ArticlePDF AvailableLiterature Review

The Role of the Microbiota in Esophageal Cancer

MDPI
Cancers
Authors:

Abstract and Figures

Simple Summary Esophageal cancer has very high mortality and morbidity rates. In this study, we reviewed the current literature on the impact of microbiota on esophageal cancer and its precursor lesions. Globally, a decrease in microbiota richness and evenness in esophageal cancer is identified, which is accompanied by an increase in the abundance of Gram-negative bacteria. Abstract Esophageal cancer is a major health problem, being the seventh most incidence cancer worldwide. Due to the often-late diagnosis and lack of efficient treatments, the overall 5-year survival is as low as 10%. Therefore, understanding the etiology and the mechanisms that drive the development of this type of cancer could improve the management of patients, increasing the chance of achieving a better clinical outcome. Recently, the microbiome has been studied as a putative etiological factor for esophageal cancer. Nevertheless, the number of studies tackling this issue is low, and the heterogeneity in the study design and data analysis has hindered consistent findings. In this work, we reviewed the current literature on the evaluation of the role of microbiota in the development of esophageal cancer. We analyzed the composition of the normal microbiota and the alterations found in precursor lesions, namely Barrett’s esophagus and dysplasia, as well as in esophageal cancer. Additionally, we explored how other environmental factors can modify microbiota and contribute to the development of this neoplasia. Finally, we identify critical aspects to be improved in future studies, with the aim of refining the interpretation of the relationship between the microbiome and esophageal cancer.
Content may be subject to copyright.
Citation: Moreira, C.; Figueiredo, C.;
Ferreira, R.M. The Role of the
Microbiota in Esophageal Cancer.
Cancers 2023,15, 2576. https://
doi.org/10.3390/cancers15092576
Academic Editor: B.P.L. (Bas)
Wijnhoven
Received: 13 April 2023
Revised: 28 April 2023
Accepted: 29 April 2023
Published: 30 April 2023
Copyright: © 2023 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
cancers
Review
The Role of the Microbiota in Esophageal Cancer
Clara Moreira 1,2, Ceu Figueiredo 1,2,3 and Rui Manuel Ferreira 1, 3, *
1Instituto de Investigação e Inovação em Saúde, Universidade do Porto (i3S), 4200-135 Porto, Portugal
2Department of Pathology, Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
3Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP),
4200-135 Porto, Portugal
*Correspondence: ruif@i3s.up.pt
Simple Summary:
Esophageal cancer has very high mortality and morbidity rates. In this study, we
reviewed the current literature on the impact of microbiota on esophageal cancer and its precursor
lesions. Globally, a decrease in microbiota richness and evenness in esophageal cancer is identified,
which is accompanied by an increase in the abundance of Gram-negative bacteria.
Abstract:
Esophageal cancer is a major health problem, being the seventh most incidence cancer
worldwide. Due to the often-late diagnosis and lack of efficient treatments, the overall 5-year
survival is as low as 10%. Therefore, understanding the etiology and the mechanisms that drive
the development of this type of cancer could improve the management of patients, increasing the
chance of achieving a better clinical outcome. Recently, the microbiome has been studied as a
putative etiological factor for esophageal cancer. Nevertheless, the number of studies tackling this
issue is low, and the heterogeneity in the study design and data analysis has hindered consistent
findings. In this work, we reviewed the current literature on the evaluation of the role of microbiota
in the development of esophageal cancer. We analyzed the composition of the normal microbiota
and the alterations found in precursor lesions, namely Barrett’s esophagus and dysplasia, as well
as in esophageal cancer. Additionally, we explored how other environmental factors can modify
microbiota and contribute to the development of this neoplasia. Finally, we identify critical aspects to
be improved in future studies, with the aim of refining the interpretation of the relationship between
the microbiome and esophageal cancer.
Keywords:
microbiota; esophageal cancer; esophageal squamous cell carcinoma; esophageal
adenocarcinoma; dysbiosis; bacteria
1. Introduction
The human microbiome comprises the whole set of microbial taxa and their genomes
that inhabit different niches in the human body, including bacteria, fungi, and viruses.
Usually, the term microbiota refers only to the collection of microorganisms themselves [
1
].
The number of bacteria colonizing the human body is frequently reported as outnumbering
the number of host cells, but updated estimates propose a ratio of 1.3 bacterial cells for every
human cell [
2
]. Although the number and abundance of bacteria are high, the diversity
of the genomic information can be even greater since each species contains thousands
of genes that contribute to a substantially more flexible and diverse metagenome than
the human genome alone [
3
]. The microbiota plays important roles in the maintenance
of human health, including metabolic activities, such as the production of vitamins and
anti-inflammatory compounds, and also contributes contributing to the maturation of the
immune system [4].
In recent years, the implementation of next-generation sequencing techniques has
enabled in-depth characterization of the gastrointestinal microbiota, circumventing the
limitations of the traditional culturing methods that only allowed the identification of
Cancers 2023,15, 2576. https://doi.org/10.3390/cancers15092576 https://www.mdpi.com/journal/cancers
Cancers 2023,15, 2576 2 of 15
cultivable species [
4
]. The study of the microbiota in a specific niche may involve the
identification and quantification of each member of the community and the estimation
of the diversity of the entire community, the latter varying from individual to individual
and between groups of individuals. The composition of the microbial communities can be
affected by diet, exercise, stress, aging, the intake of probiotics/prebiotics, antibiotics, or
other drugs, as well as by the host immune system and host genetic factors [
5
]. Importantly,
alterations in the equilibrium between the human host and the microbiota have been associ-
ated with the development of several diseases, namely cancer [
6
]. In several gastrointestinal
cancers, including the stomach and the colon, dysbiosis has been consistently associated
with cancer [
6
,
7
]. However, the mechanisms by which microbial dysbiosis can promote
cancer are far from understood. One can speculate that the microbiota has direct oncogenic
effects through the release of oncogenic toxins or secretion of oncometabolites, and/or
indirect oncogenic actions, by the promotion and maintenance of local inflammation or by
alteration of the systemic immune response.
Regarding esophageal cancer, the microbiota of the upper gastrointestinal tract has
been suggested to play a role in the etiology of this malignancy, particularly in esophageal
adenocarcinoma. Here, we review and discuss the literature that addresses the role of
microbiota in the development of esophageal cancer. For this purpose, we conducted a
search in the PubMed database for the query “esophageal cancer” AND “microbiota” in
July 2022.
2. Esophageal Cancer
Esophageal cancer occupies the sixth position on the global ranking of cancer-related
death, and it is the seventh most incident malignancy. This type of cancer was responsible
for more than 544,000 deaths worldwide in 2020 [
8
]. Esophageal cancer incidence and
mortality are unequally distributed around the world, being more incident and deadly in
Eastern Asia and Southern Africa than in Western countries, such as France, Canada, and
the USA [9].
The overall 5-year survival rate of esophageal cancer is approximately 10%, and the
5-year post-esophagectomy survival rate is between 15% to 40% [
10
]. Unfortunately, this
disease is diagnosed at an advanced stage due to the lack of clinical symptoms, which
contributes to the high mortality rate [
10
]. Since there are a small number of targeted
therapies for esophageal cancer, the main treatment options are surgery, chemotherapy, and
radiotherapy, as well as endoscopic therapies, such as radiofrequency ablation, endoscopic
mucosal resection, and endoscopic submucosal dissection [11].
The great majority of esophageal cancers are esophageal squamous cell carcinomas
(ESCC) or esophageal adenocarcinomas (EAC). ESCC is the most common type, repre-
senting 90% of esophageal cancers, having higher incidences in Asian and Eastern coun-
tries [
12
,
13
]. ESCC predominates in the upper and mid-thirds of the esophagus. It is
thought to develop through the progression of premalignant precursor lesions of the squa-
mous lining of the esophagus (squamous dysplasia), which result from the presence of
chronic inflammation [
14
,
15
] that causes architectural changes such as disordered cellular
stratification, loss of cellular polarity, and premature keratinization. Several risk factors
have been associated with the development of ESCC, namely smoking, tobacco chewing,
alcohol consumption, intake of hot beverages, and diets with reduced consumption of fresh
fruits and vegetables [1618].
At the molecular level, the ESCC subtype contains frequent mutations in TP53, NFE2L2,
MLL2, ZNF750, NOTCH1, and TGFBR2 genes and is characterized by upregulation of Wnt,
syndecan, and p63 pathways, which are associated with the development and differen-
tiation of squamous epithelial cells [
19
]. Epidermal growth factor receptor (EGFR) and
receptor tyrosine kinase, or RAS signaling alterations, have been associated with ESCC and
with a worse prognosis. In addition, vascular endothelial growth factor (VEGF) signaling
pathway activation was also linked to ESCC and correlated with higher tumor stages and
lymph node metastasis [10].
Cancers 2023,15, 2576 3 of 15
Although in Western countries, the incidence of EAC is typically low, EAC has been
rapidly increasing, and in countries such as the United Kingdom, the Netherlands, the
United States, Denmark, Canada, and Sweden, it has already overtaken ESCC [
20
,
21
]. The
male sex, the Caucasian race, tobacco smoking, and obesity are the main risk factors for
EAC. Helicobacter pylori infection in the stomach appears to be inversely associated with
the incidence of EAC [
22
,
23
]. EAC occurs predominantly in the lower esophagus near the
gastric junction and develops in the context of Barrett’s esophagus [
24
]. This condition in
the esophagus arises from gastroesophageal reflux disease, which is a chronic digestive
condition in which acidic contents from the stomach, frequently mixed with duodenal bile,
enter the esophagus leading to esophageal tissue injury. At the cellular level, progression to
EAC is underlined by continuous DNA damage caused by reflux and chronic inflammatory
factors, which likely increase the mutation rate and promote genomic instability. TP53 is the
most frequently mutated gene in EAC tumors. A high frequency of mutations is also found
in CDKN2, which is known to regulate the cell cycle [
20
]. These molecular alterations are
consistent with the predominance of CDKN2A and TP53 mutations in dysplastic Barrett’s
esophagus, a precursor lesion of EAC. In addition, in EAC, mutations are frequently present
in ARID1A,SMAD4, and ERBB2 genes. Increased E-cadherin signaling and overexpression
of FOXA and ARF6, which regulate E-cadherin during the endocytic pathway, may also be
present in EAC [25].
3. Esophagus Microbiota in Healthy Conditions
The analysis of the bacterial community in the normal esophagus has been explored
in a small number of studies (Table 1), probably due to difficulties in the recruitment of
healthy volunteers for invasive sampling by upper endoscopy.
Table 1. Summary of the studies evaluating the esophageal microbiota in healthy individuals.
Reference Country No. Participants Specimen and Measurement Taxonomic Findings in Healthy Individuals *
Vuik et al. 2019 [26] The Netherlands HC (13)
RE (1)
Biopsy specimens
16S rRNA gene sequencing
Veillonellacea (F)
Psudomonadaceae (F)
Streptoccaceae (F)
Peter et al. 2020 [27] USA
HC (10)
IM (10)
HGD (10)
LGD (10)
EAC (12)
Biopsy specimens
16S rRNA gene sequencing
At the phylum-level:
Firmicutes
Proteobacteria
Bacteriodetes
Actinobacteria
Fusobacteria
At the genus-level:
Tissierella
Streptococcus
Lactobacillus
Acinetobacter
Prevotela
Fusobacterium
Staphylococcus
Akkermansia
Blautia
Yin et al. 2020 [28] China HC (27) Esophageal brush specimens
16S rRNA gene sequencing
At the phylum-level:
Proteobacteria
Firmicutes
Bacteroidetes
Actinobacteria
Fusobacteria
TM7
At the genus-level:
Streptococcus
Actinobacillus
Sphingomonas
unclassified Enterobacteriaceae
Neisseria
Haemophilus
Prevotella
Veillonella
Porphyromonas
Cancers 2023,15, 2576 4 of 15
Table 1. Cont.
