cell carcinoma [22]. Screen-
and novel treatments addressing this devastating disease. ings can prevent progression to more aggressive tumor growth and
thereby aid the reduction of mortality [23].
2. Epidemiology As Barrett’s esophagus is considered a risk factor for esophageal
adenocarcinoma, patients with gastroesophageal reflux diseases
Two major histological subtypes arise: esophageal squamous (GERD), who are at risk for high grade dysplasia ot Barrett’s esoph-
cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC). Over- agus, undergo endoscopy for screening [24]. Barrett’s esophagus
all, esophageal cancer incidence is three times higher in men than patients undergo surveillance endoscopy yet less than 15% of EAC
women and varies geographically by subtype [3]. are detected and most Barrett’s patients will never progress to
develop esophageal adenocarcinoma [25]. Subjecting biopsy mate-
2.1. ESCC rial to next-generation sequencing has been shown to identify
patients with Barrett’s esophagus who might have an increased
ESCC prevalence is highest in East Asia, Eastern and Southern risk for EAC, based on common mutations for example in tumor
Africa and some parts of Europe [4]. ESCC incidence has decreased suppressors such as TP53, as well as APC and CDKN2A [26]. So far,
over the past three decades in developed countries, where annual screening, counseling on lifestyle changes and preventative treat-
incidence rates are generally less than 10 per 100,000 per year ments are recommended [27,28].
[5], on the other hand, ESCC incidence has remained high in
less-developed countries with incidence rates exceeding 100 per 4. Prevention
100,000 per year, particularly in high-risk areas of China [6], Iran
[7,8] and South Africa [9]. The use of tobacco products, including Acid reflux is a risk factor for Barrett’s esophagus and for the pro-
cigarettes, cigars, pipes, and chewing tobacco, is a major risk fac- gression to esophageal cancer. Therefore, the use of proton pump
tor together with alcohol consumption for developing esophageal inhibitors is recommendedfor the reduction of gastric acid produc-
cancer [10,11]. ESCC is also a disease with health disparities affect- tion. Yet, recent studies also found a reduction in cancer risk in
ing more black males in Western countries and the United States, people with Barrett’s esophagus who take aspirin or similar. Some
especially in the context of high risk behavior [12]. studies identified statins, which are commonly used to treat high
cholesterol, to be effective in lowering cancer risk but are not pre-
2.2. EAC scribed as a preventative as they have serious side effects.

With ESCC rates declining, EAC now constitutes half of the


esophageal cancer cases in Western countries [13]. The increased 4.1. Proton pump inhibitors (PPI)
incidence rates are a reflection of life-style changes leading to
high rates of obesity and acid reflux, major factors contributing Proton pump inhibitors reduce stomach acid secretion through
to Barrett’s esophagus, a premalignant lesion, which predisposes inhibition of the H+/K+-ATPase in the stomach. This treatment tar-
the patients for a higher risk of developing EAC [14]. Overall, inci- gets patients with GERD as a risk factor for Barrett’s esophagus
dence rates rise steadily with advancing age [15]. A meta-analysis and esophageal adenocarcinoma. A significant association of PPI
of population-based studies showed a 5-fold increased risk for treatment with a decreased risk of high-grade dysplasia and ade-
EAC for weekly acid reflux symptoms [16]. Obesity, in particular nocarcinoma has been shown in clinic-based cohort studies for
a body mass index of >30 is associated with an approximate 2- patients with Barrett’s esophagus [29–31]. The indirect evidence
fold increased risk for EAC [17]. Strong epidemiologic risk factors supporting PPIs as a cancer-preventative promises a cost-effective
include aging, male gender, obesity and smoking [18]. The regional treatment for all patients with Barrett’s esophagus after they have
differences observed in the incidence of EAC indicate that race is been informed of the potential risks of long-term PPI therapy [32].
a strong risk factor for EAC. In the United Kingdom, the incidence There is some controversy as prolonged PPI use possibly promotes
of EAC is much lower among people of Asian and African descent progression to EAC through increased gastric pH associated with
compared with Caucasians [19]. In the United States, Asian and bile salt toxicity [33].
African Americans have a greatly decreased risk of EAC compared
238 G.F. Le Bras et al. / Pharmacological Research 113 (2016) 236–244

4.2. Aspirin and NSAIDs type II microbiome by killing acid- sensitive bacteria in the esoph-
agus. The use of proton pump inhibitors therefore acts on several
Results from observational studies demonstrated a protective levels of chemoprevention: inhibiting acid-related injury, and pre-
association between aspirin or non-steroidal anti-inflammatory venting changes in the microbiome [50,51]. The changes in the
drugs (NSAID) and esophageal cancer [34], which prompted a large microbiome hint at a potential use of antibiotics in the chemo-
ongoing randomized, phase III study of aspirin and esomepra- prevention of esophageal adenocarcinoma. However, conversion
zole, a PPI, in the prevention of esophageal cancer in patients of the altered “back to a normal” microbiota has not been tested
with Barrett’s esophagus. Long-term data are needed on the use yet. Invasion of pathogens is met with the innate immune response
of aspirin in Barrett esophagus patients. The ongoing AspECT trial upon sensing of a variety of microbial components through Toll-like
will be completed in 2017 and will give more information on clin- receptor (TLR) signaling. Due to the evidence for activation of the
ical outcomes. Aspirin has been proven effective as an analgesic TLR–NF-␬B pathway, which is responsible for the host immune and
and anti-inflammatory agent. Inflammation is frequently associ- inflammatory response to bacterial stimuli, it is plausible to utilize
ated with tumorigenesis, and inflammatory cells are often seen in anti-inflammatory strategies in chemoprevention of esophageal
Barrett’s esophagus as a result of COX-2 upregulation. Furthermore, adenocarcinoma, even in the context of microbial immune mod-
inflammatory signaling through IL-6 and IL-1␤ can be induced in ulation. However, as little is known about the mechanisms of
response to acid reflux [35]. However, aspirin is a key antiplatelet controlling the microbiome, more research in this area is war-
drug in the pharmacological prevention of cardiovascular diseases ranted.
at low doses, with the caveat that it could put patients at risk
for gastrointestinal bleeding due to damage of the mucosa [36].
5. Tumor staging
Recently, a study using an acute animal model of reflux esophagitis,
which mimicks the esophageal iniury of acid reflux, has shown that
The stage of esophageal cancer is determined by the depth of
nitric oxide derivatives of aspirin exerted protective effects against
infiltration of the tumor into the esophageal wall (T stage), by
reflux disease, while conventional NSAIDs like aspirin augmented
the presence of tumor cells in regional lymph nodes (N stage)
esophagitis [37].
and by distant metastases (M stage) [52]. Imaging techniques such
as endoscopic ultrasonography and 18 F-fluoro-2-deoxy-d-glucose
4.3. Statins
positron emission tomography (FDG-PET) have helped improve
staging and detect the status of tumor and lymph-nodes infil-
Considerable research has focused on the potential anti-cancer
tration with a relatively good accuracy [53,54]. In addition, the
effects of statins (3-hydroxy-3-methylglutaryl coenzyme A [HMG-
combination of computed tomography (CT) and FDG-PET can guide
CoA] reductase inhibitors) [38]. Commonly prescribed for the
surgical decision making [55,56]. Current standard therapy for
primary and secondary prevention of cardiovascular disease [39],
Stage I esophageal cancer, in which the tumor is localized to the
recent research studies have demonstrated that statins promote
esophagus is chemoradiation followed by surgery or surgery alone,
apoptosis and limit proliferation in EAC and ESCC cell lines [40–43].
while patients with advanced tumors (Stage IV), where the can-
In Demark, a population-based cohort showed that statin use
cer has spread, will additionally undergo post-operative radiation
prior to diagnosis of esophageal cancer was associated with a 19%
therapy.
decrease in cancer-specific mortality [44].
Esophageal squamous cell carcinoma and esophageal adenocar-
Observational studies have demonstrated that statin use in Bar-
cinoma are now considered separately. EAC staging is based on its
rett’s patients is associated with a lower incidence in EAC [45,46].
anatomical specificities as it is now included in a subset of tumors
Multivariable-adjusted hazard ratios were 0.85 for death unrelated
originating in close proximity to the gastro-esophageal junction
to cancer (95% confidence interval [CI], 0.83–0.87) and 0.85 for
[57,58], Fig. 1. Ultimately, the staging will determine the choice
cancer-related death (95% CI, 0.82–0.87) for statin users, when com-
treatment.
pared with patients who had never used statins [44]. A cohort study
conducted in the UK reported an increased median survival of 14.9
month in subjects treated with statins after diagnosis compared to 6. Treatment
8.1 month in non-users. However, while patients with esophageal
adenocarcinoma had a reduced risk of esophageal cancer-specific Much effort has been dedicated to improving treatment of
mortality (HR, 0.61; 95% CI 0.38–0.96) and all-cause mortality (HR, patients suffering from ESCC and EAC. Treatment of esophageal
0.63; 95% 0.43–0.92) upon statin use after diagnosis, no such effect tumors has traditionally involved surgery and esophagectomy,
was observed for ESCC [47]. which is associated with a high risk of morbidity in elderly patients
and patients suffering from concomitant diseases. Improvement in
4.4. Antibacterial drugs staging and in the evaluation of a patient’s fitness for resection have
reduced the perioperative risks, while the experience of physicians
The normal esophageal microbiome is composed of six major and high volume center are strongly associated with better surgery
phyla (Firmicutes, Bacteroides, Actinobacteria, Proteobacteria, outcome [59].
Fusobacteria, and TM7) and is dominated by the genus Streptococ- Ablation therapies such as radiofrequency ablation and pho-
cus [48]. The microbiome of the distal esophagus can be classified todynamic therapy are the new standard to treat patients with
into two groups, type I and type II [49]. Barrett’s esophagus or esophageal dysplasia. Endoscopic mucosal
The type I microbiome is composed primarily of Gram-positive resection and submucosal dissection have been shown as effective
bacteria and is more closely associated with a normal esophagus and safe new methods to remove small and superficial lesions [60].
whereas the type II microbiome is enriched with Gram-negative Aside from surgical resection the treatment options for
bacteria and is generally associated with an abnormal esopha- advanced stage cancer have been limited to chemoradiotherapy
gus. Streptococcus is the most dominant genus in the esophageal as an alternative for patients unable to receive surgery. Combi-
microbiome and its relative abundance is significantly higher in nation of radiation and chemotherapy may be associated with
the type I microbiome than in the type II microbiome. GERD plays improved survival if used in early stages of esophageal cancer
a role in the change of the esophageal microbiome as acidic gas- [52,61]. The benefit of neoadjuvant chemoradiation at later stage
tric juice can convert the type I microbiome into the abnormal remains unclear [62]. To improve survival outcomes adjuvant
G.F. Le Bras et al. / Pharmacological Research 113 (2016) 236–244 239

