ArticlePDF Available

Porphyromonas gingivalis Promotes Oral Squamous Cell Carcinoma Progression in an Immune Microenvironment

Authors:

Abstract and Figures

Increasing evidence has revealed a significant association between microorganisms and oral squamous cell carcinoma (OSCC). Porphyromonas gingivalis, the keystone pathogen in chronic periodontitis, is considered an important potential etiologic agent of OSCC, but the underlying immune mechanisms through which P. gingivalis mediates tumor progression of the oral cancer remain poorly understood. Our cohort study showed that the localization of P. gingivalis in tumor tissues was related to poor survival of patients with OSCC. Moreover, P. gingivalis infection increased oral lesion multiplicity and size and promoted tumor progression in a 4-nitroquinoline-1 oxide (4NQO)–induced carcinogenesis mouse model by invading the oral lesions. In addition, CD11b ⁺ myeloid cells and myeloid-derived suppressor cells (MDSCs) showed increased infiltration of oral lesions. Furthermore, in vitro observations showed that MDSCs accumulated when human-derived dysplastic oral keratinocytes (DOKs) were exposed to P. gingivalis, and CXCL2, CCL2, interleukin (IL)–6, and IL-8 may be potential candidate genes that facilitate the recruitment of MDSCs. Taken together, our findings suggest that P. gingivalis promotes tumor progression by generating a cancer-promoting microenvironment, indicating a close relationship among P. gingivalis, tumor progression of the oral cancer, and immune responses.
Content may be subject to copyright.
https://doi.org/10.1177/0022034520909312
Journal of Dental Research
1 –10
© International & American Associations
for Dental Research 2020
Article reuse guidelines:
sagepub.com/journals-permissions
DOI: 10.1177/0022034520909312
journals.sagepub.com/home/jdr
Research Reports: Biological
Introduction
Oral squamous cell carcinoma (OSCC) is the most common
malignant cancer of the head and neck (Chi et al. 2015), which
accounted for approximately 180,000 deaths worldwide in
2018 (1.9% of total cancer cases), and is among the top 15
common cancers worldwide (Bray et al. 2018). OSCC is often
preceded by oral potentially malignant disorders (OPMDs),
but the mechanisms by which OPMDs progress into cancer
remains largely uncertain. Among the major risk factors for
oral cancers, alcohol consumption, areca nut (betel quid) chew-
ing, and human papillomavirus infection were extensively
studied (Mehanna et al. 2013; Winn et al. 2015; Mehrtash et al.
2017). However, around 15% cases of oral carcinogenesis
remain unexplained (Chocolatewala et al. 2010). Thus, it is
urgent to find new pathogenic factors so that we can prevent
the tumor progression of oral cancer.
An increasing and substantial number of microorganisms
have been confirmed to be strongly associated with human car-
cinogenesis, including Helicobacter pylori in gastric cancer
(Amieva and Peek 2016), Fusobacterium nucleatum in
colorectal cancer (Yu et al. 2017), and so on. Porphyromonas
gingivalis, a Gram-negative oral bacteria as “keystone patho-
gen” in periodontitis (Hajishengallis et al. 2012), shows strong
carcinogenic potential in gastrointestinal tract cancers, such as
colon cancer (Ahn et al. 2012), pancreatic cancer (Michaud
et al. 2013), esophageal cancer (Peters et al. 2017), and OSCC.
The relative abundance of P. gingivalis in gingival carcinoma
has been reported to be 33% higher than that in normal gingival
909312JDRXXX10.1177/0022034520909312Journal of Dental ResearchP. gingivalis Promotes OSCC Progression in an Immune Microenvironment
research-article2020
1Hospital of Stomatology, Guanghua School of Stomatology, Guangdong
Provincial Key Laboratory of Stomatology, Sun Yat-Sen University,
Guangzhou, Guangdong, China
2State Key Laboratory of Oncology in South China, MOE Key
Laboratory of Gene Function and Regulation, School of Life Sciences,
Sun Yat-sen University, Guangzhou, Guangdong, China
3Department of Microbiology, Zhongshan School of Medicine, Key
Laboratory for Tropical Diseases Control of the Ministry of Education,
Sun Yat-sen University, Guangzhou, Guangdong, China
4Discipline of Oral Bioscience, Sydney Dental School, Faculty of Medicine
and Health, The University of Sydney, Westmead, NSW, Australia
A supplemental appendix to this article is available online.
Corresponding Authors:
Z. Wang, Hospital of Stomatology, Guanghua School of Stomatology,
Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen
University, No. 56, Lingyuan West Road, Yuexiu District, Guangzhou,
Guangdong 510055, China.
Email: wangzh75@mail.sysu.edu.cn
B. Cheng, Hospital of Stomatology, Guanghua School of Stomatology,
Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen
University, No. 56, Lingyuan West Road, Yuexiu District, Guangzhou,
Guangdong 510055, China.
Email: chengbin@mail.sysu.edu.cn
Porphyromonas gingivalis Promotes Oral
Squamous Cell Carcinoma Progression
in an Immune Microenvironment
L. Wen1, W. Mu1, H. Lu1, X. Wang1, J. Fang1, Y. Jia1, Q. Li1, D. Wang1, S. Wen1,
J. Guo1, W. Dai1, X. Ren1, J. Cui2, G. Zeng3, J. Gao4, Z. Wang1, and B. Cheng1
Abstract
Increasing evidence has revealed a significant association between microorganisms and oral squamous cell carcinoma (OSCC).
Porphyromonas gingivalis, the keystone pathogen in chronic periodontitis, is considered an important potential etiologic agent of OSCC,
but the underlying immune mechanisms through which P. gingivalis mediates tumor progression of the oral cancer remain poorly
understood. Our cohort study showed that the localization of P. gingivalis in tumor tissues was related to poor survival of patients with
OSCC. Moreover, P. gingivalis infection increased oral lesion multiplicity and size and promoted tumor progression in a 4-nitroquinoline-1
oxide (4NQO)–induced carcinogenesis mouse model by invading the oral lesions. In addition, CD11b+ myeloid cells and myeloid-
derived suppressor cells (MDSCs) showed increased infiltration of oral lesions. Furthermore, in vitro observations showed that MDSCs
accumulated when human-derived dysplastic oral keratinocytes (DOKs) were exposed to P. gingivalis, and CXCL2, CCL2, interleukin
(IL)–6, and IL-8 may be potential candidate genes that facilitate the recruitment of MDSCs. Taken together, our findings suggest that
P. gingivalis promotes tumor progression by generating a cancer-promoting microenvironment, indicating a close relationship among
P. gingivalis, tumor progression of the oral cancer, and immune responses.
Keywords: microbiology, bacteria, 4-Nitroquinoline-1-oxide, precancerous conditions, mucosal immune response, retrospective
studies
2 Journal of Dental Research 00(0)
tissues, suggesting an association between P. gingivalis and
OSCC (Katz et al. 2011). More recently, a meta-analysis indi-
cated that the existence of P. gingivalis increased OSCC devel-
opment by 1.36 times (Sayehmiri et al. 2015). These findings
indicated that P. gingivalis might be a new candidate that pro-
motes the tumor progression of oral cancer. However, few
studies have explored the molecules and mechanisms underly-
ing these associations.
Importantly, while P. gingivalis infection is strongly corre-
lated with chronic periodontitis, the infection result in tissue
destruction attributes to host immune response but not bacteria
itself (Hajishengallis 2015). The interaction of the host immune
system with the oral bacteria in healthy states and in diseased
states has been widely described; nevertheless, the possible
interrelation of infection of P. gingivalis, immune microenvi-
ronment development, and oral cancer is rarely explored.
The current study investigated the mechanisms by which
P. gingivalis participates in tumor progression of oral cancer
and its association with the immune microenvironment.
Therefore, we first examined whether the existence of P. gingi-
valis was correlated with survival in patients with OSCC. To
delineate the role of P. gingivalis in oral carcinogenesis, we
determine whether P. gingivalis invades the oral precancerous
and cancer lesions of an established 4-nitroquinoline-1 oxide
(4NQO)–induced carcinogenesis mouse model. Furthermore,
our results show that myeloid cells and myeloid-derived sup-
pressor cells (MDSCs) were more infiltrated in the P. gingiva-
lis infection group, which might be associated with interleukin
(IL)–6, IL-8, CCL2, and CXCL2 expression. This study pro-
vides novel insights and experimental basis in vivo and in vitro
to support the role of P. gingivalis in the tumor progression of
oral cancer.
Materials and Methods
Collection of Clinical Samples
Fifty-six formalin-fixed, paraffin-embedded (FFPE) blocks of
OSCC tissues were collected from the Stomatological Hospital,
Sun Yat-Sen University (SYSU). The patients were pathologi-
cally with OSCC and over 18 y of age. Overall survival,
defined as the time from the first disease diagnosis to the date
of death or last contact, was designated as the end point.