Reference Country No. Participants Specimen and Measurement Taxonomic Findings in Healthy Individuals *
Li et al. 2020 [29] China
HC (16)
ESCC (17)
EGJ (11)
Esophageal brush specimens
16S rRNA gene sequencing
At the phylum-level:
Proteobacteria
Firmicutes
Bacteroidetes
Actinobacteria
At the genus-level:
Streptococcus
Ralstonia
Burkholderia–Caballeronia–Paraburkholderia
Fusobacterium
Li et al. 2021 [30] China
HC (82)
LGD (60)
HGD (64)
ESCC (70)
Esophageal brush specimens
16S rRNA gene sequencing
At the phylum-level:
Firmicutes
Proteobacteria
Bacteroidetes
Actinobacteria
Fusobacteria
At the genus-level:
Streptococcus
Neisseria
Haemophilus
Prevotella
EAC—esophageal adenocarcinoma; EGJ—esophagogastric junction cancer, F—family; HC—Healthy controls;
HGD—high-grade dysplasia; IM—Intestinal metaplasia, LGD—low-grade dysplasia; RE—reflux esophagitis.
* When data are available, taxa are displayed in descending order of abundance.
A Dutch study analyzed the mucosal microbiota composition along the entire gastroin-
testinal tract to find that the esophageal microbiota closely resembles the stomach micro-
biota rather than other communities in the gastrointestinal tract [
26
]. Proteobacteria were
the most abundant phylum in the esophagus, followed by Firmicutes and Bacteroidetes [
26
].
Accordingly, the families Veillonellaceae, Streptococcaceae of the phylum Firmicutes, and
Pseudomonadaceae of the phylum Proteobacteria were identified as the most abundant in
the distal esophagus [
26
]. Subsequent studies that characterized the esophageal microbiota
in biopsy specimens of American individuals and in mucosal brushings of Chinese subjects
confirmed the high abundance of Firmicutes, followed by Proteobacteria, Bacteroidetes,
Actinobacteria, and Fusobacteria [
27
,
28
,
30
]. Analysis of the esophageal mucosa microbiota
of American individuals revealed that the top-5 most abundant genera were Tissierella,
Lactobacillus,Streptococcus,Acinetobacter, and Prevotella [
27
]. Streptococcus,Actinobacillus,
Sphingomonas,Enterobacteriaceae,Neisseria,Haemophilus,Prevotella, Veillonella, and Porphyromonas
were identified as the most abundant members of the esophageal microbial community in a Chi-
nese study that characterized the microbiota in brush specimens from 27 healthy individuals [
28
].
Of note, all the studies that profiled the microbiome at the genus level consistently identified
the genus Streptococcus as one of the most abundant genera in the normal esophagus [
28
30
].
Despite the effort to characterize the normal esophagus microbiota, most publications
included a low number of individuals and presented limitations regarding sequencing and
data analysis sensitivity.
4. Modulators of the Esophageal Microbiota and of the Esophageal Cancer Risk
Several factors can disturb the esophageal microbiota, including alcohol consumption,
dietary habits, smoking, obesity, and the intake of drugs, such as antibiotics or proton-
pump inhibitors (PPIs) [
31
,
32
]. These elements are also important risk factors for the
development of esophageal cancer, despite the fact that their association is restricted to
specific cancer subtypes. For example, the intake of alcohol has been consistently associated
with ESCC, but its association with EAC is unclear [
33
]. Regarding the esophageal micro-
biota, a recent study that analyzed 120 patients with ESCC, 60 of which were drinkers and
60 were non-drinkers, found that alcohol consumption was associated with alterations
in the diversity and composition of the esophageal microbiota in ESCC patients. The
microbiota of drinkers showed significantly lower alpha-diversity (within sample diversity)
than that of non-drinkers, and their microbial profiles could be distinguished. Drinkers
showed an enrichment of the order Pasteurellales, particularly the family Pasteurellaceae.
Cancers 2023,15, 2576 5 of 15
In contrast, non-drinkers had a higher abundance of the class Clostridia, with specific en-
richment of the bacterial families Clostridiaceae, Lanchnospiraceae, and Helicobacteraceae,
and of the genera Clostridium,Helicobacter, and Catonella [34].
Diet can rapidly alter the structure and activity of the human gut microbiome [
35
].
Diets rich in lipids and sugars result in an increased body mass index and obesity, which
are strongly associated with an increased risk of EAC [
36
]. Whether obesity modulates
the human esophageal microbiota has not yet been addressed, but a study conducted in a
mouse model suggests that high-fat diets influence the gut microbiota, contributing to the
development of esophageal cancer [
37
]. Accordingly, the study reported that mice fed with
a high-fat diet developed esophageal dysplasia and tumors more rapidly than those fed
with a control diet. The high-fat diet resulted in an increased ratio of neutrophils to natural
killer cells, which favors cancer development. Moreover, a high-fat diet led to a shift in
the gut microbiota in comparison with a control diet. Analysis of microbial beta-diversity
(between sample diversity) showed separation between mice fed with a high-fat diet and
mice fed with the control diet. This differentiation was characterized by an altered ratio of
Firmicutes to Bacteroidetes.
The exposure of fecal microbiota from mice fed the high-fat diet to mice fed the control
diet accelerated tumor development in the esophagus. The relevance of the microbiota in
the development of esophageal cancer was further demonstrated in animals raised in germ-
free housing, which developed less dysplasia than mice grown under specific pathogen-free
conditions [
37
]. In contrast to high-fat diets, the consumption of dietary fibers can modu-
late the microbial composition of the esophagus and have a protective role in esophageal
neoplasia. A study comprising 47 patients demonstrated that fiber intake was positively
associated with the relative abundance of Firmicutes [
38
]. In contrast, fiber intake was
inversely associated with the relative abundance of Gram-negative Proteobacteria, which
can be found in the abnormal esophagus, including reflux esophagitis and Barrett’s esopha-
gus [
39
41
]. Altogether, these results are in line with those observed in the colon cancer
model, where changes in the dietary content of fiber and fat have an impact on the colonic
microbiome and metabolome, resulting in significant changes in mucosal inflammation
and proliferation, thereby increasing colon cancer risk [
42
]. Smoking is a well-known risk
factor for the development of cancer, but it can also shape the esophageal microbial commu-
nities through chemicals, heavy metals, and other constituents of tobacco [
43
]. A study of
278 male Chinese participants argued that current smokers tended to have increased micro-
bial alpha-diversity compared to never smokers. The same study identified the enrichment
of two anaerobes, Dialister invisus, and Megasphaera micronuciformis, in current smokers in
comparison with never smokers [44].
PPIs can also modulate microbiota. These drugs are used to relieve symptoms caused
by excessive acid production in the stomach, such as heartburn or gastroesophageal reflux
disease. Despite anti-acid drugs targeting the acid-producing glands of the stomach, they
can affect not only the gastric bacterial community but also the microbiota of the esoph-
agus and the intestine [
45
]. It has been proposed that anti-acid drugs directly target the
proton pumps containing P-type ATPase enzymes of specific bacteria, such as Streptococcus
pneumonia, or indirectly increase the pH to levels that cause the death of certain bacterial
species [
46
]. An investigation comparing the microbiota before and after PPI treatment
in 16 patients with heartburn and normal-appearing mucosa and in 29 patients with ab-
normal mucosa due to esophagitis and Barrett’s esophagus found significantly different
microbial profiles [
47
]. The treatment with PPIs was associated with increased abundance
of the Comamonadaceae family and decreased abundance of the families Clostridiaceae,
Lachnospiraceae, and two unclassified families of the orders Clostridia, Lactobacillales,
and Gemelalles [47].
Antibiotics have been used to treat bacterial infections, but they can also disturb the
resident bacteria. Despite this negative effect, in the context of cancer, specific antibiotics
have been used to eradicate opportunistic microbes in cancer patients, resulting in poten-
tially beneficial clinical outcomes [
48
]. Whether antibiotics have a protective effect against
Cancers 2023,15, 2576 6 of 15
the development of esophageal cancer is poorly explored. In fact, one study using a rat
mouse model showed a slight but non-significant reduction in the incidence of esophageal
adenocarcinoma in animals treated with penicillin G and streptomycin in comparison
with the control group [
49
]. This reduction was accompanied by an increased proportion
of Clostridium clusters XIVa and XVIII and a decreased proportion of the Lactobacillales
order [49].
5. Alterations in the Microbiota of Esophageal Premalignant Lesions
The role of the esophageal microbiota in the transition of normal epithelium to prema-
lignant lesions and in the progression towards esophageal cancer is mostly unknown. So
far, only a few studies have addressed this issue, and most of them presented limitations
in sensitivity and depth of coverage, impairing the ability to draw clear and statistically
based conclusions (Table 2). One of the first studies used an oral microbe identification
microarray to characterize the presence of 272 microbial species in 142 Chinese patients
with esophageal dysplasia and in 191 patients without esophageal dysplasia [
50
]. The
study revealed an association between microbial richness and esophageal squamous dys-
plasia [
50
]. These results were confirmed in a more recent study that profiled the esophageal
microbiota in 227 patients with normal esophageal function, low-grade dysplasia, high-
grade dysplasia, and ESCC [
30
]. Disease progression was associated with decreased
alpha-diversity and with a small but significant beta-diversity. Comparing the microbiota
of dysplastic lesions with that of the normal esophagus, the genera Atopobium,Enterococcus,
Granulicatella,Lachnoanaerobaculum,Rothia, Solobacterium, and Streptococcus were found to
be enriched in low-grade dysplasia, while the genera Bacillus,Lactobacillus and Streptococcus
were identified as markers of high-grade dysplasia [30].
Table 2. Studies characterizing the esophageal microbiota in esophageal premalignant lesions.
Reference Country (N) Specimen and Measurement Diversity Taxonomic Findings
Alpha Beta
Macfarlane et al.
2007 [51]
USA
No BE (7)
BE (7)
Biopsy; Aspirate
16S rRNA gene sequencing
BE NS
Veillonella (G)
Neisseria (G)
Camplylobacter (G)
Fusobacterium (G)
Megasphaera (G)
Staphylococcus (G)
Lactobacillus (G)
Yang et al.
2009 [40]
USA
HC (12)
ES (12)
BE (10)
Biopsy
16S rRNA gene sequencing
Type II compared to
type I
Distinguished
Type I and Type
II microbiome
Type I microbiome:
Normal esophagus
Type II microbiome: Abnormal
esophagus
Type II vs. Type I:
Gram-negative bacteria
Veillonella (G)
Prevotella (G)
Haemophilus (G)
Neisseria (G)
Rothia (G)
Granulicatella (G)
Campylobacter (G)
Porphyromonas (G)
Fusobacterium (G)
Actinomyces (G)
Firmicutes (P)
Streptoccocus (G)
Liu et al.