Fig. 1. Esophageal cancer risks and etiology. A. Alcohol and smoking are risk factors mainly for squamous cell carcinoma of the upper esophagus, yet also play a role in
esophageal adenocarcinoma, which is confined to the lower esophagus or the gastro-esophageal junction. Gastroesophageal acid reflux disease induces injury in the lower
esophagus resulting in metasplasia of the normal squamous epithelium. B. Morphology of the normal squamous epithelium compared to a well-differentiated esophageal
squamous cancer and the glandular morphology of the esophageal adenocarcinoma. Immunohistochemistry staining using antibody against E-cadherin, a cell adhesion
molecule, marking the epithelial and differentiated tumor tissue.

or neo-adjuvant therapies are used in association with surgery. ments, notably in the MAGIC trial, which was conducted using
Chemotherapy is more often used in Europe and Asia, while established guidelines for the administration of perioperative
chemoradiation is preferred in the United States. Combination chemotherapy (cisplatin, 5-fluorouracil (5-FU), and epirubicin) in
treatments are standard therapy now as intensifying conventional the surgical management of lower esophageal cancer; as well
chemotherapy drugs or increasing radiation dose have not been as in the “Chemoradiotherapy for Oesophageal Cancer Followed
successful (Tables 1 and 2 ). by Surgery Study” (CROSS) [75–78]. Similar effects were earlier
demonstrated by Tepper et al., although the number of patients
enrolled was low [79].
6.1. Multimodal treatment for ESCC
In a follow up to the CROSS study, Oppedijk et al. reported
that patients who received chemoradiation therapy had reduced
Data on neoadjuvant radiotherapy are controversial as com-
locoregional recurrence [80]. This has been confirmed in a recent
pared to surgery alone a meta-analysis of the literature reported
article by Thoen et al. showing low recurrence when neoadjuvant
only a limited effect [63], while neoadjuvant radiotherapy before
chemoradiotherapy was followed by surgery [81].
esophagectomy has been associated with increased overall survival
[64].
The aim of chemotherapy is to reduce tumor size prior to surgery 6.3. Positron emission tomography (PET): a role in evaluating
and target micrometastasis to avoid tumor spread. Chemother- prognosis
apy, however, is associated with toxicity and the risk of selecting
drug-resistant clones and delaying surgical treatment. Prior to Positron emission tomography (PET) is considered as a method
surgery, chemotherapy has been shown to provide an overall and to evaluate the response to chemotherapy in ESCC. The maximum
disease-free survival benefit over surgery alone as reported in a stand uptake value (SUVmax) together with the tumor/normal
meta-analysis based on 12 randomized trials [65]. E.g., preopera- esophageal ratio could be used to evaluate the effect of neoadju-
tive chemotherapy using two cycles of cisplatin and fluorouracil vant chemotherapy [82]. Additionally, [(18)F] fluorodeoxyglucose
improved survival for patients with resectable esophageal cancer (FDG)-PET positive lymph node analysis can be used to predict sur-
[66], yet the overall benefit of neoadjuvant or adjuvant treatments vival of patients who underwent neoadjuvant chemotherapy [83].
over surgery is still unclear in the case of resectable ESCC [67]. While Additional factors predicting outcome of neoadjuvant therapy can
few studies have shown a small benefits of the adjuvant treatment, be found in a recent review by Tao et al. [84].
but the effect on 5-year overall survival has not been consistently
demonstrated [67–71]. Similarly, overall survival after neoadjuvant
6.4. Chemotherapy advances: improvements of the classics
chemotherapy versus upfront surgery for patients with late stage
ESCC has not been demonstrated [72]. Interestingly, immediate
Management of esophageal cancer has evolved to include new
surgery might hold better outcome over neoadjuvant chemother-
chemotherapy regimen, multi-model treatments and promising
apy or definitive chemoradiotherapy for patients with stage III
new approaches such as immunotherapy. However, most of the
disease [73].
current protocols are based on novel combinations of standard
chemotherapy drugs such as cisplatin and 5-Fluorouracil.
6.2. Multi-modal treatments for EAC and tumors of the Cisplatin (Platinol), SP-4-2)-diamminedichloroplatinum(II),
gastro-esophageal junction (GEJ) was the first member of a class of platinum-containing anti-
cancer drugs, which now also includes carboplatin and oxaliplatin
Due to the difficulty of conducting large scale studies, the (Eloxatin). It is also called the “penicillin of cancer” because of
benefit of neoadjuvant chemoradiotherapy or chemotherapy over its wide-spread use. Cisplatin interferes with DNA replication
surgery alone has remained controversial in esophageal adenocar- and kills the proliferating cells, representing in theory the bulk
cinoma for a long time [74]. Recently, however, several studies of the tumor cells. Following administration, one of the two
have shown a consistent benefit of combining multimodal treat- chloride ligands is slowly displaced by water during aquation, cis-
240 G.F. Le Bras et al. / Pharmacological Research 113 (2016) 236–244

Table 1
Summary of recent publications of importance in the field of esophageal cancer treatments and their advances. Publication title and authors are listed.