Recurrence was monitored by clinical and pathological diag-
nosis. Informed consent was obtained from all patients. The
use of human samples for this study was approved by the ethics
committee of SYSU and was conducted according to the
STROBE (Strengthening the Reporting of Observational
Studies in Epidemiology) statement.
Bacterial Stains and Culturing
Strain P. gingivalis ATCC33277, P. gingivalis W83, and
Streptococcus mutans UA159 (S. mutans) were purchased
from ATCC. The bacterial were grown in Brain Heart Infusion
broth (BHI) supplemented with yeast extract at 5 mg/mL,
cysteine at 1 mg/mL, vitamin K1 at 0.5 µg/mL and hemin at 5
µg/mL under anaerobic conditions with oxygen concentration
<1% at 37°C (Bactrox-2; SHELLAB). Human-derived dys-
plastic oral keratinocytes (DOKs; Sigma) were grown and cul-
tured in high-glucose Dulbecco’s modified Eagle’s media
(DMEM; Gibco) supplemented with 10% fetal bovine serum
(FBS; Millipore Sigma) and 0.05% hydrocortisone (Millipore
Sigma) at 37°C in 5% CO2. DOK cells were starved in low-
glucose serum-free DMEM (Gibco) overnight prior to experi-
ments, followed by continued treatment in serum-free
low-glucose medium.
4NQO-Induced Oral Tumorigenesis
Model and Animal Experiments
Six-week-old wild-type C57BL/6 mice were purchased from
Guangzhou University of Chinese Medicine and were main-
tained in the SYSU in a specific pathogen-free facility at stan-
dard laboratory conditions. The chemical carcinogen 4NQO
(Sigma-Aldrich) was performed according to the published
methods (Chen et al. 2018). For the infected model, mice were
randomly divided into a control group (n = 6) and a P. gingiva-
lis group (n = 12). P. gingivalis ATCC33277 (1 × 108 colony-
forming units [CFU] per 100 µL) suspended in 2%
carboxymethylcellulose was orally inoculated to mice every
other day for a period of 10 wk. For the control group, the mice
were treated with BHI. Mice were euthanized at week 26, and
tongue, spleen, and draining lymph node were collected. The
gross examination of tongue lesions was performed according
to a previous study (Wu et al. 2018). All experimental handing
was conducted in compliance with the ARRIVE (Animal
Research: Reporting of In Vivo Experiments) guidelines. The
study procedure was approved by the Animal Care and Use
Committee of SYSU.
Detection of Bacteria by Immunohistochemistry
and Fluorescence In Situ Hybridization
Immunohistochemistry (IHC) and fluorescence in situ hybrid-
ization (FISH) were employed to detect the colonization of
P. gingivalis. Consecutive 4-µm-thick paraffin sections were
cut from each block, 2 for human samples and 3 for mice. The
first section was used as a negative control, and the second and
third sections were used for the detection of P. gingivalis.
Antigen retrieval was performed under high-pressure heating
with EDTA buffer (ZLI-9067, ZSGB-BIO, pH 8.0). A mono-
clonal mouse anti-RgpB antibody (gift from the laboratory of
Jinlong Gao, University of Sydney; 1:100) was employed as
primary antibody for IHC. FISH was carried out with appropri-
ate specific probes according to the manufacturer’s instruc-
tions. FFPE tissue sections were probed with 5 mg/mL
P. gingivalis 16S ribosomal RNA-specific oligonucleotide
POGI (CAATACTCGTATCGCCCGTTATTC), 5-TGCAC
AAGGCACAACGCAACAGGGCA-3 labeled with Cy3
(Takara, Japan) dye. FISH was performed according to the
published methods (Romero-Lastra et al. 2019).
P. gingivalis Promotes OSCC Progression in an Immune Microenvironment 3
Tissue Preparation, Histology,
and Immunostaining
Harvested oral lesions for hematoxylin and eosin (H&E) stain-
ing and IHC were stained as previously described (Wen et al.
2019). Samples for immunofluorescence (IF) were embedded
in OCT compound (Sakura Tissue-Tek) and sectioned into
8-µm sections. IHC was then carried out with anti-Ki67 rabbit
polyclonal antibody (ab15580, Abcam; 1:500) and anti-Gr-1 rab-
bit polyclonal antibody (GB11229, Servicebio; 1:500). IF was
performed with anti-CD11b rabbit monoclonal antibody
(ab133357, Abcam; 1:200).
Flow Cytometry
A single cell suspension was prepared from tongue, spleen, and
draining lymph nodes of the mice. Tongues were dissected,
minced, and resuspended in complete media (RPMI 1640 with
10% FBS) supplemented with Collagenase-IV (17104019;
Gibco) at 1 mg/mL and DNase I (1121MG010; BioFroxx) at
0.1 mg/mL and incubated at 37°C for 30 min with shaking to
form a single cell. Tissue were passed through a 70-µm strainer,
collected, and washed with phosphate-buffered saline (PBS)
with 1% FBS. Spleen and draining lymph nodes were dissected
and minced to form a single cell system. Cells were processed
to live/dead cell discrimination using Fixable Viability Dye
(423105; BioLegend) and were stained for 30 min at 4°C with
the following antibodies (all from BioLegend): CD45 (103112),
CD3 (100203), CD4 (100528), CD8 (100726), CD11b
(101210), and Gr-1 (108408). All flow cytometry data acquisi-
tion was done using CytExpert (2.1.0.92; Beckman Coulter)
and analyzed using FlowJo software version 10 (TreeStar).
RNA Extraction and Quantitative Real-Time
Polymerase Chain Reaction
Total RNA was isolated from cells with TRIzol reagent
(Invitrogen) in accordance with the manufacturer’s instruc-
tions, and 500 ng total RNA was reverse transcribed using the
PrimeScript RT Reagent Kit (Perfect Real Time; Takara) for
quantitative real-time polymerase chain reaction (qPCR).
qPCR was performed in triplicate with an Applied LightCycler
96 quantitative PCR system (Roche). The Ct values obtained
from different samples were compared using the 2DDCt method.
β-Actin served as an internal reference gene. Primers used in
this study are shown in Appendix Table 1.
MDSC Isolation and Chemotaxis Assays
MDSCs were isolated from mice bone marrow. Bone marrow
was dissected, minced, and resuspended in complete media
(DMEM with 10% FBS). Then, the bone marrow cells were
passed through a 70-µm strainer, collected, and washed with
PBS with 1% FBS. After staining with CD11b (101210;
BioLegend) and Gr-1 (108408; BioLegend), MDSCs were iso-
lated by BD FACSAria Fusion.
Chemotaxis assays were performed using 24-well plates
with 5-µm pore size inserts (3421; Corning) according to the
manufacturer’s instructions. Then, 2.5 × 105 DOK cells with or
without coculture with P. gingivalis 33277 (multiplicity of
infection [MOI] = 1:100, 24 h) in complete medium (DMEM +
10% FBS) were added to the lower chamber. Also, a total of 1
× 106 MDSCs in serum-free medium were loaded into the
upper chamber. After 24 h of incubation, migrated cells were
counted as previously described (Zhang et al. 2016). These
experiments were repeated 3 times.
Statistical Analysis
Kaplan-Meier survival curves and log-rank tests were per-
formed for overall survival and recurrence-free survival.
Imaging was reviewed by 2 certified pathologists. The positive
cells were counted under 400× magnification using ImageJ
software (National Institutes of Health), and 5 randomly
selected independent microscopic fields were counted for each
sample to ensure that the data were representative and homo-
geneous. Differences in quantitative data between groups were
assessed using an unpaired Student’s t test. Comparisons of
means among multiple groups were performed by 1-way anal-
ysis of variance (ANOVA) tests. Measurements are expressed
as the mean ± standard deviation (SD). All statistical analyses
and graphs were generated with GraphPad Prism version 7.0
(GraphPad Software). P values less than 0.05 were considered
statistically significant (*P < 0.05, **P < 0.01, ***P < 0.001,
****P < 0.0001).
Results
Existence of P. gingivalis Is Associated with Oral
Squamous Cell Carcinoma Patient Mortality
To examine whether P. gingivalis is related to the prognosis of
patients with OSCC, we measured the relative abundance of
P. gingivalis in 56 OSCC FFPE samples using IHC staining.
According to the staining intensity, patients were classified as
P. gingivalis negative (0%, n = 22) or P. gingivalis positive
(>0%, n = 34) (Fig. 1A). Patients diagnosed with OSCC were
reviewed by 2 certified pathologists. Among the 56 OSCC
cases, 19 deaths occurred, including 15 deaths of P. gingivalis
positive patients, during a median patient follow-up period of
5.25 y (interquartile range [IQR], 1.16–5.71). We performed
univariate regression analyses and multivariate regression
analyses of baseline information in this cohort, including
P. gingivalis infection, age, location, T stage, lymphatic metas-
tasis, smoking, and alcoholic drinking (Appendix Tables 2 and
3). In our cohort study, colonization with P. gingivalis was sig-
nificantly associated with shorter overall survival (OS; hazard
ratio [HR] = 2.928; 95% confidence interval [CI], 1.183–7.247;
P < 0.05 by log-rank test; Fig. 1B, left). We also observed
4 Journal of Dental Research 00(0)
nonsignificantly higher abundance of P. gingivalis associated
with recurrence-free survival (RFS; HR = 1.492; 95% CI,
0.4775–4.695; P > 0.05 by log-rank test; Fig. 1B, right).