2013 [52]
Japan
HC (6)
ES (6)
BE (6)
Biopsy
16S rRNA gene sequencing
NS NS
ES/HC vs. BE:
Streptococcus (G)
ES/BE vs. HC:
Veillonella (G)
Neisseria (G)
Fusobacterium (G)
Yu et al.
2014 [50]
China
No ESD (191)
ESD (142)
Brush
Human oral microbe
identification microarray
ESD Distinguished
ESD and No ESD
Inverse association
between microbial richness
and ESD when comparing with
non ESD
Cancers 2023,15, 2576 7 of 15
Table 2. Cont.
Reference Country (N) Specimen and Measurement Diversity Taxonomic Findings
Alpha Beta
Elliot et al.
2017 [24]
UK
HC (20)
NDBE (24)
BE (23)
EAC (19)
Biopsy, Brushing, Cytosponge
16S rRNA gene sequencing
NS
Distinguished
HC
and BE
Proteobacteria (P)
Deshpande et al. 2018
[53]
Australia
HC (59)
GERD (29)
GM (7)
BE (5)
EAC (1)
EoE (1)
Esophageal brush specimens
16S rRNA gene sequencing
Whole metagenome
sequencing
NS NS
GERD vs. HC:
Flavobacteriaceae (F)
Acetoanaerobium (G)
Filifactor (G)
Campylobacter (G)
Prevotella intermedia (S)
Prevotella micans (S)
Neisseria macacae (S)
Neisseria meningitidis (S)
Haemophilus parainfluenzae (S)
Treponema medium (S)
BE vs. HC:
Leptotrichia (G)
Capnocytophaga (G)
Gemella (G)
Veillonella (G)
Streptococcus sanguinis (S)
Snider et al.
2019 [54]
USA
HC (16)
NDBE (14)
LGD (6)
HGD (5)
EAC (4)
Brush
16S rRNA gene sequencing
NS NS
HGD vs. LGD:
Proteobacteria (P)
Firmicutes (P)
Veillonella (G)
Lopetuso et al.
2020 [55]
Italy
HC (10)
BE (10)
BEU (10)
EAC (6)
Biopsy
16S rRNA gene sequencing
BE Distinguished
HC and BE
BE vs. HC:
Fusobacteria (P)
Leptotrichia (G)
BE vs. BEU:
Bacteroidetes (P)
TM7 (P)
Prevotellaceae (F)
Veillonellaceae (F)
Fusobacteriaceae (F)
Lachnospiraceae (F)
Campylobacteraceae (F)
Prevotella (G)
Fusobacterium (G)
Campylobacter (G)
Selenomonas (G)
Li et al.
2021 [30]
China
HC (82)
LGD (60)
HGD (64)
ESCC (70)
Brush
16S rRNA gene sequencing
HGD compared to
HC/LGD
Distinguished
HC/LGD group
and HGD
LGD vs. HC:
Atopobium (G)
Enterococcus (G)
Granulicatella (G)
Lachnnoanaerobaculum (G)
Rothia (G)
Solobacterium (G)
Streptococcus (G)
HGD vs. HC:
Bacillus (G)
Lactobacillus (G)
Streptococcus (G)
AHT—adjacent mucosa healthy tissue; BE—Barrett’s esophagus; BEU—unaffected esophageal mucosa of Barrett’s
esophagus patient; EAC—esophageal adenocarcinoma; ES—esophagitis; ESD—esophageal squamous dysplasia;
GERD—gastroesophageal reflux disease; GM—glandular mucosa; EoE—Eosinophilic esophagitis; HC—healthy
control, HGD—High grade dysplasia; LGD—Low grade dysplasia, NDBE—non-dysplastic Barrett’s esophagus;
NS—not significant; P—phylum; F—family; G—genus; S—species; —increase; —decrease.
Initial analyses of Barrett’s esophagus microbiota revealed that the esophageal mi-
crobiota could be categorized into two main types [
40
]. Type I was dominated by Gram-
positive Streptococcus sp. and comprised cases with a morphologically normal esophagus.
Conversely, type II was characterized by a high abundance of Gram-negative anaerobes
and microaerophiles and was associated with esophagitis and Barrett’s esophagus [
40
].
Comparing the microbial community of Barrett’s esophagus with that of healthy controls,
Lopetuso et al. identified only minor taxonomic differences, including the increase in the
Cancers 2023,15, 2576 8 of 15
relative abundance of Fusobacteria at the phylum level and of Leptotrichia genus. How-
ever, this study noted a decrease in the abundance of the genera Prevotella,Fusobacterium,
Campylobacter, and Selenomonas when metaplastic mucosa of Barrett’s esophagus was com-
pared with adjacent normal areas of Barrett’s esophagus [
55
]. A larger case-control study
reported marginal differences between the microbiota of Barrett’s esophagus and the mi-
crobial community of EAC. This study pointed to a significantly higher number of taxa
but not to an overall difference in the microbial alpha-diversity of Barrett’s esophagus
compared to that of EAC [24]. Moreover, Barrett’s lesions contained a significantly higher
abundance of Proteobacteria than that found in EAC and in normal controls [
24
]. A study
that combined 16S rRNA gene sequencing with whole metagenome sequencing identified
10 microbial taxa in gastroesophageal reflux disease patients in comparison with healthy
controls, including Prevotella intermedia,Prevotella micans,Neisseria macacae,Neisseria meningitidis,
Haemophilus parainfluenzae, and Treponema medium. The comparison of Barrett’s esophagus
patients with controls revealed an increase in abundance of Leptotrichia,Capnocytophaga,
Gemella,Veillonella, and Streptococcus sanguinis. The study pointed that distinct microbial
community types are mainly defined by the relative abundance of the genera Streptococcus
and Prevotella [53].
A study that aimed to reveal the potential role of the microbiota in the development of
esophageal cancer profiled the microbial community of 45 patients, following the progres-
sion from Barrett’s esophagus to cancer [
54
]. The study reported a shift in the microbial
composition of Barrett’s esophagus at the transition from low- to high-grade dysplasia.
This was characterized by decreased abundance of Firmicutes, namely of Veillonella sp., an
increased abundance of Proteobacteria of the Enterobacteriaceae family, and an increased
proportion of samples with Akkermansia muciniphila in high-grade dysplasia.
6. The Esophageal Microbiota in Esophageal Cancer
Considering that esophageal cancer develops in the context of chronic inflammation,
which is influenced by genetic and environmental factors, the esophageal microbiota is a
possible etiological factor of esophageal cancer (Table 3).
Table 3. Summary of the studies evaluating esophageal microbiota in esophageal cancer.
Reference Country (N) Specimen and Measurement Diversity Taxonomic Findings
Alpha Beta
Elliott et al.
2017 [24] *
UK
HC (20)
NDBE (24)
BE (23)
EAC (19)
Biopsy, Brushing, Cytosponge
16S rRNA gene sequencing
EAC in
comparison with HC
Distinguished EAC
and HC
EAC vs. HC:
Lactobacillales (O)
Coriobacteriacea (F)
Lactobacillaceae (F)
Streptococcus (G)
Lactobacillus (G)
Shao et al.
2019 [56] **
China
ESCC (67)
NAM (67)
Biopsy
16S rRNA gene sequencing
NS Distinguished ESCC
and NAM
ESCC vs. NAM:
Fusobacteria (P)
Firmicutes (P)
Fusobacterium (G)
Streptococcus (G)
Snider et al.
2019 [54] *
USA
HC (16)
NDBE (14)
LGD (6)
HGD (5)
EAC (4)
Brush
16S rRNA gene sequencing
EAC in
comparison with
NDBE, LGD
and HGD
NS
HGD/EAC vs. NDBE/LGD:
Firmicutes (P)
Proteobacteria (P)
Enterobacteriacea (F)
Akkermansia muciniphila (S)
Veillonella (G)
Lopetuso et al.
2020 [55] *
Italy
HC (10)
BE (10)
EAC (6)
Biopsy
16S rRNA gene sequencing
EAC in
comparison with HC
Distinguished EAC
and HC
EAC vs. HC:
Prevotella (G)
Leptotrichia (G)
Veillonella (G)
Bacteroidetes (G)
Streptococcus (G)
Granulicatella (G)
Cancers 2023,15, 2576 9 of 15
Table 3. Cont.
Reference Country (N) Specimen and Measurement Diversity Taxonomic Findings
Alpha Beta
Li et al.
2020 [29] *
China
HC (16)
RE (15)
EGJ (11)
ESCC (17)
Brush
16S rRNA gene sequencing
ESCC and EGJ
in comparison
with HC
Distinguished ESCC
and HC
ESCC vs. HC:
Fusobacteria (P)
Actinobacteria (P)
Peter et al.
2020 [27] *
USA
IM (10)
LGD (10)
HGD (10)
EAC (12)
HC (10)
Biopsy
16S rRNA gene sequencing
NS NS
EAC vs. HC:
Planctomycetes (P)
Crenachaeota (P)
Siphonobacter (G)
Nitrosopumilus (G)
Planctomyces (G)
Li et al.
2021 [30] *
China
HC (82)
LGD (60)
HGD (64)
ESCC (70)
Brush
16S rRNA gene sequencing
ESCC in
comparison with HC
Distinguished ESCC
and HC
ESCC vs. HC:
Peptoniphilus (G)
Petostreptococcus (G)
Lachnospiraceae_[G9] (G)
Bosea (G)
Gemella (G)
Solabacterium (G)
Streptococcus (G)
Li et al.
2021 [57] **
China
ESCC (41)
NAM (41)
Biopsy
16S rRNA gene sequencing
ESCC in
comparison
with NAM
Distinguished ESCC
and NAM
ESCC vs. NAM:
Bacteroidetes (P)
Fusobacteria (P)
Spirochaetae (P)
Proteobacteria (P)
Firmicutes (P)
Actinobacteria (P)
Streptococcus (G)
Fusobacterium (G)
Prevotella (G)
Butyrivibrio (G)
Lactobacillus (G)
Jiang et al.
2021 [15] *
China
HC (21)
ES (15)
ESCC (32)
Biopsy
16S rRNA gene sequencing
ESCC in
comparison with HC
Distinguished ESCC
and HC
ESCC vs. HC:
Fusobacteria (P)
Streptococcus (G)
Actinobacillus (G)
Peptostreptococcus (G)
Fusobacterium (G)
Prevotella (G)
Bacteroides (G)
Faecalibacterium (G)
Curvibacter (G)
Blautia (G)
Shen
2022 et al. [58] **
China
ESCC (51)
NAM (51)
Biopsy
16S rRNA gene sequencing
Species-specific qPCR
NS NS
ESCC vs. NAM:
Deinococcus-Thermus (P)
Spirochaetes (P)
Tenericutes (P)
Actinobacteria (P)
Verrumicrobia (P)
Lentimicrobiaceae (F)
Treponema (G)
Selenomonas (G)
Peptonaerobacter (G)
Methylobacter (G)
Akkermansia (G)
Blautia (G)
Labrys ginsengisoli (S)
Peptoanaerobacter stomatis (S)
Selenomonas sputigena (S)
Streptococcus constellatus (S)
Fusobacterium periodonticum (S)
Lactobacillus murinus (S)
Escherichia coli (S)
* Studies comparing esophageal cancer with healthy subjects. ** Studies comparing paired normal adjacent mucosa
with tumor tissue. BE—Barrett’s esophagus; C—class; EAC—esophageal adenocarcinoma; EC—esophageal cancer;
EGJ—esophagogastric junction cancer; ES—esophagitis; ESCC—esophageal squamous cell carcinoma; ESD—
esophageal squamous dysplasia; F—family; G—genus; HC—healthy controls; HGD—high grade dysplasia;
IM—intestinal metaplasia; LGD—low grade dysplasia; MEE—mid-esophageal esophagitis; NAM—normal
adjacent mucosa; NDBE—non-dysplastic Barrett’s esophagus; NS—no significant difference; RE—after radical
esophagectomy; P—phylum; O—order; F—family; S—species; —increase; —decrease.