Recent publications of importance in the field of esophageal cancer treatments

Title Refs.

Neoadjuvant Chemotherapy with Divided-dose Docetaxel, Cisplatin and Ojima T et al., Anticancer Res 2016, 36:829–34.
Fluorouracil for Patients with Squamous Cell Carcinoma of the Esophagus
Nimotuzumab Plus Paclitaxel and Cisplatin as the1st Line Treatment for Lu M et al., Cancer Sci 2016, 107:486–90.
Advanced Esophageal Squamous Cell Cancer(ESCC): a Single Centre
Prospective Phase II Trial
A randomized clinical trial of neoadjuvant chemotherapy versus neoadjuvant Klevebro F et al, Ann Oncol 2016, 27:660–7.
chemoradiotherapy for cancer of the esophagus or gastro-oesophageal
junction.
Association Between Statin Use After Diagnosis of Esophageal Cancer and [47]
Survival: a Population-based Cohort Study.
High-dose nimotuzumab improves the survival rate of esophageal cancer Wang C e tal., Onco Targets Ther 2015, 9:117–22.
patients who underwent radiotherapy.
Phase I/II study of divided-dose docetaxel, cisplatin and fluorouracil for Ojima T et al., Dis Esophagus, 2016 Jan 3. doi: 10.111.
patients with recurrent or metastatic squamous cell carcinoma of the
esophagus.
Preoperative low-dose weekly cisplatin and continuous infusion fluorouracil Caro M et al., Clin Transl Oncol 2016 Feb 8. doi:10.1007
plus hyperfractionated radiotherapy in stage II–III esophageal carcinoma.
Combination of cetuximab with radio-chemotherapy in patients with Hofheinz RD et al., J Thorac Dis. 2015, 7:e629–32.
esophageal cancer: less is more!
Possible prediction of the response of esophageal squamous cell carcinoma to Shen LY et al., Oncotarget 2016, 7:4531–41.
neoadjuvant chemotherapy based on gene expression profiling.
Adjuvant sunitinib following chemoradiotherapy and surgery for locally Horgan AM et al., Dis Esophagus 2015 Dec 10, doi:10.1111
advanced esophageal cancer: a phase II trial.
Changes in tumor expression of programmed death-ligand 1 after neoadjuvant Lim SH et al., Eur J Cancer 2016, 52:1–9.
concurrent chemoradiotherapy in patients with squamous oesophageal
cancer.
A randomized Phase II trial of systemic chemotherapy with and without Kataoka K, Jpn J Clin Oncol 2015, 45:1082-6.
trastuzumab followed by surgery in HER2-positive advanced gastric or
esophagogastric junction adenocarcinoma with extensive lymph node
metastasis: Japan Clinical Oncology Group study JCOG1301 (Trigger Study)
Metformin Use During Treatment of Potentially Curable Esophageal Cancer Spierings LE, Ann Surg Oncol 2015, suppl 3:766–71.
Patients is not Associated with Better Outcomes.
Neoadjuvant chemoradiotherapy with cisplatin plus vinorelbine versus Liu SL, Radiother Oncol 2015, 116;262–8.
cisplatin plus fluorouracil for esophageal squamous cell carcinoma: A matched
case-control study.
Neoadjuvant chemoradiotherapy plus surgery versus surgery alone for Shapiro J et al., Lancet Oncol 2015, 16:1090–8.
oesophageal or junctional cancer (CROSS): long-term results of a randomized
controlled trial.
Association Between Clinically Staged Node-Negative Esophageal Gabriel E, JAMA Surg 2016, 151:234–45.
Adenocarcinoma and Overall Survival Benefit From Neoadjuvant
Chemoradiation.
Cachexia in patients with oesophageal cancer Anandavadivelan P and Lagergren P. Nat Rev Clin Oncol 2016, 13:185–98.
Current concepts and future potential in neoadjuvant chemotherapy for Blum Murphy MA et al., Expert Rev Gastroenterol Hepatol 2016, 10:383–92.
esophageal cancer.
Current and Emerging Systemic Therapy in Gastro-Esophageal Cancer “The Old Lim B, Curr Clin Pharmcol 2015, 10: 267–78.
and New Therapy for Metastatic Disease, The Role of Adjuvant and
Neoadjuvant Therapy for Localized Disease”
A germline predictive signature of response to platinum chemotherapy Rumiato E, Transl Res 2015, S1931-5244 (15) 00453–3.
inesophageal cancer
Predictive Value of Ercc1 and Xpd Polymorphisms for Clinical Outcomes of Qixing M et al., Medicine 2015, 94:e1593.
Patients Receiving Neoadjuvant Therapy: A Prisma-Compliant Meta-Analysis.
Molecular Marker Expression Is Highly Heterogeneous in Esophageal Bronson NW, J Gastrointest Surg 2015, 19:2105–10.
Adenocarcinoma and Does Not Predict a Response to Neoadjuvant Therapy.
CD44, SHH and SOX2 as novel biomarkers in esophageal cancer patients Honing J et al., Radiother Oncol 2015, 117:152-8.
treated with neoadjuvant chemoradiotherapy.
IGFBP2 modulates the chemoresistant phenotype in esophageal Myers AL et al., Oncotarget 2015, 6:25897–916.
adenocarcinoma.
MiRNA Expression Analysis of Pretreatment Biopsies Predicts the Pathological Wen J et al., Ann Surg 2016, 263:942–8.
Response of Esophageal Squamous Cell Carcinomas to Neoadjuvant
Chemoradiotherapy.
Reevaluation of Neoadjuvant Chemotherapy for Esophageal Squamous Cell Zhen Y, eMedicine 2015, 94:e1102.
Carcinoma: A Meta-Analysis of Randomized Controlled Trials Over the Past 20
Years

[PtCl(NH3 )2 (H2 O)]+ . Dissociation of the chloride ligand is favored achieve synergistic effects. Together with folinic acid (Leucov-
due to gradients between the intra-and extracellular chloride orin) and 5 FU, oxaliplatin is known as FOLXFOX, a combi-
concentration [85]. nation chemotherapy regimen used as first line treatment in
Newer platinum-based drugs were introduced in the 1980s and advanced and metastatic esophageal cancer. Positive outcomes of
show fewer side effects, such as nephrotoxicity, ototoxicity, nausea oxaliplatin/fluoropyrimidine-based combinations have spured fur-
and vomitting. Oxaliplatin (Eloxatin), [(1R,2R)-cyclohexane-1,2- ther testing of these regimens.
diamine](ethanedioato-O,O’)platinum(II), a new platinum-based 5-Fluorouracil, (5-FU or Adrucil), 5-Fluoro-1H,3H-pyrimidine-
drug, is frequently used in combination with other drug to 2,4-dione, is a pyrimidine analogue, member of the anti-metabolite
G.F. Le Bras et al. / Pharmacological Research 113 (2016) 236–244 241

Table 2
A short list of currently ongoing trials reflecting some of the recent combination and novel targeted therapies.