Consequently, our results showed that infection with P. gingi-
valis was associated with mortality in the patients with OSCC.
P. gingivalis Promotes Tumor Progression of
Oral Mucosa Epithelium in 4NQO-Induced Mice
P. gingivalis infection was related to a poor outcome in patients
with OSCC, which prompted us to investigate whether chronic
P. gingivalis infection could promote the progression of oral
squamous cell carcinoma. In our preliminary experiment, we
administered P.g ATCC33277 (P. gingivalis group),
Streptococcus mutans UA159 (S. mutans group), or BHI broth
(control group) to 4NQO-inducted mice by oral gavage (Appendix
Fig. 1). S. mutans was used as a negative control since
S. mutans has not been shown to have an obvious effect on oral
tumor progression (Gholizadeh et al. 2016; Orlandi et al. 2019).
A diagram of our experimental protocol is shown in Figure
2A. As shown in Figure 2B, 10 wk after P. gingivalis adminis-
tration, lesions in 4NQO-induced mice progressed to white
masses with a cauliflower-like or verrucous appearance, while
the control group still exhibited a wrinkled paper-like lesion
appearance. Notably, we found that the P. gingivalis group
showed a significant increase in lesion number (2.50 ± 1.22 vs.
1.17 ± 0.41, P < 0.05) and lesion diameter (2.61 ± 2.40 mm vs.
1.20 ± 0.29 mm, P < 0.05) (Fig. 2C). In addition, during the
course of administration, we observed that the P. gingivalis
group had significant weight loss compared to
the control group (Fig. 2D).
Then, H&E staining showed that mice with
or without introduction of P. gingivalis exhib-
ited different stages of oral carcinogenesis (Fig.
2E). A 2-category system was used to analyze
the oral cancer risk (normal/hyperplasia/mild
or moderate dysplasia: low risk; severe dyspla-
sia/invasive cancer: high risk) of mice accord-
ing to a previous study (Wu et al. 2018). In the
P. gingivalis group, 9 of 12 of mice showed a
high risk of oral carcinogenesis, compared to 1
of 6 in the control group (Appendix Table 4).
Moreover, P. gingivalis–treated mice showed
increased basal proliferation of hyperplastic
regions in the tongue (Fig. 2F). These data sug-
gest that P. gingivalis infection of oral mucosa
can promote oral carcinogenesis.
P. gingivalis Promotes Tumor
Progression by Colonizing Oral Tissue
A previous study reported that P. gingivalis is a
type of intracellular colonizing bacteria that
invades, replicates, and survives in human pri-
mary gingival epithelial cells. Therefore, we
hypothesized that P. gingivalis could accelerate
tumorigenesis by invading and colonizing oral tissues in mice.
To test our hypothesis, we detected the existence of P. gingiva-
lis by IHC and FISH. As shown in Figure 3A and 3B, P. gingi-
valis was always found in the P. gingivalis group (12/12) and
was not found in the control group (0/6) (P < 0.0001). In addi-
tion, we observed P. gingivalis was present not only in the epi-
thelial layer in mouse tissue (Fig. 3A, red arrows) but also in
the lamina propria (Fig. 3A, blue arrows), which was also
shown in human OSCC samples (Fig. 1A, red and blue arrows).
P. gingivalis Infection Expands CD11b+ Myeloid
Cells and MDSCs in Local Lesions
Immune cells and their effectors are key components of tumors
and promote neoplastic progression. To determine whether
P. gingivalis contributes to tumorigenesis by affecting intratu-
moral immune cells, we characterized infiltrating immune
cells from lesions, the spleen, and draining lymph nodes of the
P. gingivalis group and control group. Appendix Fig. 2A shows
the gating strategy for analysis of immune cells in oral lesions.
Interestingly, we observed an increase in infiltrating CD11b+
myeloid cells (P < 0.001) and MDSCs (P < 0.01) in the lesions
of the P. gingivalis group (Fig. 4A), while the populations of
CD3+, CD4+, and CD8+ T lymphocytes were not significantly
different (Fig. 4B). The populations of CD3+, CD4+, and CD8+
T lymphocytes; CD11b+ myeloid cells; and MDSCs in spleen
and draining lymph nodes showed no differences between the
2 groups (Appendix Fig. 2B, C). To further confirm the changes
in the immune microenvironment induced by P. gingivalis, we
Figure 1. Intratumoral Porphyromonas gingivalis infection is associated with
outcome in patients with oral squamous cell carcinoma (OSCC). (A) Representative
immunohistochemistry-stained paraffin sections of P. gingivalis–negative and P. gingivalis
positive group using monoclonal mouse anti-RapB antibody in human OSCC samples (red
arrows, P. gingivalis infection at the epithelial layer; blue arrows, P. gingivalis infection at the
lamina propria). Scale bar = 20 μm. (B) Overall survival (left) and recurrence-free survival
(right) were compared between the patients with or without P. gingivalis infection.
P. gingivalis Promotes OSCC Progression in an Immune Microenvironment 5
Figure 2. Porphyromonas gingivalis infection promoted oral carcinogenesis in 4-nitroquinoline-1 oxide (4NQO)–induced mice. (A) Diagram of the
experimental protocol. (B) Representative images of tongue tissues of control group (n = 6) and P. gingivalis group (n = 12). (C) The average oral lesion
numbers and average lesion size per mice in the control group and P. gingivalis group. (D) The body weights of mice in the control and P. gingivalis
group. The data are represented as mean ± SEM. (E) Representative hematoxylin and eosin sections of pathology, including mild dysplasia, moderate
dysplasia, severe dysplasia, and invasive carcinoma. Scale bar = 50 μm. (F) Representative immunostaining for Ki67 in oral lesions. The percentage of
proliferating cells was evaluated in severe dysplasia from the control and P. gingivalis groups using a Ki67 rabbit polyclonal antibody. Scale bar = 20 μm.
*P < 0.05. **P < 0.01. ***P < 0.001.
6 Journal of Dental Research 00(0)
Figure 3. Porphyromonas gingivalis could accelerate tumorigenesis by invading and colonizing oral tissues in mice. (A) Representative
immunohistochemistry images and analysis of paraffin tissue sections in oral lesions from the control group and P. gingivalis group (yellow-brown,
P. gingivalis; red arrows, P. gingivalis infection in epithelial layer; blue arrows, infection in lamina propria). Scale bars = 20 μm. (B) Representative
fluorescence in situ hybridization images and analysis of paraffin tissue sections of 2 groups (red, P. gingivalis). Scale bars = 20 μm. ****P < 0.0001.
P. gingivalis Promotes OSCC Progression in an Immune Microenvironment 7
Figure 4. Porphyromonas gingivalis infection selectively recruits CD11b+ myeloid cells and myeloid-derived suppressor cells (MDSCs) in oral lesions of
4-nitroquinoline-1 oxide (4NQO)–induced mice. (A) The proportion of CD11b+ myeloid cells and MDSCs in oral lesions from the control and
P. gingivalis groups. (B) The proportion of CD3+, CD4+, and CD8+ T cells in oral lesions from the control and P. gingivalis groups. (C) Representative
immunofluorescence images and analysis of CD11b expression in oral lesions between the 2 groups. And representative immunohistochemistry
staining images and analysis of Gr-1 expression in oral lesions between the 2 groups. Scale bars = 20 μm. *P < 0.05. **P < 0.01. ***P < 0.001.
8 Journal of Dental Research 00(0)
subjected FFPE samples from the P. gingivalis group and con-
trol group to IF (with an anti-CD11b antibody) and IHC stain-
ing (with an anti-Gr-1 antibody). As shown in Figure 4C,
infiltration of CD11b+ myeloid cells and MDSCs was increased
in the P. gingivalis group, which was consistent with the pri-
mary results. We also found that in the clinical OSCC samples,
the presence of P. gingivalis was associated with more MDSC
influx than those tumors free of P. gingivalis (P < 0.05)
(Appendix Fig. 3).