Cancers 2023,15, 2576 10 of 15
In comparison with healthy controls, the microbial community of ESCC is characterized
by reduced bacterial diversity [
15
,
29
,
30
] and by the enrichment of several genera, including
Streptococcus,Peptostreptococcus,Prevotella,Fusobacterium,Actinobacillus,Gemella,Rothia, and
Prevotella [
15
,
29
,
30
]. However, the analysis of the microbial community of tumor tissues
and paired adjacent normal tissues did not identify consistent significant differences in the
bacterial richness and diversity [
56
,
57
] but revealed a significant enrichment of Fusobacterium
and Streptococcus and a decrease in the abundance of Lactobacillus in the ESCC tissue [
57
59
].
Interestingly, one study that included patients with ESCC and combined several genomics
methods showed that the abundance of Fusobacterium nucleatum was significantly associated
with the tumor tissue and not with the normal adjacent epithelium. This study pointed
out that F. nucleatum was closely related to increased tumor staging and to the presence of
mutations in genes such as TP53,COL22A1,TRBV10–1, CSMD3,SCN7A, and PSG11 [57].
The microbial community of EAC is less characterized than ESCC. In fact, so far,
only four studies evaluated the microbiota in EAC, with inconsistent results, proba-
bly due to differences in methodology, study design, and the limited number of cases
included [
24
,
27
,
54
,
55
]. A study that performed a microbiome survey in only four patients
with EAC, 25 with Barrett’s esophagus, and 16 healthy controls showed a decrease in
the alpha-diversity in cases with non-dysplastic Barrett’s esophagus and EAC [
54
]. This
study identified the enrichment of Enterobacteriaceae and Akkermansia municiphila and the
decrease in Veillonella in cancer cases [
54
]. Another study that compared six EAC patients
with 10 healthy controls identified a decrease in the microbial alpha-diversity in cancer
in comparison with healthy controls, which was accompanied by a significant increase
in the abundance of Prevotella sp., Veillonella sp., and Leptotrichia sp. [
55
]. On the other
hand, Peter et al. identified in the EAC mucosa a significant decrease in the abundance of
Siphonobacter,Nitrosopumilus, and Planctomyces in comparison tissue of healthy controls [
27
].
A larger study that compared the mucosal microbiota of 19 EAC with 20 healthy controls
detected a significant increase in the abundance of Streptococcus and Lactobacillus in cancer
cases [
24
]. This study suggested that Lactobacillus sp. dominate the niche of EAC due to
their capability to resist low pH and to inhibit the growth of competitor bacteria through
the production of lactic acid [24].
7. Non-Esophageal Microbiota and Esophageal Cancer
In recent years, several studies reported associations between the oral or the gut mi-
crobiota and increased risk of esophageal cancer. Given the invasive nature of sampling the
esophagus through upper endoscopy for microbiome analysis, access to non-invasive oral
or fecal specimens can be viewed as an opportunity to easily detect microbial biomarkers
of esophageal cancer (Table 4).
Table 4. Studies show relationships between non-esophageal microbiota and esophageal cancer.
Reference Country (N) Specimen and Measurement Diversity Taxonomic Findings
Alpha Beta
Peters et al.
2017 [60]
USA
HC (210)
ESCC (25)
EAC (81)
Mouthwash
16S rRNA gene sequencing
NS NS
EAC:
Selenomonas (G)
Veillonella (G)
Tannerella forsythia (S)
Actinomyces cardiffensis (S)
ESCC:
Bergeyella (G)
Porphyromonas gingivales (S)
Prevotella nanceiensis (S)
Neisseria weaver (S)
Treponema vincentii (S)
Wang et al.
2019 [61]
China
HC (21)
ESCC (20)
Saliva
16S rRNA gene sequencing
NS Distinguished HC
and ESCC
ESCC vs. HC:
Firmicutes (P)
Gammaproteobacteria (C)
Bacillus (G)
Lactobacillus (G)
Cancers 2023,15, 2576 11 of 15
Table 4. Cont.
Reference Country (N) Specimen and Measurement Diversity Taxonomic Findings
Alpha Beta
Ishaq et al.
2021 [62]
China
HC (10)
EC (15)
Biopsy
DGGE
16S rRNA gene sequencing
EC Distinguished HC
and EC
EC vs. HC:
Bacteroidetes (P)
Bacteroidaceae (F)
Enterobacteriaceae (F)
Bacteroides (G)
Escherichia-Shigella (G)
Prevotellaceae (F)
Veillonellaceae (F)
Prevotella (G)
Dialister (G)
Chen et al.
2021 [6]
Taiwan
HC (18)
ESCC (34)
Oral biofilm; Biopsy
P. gingivalis qPCR;
16S rRNA gene sequencing
ESCC Distinguished HC
and ESCC
ESCC vs. HC:
Oral biofilm:
Streptococcus (G)
Veillonella (G)
Porphyromonas gingivalis (S)
Biopsy:
P. gingivalis (S)
Zhao et al.
2020 [63]
China
HC (51)
EC (49)
Saliva
16S rRNA gene sequencing
NS Distinguished HC
and EC
EC vs. HC:
Firmicutes (P)
Proteobacteria (P)
Negativicutes (C)
Betaproteobacteria (C)
Selenomonadales (O)
Neisseriales (O)
Veillonellaceae (F)
Prevotellaceae (F)
Neisseriaceae (F)
Prevotella (G)
Neisseria (G)
Deng et al.
2021 [64]
China
HC (23)
EC (23)
Stool
16S rRNA gene sequencing
richness in EC Distinguished HC
and EC
EC vs. HC:
Bacteroides (P)
Streptococcus (G)
Bifidobacterium (G)
Subdoligranulum (G)
Blautia (G)
Romboutsia (G)
Collinsella (G)
Paeniclostridium (G)
Dorea (G)
Atopobium (G)
Lachnospira (G)
Bacteroides (G)
Agathobacter (G)
Lachnoclostridium (G)
Parabacteroides (G)
Paraprevotella (G)
Butyricicoccus (G)
Tyzzerella (G)
Fusicatenibacter (G)
Sutterella (G)
Wu et al.
2022 [65]
China
HC (40)
EC (40)
Stool
Culture
EC vs. HC:
Enterococcus (G)
Escherichia coli (S)
Bifidobacterium (G)
Lactobacillus (G)
C—class; DS—dysplasia; EAC—esophageal adenocarcinoma; EC—esophageal cancer; ESCC—esophageal squa-
mous cell carcinoma; F—family; G—genus; NS—no significant difference; O—order; P—phylum,
—increase;
—decrease.
Comparing the salivary bacterial community of 34 ESCC patients and 18 healthy
controls from China, Chen et al. showed an overall decrease in alpha-diversity and clear
separation of patient groups in beta-diversity matrices [
6
]. In accordance with these data,
a Taiwanese study reported a decrease in bacterial richness in the saliva of patients with
ESCC [
6
]. In contrast, two studies that also included Chinese patients did not find significant
differences in the oral microbial alpha-diversity between ESCC and healthy controls [
61
,
63
].
When evaluating taxonomic differences, the relative abundance of Prevotella,Streptococcus,
Porphyromonas [
6
], and Neisseria [
63
] was significantly higher in patients than in controls.
In another study that examined the oral microbiome of 25 ESCC, 81 EAC patients, and
50 controls from the United States, Peters et al. did not find significant differences in the
Cancers 2023,15, 2576 12 of 15
alpha-diversity [
60
]. The same study reported an association between the presence of
Porphyromonas gingivalis, a pathogen associated with periodontal disease, and an increased
risk of ESCC [
60
]. Moreover, the presence of the pathogen Tannerella forsythia and the enrich-
ment of Actinomyces cardiffensis,Selenomonas, and Veillonella were significantly associated
with an increased risk of EAC [
60
]. Zhao et al. compared the microbiota of 49 esophageal
cancers with 51 healthy controls and detected higher abundances of Prevotella nanceiensis,
Neisseria weaveri, and Treponema vincentii in cancer cases [63].
The relationship between the gut microbiome and esophageal cancer has been poorly
explored [
62
,
64
]. In a case-control study that characterized the microbiome in fecal samples
from 15 patients with esophageal cancer and 10 healthy controls, significant alterations
in the microbiota were identified, which are compatible with gut microbial dysbiosis in
cancer patients [
62
]. This study detected increased diversity in the fecal microbiota of
cancer patients in comparison with that of healthy controls. Moreover, significant en-
richment in the abundances of Bacteroidetes, Bacteroidaceae, Enterobacteriaceae, and of
Escherichia-Shigella, was identified in esophageal cancer patients [
62
]. In another study that
included 23 esophageal cancers and 23 healthy controls, a significant increase in microbial
richness was detected in the gut microbiota of cancer patients [
64
]. Analysis of the microbial
profile by hierarchical clustering revealed a clear separation of the microbial community be-
tween esophageal cancer patients and controls. These differences were mainly attributed to
the enrichment in cancer patients of several genera, including Streptococcus,Bifidobacterium,
Subdoligranulum,Blautia,Romboutsia,Collinsella,Paeniclostridium,Dorea, and Atopobium [
64
].
8. Conclusions and Future Directions
Esophageal cancer is a very relevant pathology both in terms of mortality and morbidity.
It is now accepted that the esophageal microbiome is altered in esophageal carcinogenesis.
Some of the classical risk factors associated with esophageal cancer appear to be
associated with changes in the normal microbiota of the esophagus. Alcohol consumption
has been linked with alterations of the diversity of the microbiota in ESCC patients. Diets
rich in high-fat contents in animal studies have been linked with esophageal dysplasia and
alterations of the microbiota. The low intake of fibers was also associated with an increase
in Gram-negative species, which can be found in esophageal cancer precursor lesions.
Smoking, and drugs such as PPIs and antibiotics, have been associated to changes in the
esophageal bacterial population. Overall, the microbiota of the normal esophagus is mainly
composed of Firmicutes, Proteobacteria, Bacteriodetes, Actinobacteria, and Fusobacteria. In
contrast, esophageal cancer is characterized by reduced microbial diversity, which appears
to begin in the precursor stages of esophageal cancer. At the taxonomic level, the esophageal
cancer microbiota is characterized by a shift from Gram-positive to Gram-negative bacteria.
The genera most commonly enriched in esophageal cancer are Fusobacterium,Streptococcus,
Veilonella, and Prevotella.
Even though one can pinpoint specific alterations in the esophageal cancer microbiota,
a consistent esophageal cancer-associated microbiota profile has not yet been identified.