Current Clinical Trials


Memorial Sloan Kettering Cancer Center, New York:
A Phase I Study of ADI-PEG 20 plus FOLFOX in Patients with Advanced Digestive Cancers
A Phase II Study of Afatinib (BIBW 2992) in Patients with Advanced HER2-Positive Esophagogastric Cancer Refractory to Trastuzumab
A Phase II Study of Neratinib in Patients with Solid Tumors Containing Mutated HER2
A Phase II Study of Nintedanib in Patients with Advanced Esophagogastric Cancer
A Phase II Study of Regorafenib versus Placebo in Patients with Node-Positive Esophageal Cancer Who Completed Chemotherapy, Radiation Therapy, and Surgery
A Phase III Study of Chemotherapy with or without Ramucirumab as Initial Treatment for Cancer of the Stomach or Gastroesophageal Junction
A Pilot Trial of PET Imaging with (89) Zr-DFO-Trastuzumab in Esophagogastric Cancer
A Prospective Clinical Trial to Evaluate Mesothelin as a Biomarker for the Clinical Management of Esophageal Cancer
M.D. Anderson, Texas:
A Phase I Study of LDE225 in Combination With Everolimus in Patients With Advanced Gastroesophageal Adenocarcinoma
Emory, Georgia; and Vanderbilt University, Tennessee:
A Phase I/IIB Study of Induction Chemotherapy with XELOX, Followed by Radiation Therapy and Dose Escalation of RAD001 in Patients With Esophageal Cancer
Multiple locations:
A Phase I/II Study of IMMU-132 (hRS7-SN38 Antibody Drug Conjugate) in Patients with Epithelial Cancers

class of drugs, which irreversibly inhibits thymidylate synthase. The aim of new combination chemotherapies is to account
By blocking thymidylate synthase, which is required for methyla- for tumor cell heterogeneity and the role it plays for drug resis-
tion of deoxyuridine monophosphate (dUMP) to form thymidine tance. Ample clinical precedent for using multiple agents has been
monophosphate (dTMP), it prevents synthesis of the pyrimi- presented in the past demonstrating evidence for synergy or an
dine thymidine needed for DNA replication. Administration of additive effect between established chemotherapeutic agents and
5-FU reduces dTMP, thereby inducing cell death [86]. Biomedi- agents representing other classes. The overall supportive care has
cal modifications and pharmacomodulation resulted in increased allowed patients to tolerat the toxicity better and retain a higher
therapeutic efficacy over a drug that has been reported first quality of life during treatment. Yet, given that every tumor is
in 1957 as a new class of tumor inhibitors. Other pyrimidine unique, we should aim to personalize treatments and reduce tox-
antagonist are Foxuridine, Cytarabine, Capecitabine, and Gemc- icity by targeting therapy. The discovery of new drugs as well
itabine. Combination of 5-FU with folinic acid (leucovorin) has as identification of targeted therapies that are more effective for
been shown to have synergistic effects as adjuvant chemother- esophageal cancer is ongoing (Tables 1 and 2).
apy. Folinic acid is a folate, which constitutes a group of vitamins.
It enhance the binding of 5-FU to thymidylate synthase increas- 6.5. Targeted therapies: challenges
ing its action in the cell [87]. A variant of 5-FU is capecitabine
(Xeloda), entyl [1-(3,4-dihydroxy-5-methyltetrahydrofuran-2-yl)- Several types of targeted therapies are currently being studied
5-fluoro-2-oxo-1H-pyrimidin-4-yl]carbamate. It is approved to for esophageal cancer. The most well-known targeted approach to
treat advanced breast cancer and colorectal cancer and used esophageal cancer has been the inhibition of epidermal growth fac-
off-label for esophageal and gastric cancer. It is a pro-drug, tor receptor (EGFR). EGFR is a member of the ErbB family of receptor
which is enzymatically converted into 5-FU, and classified as and consists of four closely related receptor tyrosine kinases (EGFR
an anti-metabolite drug. Xeloda has been described as an oral and Her2 through Her4). Cetuximab, a monoclonal immunoglobu-
chemotherapy drug, which could replace the use of cisplatin and lin against EGFR, is effective against head and neck squamous cell
5-FU. carcinoma in combination with radiotherapy [92]. However, addi-
Paclitaxel (Taxol) is one of several drugs targeting the cytoskele- tion of cetuximab to chemoradiation therapy showed no significant
ton, more specifically tubulin. Paclitaxel-treated cells have mitotic survival benefit for esophageal cancer, including ESCC, in a phase III
spindle assembly and cell division defects, as paclitaxel stabilizes study [93]. Similarly, aphase III study of gefitinib (Iressa), a tyrosine
the microtubules and prevent them from disassembly. This leads to kinase EGFR inhibitor, did not show an increase in overall survival
disruption of mitosis and the cells undergo apoptosis [88]. Another in patients with ESCC [94]. While more successful in small cell lung
anti-mitotic chemotherapeutic is docetaxel (Taxotere). Taxol is one cancer and head-and neck cancer, the majority of patients does not
of the most successful and bestselling drugs since its FDA approval respond to EGFR inhibitors. Resistance due to the accumulation of
in 1984 for use in a number of cancer such as breast, lung and ovar- mutations in EGFR or pathway targets as well as overexpression of
ian as reported by the NCI and is used in esophageal combination signaling targets. EGFR inhibitors and the mechanisms of resistance
therapy to improve patient outcome. appear to be diverse between and within cancer tissue types and
Irinotecan (Camptosar), is a topoisomerase I inhibitor, a have failed to give hope to esophageal cancer patients.
semisynthetic analogue of the natural alkaloid acamptothecin [89]. Her2-receptor targeting has been mainly used for breast cancer,
This is a more novel drug and, while it has a modified toxicity pro- but has recently been applied to treat EAC and adenocarcinoma
file when combined with radiation, its progonistic effectiveness has at the GEJ. Trastuzumab, sold under the brandname Herceptin
not yet been demonstrated. For colon cancer a combination regi- among others is a monoclonal antibody that interferes mainly
ment called FLFIRI, is used which consists of 5-FU, leucovorin and with the HER2/neu receptor [95]. Additionally, researchers are
irinotecan and could potential benefit esophageal cancer patients investigating novel HER2 target drugs for use in patients with
as well. advanced esophageal adenocarcinomas, including combinations of
A Phase I/II trial measuring the effectiveness of S-1, a novel trastuzumab and radiation therapy.
oral fluoropyrimidine derivative that consists of tegafur, gimeracil At the same time, researchers and clinicians are looking into
and potassium oxonate, with cisplatin (JCOG 0604) showed favor- drugs that block vascular endothelial growth factor (VEGF) sig-
able 3-year survival but did not meet its primary endpoint due to naling to inhibit neoangiogenesis. Particularly the use of the
low patient enrollment numbers [90]. A review by Tai et al. [91] anti-VEGFR2 antibody ramucirumab combined with chemother-
describes how ongoing studies, ZTOG1201 and QUINTETT, explor- apy as the first treatment has become a novel option. In 2014, the
ing neoadjuvant vs. adjuvant treatments in detail. FDA approved ramucirumab, as a single agent or in combination
with paclitaxel for the treatment of advanced gastric or gastro-
242 G.F. Le Bras et al. / Pharmacological Research 113 (2016) 236–244