P. gingivalis Infection Is Associated
with the Recruitment of MDSCs In Vitro
MDSCs are immature myeloid cells that have potent immuno-
suppressive activity. Given our findings of P. gingivalis
induced MDSC expansion in mouse oral precancerous lesions,
we investigated the potential mechanisms. First, to explore the
relationship between P. gingivalis infection and MDSC recruit-
ment, we established a coculture system with Transwell mem-
branes (5 µm) in vitro. MDSCs were added to medium without
FBS in the upper chamber, and P. gingivalis, DOK cells, or
P. gingivalis–infected DOK cells (MOI = 1:100, 24 h) in 10%
FBS were added to the lower chamber (Fig. 5A). After 24 h of
incubation, the number of migrating MDSCs was significantly
elevated following coculture with P. gingivalis–infected DOK
cells compared to those cultured only with P. gingivalis or
DOK cells (Fig. 5B). In addition, we extracted RNA from
DOK cells infected with P. gingivalis for 24 h (MOI = 1:100)
and analyzed the expression levels of CCL2, CXCL2, CXCL5,
CCL5, IL-6, and IL-8, which have been described as potential
candidate genes for oral cancer in response to chronic infection
with P. gingivalis (Geng et al. 2019) and the recruitment and
expansion of MDSCs (Zhang et al. 2016). DOK cells without
P. gingivalis infection were cultured at the same time as con-
trol. DOK cells exposed to P. gingivalis showed significantly
increased expression of CCL2 (P < 0.05),
CXCL2 (P < 0.01), IL-6 (P < 0.01), and
IL-8 (P < 0.0001) compared to uninfected
DOK cells (Fig. 5C).
Discussion
P. gingivalis is regarded as a keystone
pathogen of periodontitis because of its
ability to disrupt the host immune
response, and it has been reported to be
associated with OSCC progression.
Therefore, we investigated whether it does
play an important role in OSCC progres-
sion. First, our cohort study showed that
the colonization of tumors by P. gingivalis
was negatively correlated with overall
survival in patients with OSCC. Recently,
many in vitro studies have provided
insights into the effects of P. gingivalis in
promoting oral cancer progression, including activation of cell
proliferation, inhibition of apoptosis, and promotion of cellular
invasion (Perera et al. 2016). However, the potential effect of P.
gingivalis on the immune microenvironment during tumor pro-
gression of oral cancer is poorly understood. In the present
study, we found that P. gingivalis invades oral precancerous
lesions and recruits the myeloid-derived suppressor cells by
expressing chemokines such as CCL2 and CXCL2 and cyto-
kines such as IL-6 and IL-8.
Entry of microbes and/or microbial metabolites into the
tumor microenvironment promotes neoplastic progression by
eliciting tumor-potentiating immune cell responses (Jobin
2012). Studies in mice using micro-osmotic pumps implanted
into the lumbodorsal region to deliver P. gingivalis showed that
the percentage of MDSCs was increased in the bone marrow
and spleen (Su et al. 2017). Interestingly, we observed a sig-
nificant increase in infiltrating CD11b+ myeloid cells and
MDSCs in local sites of oral lesions resulting from infection of
P. gingivalis in mice exposed to 4NQO. Moreover, P. gingiva-
lis presence was associated with more MDSC influx than those
tumors free of P. gingivalis in the OSCC samples. An in vitro
study showed that P. gingivalis can upregulate the MDSC
recruitment-related genes and recruit MDSCs. The main fea-
ture of MDSCs in the tumor environment is their potent immu-
nosuppressive activity, which is often related to poor patient
survival (Kumar et al. 2016). This indicates that during the
process of oral tumorigenesis, myeloid cells, especially
MDSCs, create an immunosuppressive microenvironment that
favors tumor progression.
It is well known that tumor-produced chemokines or cyto-
kines cause MDSCs to be trafficked through the circulatory
system or migrate into solid tumors. Among cytokines, IL-6
and IL-8 have been identified as major cytokines involved in
P. gingivalis infection (Liu et al. 2014; Yee et al. 2014). IL-6
has been reported as a promising predictor for the early diagno-
sis of tongue squamous cell carcinoma (Hussein et al. 2018). In
Figure 5. Porphyromonas gingivalis infection promoted myeloid-derived suppressor cell (MDSC)
migration in vitro, and it was associated with interleukin (IL)–6, IL-8, CCL2, and CXCL2
expression. (A) Schematic diagram of MDSC chemotaxis assays. MDSCs were seeded into the
upper chamber of a Transwell system, and P. gingivalis and dysplastic oral keratinocyte (DOK) cells
with or without P. gingivalis infection were added to the lower chamber. (B) The analysis of MDSC
chemotaxis assays. (C) The relative expression level of CCL2, CXCL2, CXCL5, CCL5, IL-6, and
IL-8 between uninfected DOK cells and DOK cells infected with P. gingivalis. *P < 0.05. **P < 0.01.
***P < 0.001. ****P < 0.0001.
P. gingivalis Promotes OSCC Progression in an Immune Microenvironment 9
addition, IL-6 has been reported as a candidate to illustrate the
role of P. gingivalis infection in promoting OSCC initiation
and progression (Geng et al. 2019). Furthermore, IL-6 and
IL-1β play an important role in driving both the accumulation
and suppressive potency of murine MDSCs (Bunt et al. 2007).
IL-8, which is frequently secreted in the tumor microenviron-
ment, promotes tumor progression through chemotaxis of
MDSCs (Alfaro et al. 2017). Whether P. gingivalis may have a
direct and an indirect impact on the production of IL-8 in our
study remains unknown. P. gingivalis expresses a variety of
virulence factors that play different roles in subverting the host
immune response (Zenobia and Hajishengallis 2015). Different
virulence factors of P. gingivalis may exert contrasting influ-
ence on the production of IL-8 through various mechanisms
(Isberg et al. 2013; Zhang and Li 2015), and our study shows
that P. gingivalis can promote IL-8 production of DOKs in
vitro. Therefore, whether P. gingivalis may have a direct and
indirect impact on the production of IL-8 may depend on which
virulence factor play a leading role in our model. Regarding
chemokines, CCL2 has been reported to promote colorectal
carcinogenesis by enhancing the population and function of
polymorphonuclear MDSCs (Chun et al. 2015). Another che-
mokine, CXCL2, promotes the generation of monocytic
MDSCs (Shi et al. 2018). Furthermore, CXCL5 and CCL5 are
also associated with tumor progression (Ban et al. 2017; Najjar
et al. 2017). Our results showed that CCL2, CXCL2, IL-6, and
IL-8 were significantly increased in DOK cells after exposure
to P. gingivalis in vitro, indicating that these chemokines and
cytokines are involved in the recruitment of MDSCs, which
contributes to tumor progression of oral cancer.
To our knowledge, this is the first report that illustrates that
P. gingivalis mediates oral carcinogenesis by recruiting
MDSCs, and we realize that our current study has some limita-
tions. The mechanism by which P. gingivalis affects the func-
tion of MDSCs in the microenvironment needs to be explored.
In conclusion, P. gingivalis promotes tumor progression by
recruiting MDSCs via increasing secretion of IL-6, IL-8,
CCL2, and CXCL2 from infected oral dysplastic keratino-
cytes. These findings suggest that P. gingivalis is another
potential target for the management of tumor progression of
oral cancer.
Author Contributions
L. Wen, W. Mu, H. Lu, contributed to design, data acquisition, and
analysis, drafted and critically revised the manuscript; X. Wang, J.
Fang, Y. Jia, contributed to data analysis and interpretation, criti-
cally revised the manuscript; Q. Li, D. Wang, S. Wen, J. Guo, W.
Dai, contributed to data acquisition and analysis, critically revised
the manuscript; X. Ren, J. Cui, G. Zeng, contributed to data inter-
pretation, critically revised the manuscript; J. Gao, contributed to
conception and design, critically revised the manuscript; Z. Wang,
B. Cheng, contributed to conception and design, drafted and criti-
cally revised the manuscript. All authors gave final approval and
agree to be accountable for all aspects of the work.
Acknowledgments
This project was supported by grants from the National Natural
Science Foundation of China (No. 81772896, 81630025, and
81972532), the Science and Technology Planning Project of
Guangzhou City of China (No. 2017004020102), the Science and
Technology Program of Guangzhou city of China (No. 20180
4010144), and the China Postdoctoral Science Foundation (No.
2019TQ0388). The authors declare no potential conflicts of inter-
est with respect to the authorship and/or publication of this
article.
References
Ahn J, Segers S, Hayes RB. 2012. Periodontal disease, Porphyromonas
gingivalis serum antibody levels and orodigestive cancer mortality.
Carcinogenesis. 33(5):1055–1058.
Alfaro C, Sanmamed MF, Rodríguez-Ruiz ME, Teijeira Á, Oñate C, González
Á, Ponz M, Schalper KA, Pérez-Gracia JL, Melero I. 2017. Interleukin-8 in
cancer pathogenesis, treatment and follow-up. Cancer Treat Rev. 60:24–31.
Amieva M, Peek RM Jr. 2016. Pathobiology of helicobacter pylori-induced
gastric cancer. Gastroenterology. 150(1):64–78.