This lack of consistency can be due to the different technical approaches, such as the
sampling method, the type of samples analyzed, and the analytic strategy used to profile
the microbiome, as well as to the relatively small number of individuals included in each
study. Future studies should consider the standardization of the methods, namely DNA
isolation, 16S rRNA amplification and sequencing, and data analysis, which will positively
impact the reliability of the results. In addition, the inclusion of blanks to control potential
microbial contaminations, which can arise during sample processing, will improve the
quality of the final datasets. The use of molecular approaches alternative to 16S rRNA gene
sequencing, such as whole metagenome sequencing, may help disclose in greater detail the
taxonomic profile to the species level and also allow the evaluation of the functional content
of the microbiome. Although these methods have never been applied to the mucosal
esophageal microbiome, they have the potential to identify microbial toxins, virulence
factors, and enzymes that can influence carcinogenesis in the esophagus.
Cancers 2023,15, 2576 13 of 15
Additional relevant aspects need to be considered when addressing the relationship
between the microbiome and esophageal cancer. The inclusion of patients of distinct
geographic origins will be important to control the influences of the host genetic diversity
and distinct cultural lifestyles. Furthermore, the collection of data on dietary habits, use
of medication, and lifestyle factors of each individual will be relevant to control potential
confounding variables in the observed microbiota profiles. In future studies in the setting
of esophageal carcinogenesis associated with the microbiome, multidisciplinary research
efforts will be fundamental to analyze and integrate the effects of multiple exposures,
including those of the diet, with the microbiome and with the changes that occur along
the process of carcinogenesis [
66
]. The dissection of such complex interactions would
benefit from large and well-characterized prospective cohort studies and would certainly
contribute to designing novel prevention and treatment strategies to reduce the burden of
esophageal cancer.
Author Contributions:
Conceptualization: R.M.F.; Literature search and data retrieval: C.M, C.F.
and R.M.F.; Data retrieval and interpretation: C.M., C.F. and R.M.F.; Writing—original draft: C.M.
and R.M.F.; Writing—critical review and editing: all authors; Visualization: all authors; Supervision:
R.M.F. All authors have read and agreed to the published version of the manuscript.
Funding:
R.M.F. has a Fundação para a Ciência e a Tecnologia (FCT) researcher position under the
Individual Call to Scientific Employment Stimulus (CEECIND/01854/2017). The team is also funded
by national funds through FCT (PTDC/BTM-TEC/0367/2021 and 2022.02141.PTDC).
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Neto, A.G.; Whitaker, A.; Pei, Z. Microbiome and potential targets for chemoprevention of esophageal adenocarcinoma. Semin.
Oncol. 2016,43, 86–96. [CrossRef] [PubMed]
2.
Sender, R.; Fuchs, S.; Milo, R. Are We Really Vastly Outnumbered? Revisiting the Ratio of Bacterial to Host Cells in Humans. Cell
2016,164, 337–340. [CrossRef] [PubMed]
3.
Bahram, M.; Netherway, T.; Frioux, C.; Ferretti, P.; Coelho, L.P.; Geisen, S.; Bork, P.; Hildebrand, F. Metagenomic assessment of the
global diversity and distribution of bacteria and fungi. Environ. Microbiol. 2021,23, 316–326. [CrossRef]
4. Gilbert, J.A.; Blaser, M.J.; Caporaso, J.G.; Jansson, J.K.; Lynch, S.V.; Knight, R. Current understanding of the human microbiome.
Nat. Med. 2018,24, 392–400. [CrossRef] [PubMed]
5. Kataoka, K. The intestinal microbiota and its role in human health and disease. J. Med. Investig. 2016,63, 27–37. [CrossRef]
6.
Chen, C.; Chen, L.; Lin, L.; Jin, D.; Du, Y.; Lyu, J. Research progress on gut microbiota in patients with gastric cancer, esophageal
cancer, and small intestine cancer. Appl. Microbiol. Biotechnol. 2021,105, 4415–4425. [CrossRef]
7.
Sun, C.H.; Li, B.B.; Wang, B.; Zhao, J.; Zhang, X.Y.; Li, T.T.; Li, W.B.; Tang, D.; Qiu, M.J.; Wang, X.C.; et al. The role of Fusobacterium
nucleatum in colorectal cancer: From carcinogenesis to clinical management. Chronic Dis. Transl. Med.
2019
,5, 178–187. [CrossRef]
8.
Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN
Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin.
2021
,71, 209–249. [CrossRef]
9.
Global Burden of Disease Cancer, C.; Fitzmaurice, C.; Dicker, D.; Pain, A.; Hamavid, H.; Moradi-Lakeh, M.; MacIntyre, M.F.;
Allen, C.; Hansen, G.; Woodbrook, R.; et al. The Global Burden of Cancer 2013. JAMA Oncol. 2015,1, 505–527. [CrossRef]
10.
Huang, F.L.; Yu, S.J. Esophageal cancer: Risk factors, genetic association, and treatment. Asian J. Surg.
2018
,41, 210–215. [CrossRef]
11. Lagergren, J.; Smyth, E.; Cunningham, D.; Lagergren, P. Oesophageal cancer. Lancet 2017,390, 2383–2396. [CrossRef] [PubMed]
12.
Wang, Q.L.; Xie, S.H.; Wahlin, K.; Lagergren, J. Global time trends in the incidence of esophageal squamous cell carcinoma. Clin.
Epidemiol. 2018,10, 717–728. [CrossRef] [PubMed]
13.
Thrift, A.P.; Whiteman, D.C. The incidence of esophageal adenocarcinoma continues to rise: Analysis of period and birth cohort
effects on recent trends. Ann. Oncol. 2012,23, 3155–3162. [CrossRef]
14.
Jain, S.; Dhingra, S. Pathology of esophageal cancer and Barrett’s esophagus. Ann. Cardiothorac. Surg.
2017
,6, 99–109. [CrossRef]
[PubMed]
15.
Jiang, Z.; Wang, J.; Shen, Z.; Zhang, Z.; Wang, S. Characterization of Esophageal Microbiota in Patients With Esophagitis and
Esophageal Squamous Cell Carcinoma. Front. Cell Infect. Microbiol. 2021,11, 774330. [CrossRef]
16.
Akhtar, S.; Sheikh, A.A.; Qureshi, H.U. Chewing areca nut, betel quid, oral snuff, cigarette smoking and the risk of oesophageal
squamous-cell carcinoma in South Asians: A multicentre case-control study. Eur. J. Cancer 2012,48, 655–661. [CrossRef]
17.
Morita, M.; Kumashiro, R.; Kubo, N.; Nakashima, Y.; Yoshida, R.; Yoshinaga, K.; Saeki, H.; Emi, Y.; Kakeji, Y.; Sakaguchi, Y.; et al.
Alcohol drinking, cigarette smoking, and the development of squamous cell carcinoma of the esophagus: Epidemiology, clinical
findings, and prevention. Int. J. Clin. Oncol. 2010,15, 126–134. [CrossRef]
18. Alsop, B.R.; Sharma, P. Esophageal Cancer. Gastroenterol. Clin. N. Am. 2016,45, 399–412. [CrossRef]
Cancers 2023,15, 2576 14 of 15
19.
Song, Y.; Li, L.; Ou, Y.; Gao, Z.; Li, E.; Li, X.; Zhang, W.; Wang, J.; Xu, L.; Zhou, Y.; et al. Identification of genomic alterations in
oesophageal squamous cell cancer. Nature 2014,509, 91–95. [CrossRef]
20.
Caspa Gokulan, R.; Garcia-Buitrago, M.T.; Zaika, A.I. From genetics to signaling pathways: Molecular pathogenesis of esophageal
adenocarcinoma. Biochim. Biophys. Acta Rev. Cancer 2019,1872, 37–48. [CrossRef]
21.
Coleman, H.G.; Xie, S.H.; Lagergren, J. The Epidemiology of Esophageal Adenocarcinoma. Gastroenterology
2018
,154, 390–405.
[CrossRef] [PubMed]
22.
Mansour, N.M.; Groth, S.S.; Anandasabapathy, S. Esophageal Adenocarcinoma: Screening, Surveillance, and Management. Annu.
Rev. Med. 2017,68, 213–227. [CrossRef] [PubMed]
23.
Schneider, J.L.; Corley, D.A. A review of the epidemiology of Barrett’s oesophagus and oesophageal adenocarcinoma. Best Pract.
Res. Clin. Gastroenterol. 2015,29, 29–39. [CrossRef]
24.
Elliott, D.R.F.; Walker, A.W.; O’Donovan, M.; Parkhill, J.; Fitzgerald, R.C. A non-endoscopic device to sample the oesophageal
microbiota: A case-control study. Lancet Gastroenterol. Hepatol. 2017,2, 32–42. [CrossRef]
25.
The Cancer Genome Atlas Research Network; Kim, J.; Bowlby, R.; Mungall, A.J.; Robertson, A.G.; Odze, R.D.; Cherniack, A.D.;
Shih, J.; Pedamallu, C.S.; Cibulskis, C.; et al. Integrated genomic characterization of oesophageal carcinoma. Nature
2017
,541,
169–175. [CrossRef]
26.
Vuik, F.; Dicksved, J.; Lam, S.Y.; Fuhler, G.M.; van der Laan, L.; van deWinkel, A.; Konstantinov, S.R.; Spaander, M.; Peppelenbosch,
M.P.; Engstrand, L.; et al. Composition of the mucosa-associated microbiota along the entire gastrointestinal tract of human
individuals. United Eur. Gastroenterol. J. 2019,7, 897–907. [CrossRef] [PubMed]
27.
Peter, S.; Pendergraft, A.; VanDerPol, W.; Wilcox, C.M.; Kyanam Kabir Baig, K.R.; Morrow, C.; Izard, J.; Mannon, P.J. Mucosa-
Associated Microbiota in Barrett’s Esophagus, Dysplasia, and Esophageal Adenocarcinoma Differ Similarly Compared With
Healthy Controls. Clin. Transl. Gastroenterol. 2020,11, e00199. [CrossRef]
28.
Yin, J.; Dong, L.; Zhao, J.; Wang, H.; Li, J.; Yu, A.; Chen, W.; Wei, W. Composition and consistence of the bacterial microbiome
in upper, middle and lower esophagus before and after Lugol’s iodine staining in the esophagus cancer screening. Scand. J.
Gastroenterol. 2020,55, 1467–1474. [CrossRef]
29.
Li, D.; He, R.; Hou, G.; Ming, W.; Fan, T.; Chen, L.; Zhang, L.; Jiang, W.; Wang, W.; Lu, Z.; et al. Characterization of the Esophageal
Microbiota and Prediction of the Metabolic Pathways Involved in Esophageal Cancer. Front. Cell Infect. Microbiol.
2020
,10, 268.
[CrossRef] [PubMed]
30.
Li, Z.; Dou, L.; Zhang, Y.; He, S.; Zhao, D.; Hao, C.; Song, G.; Zhang, W.; Liu, Y.; Wang, G. Characterization of the Oral and
Esophageal Microbiota in Esophageal Precancerous Lesions and Squamous Cell Carcinoma. Front. Cell Infect. Microbiol.
2021
,
11, 714162. [CrossRef]
31. Su, Q.; Liu, Q. Factors Affecting Gut Microbiome in Daily Diet. Front. Nutr. 2021,8, 644138. [CrossRef] [PubMed]
32.