esophageal junction adenocarcinoma. Especially, for cases in which References


the disease has progressed despite fluoropyrimidine- or platinum-
based therapy [96]. [1] A. Pennathur, M.K. Gibson, B.A. Jobe, et al., Oesophageal carcinoma, Lancet
381 (2013) 400–412.
Precision medicine based on genomic data could lead to new [2] F. Kamangar, G.M. Dores, W.F. Anderson, Patterns of cancer incidence,
methods for prevention, diagnosis, and treatment of esophageal mortality, and prevalence across five continents: defining priorities to reduce
cancer. Currently, the promise of personalized medicine has failed cancer dispar- ities in different geographic regions of the world, J. Clin. Oncol.
24 (2006) 2137–2150.
to give hope to improve the dire patient situation. [3] J. Ferlay, I. Soerjomataram, R. Dikshit, et al., Cancer inci- dence and mortality
worldwide: sources, methods and major patterns in GLOBOCAN 2012, Int. J.
Cancer 136 (2015) E359–E386.
[4] A. Pickens, M.B. Orringer, Geographical distribution and racial disparity in
6.6. Immunotherapy: failed hope and uncertainties esophageal cancer, Ann. Thorac. Surg. 76 (2003) S1367–S1369.
[5] L.M. Brown, S.S. Devesa, Epidemiologic trends in esophageal and gastric
cancer in the United States, Surg. Oncol. Clin. N. Am. 11 (2002) 235–256.
While monoclonal antibodies used as drugs against specific [6] W.J. Blot, J.Y. Li, P.R. Taylor, et al., Nutrition intervention trials in Linxian,
target in the bodies can be considered immunotherapy as they China: supplementation with specific vitamin/mineral combinations, cancer
cause an immune response to the cancer cell, there are other types incidence, and disease-specific mortality in the general population, J. Natl.
Cancer Inst. 85 (1993) 1483–1492.
of immunotherapy. Immunotherapy, also referred to as biologi- [7] F. Islami, F. Kamangar, K. Aghcheli, et al., Epidemiologic features of upper
cal therapy, is designed to improve, target, or restore the patient gastrointestinal tract cancers in Northeastern Iran, Br. J. Cancer 90 (2004)
immune function to target cancer cells. Adoptive cell transfer is 1402–1406.
[8] E. Mahboubi, J. Kmet, P.J. Cook, et al., Oesophageal cancer studies in the
one of them, in which T cells from a patient’s tumor are isolated and Caspian Littoral of Iran: the Caspian cancer registry, Br. J. Cancer 28 (1973)
the most tumor-reactive ones applied to destroy the tumor cells. 197–214.
Another approach is to use cytokines such as interferons and inter- [9] N.I. Somdyala, W.F. Marasas, F.S. Venter, et al., Cancer patterns in four districts
of the Transkei region—1991–1995, S. Afr. Med. J. 93 (2003) 144–148.
leukins to regulate immune responses. Interferon-alpha treatment [10] L.M. Brown, R.N. Hoover, R.S. Greenberg, et al., Are racial differences in
in melanoma was a recent hot topic due to price of the treatment squamous cell esophageal cancer explained by alcohol and tobacco use? J.
and the marginal improvement in overall survival. Natl. Cancer Inst. 86 (1994) 1340–1345.
[11] A.J. Tuyns, Oesophageal cancer in non-smoking drinkers and in non-drinking
A current highlight are the use of immune checkpoint block- smokers, Int. J. Cancer 32 (1983) 443–444.
ades, which have provided new therapeutic avenues in melanoma. [12] L.M. Brown, R. Hoover, D. Silverman, et al., Excess incidence of squamous cell
Ongoing investigations will hopefully allow treatment of other esophageal cancer among US black men: role of social class and other risk
factors, Am. J. Epidemiol. 153 (2001) 114–122.
tumor types including gastrointestinal malignancies. So far,
[13] H. Rubenstein, N.J. Shaheen, Epidemiology, diagnosis, and management of
esophageal and gastriccancers have represented a minority in esophageal adenocarcinoma, Gastroenterology 149 (2015) 302–317.
early phase immune checkpoint inhibitor trials. Major advances [14] F. Hvid-Jensen, L. Pedersen, A.M. Drewes, H.T. Sørensen, P. Funch-Jensen,
have come from inhibitory antibodiy development, which can Incidence of adenocarcinoma among patients with Barrett’s esophagus, N.
Engl. J. Med. 365 (2011) 1375–1383.
modulate immune checkpoints. Three checkpoint inhibitors, pem- [15] F. Kamangar, G.M. Dores, W.F. Anderson, Patterns of cancer incidence,
brolizumab, nivolumab, and ipilimumab, are now approved by the mortality and prevalence across five continents: defining priorities to reduce
FDA. Ipilimumad is a human monoclonal antibody that binds cyto- cancer disparities in different geographic regions of the world, J. Clin. Oncol.
24 (2006) 2137–2150.
toxic T-lymphocyte antigen 4 (CTLA-4), programmed cell death [16] J.H. Rubenstein, J.B. Taylor, Meta-analysis: the association of oesophageal
protein 1 (PD-1), and programmed death ligand 1 (PD-L1). The adenocarcinoma with symptoms of gastro-oesophageal reflux, Aliment.
outlook for their use in gastrointestinal cancer is uncertain as Pharmacol. Ther. 32 (2010) 1222–1227.
[17] C. Hoyo, M.B. Cook, F. Kamangar, et al., Body mass index in relation to
numbers in the multicenter phase I trial (e.g. for anti-PD-L1 mAb oesophageal and oesophagogastric junc- tion adenocarcinomas: a pooled
BMS-936559) are small with only 7 gastric cancer patients of 207 analysis from the in- ternational BEACON consortium, Int. J. Epidemiol. 41
enrolled patients overall. A second line gastroesophageal-specific (2012) 1706–1718.
[18] G.W. Falk, Barrett’s oesophagus: frequency and [rediction of dysplasia and
phase II trial (n = 18) showed even discouraging results as the cancer, Best Pract. Res. Clin. Gastroenterol. 29 (2015) 125–138.
observed response rate to tremelimumab (anti-CTLA4 mAb) was [19] R. Ali, I. Barnes, B.J. Cairns, et al., Incidence of gastroin- testinal cancers by
only 5%, well below the response rate for cytotoxic chemotherapy ethnic group in England, 2001–2007, Gut 62 (2013) 1692–1703.
[20] A. Kubo, D.A. Corley, Marked multi-ethnic variation of esophageal and gastric
[97].
cardia carcinomas within the United States, Am. J. Gastroenterol. 99 (2004)
Very few reports describe vaccine-based or adoptive cell trans- 582–588.
fer immunotherapy for esophageal carcinoma. A phase I trial was [21] Uptodate.
conducted with a peptide vaccine administered to 10 patients with [22] A.K. Rustgi, H.B. El-Serag, Esophageal carcinoma, N. Engl. J. Med. 371 (2014)
2499–2509.
stage III or IV ESCC whose disease had progressed on conven- [23] W.Q. Wei, Z.F. Chen, Y.T. He, et al., Long-term follow-up of a community
tional treatment, with 1 complete response and stable disease in assignment, one-time endoscopic screening study of esophageal cancer in
3 patients [98]. China, J. Clin. Oncol. 33 (2015) 1951–1957.
[24] N.J. Shaheen, G.W. Falk, P.G. Iyer, L.B. Gerson, American college of
One phase I/II trial has been conducted for esophageal SCC gastroenterology. ACG clinical guideline: diagnosis and management for
with adoptive cell therapy [99]. Additional trials are required to Barrett’s esophagus, Am. J. Gastroenterol. 111 (2016) 30–50.
determine the efficacy of vaccine and T-cell–based therapies for [25] R.E. Verbeek, M. Leenders, F.J. Ten Kate, et al., Surveillance of Barrett’s
esophagus and mortality from esophageal adenocarcinoma: a
esophageal carcinoma. population-based cohort study, Am. J. Gastroenterol. 109 (2014) 1215–1222.
Today, researchers and clinicians are conducting a diverse range [26] A. Del Portillo, S.M. Lagana, Y. Yao, T. Uehara, N. Jhala, T. Ganguly, P. Nagy, J.
of studies aimed at developing the next generation therapies, Gutierrez, A. Luna, J. Abrams, Y. Liu, R. Brand, J.L. Sepulveda, G.W. Falk, A.R.
Sepulveda, Evaluation of mutational testing of preneoplastic Barrett’s mucosa
including targeted and immune-based treatments. by next-generation sequencing of formalin-fixed, paraffin-embedded
endoscopic samples for detection of concurrent dysplasia and
adenocarcinoma in Barrett’s esophagus, J. Mol. Diagn. 17 (2015) 412–419.
[27] B.C. Jacobson, S.C. Somers, C.S. Fuchs, C.P. Kelly, C.A. Camargo Jr., Body-mass
Acknowledgements index and symptoms of gastroesophageal reflux in women, N. Engl. J. Med.
354 (2006) 2340–2348.
[28] G. Boeckxstaens, H.B. El-Serag, A.J. Smout, P.J. Kahrilas, Symptomatic reflux
This work was supported by grant NIH-DK094900 from the disease: the present, the past and the future, Gut 63 (2014) 1185–1193.
National Institute of Diabetes and Digestive and Kidney Disease. [29] D.M. Nguyen, H.B. El-Serag, L. Henderson, D. Stein, A. Bhattacharyya, R.E.
We like to thank Dr. Thomas Andl for discussion and editing as Sampliner, Medication usage and the risk of neoplasia in patients with
Barrett’s esophagus, Clin. Gastroenterol. Hepatol. 7 (2009) 1299–1304.
well as all the contributions of other studies and authors which we
could not cite in this review.
G.F. Le Bras et al. / Pharmacological Research 113 (2016) 236–244 243