Ban Y, Mai J, Li X, Mitchell-Flack M, Zhang T, Zhang L, Chouchane L, Ferrari
M, Shen H, Ma X. 2017. Targeting autocrine CCLl5-CCR5 axis reprograms
immunosuppressive myeloid cells and reinvigorates antitumor immunity.
Cancer Res. 77(11):2857–2868.
Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. 2018. Global
cancer statistics 2018: Globocan estimates of incidence and mortality world-
wide for 36 cancers in 185 countries. CA Cancer J Clin. 68(6):394–424.
Bunt SK, Yang L, Sinha P, Clements VK, Leips J, Ostrand-Rosenberg S. 2007.
Reduced inflammation in the tumor microenvironment delays the accumu-
lation of myeloid-derived suppressor cells and limits tumor progression.
Cancer Res. 67(20):10019–10026.
Chen Y, Li Q, Li X, Ma D, Fang J, Luo L, Liu X, Wang X, Lui VWY, Xia J,
et al. 2018. Blockade of PD-1 effectively inhibits in vivo malignant trans-
formation of oral mucosa. Oncoimmunology. 7(2):e1388484.
Chi AC, Day TA, Neville BW. 2015. Oral cavity and oropharyngeal squamous
cell carcinoma—an update. CA Cancer J Clin. 65(5):401–421.
Chocolatewala N, Chaturvedi P, Desale R. 2010. The role of bacteria in oral
cancer. Indian J Med Paediatr Oncol. 31(4):126–131.
Chun E, Lavoie S, Michaud M, Gallini CA, Kim J, Soucy G, Odze R, Glickman
JN, Garrett WS. 2015. CCL2 promotes colorectal carcinogenesis by
enhancing polymorphonuclear myeloid-derived suppressor cell population
and function. Cell Rep. 12(2):244–257.
Geng F, Wang Q, Li C, Liu J, Zhang D, Zhang S, Pan Y. 2019. Identification of
potential candidate genes of oral cancer in response to chronic infection with
Porphyromonas gingivalis using bioinformatical analyses. Front Oncol. 9:91.
Gholizadeh P, Eslami H, Yousefi M, Asgharzadeh M, Aghazadeh M, Kafil
HS. 2016. Role of oral microbiome on oral cancers, a review. Biomed
Pharmacother. 84:552–558.
Hajishengallis G. 2015. Periodontitis: from microbial immune subversion to
systemic inflammation. Nat Rev Immunol. 15(1):30–44.
Hajishengallis G, Darveau RP, Curtis MA. 2012. The keystone-pathogen
hypothesis. Nat Rev Microbiol. 10(10):717–725.
Hussein AA, Forouzanfar T, Bloemena E, de Visscher J, Brakenhoff RH,
Leemans CR, Helder MN. 2018. A review of the most promising biomark-
ers for early diagnosis and prognosis prediction of tongue squamous cell
carcinoma. Br J Cancer. 119(6):724–736.
Isberg RR, Takeuchi H, Hirano T, Whitmore SE, Morisaki I, Amano A, Lamont
RJ. 2013. The serine phosphatase SerB of Porphyromonas gingivalis sup-
presses IL-8 production by dephosphorylation of NF-κB RelA/p65. PLoS
Pathog. 9(4):e1003326.
Jobin C. 2012. Colorectal cancer: CRC—all about microbial products and bar-
rier function? Nat Rev Gastroenterol Hepatol. 9(12):694–696.
Katz J, Onate MD, Pauley KM, Bhattacharyya I, Cha S. 2011. Presence of
Porphyromonas gingivalis in gingival squamous cell carcinoma. Int J Oral
Sci. 3(4):209–215.
Kumar V, Patel S, Tcyganov E, Gabrilovich DI. 2016. The nature of myeloid-
derived suppressor cells in the tumor microenvironment. Trends Immunol.
37(3):208–220.
10 Journal of Dental Research 00(0)
Liu J, Wang Y, Ouyang X. 2014. Beyond Toll-like receptors: Porphyromonas
gingivalis induces IL-6, IL-8, and VCAM-1 expression through NOD-
mediated NF-κB and ERK signaling pathways in periodontal fibroblasts.
Inflammation. 37(2):522–533.
Mehanna H, Beech T, Nicholson T, El-Hariry I, McConkey C, Paleri V, Roberts
S. 2013. Prevalence of human papillomavirus in oropharyngeal and nonoro-
pharyngeal head and neck cancer—systematic review and meta-analysis of
trends by time and region. Head Neck. 35(5):747–755.
Mehrtash H, Duncan K, Parascandola M, David A, Gritz ER, Gupta PC,
Mehrotra R, Amer Nordin AS, Pearlman PC, Warnakulasuriya S, et al.
2017. Defining a global research and policy agenda for betel quid and areca
nut. Lancet Oncol. 18(12):e767–e775.
Michaud DS, Izard J, Wilhelm-Benartzi CS, You DH, Grote VA, Tjonneland A,
Dahm CC, Overvad K, Jenab M, Fedirko V, et al. 2013. Plasma antibodies
to oral bacteria and risk of pancreatic cancer in a large European prospec-
tive cohort study. Gut. 62(12):1764–1770.
Najjar YG, Rayman P, Jia X, Pavicic PG, Jr., Rini BI, Tannenbaum C, Ko J,
Haywood S, Cohen P, Hamilton T, et al. 2017. Myeloid-derived suppres-
sor cell subset accumulation in renal cell carcinoma parenchyma is associ-
ated with intratumoral expression of IL1β, IL8, CXCl5, and Mip-1α. Clin
Cancer Res. 23(9):2346–2355.
Orlandi E, Iacovelli NA, Tombolini V, Rancati T, Polimeni A, De Cecco L,
Valdagni R, De Felice F. 2019. Potential role of microbiome in oncogen-
esis, outcome prediction and therapeutic targeting for head and neck cancer.
Oral Oncol. 99:104453.
Perera M, Al-Hebshi NN, Speicher DJ, Perera I, Johnson NW. 2016. Emerging
role of bacteria in oral carcinogenesis: a review with special reference to
perio-pathogenic bacteria. J Oral Microbiol. 8:32762.
Peters BA, Wu J, Pei Z, Yang L, Purdue MP, Freedman ND, Jacobs EJ, Gapstur
SM, Hayes RB, Ahn J. 2017. Oral microbiome composition reflects pro-
spective risk for esophageal cancers. Cancer Res. 77(23):6777–6787.
Romero-Lastra P, Sanchez MC, Llama-Palacios A, Figuero E, Herrera D, Sanz
M. 2019. Gene expression of Porphyromonas gingivalis ATCC 33277 when
growing in an in vitro multispecies biofilm. PLoS One. 14(8):e0221234.
Sayehmiri F, Sayehmiri K, Asadollahi K, Soroush S, Bogdanovic L, Jalilian FA,
Emaneini M, Taherikalani M. 2015. The prevalence rate of Porphyromonas
gingivalis and its association with cancer: a systematic review and meta-
analysis. Int J Immunopathol Pharmacol. 28(2):160–167.
Shi H, Han X, Sun Y, Shang C, Wei M, Ba X, Zeng X. 2018. Chemokine
(C-X-C motif) ligand 1 and CXCL2 produced by tumor promote the
generation of monocytic myeloid-derived suppressor cells. Cancer Sci.
109(12):3826–3839.
Su L, Xu Q, Zhang P, Michalek SM, Katz J. 2017. Phenotype and function
of myeloid-derived suppressor cells induced by Porphyromonas gingivalis
infection. Infect Immun. 85(8). pii: e00213-17.
Wen L, Lu H, Li Q, Li Q, Wen S, Wang D, Wang X, Fang J, Cui J, Cheng B,
et al. 2019. Contributions of T cell dysfunction to the resistance against
anti-PD-1 therapy in oral carcinogenesis. J Exp Clin Cancer Res. 38(1):299.
Winn DM, Lee YC, Hashibe M, Boffetta P; INHANCE Consortium. 2015. The
INHANCE consortium: toward a better understanding of the causes and
mechanisms of head and neck cancer. Oral Dis. 21(6):685–693.
Wu JS, Zheng M, Zhang M, Pang X, Li L, Wang SS, Yang X, Wu JB, Tang
YJ, Tang YL, et al. 2018. Porphyromonas gingivalis promotes 4-nitroquin-
oline-1-oxide-induced oral carcinogenesis with an alteration of fatty acid
metabolism. Front Microbiol. 9:2081.
Yee M, Kim S, Sethi P, Duzgunes N, Konopka K. 2014. Porphyromonas gingi-
valis stimulates IL-6 and IL-8 secretion in GMSM-K, HSC-3 and H413 oral
epithelial cells. Anaerobe. 28:62–67.
Yu T, Guo F, Yu Y, Sun T, Ma D, Han J, Qian Y, Kryczek I, Sun D, Nagarsheth
N, et al. 2017. Fusobacterium nucleatum promotes chemoresistance to
colorectal cancer by modulating autophagy. Cell. 170(3):548–563.e16.