Hitch, T.C.A.; Hall, L.J.; Walsh, S.K.; Leventhal, G.E.; Slack, E.; de Wouters, T.; Walter, J.; Clavel, T. Microbiome-based interventions
to modulate gut ecology and the immune system. Mucosal. Immunol. 2022. [CrossRef] [PubMed]
33.
Matejcic, M.; Gunter, M.J.; Ferrari, P. Alcohol metabolism and oesophageal cancer: A systematic review of the evidence.
Carcinogenesis 2017,38, 859–872. [CrossRef] [PubMed]
34.
Rao, W.; Lin, Z.; Liu, S.; Zhang, Z.; Xie, Q.; Chen, H.; Lin, X.; Chen, Y.; Yang, H.; Yu, K.; et al. Association between alcohol
consumption and oesophageal microbiota in oesophageal squamous cell carcinoma. BMC Microbiol. 2021,21, 73. [CrossRef]
35.
David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.;
Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014,505, 559–563. [CrossRef]
36.
Long, E.; Beales, I.L. The role of obesity in oesophageal cancer development. Therap. Adv. Gastroenterol.
2014
,7, 247–268.
[CrossRef]
37.
Münch, N.S.; Fang, H.Y.; Ingermann, J.; Maurer, H.C.; Anand, A.; Kellner, V.; Sahm, V.; Wiethaler, M.; Baumeister, T.; Wein, F.; et al.
High-Fat Diet Accelerates Carcinogenesis in a Mouse Model of Barrett’s Esophagus via Interleukin 8 and Alterations to the Gut
Microbiome. Gastroenterology 2019,157, 492–506.e492. [CrossRef]
38.
Nobel, Y.R.; Snider, E.J.; Compres, G.; Freedberg, D.E.; Khiabanian, H.; Lightdale, C.J.; Toussaint, N.C.; Abrams, J.A. Increasing
Dietary Fiber Intake Is Associated with a Distinct Esophageal Microbiome. Clin. Transl. Gastroenterol. 2018,9, 199. [CrossRef]
39. Kaakoush, N.O. Microbiome and Esophageal Adenocarcinoma-Letter. Cancer Res. 2018,78, 1574. [CrossRef]
40.
Yang, L.; Lu, X.; Nossa, C.W.; Francois, F.; Peek, R.M.; Pei, Z. Inflammation and intestinal metaplasia of the distal esophagus are
associated with alterations in the microbiome. Gastroenterology 2009,137, 588–597. [CrossRef]
41.
Gall, A.; Fero, J.; McCoy, C.; Claywell, B.C.; Sanchez, C.A.; Blount, P.L.; Li, X.; Vaughan, T.L.; Matsen, F.A.; Reid, B.J.; et al. Bacterial
Composition of the Human Upper Gastrointestinal Tract Microbiome Is Dynamic and Associated with Genomic Instability in a
Barrett’s Esophagus Cohort. PLoS ONE 2015,10, e0129055. [CrossRef]
42.
O’Keefe, S.J.; Li, J.V.; Lahti, L.; Ou, J.; Carbonero, F.; Mohammed, K.; Posma, J.M.; Kinross, J.; Wahl, E.; Ruder, E.; et al. Fat, fibre
and cancer risk in African Americans and rural Africans. Nat. Commun. 2015,6, 6342. [CrossRef] [PubMed]
43.
Huang, C.; Shi, G. Smoking and microbiome in oral, airway, gut and some systemic diseases. J. Transl. Med.
2019
,17, 225.
[CrossRef] [PubMed]
44.
Vogtmann, E.; Flores, R.; Yu, G.; Freedman, N.D.; Shi, J.; Gail, M.H.; Dye, B.A.; Wang, G.Q.; Klepac-Ceraj, V.; Paster, B.J.; et al.
Association between tobacco use and the upper gastrointestinal microbiome among Chinese men. Cancer Causes Control
2015
,26,
581–588. [CrossRef]
Cancers 2023,15, 2576 15 of 15
45.
Lv, J.; Guo, L.; Liu, J.J.; Zhao, H.P.; Zhang, J.; Wang, J.H. Alteration of the esophageal microbiota in Barrett’s esophagus and
esophageal adenocarcinoma. World J. Gastroenterol. 2019,25, 2149–2161. [CrossRef] [PubMed]
46.
Vesper, B.J.; Jawdi, A.; Altman, K.W.; Haines, G.K., 3rd; Tao, L.; Radosevich, J.A. The effect of proton pump inhibitors on the
human microbiota. Curr. Drug Metab. 2009,10, 84–89. [CrossRef]
47.
Amir, I.; Konikoff, F.M.; Oppenheim, M.; Gophna, U.; Half, E.E. Gastric microbiota is altered in oesophagitis and Barrett’s
oesophagus and further modified by proton pump inhibitors. Environ. Microbiol. 2014,16, 2905–2914. [CrossRef]
48.
de Gunzburg, J.; Ghozlane, A.; Ducher, A.; Le Chatelier, E.; Duval, X.; Ruppe, E.; Armand-Lefevre, L.; Sablier-Gallis, F.; Burdet, C.;
Alavoine, L.; et al. Protection of the Human Gut Microbiome From Antibiotics. J. Infect. Dis. 2018,217, 628–636. [CrossRef]
49.
Sawada, A.; Fujiwara, Y.; Nagami, Y.; Tanaka, F.; Yamagami, H.; Tanigawa, T.; Shiba, M.; Tominaga, K.; Watanabe, T.; Gi, M.; et al.
Alteration of Esophageal Microbiome by Antibiotic Treatment Does Not Affect Incidence of Rat Esophageal Adenocarcinoma.
Dig. Dis. Sci. 2016,61, 3161–3168. [CrossRef]
50.
Yu, G.; Gail, M.H.; Shi, J.; Klepac-Ceraj, V.; Paster, B.J.; Dye, B.A.; Wang, G.Q.; Wei, W.Q.; Fan, J.H.; Qiao, Y.L.; et al. Association
between upper digestive tract microbiota and cancer-predisposing states in the esophagus and stomach. Cancer Epidemiol. Biomark.
Prev. 2014,23, 735–741. [CrossRef]
51.
Macfarlane, S.; Furrie, E.; Macfarlane, G.T.; Dillon, J.F. Microbial colonization of the upper gastrointestinal tract in patients with
Barrett’s esophagus. Clin. Infect. Dis. 2007,45, 29–38. [CrossRef] [PubMed]
52.
Liu, N.; Ando, T.; Ishiguro, K.; Maeda, O.; Watanabe, O.; Funasaka, K.; Nakamura, M.; Miyahara, R.; Ohmiya, N.; Goto, H.
Characterization of bacterial biota in the distal esophagus of Japanese patients with reflux esophagitis and Barrett’s esophagus.
BMC Infect. Dis. 2013,13, 130. [CrossRef] [PubMed]
53.
Deshpande, N.P.; Riordan, S.M.; Castano-Rodriguez, N.; Wilkins, M.R.; Kaakoush, N.O. Signatures within the esophageal
microbiome are associated with host genetics, age, and disease. Microbiome 2018,6, 227. [CrossRef] [PubMed]
54.
Snider, E.J.; Compres, G.; Freedberg, D.E.; Khiabanian, H.; Nobel, Y.R.; Stump, S.; Uhlemann, A.C.; Lightdale, C.J.; Abrams, J.A.
Alterations to the Esophageal Microbiome Associated with Progression from Barrett’s Esophagus to Esophageal Adenocarcinoma.
Cancer Epidemiol. Biomark. Prev. 2019,28, 1687–1693. [CrossRef]
55.
Lopetuso, L.R.; Severgnini, M.; Pecere, S.; Ponziani, F.R.; Boskoski, I.; Larghi, A.; Quaranta, G.; Masucci, L.; Ianiro, G.;
Camboni, T.; et al. Esophageal microbiome signature in patients with Barrett’s esophagus and esophageal adenocarcinoma. PLoS
ONE 2020,15, e0231789. [CrossRef] [PubMed]
56.
Shao, D.; Vogtmann, E.; Liu, A.; Qin, J.; Chen, W.; Abnet, C.C.; Wei, W. Microbial characterization of esophageal squamous cell
carcinoma and gastric cardia adenocarcinoma from a high-risk region of China. Cancer 2019,125, 3993–4002. [CrossRef]
57.
Li, Z.; Shi, C.; Zheng, J.; Guo, Y.; Fan, T.; Zhao, H.; Jian, D.; Cheng, X.; Tang, H.; Ma, J. Fusobacterium nucleatum predicts a high
risk of metastasis for esophageal squamous cell carcinoma. BMC Microbiol. 2021,21, 301. [CrossRef] [PubMed]
58.
Shen, W.; Tang, D.; Wan, P.; Peng, Z.; Sun, M.; Guo, X.; Liu, R. Identification of tissue-specific microbial profile of esophageal
squamous cell carcinoma by full-length 16S rDNA sequencing. Appl. Microbiol. Biotechnol. 2022,106, 3215–3229. [CrossRef]
59.
Liu, Y.; Lin, Z.; Lin, Y.; Chen, Y.; Peng, X.E.; He, F.; Liu, S.; Yan, S.; Huang, L.; Lu, W.; et al. Streptococcus and Prevotella are
associated with the prognosis of oesophageal squamous cell carcinoma. J. Med. Microbiol. 2018,67, 1058–1068. [CrossRef]
60.
Peters, B.A.; Wu, J.; Pei, Z.; Yang, L.; Purdue, M.P.; Freedman, N.D.; Jacobs, E.J.; Gapstur, S.M.; Hayes, R.B.; Ahn, J. Oral
Microbiome Composition Reflects Prospective Risk for Esophageal Cancers. Cancer Res. 2017,77, 6777–6787. [CrossRef]
61.
Wang, Q.; Rao, Y.; Guo, X.; Liu, N.; Liu, S.; Wen, P.; Li, S.; Li, Y. Oral Microbiome in Patients with Oesophageal Squamous Cell
Carcinoma. Sci. Rep. 2019,9, 19055. [CrossRef] [PubMed]
62.
Ishaq, H.M.; Mohammad, I.S.; Sher Muhammad, K.; Li, H.; Abbas, R.Z.; Din Sindhu, Z.U.; Ullah, S.; Fan, Y.; Sadiq, A.; Raza, M.A.;
et al. Gut microbial dysbiosis and its association with esophageal cancer. J. Appl. Biomed. 2021,19, 1–13. [CrossRef] [PubMed]
63.
Zhao, Q.; Yang, T.; Yan, Y.; Zhang, Y.; Li, Z.; Wang, Y.; Yang, J.; Xia, Y.; Xiao, H.; Han, H.; et al. Alterations of Oral Microbiota in
Chinese Patients With Esophageal Cancer. Front. Cell Infect. Microbiol. 2020,10, 541144. [CrossRef] [PubMed]
64.
Deng, Y.; Tang, D.; Hou, P.; Shen, W.; Li, H.; Wang, T.; Liu, R. Dysbiosis of gut microbiota in patients with esophageal cancer.
Microb. Pathog. 2021,150, 104709. [CrossRef] [PubMed]
65.
Wu, C.; Wang, M.; Zhou, Q.; Shi, H. Associations of Changes in Intestinal Flora and Inflammatory Factors with Prognosis of
Patients with Esophageal Cancer. J. Healthc. Eng. 2022,2022, 2426301. [CrossRef] [PubMed]
66.