[30] L.C. Hillman, L. Chiragakis, B. Shadbolt, G.L. Kaye, A.C. Clarke, Proton-pump [57] S.B. Edge, C.C. Compton, The American joint committee on cancer: the 7th
inhibitor therapy and the development of dysplasia in patients with Barrett’s edition of the AJCC cancer staging manual and the future of TNM, Ann. Surg.
oesophagus, Med. J. Aust. 180 (2004) 387–391. Oncol. 17 (2010) 1471–1474.
[31] F. Kastelein, M.C. Spaander, E.W. Steyerberg, et al., Proton pump inhibitors [58] L. Kleinberg, M. Brock, M. Gibson, Management of locally advanced
reduce the risk of neoplastic progression in patients with Barrett’s esophagus, adenocarcinoma of the esophagus and gastroesophageal junction: finally a
Clin. Gastroenterol. Hepatol. 11 (2013) 382–388. consensus, Curr. Treat. Options Oncol. 16 (2015) 35.
[32] K.B. Dunbar, R.F. Souza, S.J. Spechler, The effect of proton pump inhibitors on [59] E.W. Steyerberg, B.A. Neville, L.B. Koppert, V.E.P.P. Lemmens, H.W. Tilanus,
Barrett’s esophagus, Gastroenterol. Clin. N. Am. 44 (2015) 415–424. J.-W.W. Coebergh, J.C. Weeks, C.C. Earle, Surgical mortality in patients with
[33] O. Alsalahi, A.D. Dobrian, Proton pump inhibitors: the culprit for barrett’s esophageal cancer: development and validation of a simple risk score, J. Clin.
esophagus, Front. Oncol. 4 (2014) 373. Oncol. 24 (2006) 4277–4284.
[34] D.A. Corley, K. Kerlikowske, R. Verma, P. Buffler, Protective association of [60] P.M. Shah, H. Gerdes, Endocscopic options for early stage esophageal cancer, J.
aspirin/NSAIDs and esophageal cancer: a systematic review and Gastrointest. Oncol. 6 (2015) 20–30.
meta-analysis, Gastroenterology 124 (2003) 47–56. [61] C. Pöttgen, M. Stuschke, Radiotherapy versus surgery within multimodality
[35] E. Richmond, A. Umar, Mechanisms of esophageal adenocarcinoma formation protocols for esophageal cancer—a meta-analysis of the randomized trials,
and approaches to chemopreventative intervention, Semin. Oncol. 43 (2016) Cancer Treat. Rev. 38 (2012) 599–604.
78–85. [62] J.Q. Zhang, C.M. Hooker, M.V. Brock, J. Shin, S. Lee, R. How, N. Franco, H. Prevas,
[36] C. Sostres, A. Lanas, Gastrointestinal effects of aspririn, Nat. Rev. A. Hulbert, S.C. Yang, Neoadjuvant chemoradiation therapy is beneficial for
Gastroenterol. Hepatol. 8 (2011) 385–394. clinical stage T2 N0 esophageal cancer patients due to inaccurate preoperative
[37] M. Pawlik, R. Pajdo, S. Kwiecien, A. Ptak-Belowska, Z. Sliwowski, M. staging, Ann. Thorac. Surg. 93 (2012) 429–435, discussion 436–437.
Mazurkiewicz-Janik, S.J. Konturek, W.W. Pawlik, T. Brzozowski, Nitric oxide [63] S.J. Arnott, W. Duncan, M. Gignoux, H.S. Hansen, B. Launois, K. Nygaard, M.K.B.
(NO)-releasing aspirin exhibits a potent esophagoprotection in experimental Parmar, A. Rousell, G. Spilopoulos, G. Stewart, et al., Preoperative radiotherapy
models of acute reflux esophagitis. Role of nitric oxide and proinflammatory for esophageal carcinoma, Cochrane Database Syst. Rev. 4 (2005), CD001799.
cytokines, J. Physiol. Pharmacol. 62 (2011) 75–86. [64] A.L. Schwer, A. Ballonoff, R. McCammon, K. Rusthoven, R.B. D’Agostino, T.E.
[38] S. Zhong, X. Zhang, L. Chen, T. Ma, J. Tang, J. Zhao, Statin use and mortality in Schefter, Survival effect of neoadjuvant radiotherapy before esophagectomy
cancer patients: systematic review and meta-analysis of observational for patients with esophageal cancer: a surveillance, epidemiology, and
studies, Cancer Treat. Rev. 41 (2015) 554–567. end-results study, Int. J. Radiat. Oncol. Biol. Phys. 73 (2009) 449–455.
[39] A. Cooper, N. O’Flynn, Guideline Development Group, Risk assessment and [65] P. Thirion, E. Maillard, J. Pignon, Individual patient data-based meta-analysis
lipid modification for primary and secondary prevention of cardiovascular assessing the effect of preoperative chemo-radiotherapy in resectable
disease: summary of NICE guidance, BMJ 336 (2008) 1246–1248. oesophageal carcinoma, Int. J. Radiat. Oncol. Biol. Phys. 72 (2008) S71–S72.
[40] O.O. Ogunwobi, I.L. Beales, Statins inhibit proliferation and induce apoptosis [66] Medical Research Council Oesophageal Cancer Working Group. Surgical
in Barrett’s esophageal adenocarcinoma cells, Am. J. Gastroenterol. 103 (2008) resection with or without preoperative chemotherapy in oesophageal cancer:
825–837. a randomised controlled trial. The Lancet (2002) 359: 1727–1733.
[41] M.R. Sadaria, A.E. Reppert, J.A. Yu, et al., Statin therapy attenuates growth and [67] Y. Xu, X. Yu, Q. Chen, W. Mao, Neoadjuvant versus adjuvant treatment: which
malignant potential of human esophageal adenocarcinoma cells, J. Thorac. one is better for resectable esophageal squamous cell carcinoma? World J.
Cardiovasc. Surg. 142 (2011) 1152–1160. Surg. Oncol. 10 (2012) 173.
[42] P.C. Konturek, G. Burnat, E.G. Hahn, Inhibition of Barret’s adenocarcinoma cell [68] N. Ando, T. Iizuka, H. Ide, K. Ishida, M. Shinoda, T. Nishimaki, W. Takiyama, H.
growth by simvastatin: involvement of COX-2 and apoptosis-related proteins, Watanabe, K. Isono, N. Aoyama, et al., Surgery plus chemotherapy compared
J. Physiol. Pharmacol. 58 (Suppl. 3) (2007) 141–148. with surgery alone for localized squamous cell carcinoma of the thoracic
[43] J. Shi, J. Zhu, H. Zhao, C. Zhong, Z. Xu, F. Yao, Mevalonate pathway is a esophagus: a Japan Clinical Oncology Group Study—JCOG9204, J. Clin. Oncol.
therapeutic target in esophageal squamous cell carcinoma, Tumour Biol. 34 21 (2003) 4592–4596.
(2012) 429–435. [69] N. Ando, H. Kato, H. Igaki, M. Shinoda, S. Ozawa, H. Shimizu, T. Nakamura, H.
[44] S.F. Nielsen, B.G. Nordestgaard, S.E. Bojesen, Statin use and reduced cancer- Yabusaki, N. Aoyama, A. Kurita, et al., A randomized trial comparing