Zenobia C, Hajishengallis G. 2015. Porphyromonas gingivalis virulence factors
involved in subversion of leukocytes and microbial dysbiosis. Virulence.
6(3):236–243.
Zhang H, Ye YL, Li MX, Ye SB, Huang WR, Cai TT, He J, Peng JY, Duan TH,
Cui J, et al. 2016. CXCLl2/MIF-CXCR2 signaling promotes the recruit-
ment of myeloid-derived suppressor cells and is correlated with prognosis
in bladder cancer. Oncogene. 36(15):2095–2104.
Zhang Y, Li X. 2015. Lipopolysaccharide-regulated production of bone sia-
loprotein and interleukin-8 in human periodontal ligament fibroblasts: the
role of toll-like receptors 2 and 4 and the MAPK pathway. J Periodontal
Res. 50(2):141–151.
... Chronic inflammation related with presence of P. gingivalis and immunosuppressive capacity of MDSCs have been associated to cancer. Thus, the infection with this bacterium and the biological effects on MDSCs have been analysed in the context of cancer (77,78). Oral treatment with P. gingivalis in mice models of oral squamous cell carcinoma and esophageal squamous cell carcinoma can promote more malignant progression and recruitment of MDSCs (77,78). ...
... Thus, the infection with this bacterium and the biological effects on MDSCs have been analysed in the context of cancer (77,78). Oral treatment with P. gingivalis in mice models of oral squamous cell carcinoma and esophageal squamous cell carcinoma can promote more malignant progression and recruitment of MDSCs (77,78). It was suggested that P. gingivalis can colonize oral precancerous lesions and provoke that the keratinocytes express molecules such as CCL2, CXCL2, IL-6, and IL-8, which result in the MDSCs recruitment (78). ...
... Oral treatment with P. gingivalis in mice models of oral squamous cell carcinoma and esophageal squamous cell carcinoma can promote more malignant progression and recruitment of MDSCs (77,78). It was suggested that P. gingivalis can colonize oral precancerous lesions and provoke that the keratinocytes express molecules such as CCL2, CXCL2, IL-6, and IL-8, which result in the MDSCs recruitment (78). Also, P. gingivalis can colonize esophageal cancer tissues and infection in a mouse model of esophageal squamous cell carcinoma, the bacteria promote expansion of MDSCs in spleen (77). ...
Article
Full-text available
The oral cavity presents a diverse microbiota in a dynamic balance with the host. Disruption of the microbial community can promote dysregulation of local immune response which could generate oral diseases. Additionally, alterations in host immune system can result in inflammatory disorders. Different microorganisms have been associated with establishment and progression of the oral diseases. Oral cavity pathogens/diseases can modulate components of the inflammatory response. Myeloid-derived suppressor cells (MDSCs) own immunoregulatory functions and have been involved in different inflammatory conditions such as infectious processes, autoimmune diseases, and cancer. The aim of this review is to provide a comprehensive overview of generation, phenotypes, and biological functions of the MDSCs in oral inflammatory diseases. Also, it is addressed the biological aspects of MDSCs in presence of major oral pathogens. MDSCs have been mainly analyzed in periodontal disease and Sjögren’s syndrome and could be involved in the outcome of these diseases. Studies including the participation of MDSCs in other important oral diseases are very scarce. Major oral bacterial and fungal pathogens can modulate expansion, subpopulations, recruitment, metabolism, immunosuppressive activity and osteoclastogenic potential of MDSCs. Moreover, MDSC plasticity is exhibited in presence of oral inflammatory diseases/oral pathogens and appears to be relevant in the disease progression and potentially useful in the searching of possible treatments. Further analyses of MDSCs in oral cavity context could allow to understand the contribution of these cells in the fine-tuned balance between host immune system and microorganism of the oral biofilm, as well as their involvement in the development of oral diseases when this balance is altered.
... Meanwhile, P. gingivalis colonization was 6.5 times higher in the oral microbiota of cancer patients compared to the healthy controls in this study. The underlying mechanism for P. gingivalis, despite being regarded as a significant etiological agent in oral cavity malignancies, is unknown [41]. . In contrast to the situation in cancer, G. haemolysans decreases the oral microbiota of periodontitis patients and inhibits P.gingivalis by secreting protein components in vitro [42]. ...
Article
Objectives The pathogenesis of oral cavity cancers is complex. We tested the hypothesis that oral microbiota dysbiosis is associated with oral cavity cancer. Materials and methods Patients with primary oral cavity cancer who met the inclusion and exclusion criteria were included in the study. Matching healthy individuals were recruited as controls. Data on socio-demographic and behavioral factors, self-reported periodontal measures and habits, and current dental status were collected using a structured questionnaire and periodontal chartings. In addition to self-reported oral health measures, each participant received a standard and detailed clinical examination. DNA was extracted from saliva samples from patients and healthy controls. Next-generation sequenc-ing was performed by targeting V3-V4 gene regions of the 16 S rRNA with subsequent bioinformatic analyses. Results Patients with oral cavity cancers had a lower quality of oral health than healthy controls. Proteobacteria, Aggrega-tibacter, Haemophilus, and Neisseria decreased, while Firmicutes, Bacteroidetes, Actinobacteria, Lactobacillus, Gemella, and Fusobacteria increased in oral cancer patients. At the species level, C. durum, L. umeaens, N. subflava, A. massiliensis, and V. dispar were significantly lower, while G. haemolysans was significantly increased (p < 0.05). Major periodontopatho-gens associated with periodontal disease (P. gingivalis and F.nucleatum) increased 6.5-and 2.8-fold, respectively. Conclusion These data suggested that patients with oral cancer had worse oral health conditions and a distinct oral micro-biome composition that is affected by personal daily habits and may be associated with the pathogenicity of the disease and interspecies interactions. Clinical relevance This paper demonstrates the link between oral bacteria and oral cancers, identifying mechanistic interactions between species of oral microbiome.
... Meanwhile, P. gingivalis colonization was 6.5 times higher in the oral microbiota of cancer patients compared to the healthy controls in this study. The underlying mechanism for P. gingivalis, despite being regarded as a significant etiological agent in oral cavity malignancies, is unknown [41]. . In contrast to the situation in cancer, G. haemolysans decreases the oral microbiota of periodontitis patients and inhibits P.gingivalis by secreting protein components in vitro [42]. ...
Article
Full-text available
Objectives The pathogenesis of oral cavity cancers is complex. We tested the hypothesis that oral microbiota dysbiosis is associated with oral cavity cancer. Materials and methods Patients with primary oral cavity cancer who met the inclusion and exclusion criteria were included in the study. Matching healthy individuals were recruited as controls. Data on socio-demographic and behavioral factors, self-reported periodontal measures and habits, and current dental status were collected using a structured questionnaire and periodontal chartings. In addition to self-reported oral health measures, each participant received a standard and detailed clinical examination. DNA was extracted from saliva samples from patients and healthy controls. Next-generation sequencing was performed by targeting V3-V4 gene regions of the 16 S rRNA with subsequent bioinformatic analyses. Results Patients with oral cavity cancers had a lower quality of oral health than healthy controls. Proteobacteria, Aggregatibacter, Haemophilus, and Neisseria decreased, while Firmicutes, Bacteroidetes, Actinobacteria, Lactobacillus, Gemella, and Fusobacteria increased in oral cancer patients. At the species level, C. durum, L. umeaens, N. subflava, A. massiliensis, and V. dispar were significantly lower, while G. haemolysans was significantly increased (p < 0.05). Major periodontopathogens associated with periodontal disease (P. gingivalis and F.nucleatum) increased 6.5- and 2.8-fold, respectively. Conclusion These data suggested that patients with oral cancer had worse oral health conditions and a distinct oral microbiome composition that is affected by personal daily habits and may be associated with the pathogenicity of the disease and interspecies interactions. Clinical relevance This paper demonstrates the link between oral bacteria and oral cancers, identifying mechanistic interactions between species of oral microbiome.
... Fusobacterium sp. have been found to be associated with various tumorigenic processes in other organisms (Bullman et al., 2017;Fujiwara et al., 2020). Porphyromonas gingivalis, a member of Porphyromonas sp., is a well-studied periodontal pathogen (Reyes, 2021) that can promote tumor development by creating a carcinogenic microenvironment (Wen et al., 2020). Furthermore, Campylobacter and Neisseria species are known to cause various diseases, including diarrheal diseases, periodontitis, and other chronic conditions (Baral et al., 2007;Man, 2011), in both humans and animals . ...