Inamura, K.; Hamada, T.; Bullman, S.; Ugai, T.; Yachida, S.; Ogino, S. Cancer as microenvironmental, systemic and environmental
diseases: Opportunity for transdisciplinary microbiomics science. Gut 2022,71, 2107–2122. [CrossRef]
Disclaimer/Publisher’s Note:
The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
... Malfunctions of GM can initiate a spectrum of illnesses, including metabolic disorders, cardiovascular diseases, immune diseases, mental disorders, and various types of cancers [13]. In recent years, research has suggested that GM played an important role in the occurrence and progression of GERD and BE [14,15]. For instance, Zou et al [14] indicated that Gram-positive(G+) bacteria were prevalent in the normal esophagus, with Firmicutes and Streptococcus as the most common. ...
... Esophageal microbiota's relationship with GERD/BE was characterized by a transition from the Type I to the Type II flora [42]. Although the action ratio and efficacy of G + and G-bacteria in the development of GERD and BE were nearly the same [15,43], type II flora was rich in G-bacteria and mostly associated with GERD and BE, while type I flora was predominantly composed of G + bacteria and tied to normal esophageal function [44]. This signifies a transition from a condition characterized by a substantial abundance of G + bacteria to a greater abundance of G-bacteria, accompanied with a reduction in microbial diversity [45,46]. ...
Article
Full-text available
Background Gut microbiota(GM) have been proven associated with lots of gastrointestinal diseases, but its causal relationship with Gastroesophageal reflux disease(GERD) and Barrett’s esophagus(BE) hasn’t been explored. We aimed to uncover the causal relation between GM and GERD/BE and potential mediators by utilizing Mendelian Randomization(MR) analysis. Methods Summary statistics of GM(comprising 301 bacteria taxa and 205 metabolism pathways) were extracted from MiBioGen Consortium(N = 18,340) and Dutch Microbiome Project(N = 7,738), GERD and BE from a multitrait meta-analysis(NGERD=602,604, NBE=56,429). Bidirectional two-sample MR analysis and linkage disequilibrium score regression(LDSC) were used to explore the genetic correlation between GM and GERD/BE. Mediation MR analysis was performed for the risk factors of GERD/BE, including Body mass index(BMI), weight, type 2 diabetes, major depressive disorder(MDD), smoking initiation, alcohol consumption, and dietary intake(including carbohydrate, sugar, fat, protein intake), to detect the potential mediators between GM and GERD/BE. Results 11 bacterial taxa and 13 metabolism pathways were found associated with GERD, and 18 taxa and 5 pathways exhibited causal relationship with BE. Mediation MR analysis suggested weight and BMI played a crucial role in these relationships. LDSC identified 1 taxon and 4 metabolism pathways related to GERD, and 1 taxon related to BE. Specie Faecalibacterium prausnitzii had a suggestive impact on both GERD(OR = 1.087, 95%CI = 1.01–1.17) and BE(OR = 1.388, 95%CI = 1.03–1.86) and LDSC had determined their correlation. Reverse MR indicated that BE impacted 10 taxa and 4 pathways. Conclusions This study established a causal link between gut microbiota and GERD/BE, and identified the probable mediators. It offers new insights into the role of gut microbiota in the development and progression of GERD and BE in the host.
... It is now accepted that the esophageal microbiome is altered during esophageal carcinogenesis. The well-noted risk factors associated with EAC also appear to be related to modifications to the normal microbiota that inhabits the esophagus [28]. Diets rich in high-fat contents in animal studies have been linked with esophageal dysplasia and alterations of the microbiota [29]. ...
... Diets rich in high-fat contents in animal studies have been linked with esophageal dysplasia and alterations of the microbiota [29]. The low intake of fibers was also associated with an increase in Gram-negative species, which can be found in esophageal cancer precursor lesions [22,28]. Overall, the microbiota of the normal esophagus is mainly composed of Firmicutes, Proteobacteria, Bacteriodetes, Actinobacteria, and Fusobacteria [30]. ...
Article
Full-text available
Esophageal adenocarcinoma (EAC) is the consequence of longstanding gastroesophageal reflux, which leads to inflammation and could cause Barrett’s esophagus (BE), the main risk factor for EAC development. The 5 year survival rate of EAC is poor since the diagnosis occurs at the late stage of the disease. To improve patient management, a better comprehension of the mechanism undergoing the evolution through to adenocarcinoma is needed. Within this scenario, the resident microbiome investigation was studied. This study aimed to explore the esophageal microbial profile in patients affected by non-dysplastic BE, low- and high-grade dysplastic BE, and EAC to identify parameters characterizing cancer progression and to develop a score suitable for clinical practice to stratify cancer risk. The microbiota was investigated through the 16S rRNA gene sequencing of esophageal biopsies. The microbial composition was evaluated at each different taxonomic level along the disease progression. To further investigate bacteria potentially associated with cancer development, non-dysplastic and dysplastic/cancer patients were compared. The presence of the six significant microbial features with multivariate analysis was used to develop a multiparametric score (Resident Esophageal Microbial Dysbiosis Test) to predict the risk of progression toward EAC. Finally, the diagnostic ability of the test and its discrimination threshold for its ability to identify dysplastic/cancer patients were demonstrated. Since EAC has been related to obesity, the relationship between these microbial parameters and patients’ diet/lifestyle habits was also investigated. Developing microbiome-based risk prediction models for esophageal adenocarcinoma onset could open new research avenues, demonstrating that the resident microbiome may be a valid cancer risk biomarker.
... Its presence is often associated with poor tumour differentiation, advanced stages, metastasis and reduced survival outcomes, further emphasising its prognostic significance. Emerging evidence supports the role of the oesophageal microbiome in modulating responses to immune checkpoint inhibitors in EC, with different microbial compositions correlating with varying responses to radiotherapy and chemotherapy [161,169,170]. These insights underscore the intricate relationship between intratumoral bacteria and the host immune system in EC, highlighting the potential of microbiome-targeted therapies to complement traditional cancer treatments and improve therapeutic efficacy and patient outcomes [171,172]. ...
Article
Full-text available
Recently, research on the human microbiome, especially concerning the bacteria within the digestive system, has substantially advanced. This exploration has unveiled a complex interplay between microbiota and health, particularly in the context of disease. Evidence suggests that the gut microbiome plays vital roles in digestion, immunity and the synthesis of vitamins and neurotransmitters, highlighting its significance in maintaining overall health. Conversely, disruptions in these microbial communities, termed dysbiosis, have been linked to the pathogenesis of various diseases, including digestive system cancers. These bacteria can influence cancer progression through mechanisms such as DNA damage, modulation of the tumour microenvironment, and effects on the host’s immune response. Changes in the composition and function within the tumours can also impact inflammation, immune response and cancer therapy effectiveness. These findings offer promising avenues for the clinical application of intratumoral bacteria for digestive system cancer treatment, including the potential use of microbial markers for early cancer detection, prognostication and the development of microbiome-targeted therapies to enhance treatment outcomes. This review aims to provide a comprehensive overview of the pivotal roles played by gut microbiome bacteria in the development of digestive system cancers. Additionally, we delve into the specific contributions of intratumoral bacteria to digestive system cancer development, elucidating potential mechanisms and clinical implications. Ultimately, this review underscores the intricate interplay between intratumoral bacteria and digestive system cancers, underscoring the pivotal role of microbiome research in transforming diagnostic, prognostic and therapeutic paradigms for digestive system cancers.
Article
Full-text available
Gastrointestinal (GI) cancers constitute more than 33% of new cancer cases worldwide and pose a considerable burden on public health. There exists a growing body of evidence that has systematically recorded an upward trajectory in GI malignancies within the last 5 to 10 years, thus presenting a formidable menace to the health of the human population. The perturbations in GI microbiota may have a noteworthy influence on the advancement of GI cancers; however, the precise mechanisms behind this association are still not comprehensively understood. Some bacteria have been observed to support cancer development, while others seem to provide a safeguard against it. Recent studies have indicated that alterations in the composition and abundance of microbiomes could be associated with the progression of various GI cancers, such as colorectal, gastric, hepatic, and esophageal cancers. Within this comprehensive analysis, we examine the significance of microbiomes, particularly those located in the intestines, in GI cancers. Furthermore, we explore the impact of microbiomes on various treatment modalities for GI cancer, including chemotherapy, immunotherapy, and radiotherapy. Additionally, we delve into the intricate mechanisms through which intestinal microbes influence the efficacy of GI cancer treatments.
Article
Full-text available
Many studies point to an association between Helicobacter pylori (H. pylori ) infection and inflammatory bowel diseases (IBD). Although controversial, this association indicates that the presence of the bacterium somehow affects the course of IBD. It appears that H. pylori infection influences IBD through changes in the diversity of the gut microbiota, and hence in local chemical characteristics, and alteration in the pattern of gut immune response. The gut immune response appears to be modulated by H. pylori infection towards a less aggressive inflammatory response and the establishment of a targeted response to tissue repair. Therefore, a T helper 2 (Th2)/macrophage M2 response is stimulated, while the Th1/macrophage M1 response is suppressed. The immunomodulation appears to be associated with intrinsic factors of the bacteria, such as virulence factors - such oncogenic protein cytotoxin-associated antigen A, proteins such H. pylori neutrophil-activating protein, but also with microenvironmental changes that favor permanence of H. pylori in the stomach. These changes include the increase of gastric mucosal pH by urease activity, and suppression of the stomach immune response promoted by evasion mechanisms of the bacterium. Furthermore, there is a causal relationship between H. pylori infection and components of the innate immunity such as the NLR family pyrin domain containing 3 inflammasome that directs IBD toward a better prognosis.
Article
Full-text available
The gut microbiome lies at the intersection between the environment and the host, with the ability to modify host responses to disease-relevant exposures and stimuli. This is evident in how enteric microbes interact with the immune system, e.g., supporting immune maturation in early life, affecting drug efficacy via modulation of immune responses, or influencing development of immune cell populations and their mediators. Many factors modulate gut ecosystem dynamics during daily life and we are just beginning to realise the therapeutic and prophylactic potential of microbiome-based interventions. These approaches vary in application, goal, and mechanisms of action. Some modify the entire community, such as nutritional approaches or faecal microbiota transplantation, while others, such as phage therapy, probiotics, and prebiotics, target specific taxa or strains. In this review, we assessed the experimental evidence for microbiome-based interventions, with a particular focus on their clinical relevance, ecological effects, and modulation of the immune system.