related mortality, N. Engl. J. Med. 367 (2012) 1792–1802. postoperative adjuvant chemotherapy with cisplatin and 5-fluorouracil
[45] I.L. Beales, A. Hensley, Y. Loke, Reduced esophageal cancer incidence in statin versus preoperative chemotherapy for localized advanced squamous cell
users, particular with cyclo-oxygenase inhibition, World J. Gastrointest. carcinoma of the thoracic esophagus (JCOG9907), Ann. Surg. Oncol. 19 (2012)
Pharmacol. Ther. 4 (2013) 69–79. 68–74.
[46] S. Singh, A.G. Singh, P.P. Singh, M.H. Murad, P.G. Iyer, Statins are associated [70] B.H. Burmeister, Role of radiotherapy in the pre-operative management of
with reduced risk of esophageal cancer, particularly in patients with Barrett’s carcinoma of the esophagus, World J. Gastrointest. Oncol. 7 (2015) 1–5.
esophagus: a systematic review and meta-analysis, Clin. Gastroenterol. [71] A. Hanna, R. Birla, C. Iosif, M. Boeriu, S. Constantinoiu, Benefits and
Hepatol. 11 (2013) 620–629. disadvantages of neoadjuvant radiochemotherapy (RCT) in the multimodal
[47] L. Alexandre, A.B. Clark, H.Y. Bhutta, S.S.M. Chan, M.P.N. Lewis, A.R. Hart, therapy of squamous esophageal cancer (ESC), Chirurgia (Bucur) 111 (2016)
Association between statin use after diagnosis of esophageal cancer and 12–25.
survival: a population-based cohort study, Gastroenterology 150 (2016) [72] B.H. Burmeister, B.M. Smithers, V. Gebski, L. Fitzgerald, R.J. Simes, P. Devitt, S.
854–865, e1. Ackland, D.C. Gotley, D. Joseph, J. Millar, et al., Surgery alone versus
[48] Z. Pei, E.J. Bini, L. Yang, M. Zhou, F. Francois, M.J. Blaser, Bacterial biota in the chemoradiotherapy followed by surgery for resectable cancer of the
human distal esophagus, Proc. Natl. Acad. Sci. U. S. A. 101 (2004) 4250–4255. oesophagus: a randomised controlled phase III trial, Lancet Oncol. 2005 (6)
[49] L. Yang, X. Lu, C.W. Nossa, F. Francois, R.M. Peek, Z. Pei, Inflammation and (2005) 659–668.
intestinal metaplasia of the distal esophagus are associated with alterations [73] M. Nomura, I. Oze, T. Kodaira, T. Abe, A. Komori, Y. Narita, T. Masuishi, H.
in the microbiome, Gastroenterology 137 (2009) 588–597. Taniguchi, S. Kadowaki, T. Ura, et al., Comparison between surgery and
[50] S. Singh, S.K. Garg, P.P. Singh, P.G. Iyer, H.B. El-Serag, Acid-suppressive definitive chemoradiotherapy for patients with resectable esophageal
medications and risk of oesophageal adenocarcinoma in patients with squamous cell carcinoma: a propensity score analysis, Int. J. Clin. Oncol.
Barrett’s oesophagus: a systematic review and meta-analysis, Gut 63 (2014) (2016), March 15, (epub ahead of print).
1229–12370. [74] B.P.L. Wijnhoven, J.J.B. Lanschot, Tilanus van, H.W. Steyerberg, E.W. Gaast, A.
[51] Z. Pei, L. Yang, R.M. Peek Jr., S.M. Levine, D.T. Pride, M.J. Blaser, Bacterial biota van der, Neoadjuvant chemoradiotherapy for esophageal cancer: a review of
in reflux esophagitis and Barrett’s esophagus, World J. Gastroenterol. 11 meta-analyses, World J. Surg. 33 (2009) 2606–2614.
(2005) 7277–7283. [75] D. Cunningham, W.H. Allum, S.P. Stenning, J.N. Thompson, C.J.H. Van de Velde,
[52] J.A. Ajani, T.A. D’Amico, K. Almhanna, D.J. Bentrem, S. Besh, J. Chao, P. Das, C. M. Nicolson, J.H. Scarffe, F.J. Lofts, S.J. Falk, T.J. Iveson, et al., Perioperative
Denlinger, P. Fanta, C.S. Fuchs, et al., Esophageal and esophagogastric junction chemotherapy versus surgery alone for resectable gastroesophageal cancer,
cancers, version 1.2015, J. Natl. Compr. Cancer Netw. JNCCN 13 (2015) N. Engl. J. Med. 355 (2006) 11–20.
194–227. [76] P. van Hagen, M.C.C.M. Hulshof, J.J.B. van Lanschot, E.W. Steyerberg, M.I.
[53] J. Lin, S. Kligerman, R. Goel, P. Sajedi, M. Suntharalingam, M.D. Chuong, Henegouwen, B. van, B.P.L. Wijnhoven, D.J. Richel, G.A.P. Nieuwenhuijzen,
State-of-the-art molecular imaging in esophageal cancer management: G.A.P. Hospers, J.J. Bonenkamp, et al., Preoperative chemoradiotherapy for
implications for diagnosis, prognosis, and treatment, J. Gastrointest. Oncol. 6 esophageal or junctional cancer, N. Engl. J. Med. 366 (2012) 2074–2084.
(2015) 3–19. [77] K.M. Sjoquist, B.H. Burmeister, B.M. Smithers, J.R. Zalcberg, R.J. Simes, A.
[54] E.P.M. van Vliet, M.H. Heijenbrok-Kal, M.G.M. Hunink, E.J. Kuipers, P.D. Barbour, V. Gebski, Australasian Gastro-Intestinal Trials Group, Survival after
Siersema, Staging investigations for oesophageal cancer: a meta-analysis, Br. neoadjuvant chemotherapy or chemoradiotherapy for resectable oesophageal
J. Cancer 98 (2008) 547–557. carcinoma: an updated meta-analysis, Lancet Oncol. 12 (2011) 681–692.
[55] M.C.J. Anderegg, E.J. de Groof, S.S. Gisbertz, R.J. Bennink, S.M. Lagarde, J.H.G. [78] S.O. van der Woude, M.C.C.M. Hulshof, H.W.M. van Laarhoven, CROSS and
Klinkenbijl, M.G.W. Dijkgraaf, J.J.G.H.M. Bergman, M.C.C.M. Hulshof, H.W.M. beyond: a clinical perspective on the results of the randomized
van Laarhoven, et al., 18F-FDG PET-CT after neoadjuvant chemoradiotherapy ChemoRadiotherapy for Oesophageal cancer followed by Surgery Study, Chin.
in esophageal cancer patients to optimize surgical decision making, PLoS One Clin. Oncol. 5 (2016) 13.
10 (2015) e0133690. [79] J. Tepper, M.J. Krasna, D. Niedzwiecki, D. Hollis, C.E. Reed, R. Goldberg, K. Kiel,
[56] S. Liu, H. Zhu, W. Li, B. Zhang, L. Ma, Z. Guo, Y. Huang, P. Song, J. Yu, H. Guo, C. Willett, D. Sugarbaker, R. Mayer, Phase III trial of trimodality therapy with
Potential impact of 18FDG-PET/CT on surgical approach for operable cisplatin, fluorouracil, radiotherapy, and surgery compared with surgery
squamous cell cancer of middle-to-lower esophagus, OncoTargets Ther. 9 alone for esophageal cancer: CALGB 9781, J. Clin. Oncol. 26 (2008) 1086–1092.
(2016) 855–862.
244 G.F. Le Bras et al. / Pharmacological Research 113 (2016) 236–244