Article
Full-text available
Introduction Microbial community composition is closely associated with host disease onset and progression, underscoring the importance of understanding host–microbiota dynamics in various health contexts. Methods In this study, we utilized full-length 16S rRNA gene sequencing to conduct species-level identification of the microorganisms in the oral cavity of a giant panda (Ailuropoda melanoleuca) with oral malignant fibroma. Results We observed a significant difference between the microbial community of the tumor side and non-tumor side of the oral cavity of the giant panda, with the latter exhibiting higher microbial diversity. The tumor side was dominated by specific microorganisms, such as Fusobacterium simiae, Porphyromonas sp. feline oral taxon 110, Campylobacter sp. feline oral taxon 100, and Neisseria sp. feline oral taxon 078, that have been reported to be associated with tumorigenic processes and periodontal diseases in other organisms. According to the linear discriminant analysis effect size analysis, more than 9 distinct biomarkers were obtained between the tumor side and non-tumor side samples. Furthermore, the Kyoto Encyclopedia of Genes and Genomes analysis revealed that the oral microbiota of the giant panda was significantly associated with genetic information processing and metabolism, particularly cofactor and vitamin, amino acid, and carbohydrate metabolism. Furthermore, a significant bacterial invasion of epithelial cells was predicted in the tumor side. Discussion This study provides crucial insights into the association between oral microbiota and oral tumors in giant pandas and offers potential biomarkers that may guide future health assessments and preventive strategies for captive and aging giant pandas.
... gingivalis) and Clostridium proved to be significantly elevated in OSCC instances compared to their benign mucosal counterparts. Intriguingly, the survival rate of OSCC patients was correlated with the localization of P. gingivalis within tumor tissues (Nagy et al., 1998;Wen et al., 2020). Lactobacillus has a dual role in the development and advancement of OSCC. ...
Article
Full-text available
The oral cavity stands as one of the pivotal interfaces facilitating the intricate interaction between the human body and the external environment. The impact of diverse oral microorganisms on the emergence and progression of various systemic cancers, typified by oral cancer, has garnered increasing attention. The potential pathogenicity of oral bacteria, notably the anaerobic Porphyromonas gingivalis and Fusobacterium nucleatum, has been extensively studied and exhibits obvious correlation with different carcinoma types. Furthermore, oral fungi and viruses are closely linked to oropharyngeal carcinoma. Multiple potential mechanisms of oral microbiota-induced carcinogenesis have been investigated, including heightened inflammatory responses, suppression of the host immune system, influence on the tumor microenvironment, anti-apoptotic activity, and promotion of malignant transformation. The disturbance of microbial equilibrium and the migration of oral microbiota play a pivotal role in facilitating oncogenic functions. This review aims to comprehensively outline the pathogenic mechanisms by which oral microbiota participate in carcinogenesis. Additionally, this review delves into their potential applications in cancer prevention, screening, and treatment. It proves to be a valuable resource for researchers investigating the intricate connection between oral microbiota and systemic cancers.
... IHC staining of P. gingivalis, downstream of kinase 3 (DOK3), and M2-TAM was consistent as follows: anti-P. gingivalis monoclonal antibody (#ab225982, Abcam, Cambridge, UK) at 1:100 dilution [21]; anti-DOK3 monoclonal antibody (#ab236609, Abcam, Cambridge, UK) at 1:500 dilution [20]; and anti-M2-TAM monoclonal antibody (CD206 + ; #MA5-44,409, ThermoFisher Scientific, Waltham. MA, USA) at 1:200 dilution of incubation. ...
Article
Full-text available
Background While there is an understanding of the association between the expression of Porphyromonas gingivalis (P. gingivalis) and prognosis of oral squamous cell carcinoma (OSCC), significance specially to address the relevance between different immunohistochemical intensities of P. gingivalis and tumor-associated macrophages (TAMs) in OSCC tissue and related clinicopathologic characteristics has not been well investigated. The present study aimed to investigate the pathological features related to M2-TAM in P. gingivalis-infected OSCC and ascertain its clinical relevance with patients’ prognosis. Methods A prospective cohort study was designed to comparatively analyze 200 patients from June 2008 to June 2020. Bioinformatics analyses were implemented to identify DOK3 as a key molecule and to appraise immunocyte infiltration using Gene Expression Omnibus and The Cancer Genome Atlas databases. Immunohistochemical evaluation was performed to analyze the association between the expression levels of P. gingivalis, DOK3, and M2-TAM and clinicopathological variables using Fisher’s exact test or Pearson’s chi-square test. Cox analysis was used to calculate hazard ratios (HR) with corresponding 95% confidence interval (CI) for various clinicopathological features. The Kaplan–Meier approach and log-rank test were used to plot the survival curves. Results The expression level of P. gingivalis was positively associated with DOK3 and M2-TAMs expression level (P < 0.001). Parameters, including body mass index, clinical stage, recurrence, tumor differentiation, and P. gingivalis, DOK3, and M2-TAM immunoexpression levels, affected the prognosis of patients with OSCC (all P < 0.05). In addition, P. gingivalis (HR = 1.674, 95%CI 1.216–4.142, P = 0.012), DOK3 (HR = 1.881, 95%CI 1.433–3.457, P = 0.042), and M2-TAM (HR = 1.649, 95%CI 0.824–3.082, P = 0.034) were significantly associated with the 10-year cumulative survival rate. Conclusions Elevated expression of P. gingivalis and DOK3 indicates M2-TAM infiltration and unfavorable prognosis of OSCC, and could be considered as three novel independent risk factors for predicting the prognosis of OSCC.
... The promoting or suppressing effects of oral microbes on OSCC have been studied. In particular, some periodontitis-related bacteria have been suggested to be closely associated with the development of OSCC (Polak et al. 2009;Wen et al. 2020). P. intermedia, a Gram-negative anaerobic bacterium, is recognized as a common periodontal pathogen. ...
Article
Full-text available
Purpose Periodontitis-associated bacteria, such as Porphyromonas gingivalis and Fusobacterium nucleatum, are closely linked to the risk of oral squamous cell carcinoma (OSCC). Emerging studies have indicated that another common periodontal pathogen, Prevotella intermedia (P. intermedia), is enriched in OSCC and could affect the occurrence and progression of OSCC. Our aim is to determine the effects of P. intermedia on the progression of OSCC and the role of antibiotics in reversing these effects. Methods In this study, a murine xenograft model of OSCC was established, and the mice were injected intratumorally with PBS (control group), P. intermedia (P.i group), or P. intermedia combined with an antibiotic cocktail administration (P.i + ABX group), respectively. The effects of P. intermedia and ABX administration on xenograft tumor growth, invasion, angiogenesis, and metastasis were investigated by tumor volume measurement and histopathological examination. Enzyme-linked immunosorbent assay (ELISA) was used to investigate the changes in serum cytokine levels. Immunohistochemistry (IHC) was adopted to analyze the alterations in the levels of inflammatory cytokines and infiltrated immune cells in OSCC tissues of xenograft tumors. Transcriptome sequencing and analysis were conducted to determine differential expression genes among various groups. Results Compared with the control treatment, P. intermedia treatment significantly promoted tumor growth, invasion, angiogenesis, and metastasis, markedly affected the levels of inflammatory cytokines, and markedly altered M2 macrophages and regulatory T cells (Tregs) infiltration in the tumor microenvironment. However, ABX administration clearly abolished these effects of P. intermedia. Transcriptome and immunohistochemical analyses revealed that P. intermedia infection increased the expression of interferon-stimulated gene 15 (ISG15). Correlation analysis indicated that the expression level of ISG15 was positively correlated with the Ki67 expression level, microvessel density, serum concentrations and tissue expression levels of inflammatory cytokines, and quantities of infiltrated M2 macrophages and Tregs. However, it is negatively correlated with the quantities of infiltrated CD4⁺ and CD8⁺ T cells. Conclusion In conclusion, intratumoral P. intermedia infection aggravated OSCC progression, which may be achieved through upregulation of ISG15. This study sheds new light on the possible pathogenic mechanism of intratumoral P. intermedia in OSCC progression, which could be a prospective target for OSCC prevention and treatment.
... While further validation and largerscale studies are needed, this finding holds the potential to improve diagnostic accuracy (36). In addition, it has been suggested that chronic inflammation in the esophagus or colon and bacterial infection, such as Porphyromonas gingivalis, in the oral cavity may promote the proliferation and survival of malignant cells by modulating the dysplasia-related immune response (42,43). ...
Article
Full-text available
Oral leukoplakia is the most frequent and potentially malignant lesion of the oral cavity. Although dysplasia grading remains the main factor for risk assessment, challenges persist in determining the exact risk of transformation, and the literature has focused on studying alternative biomarkers. The interaction between dysplastic epithelial cells and the microenvironment starts early, and the communication is mainly mediated by lymphocytes, inflammatory factors, fibroblasts, and the extracellular matrix, leading to dysplastic progression. Leukoplakia-infiltrating leukocytes (LILs) and leukoplakia-associated fibroblasts (LAFs) play crucial roles in the dysplastic microenvironment. The immune response is related to intraepithelial T lymphocyte infiltration, mechanisms of immunosuppression coordinated by regulatory T cells, M2 macrophage polarization, and increased numbers of Langerhans cells; in contrast, fibroblastic and extracellular matrix factors are associated with increased numbers of pro-tumorigenic myofibroblasts, increased expression of metalloproteinases vs. decreased expression of TIMPs, and increased expression of chemokines and other inflammatory mediators. The microenvironment offers insights into the progression of leukoplakia to carcinoma, and understanding the complexity of the oral microenvironment in potentially malignant diseases aids in determining the risk of malignant transformation and proposing new therapeutic alternatives.