Article
Full-text available
It was previously believed that the microbial community in the esophagus was relatively stable, but it has been reported that different esophageal diseases have different microbial community characteristics. In this study, we recruited patients with esophageal squamous cell carcinoma (ESCC) and collected 51 pairs of tumor and adjacent non-tumor tissues for full-length 16S rDNAsequencing and qPCR to compare the differences in microbial community structure. The results of sequencing in 19 pairs of tissues showed that Proteobacteria, Firmicutes, Bacteroidetes, Deinococcus-Thermus, and Actinobacteria were the main bacteria in tumor and adjacent non-tumor tissues. At the genus level, the bacteria with the highest relative proportion in tumor and adjacent non-tumor tissues were Streptococcus and Labrys, respectively. At the same time, it was observed that the complexity of microbial interactions in tumor tissues was weaker than that of adjacent non-tumor tissues. The results also found that the relative abundance of 24 taxa was statistically different between tumor and adjacent non-tumor tissues. The findings of qPCR in 32 pairs of tissues further evidence that the relative proportions of Blautia, Treponema, Lactobacillus murinus, Peptoanaerobacter stomatis, and Fusobacteria periodonticum were statistically different in tumor and adjacent non-tumor tissues. The findings of PIRCUSt2 indicated the lipopolysaccharide biosynthesis and biotin metabolism in the microbiome of cancer tissues are more significant. This study supplements the existing information on the structure, function, and interaction of microorganisms in the esophagus in situ and provides a direction for the further exploration of the relationship between esophageal in situ microorganisms and esophageal squamous cell carcinoma. Key points • The structure of the microbial community in esophageal cancer tissue and adjacent non-tumor tissues at the phylum level is similar • Streptococcus and Labrys are the most important bacteria in esophageal tumor tissues and adjacent non-tumor tissues, respectively • Microbial interactions in tumor tissues are stronger than in adjacent non-tumor tissues
Article
Full-text available
This study aims to explore the associations of changes in intestinal flora and inflammatory factors with the prognosis of patients with esophageal cancer (EC). A total of 40 EC patients treated and 40 normal people who underwent gastroscopy and CT examination for gastrointestinal discomfort during the same period were selected as the participants of the study. The endotoxin level, colonization ability of intestinal flora, and distribution of intestinal flora (Bifidobacterium, Lactobacillus, Escherichia coli, and Enterococcus) were compared between the two groups. The levels of inflammatory factors interleukin-6 (IL-6), high-sensitivity C-reactive protein (hs-CRP), and tumor necrosis factor-α (TNF-α) were also compared between the two groups. All participants were followed up for 3 years, and the associations of survival time with colonization ability of intestinal flora and changes in hs-CRP were analyzed. Finally, the univariate and multivariate logistic regression analyses were performed for related factors affecting the survival time of EC patients. In the observation group, the endotoxin level was significantly higher (P
Article
Full-text available
Microbial imbalances have been well elucidated in esophageal adenocarcinoma. However, few studies address the microbiota in esophageal squamous cell carcinoma (ESCC) and esophagitis (ES). We aimed to explore the association of esophageal microbiota with these patients. Esophageal tissues were obtained from healthy controls and ES and ESCC patients undergoing upper endoscopy. 16S rRNA gene sequencing was applied to analyze the microbiome. The α and β diversity differences were tested by Tukey test and partial least squares-discriminant analysis (PLS-DA), respectively. Linear discriminant analysis effect size (LEfSe) analysis was performed to assess taxonomic differences between groups. A total of 68 individuals were enrolled (control = 21, ES = 15, ESCC = 32). Microbial diversity was significantly different between the ESCC patients and healthy controls by Chao1 index, Shannon index, and PLS-DA. Firmicutes, Proteobacteria, Bacteroidetes, Actinobacteria, and Fusobacteria were the five dominant bacterial phyla among the three groups. Megamonas, Collinsella, Roseburia, and Ruminococcus_2 showed a significantly continuous decreasing trend from the control group to the ESCC group at the genus level. When compared with the control group, decreased Fusobacteria at phylum level and Faecalibacterium, Bacteroides, Curvibacter, and Blautia at genus level were detected. ESCC samples also displayed a striking reduction of Bacteroidetes, Faecalibacterium, Bacteroides, and Blautia in comparison with the ES patients. LEfSe analysis indicated a greater abundance of Streptococcus, Actinobacillus, Peptostreptococcus, Fusobacterium, and Prevotella in the ESCC group. Our study suggests a potential association between esophageal microbiome dysbiosis and ESCC and provides insights into potential screening markers for esophageal cancer.
Article
Full-text available
Background Esophageal squamous cell carcinoma (ESCC) is the major type of esophageal cancer in China. The role of the bacteria present in ESCC tissue in neoplastic progression has not been fully elucidated. This study aimed to uncover different bacterial communities in ESCC tissues and examine the correlation between the abundance of the esophageal flora and clinicopathologic characteristics of ESCC. Results Microorganisms in tumors and normal tissues showed obvious clustering characteristics. The abundance of Fusobacterium ( P = 0.0052) was increased in tumor tissues. The high level of Fusobacterium nucleatum was significantly associated with pT stage ( P = 0.039) and clinical stage ( P = 0.0039). The WES data showed that COL22A1, TRBV10–1, CSMD3, SCN7A and PSG11 were present in only the F. nucleatum -positive ESCC samples. GO and protein domain enrichment results suggested that epidermal growth factor might be involved in the regulation of cell apoptosis in F. nucleatum -positive ESCC. Both a higher mutational burden and F. nucleatum -positive was observed in tumors with metastasis than in tumors without metastasis. Conclusion F. nucleatum is closely related to the pT stage and clinical stage of ESCC. The abundance of F. nucleatum and tumor mutation burden may be used in combination as a potential method to predict metastasis in ESCC.
Article
Full-text available
Important evidence indicates that the microbiota plays a key role in esophageal squamous cell carcinoma (ESCC). Here, paired saliva and brush specimens were obtained from 276 participants undergoing upper gastrointestinal endoscopic examination before or during screening for upper gastrointestinal (UGI) cancer. The esophageal microbiota was investigated by 16S rRNA gene profiling and next-generation sequencing. We observed that as the disease progressed, the α diversity in the saliva and cell brush samples decreased. Linear discriminant analysis effect size (LEfSe) results showed that in both the saliva and cell brush specimens, Granulicatella, Rothia, Streptococcus, Gemella, Leptotrichia and Schaalia were common biomarkers in patients with low-grade dysplasia, Lactobacillus was a common biomarker in patients with high-grade dysplasia, and Bosea, Solobacterium, Gemella, and Peptostreptococcus were common biomarkers in patients with esophageal cancer. The top 3 genera in the saliva and cell brush specimens had areas under the curve (AUCs) of 87.16 and 89.13%, respectively, to distinguish ESCC patients from normal people. The PICRUSt2 results identified in brush samples that patients with ESCC had decreased nitrate reductase functions. Our results suggest that future studies can focus on the function of the characteristic bacteria in ESCC.
Article
Full-text available
The pathogenesis of gut microbiota in humans can be indicated due to the wide application of techniques, such as 16S rRNA sequencing. Presently, several studies have found a significant difference in fecal flora between normal individuals and patients with gastric cancer. Although clinical research on the feedback mechanism of gastric flora and gut microbiota is lacking, clarifying the relationship between gut microbiota and the characteristics of cancer is significant for the early diagnosis of gastric cancer. This study was conducted to review the results of several studies in the past 5 years and analyze the intestinal bacteria in patients with gastric cancer and compare them with those in patients with esophageal and small intestine cancers. It was found that the gut microbiota in patients with gastric cancer was similar to that in patients with esophageal cancer. However, making an analysis and comparing the gut microbiota in patients with small intestine and gastric cancers was impossible due to the low incidence of small intestinal cancer. Our review summarized the research progress on using the gut microbiota for early screening for gastric cancer, and the results of this study will provide a further direction in this field. Key points • We reviewed several relative mechanisms of the gut microbiota related to gastric cancer. • The gut microbiota in gastric, esophageal, and small intestine cancers are significantly different in types and quantity, and we have provided some tips for further research. • A prospective review of sequencing methods and study results on the gut microbiota in gastric, esophageal, and small intestine cancers was described.
Article
Full-text available
There is a growing recognition that a good diet can help people maintain mental and physical health, while a bad one will cause the disorder of body function, and even lead to several diseases. A lot of attentions have been devoted to analyze every possible health-related factor in the daily diet, including food ingredients, additives, and cooking process. With the support of high-throughput sequencing technology, there is accumulating evidence gradually clarifying that most of these factors are mainly through the interactions with gut microbiome to trigger downstream effects. The gut microbiome may be able to act as a very sensitive mirror in response to human daily diet. A complex network of interactions among diet, gut microbiome, and health has been gradually depicted, but it is rarely discussed from a more comprehensive perspective. To this end, this review summarized the latest updates in diet-gut microbiome interactions, analyzed most identified factors involved in this process, showed the possibility of maintaining health or alleviating diseases by diet intervention, aiming to help people choose a suitable recipe more accurately.
Article
Full-text available
Background Microbiota has been reported to play a role in cancer patients. Nevertheless, little is known about the association between alcohol consumption and resultant changes in the diversity and composition of oesophageal microbiota in oesophageal squamous cell carcinoma (ESCC). Methods We performed a hospital-based retrospective study of 120 patients with pathologically diagnosed primary ESCC. The relevant information for all study participants were collected through a detailed questionnaire. The differences in adjacent tissues between non-drinkers and drinkers were explored using 16S rRNA gene sequencing. Raw sequencing data were imported into QIIME 2 to analyse the diversity and abundance of microbiota. Linear discriminant analysis effect size (LEfSe) and unconditional logistic regression were performed to determine the bacterial taxa that were associated with drinking. Results The Shannon diversity index and Bray-Curtis distance of oesophageal microbiota were significantly different among drinkers(P < 0.05). The alcohol-related bacteria were primarily from the orders Clostridiales, Gemellales and Pasteurellales, family Clostridiaceae, Lanchnospiraceae, Helicobacteraceae, Alcaligenaceae, Bacteroidaceae, Pasteurellaceae and Gemellaceae; genus Clostridium, Helicobacter, Catonella, Bacteroides, Bacillus, Moraxella, and Bulleidia; and species B. moorei and longum (genus Bifidobacterium). In addition, the diversity and abundance of these microbiota were observed to be affected by the age, residential districts of the patients, and sampling seasons. Moreover, the higher the frequency and years of alcohol consumption, the lower was the relative abundance of genus Catonella that was observed. Conclusion Alcohol consumption is associated with alterations in both the diversity and composition the of the oesophageal microbiota in ESCC patients.
Article
Cancer is generally regarded as a localised disease, with the well-established role of the tumour microenvironment. However, the realm of cancer goes beyond the tumour microenvironment, and cancer should also be regarded as a systemic and environmental disease. The exposome ( ie, the totality of exposures), which encompasses diets, supplements, smoking, alcohol, other lifestyle factors, medications, etc , likely alters the microbiome (inclusive of bacteria, viruses, archaea, fungi, parasites, etc ) and immune system in various body sites and influences tumour phenotypes. The systemic metabolic/inflammatory status, which is likely influenced by exposures and intestinal physiological changes, may affect tissue microenvironment of colorectum and any other organs. Germline genomic factors can modify disease phenotypes via gene-by-environment interactions. Although challenges exist, it is crucial to advance not only basic experimental research that can analyse the effects of exposures, microorganisms and microenvironmental components on tumour evolution but also interdisciplinary human population research that can dissect the complex pathogenic roles of the exposome, microbiome and immunome. Metagenomic, metatranscriptomic and metabolomic analyses should be integrated into well-designed population research combined with advanced methodologies of artificial intelligence and molecular pathological epidemiology. Ideally, a prospective cohort study design that enables biospecimen (such as stool) collection before disease detection should be considered to address reverse causation and recall biases. Robust experimental and observational research together can provide insights into dynamic interactions between environmental exposures, microbiota, tumour and immunity during carcinogenesis processes, thereby helping us develop precision prevention and therapeutic strategies to ultimately reduce the cancer burden.