[80] V. Oppedijk, A. van der Gaast, J.J.B. van Lanschot, P. van Hagen, R. van Os, C.M. [90] M. Tahara, N. Fuse, J. Mizusawa, A. Sato, K. Nihei, K. Kanato, K. Kato, K.
van Rij, M.J. van der Sangen, J.C. Beukema, H. Rütten, P.H. Spruit, et al., Patterns Yamazaki, K. Muro, H. Takaishi, et al., Phase I/II trial of chemoradiotherapy
of recurrence after surgery alone versus preoperative chemoradiotherapy and with concurrent S-1 and cisplatin for clinical stage II/III esophageal carcinoma
surgery in the CROSS trials, J. Clin. Oncol. 32 (2014) 385–391. (JCOG 0604), Cancer Sci. 106 (2015) 1414–1420.
[81] H. Thoen, W. Ceelen, T. Boterberg, E. Van Daele, P. Pattyn, Tumor recurrence [91] P. Tai, E. Yu, Esophageal cancer management controversies: radiation
and in-field control after multimodality treatment of locally advanced oncology point of view, World J. Gastrointest. Oncol. 6 (2014) 263–274.
esophageal cancer, Radiother. Oncol. J. Eur. Soc. Ther. Radiol. Oncol. 115 [92] B. Burtness, M.A. Goldwasser, W. Flood, et al., Phase III randomized trial of
(2015) 16–21. cisplatin plus placebo compared with cisplatin plus cetuximab in
[82] D. Izumi, N. Yoshida, M. Watanabe, S. Shiraishi, T. Ishimoto, K. Kosumi, R. metastatic/recurrent head-and-neck cancer: an Eastern Cooperative Oncology
Tokunaga, K. Taki, T. Higashi, K. Harada, et al., Tumor/normal esophagus ratio Group study, J. Clin. Oncol. 23 (2005) 8646–8654.
in (18)F-fluorodeoxyglucose positron emission tomography/computed [93] T. Crosby, C.N. Hurt, S. Falk, et al., Chemoradiotherapy with or without
tomography for response and prognosis stratification after neoadjuvant cetuximab in patients with oesophageal can- cer (SCOPE1): a multicentre,
chemotherapy for esophageal squamous cell carcinoma, J. Gastroenterol. 8 phase 2/3 randomised trial, Lancet Oncol. 14 (2013) 627–637.
(2015), Dec 15. [94] S.J. Dutton, D.R. Ferry, J.M. Blazeby, et al., Gefitinib for oesophageal cancer
[83] H. Miyata, M. Yamasaki, T. Makino, M. Tatsumi, Y. Miyazaki, T. Takahashi, Y. progressing after chemotherapy (COG): a phase 3, multicentre, double-blind,
Kurokawa, S. Takiguchi, M. Mori, Y. Doki, Impact of number of placebo- controlled randomised trial, Lancet Oncol. 15 (2014) 894–904.
[18F]fluorodeoxyglucose-PET-positive lymph nodes on survival of patients [95] C.A. Hudis, Trastuzumab—mechanism of action and use in clinical practice, N.
receiving neoadjuvant chemotherapy and surgery for oesophageal cancer: Engl. J. Med. 357 (1) (2007) 39–51.
PET-positive nodes in oesophageal cancer, Br. J. Surg. 103 (2016) 97–104. [96] H. Wilke, K. Muro, E. Van Cutsem, S.C. Oh, G. Bodoky, Y. Shimada, S. Hironaka,
[84] C.-J. Tao, G. Lin, Y.-P. Xu, W.-M. Mao, Predicting the response of neoadjuvant N. Sugimoto, O. Lipatov, T.Y. Kim, D. Cunningham, P. Rougier, Y. Komatsu, J.
therapy for patients with esophageal carcinoma: an in-depth literature Ajani, M. Emig, R. Carlesi, D. Ferry, K. Chandrawansa, J.D. Schwartz, A. Ohtsu,
review, J. Cancer 6 (2015) 1179–1186. RAINBOW Study Group Ramucirumab plus paclitaxel versus placebo plus
[85] Timothy C. Johnstone, Kogularamanan Suntharalingam, Stephen J. Lippard, paclitaxel in patients with previously treated advanced gastric or
The next generation of platinum drugs: targeted Pt(II) agents, nanoparticle gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind,
delivery, and Pt(IV), Prodrugs Chem Rev. 116 (2016) 3436–3486. randomised phase 3 trial, Lancet Oncol. 15 (2014) 1224–1235.
[86] D.B. Longley, D.P. Harkin, P.G. Johnston, 5-Fluorouracil: mechanisms of action [97] A.G. Raufi, S.J. Klempner, Immunotherapy for advanced gastric and
and clinical strategies, Nat. Rev. Cancer 3 (5) (2003) 330–338. esophageal cancer: preclinical rationale and ongoing clinical investigations, J.
[87] P. Noordhuis, U. Holwerda, C.L. Van der Wilt, C.J. Van Groeningen, K. Smid, S. Gastrointest. Oncol. 6 (October (5)) (2015) 561–569.
Meijer, H.M. Pinedo, G.J. Peters, 5-Fluorouracil incorporation into RNA and [98] K. Kono, Y. Mizukami, Y. Daigo, et al., Vaccination with multiple peptides
DNA in relation to thymidylate synthase inhibition of human colorectal derived from novel cancer-testis antigens can induce specific T-cell responses
cancers, Ann. Oncol. 15 (2004) 1025–1032. and clinical responses in advanced esophageal cancer, Cancer Sci. 100 (2009)
[88] M.A. Jordan, L. Wilson, Microtubules as a target for anticancer drugs, Nat. Rev. 1502–1509.
Cancer 4 (2004) 253–265. [99] U. Toh, H. Yamana, S. Sueyoshi, et al., Locoregional cellular immunotherapy
[89] Y. Pommier, E. Leo, H. Zhang, C. Marchand, DNA topoisomerases and their for patients with advanced esophageal cancer, Clin. Cancer Res. 6 (12) (2000)
poisoning by anticancer and antibacterial drugs, Chem. Biol. 17 (2010) 4663–4673.
421–433.

You might also like