... Moreover, P. gingivalis is a known keystone pathogen related to chronic periodontitis. Clinical studies found that P. gingivalis was widely present in gingival squamous cell carcinoma tissue compared to normal gum tissue, suggesting a potential association between P. gingivalis and squamous cell gum cancer [82]. Indeed, in a specific oral microbiome, the interactions of microbial communities can also influence oral cancer. ...
Article
Full-text available
Human genital papilloma virus infection is the most prevalent sexually transmitted infection in the world. It is estimated that more than 75% of sexually active women contract this infection in their lifetime. In 80% of young women, there is the clearance of the virus within 18-24 months. In developed countries, oral squamous cell carcinoma (OSCC) is now the most frequent human papilloma virus (HPV)-related cancer, having surpassed cervical cancer, and it is predicted that by 2030 most squamous cell carcinomas will be the HPV-related rather than non-HPV-related form. However, there are currently no screening programs for oral cavity infection. While the natural history of HPV infection in the cervix is well known, in the oropharynx, it is not entirely clear. Furthermore, the prevalence of HPV in the oropharynx is unknown. Published studies have found wide-ranging prevalence estimates of 2.6% to 50%. There are also conflicting results regarding the percentage of women presenting the same type of HPV at two mucosal sites, ranging from 0 to 60%. Additionally, the question arises as to whether oral infection can develop from genital HPV infection, through oral and genital contact or by self-inoculation, or whether it should be considered an independent event. However, there is still no consensus on these topics, nor on the relationship between genital and oral HPV infections. Therefore, this literature review aims to evaluate whether there is evidence of a connection between oral and cervical HPV, while also endorsing the usefulness of the screening of oral infection in patients with high-risk cervical HPV as a means of facilitating the diagnosis and early management of HPV-related oral lesions. Finally, this review emphasizes the recommendation for the use of the HPV vaccines in primary prevention in the male and female population as the most effective means of successfully counteracting the increasing incidence of OSCC to date.
Article
Full-text available
Periodontitis is linked to the onset and progression of oral squamous cell carcinoma (OSCC), an epidemiologically frequent and clinically aggressive malignancy. In this context, Fusobacterium (F.) nucleatum and Porphyromonas (P.) gingivalis, two bacteria that cause periodontitis, are found in OSCC tissues as well as in oral premalignant lesions, where they exert pro-tumorigenic activities. Since the two bacteria are present also in endodontic diseases, playing a role in their pathogenesis, here we analyze the literature searching for information on the impact that endodontic infection by P. gingivalis or F. nucleatum could have on cellular and molecular events involved in oral carcinogenesis. Results from the reviewed papers indicate that infection by P. gingivalis and/or F. nucleatum triggers the production of inflammatory cytokines and growth factors in dental pulp cells or periodontal cells, affecting the survival, proliferation, invasion, and differentiation of OSCC cells. In addition, the two bacteria and the cytokines they induce halt the differentiation and stimulate the proliferation and invasion of stem cells populating the dental pulp or the periodontium. Although most of the literature confutes the possibility that bacteria-induced endodontic inflammatory diseases could impact on oral carcinogenesis, the papers we have analyzed and discussed herein recommend further investigations on this topic.
Article
Full-text available
Background and objectivePorphyromonas gingivalis, an oral microorganism residing in the subgingival biofilm, may exert diverse pathogenicity depending on the presence of specific virulence factors, but its gene expression has not been completely established. This investigation aims to compare the transcriptomic profile of this pathogen when growing within an in vitro multispecies biofilm or in a planktonic state.Materials and methodsP. gingivalis ATCC 33277 was grown in anaerobiosis within multi-well culture plates at 37°C under two conditions: (a) planktonic samples (no hydroxyapatite discs) or (b) within a multispecies-biofilm containing Streptococcus oralis, Actinomyces naeslundii, Veillonella parvula, Fusobacterium nucleatum and Aggregatibacter actinomycetemcomitans deposited on hydroxyapatite discs. Scanning Electron Microscopy (SEM) and Confocal Laser Scanning Microscopy (CLSM) combined with Fluorescence In Situ Hybridization (FISH) were used to verify the formation of the biofilm and the presence of P. gingivalis. Total RNA was extracted from both the multispecies biofilm and planktonic samples, then purified and, with the use of a microarray, its differential gene expression was analyzed. A linear model was used for determining the differentially expressed genes using a filtering criterion of two-fold change (up or down) and a significance p-value of
Article
Full-text available
Background: Programmed death 1 (PD-1) blockade has great effect in the prevention of oral precancerous lesions, but the drug resistance has also been observed. The determinants of immune resistance during the malignant transformation are poorly understood. Methods: Anti-PD-1 antibody was administered in the 4NQO-induced carcinogenesis mouse models. The mice were then subdivided into PD-1 resistance(PD-1R) group and PD-1 sensitive(PD-1S) group according to the efficacy. The expression of PD-1 and PD-L1, and the abundance of CD3+ T cells in tumor microenvironment between the two groups was tested by immunohistochemistry. In addition, the activation and effector functions, as well as the accumulation of immunosuppressive cells and expression of immune checkpoints of T cells in the draining lymph nodes and spleen between PD-1R and PD-1S group were analyzed by flow cytometry. Results: Our results showed that T cell infiltration in tumor microenvironment, effector T cell cytokine secretion and central memory T cell accumulation in peripheral lymphoid organs were all inhibited in the anti-PD-1 resistance group. Furthermore, we found that an increase of regulatory T cell (Treg) population contributed to the resistance of the anti-PD-1 therapy. Notably, TIM-3 was found to be the only immunosuppressive molecule that mediated the resistance to anti-PD-1 therapy in the oral malignant transformation model. Conclusions: Our findings identified a novel mechanism that T cell dysfunction contributes to the immune resistance during the malignant transformation of the oral mucosa. This study provides new targets for improving the efficacy of immunotherapy for early stage of tumorigenesis.
Article
Full-text available
Recent investigations revealed the relationship between chronic periodontitis, Porphyromonas gingivalis and cancer. However, host genes that change in response to chronic infection with P. gingivalis and may contribute to oral cancer have remained largely unknown. In the present study, we aimed to comprehensively analyze microarray data obtained from the chronic infection model of immortalized oral epithelial cells that were persistently exposed to P. gingivalis for 15 weeks. Using protein-protein interaction (PPI) networks and Ingenuity Pathway Analysis (IPA), we identified hub genes, major biological processes, upstream regulators and genes potentially involved in tumor initiation and progression. We also validated gene expression and demonstrated genetic alteration of hub genes from clinical samples of head and neck cancer. Overall, we utilized bioinformatical methods to identify IL6, STAT1, LYN, BDNF, C3, CD274, PDCD1LG2, and CXCL10 as potential candidate genes that might facilitate the prevention and treatment of oral squamous cell carcinoma (OSCC), the most common type of head and neck squamous cell carcinoma (HNSCC).
Article
Full-text available
The accumulation of myeloid‐derived suppressor cells (MDSCs) in tumor‐bearing hosts is a hallmark of tumor‐associated inflammation, which is thought to be a barrier to immunosurveillance. Multiple factors secreted by tumor cells and tumor stromal cells are reported to be involved in promoting the expansion of MDSCs. Here, we showed that the subcutaneous inoculation of tumor cells and intravenous injection of tumor‐conditioned medium increased the number of MDSCs. Subsequent investigation elucidated that CXCL1 and CXCL2, which were originally characterized as the chemokines of neutrophils, specifically promoted the expansion of mo‐MDSCs, a subtype of MDSCs, in the presence of GM‐CSF. Depletion of CXCL1 or CXCL2 in B16F10 cells or in B16F10‐bearing mice noticeably decreased the generation of mo‐MDSCs in bone marrow. Moreover, we found that, in addition to the tumor cells, the tumor‐infiltrated CD11b⁺ myeloid cells also expressed CXCL1 and CXCL2. Furthermore, the CXCL1 and CXCL2‐induced increase of mo‐MDSCs was not correlated with the chemotaxis, proliferation or apoptosis of mo‐MDSCs. These findings illuminate a novel role of CXCL1 and CXCL2 in promoting mo‐MDSC generation by favoring the differentiation of bone marrow cells in tumor‐bearing conditions, which suggests that inhibition of CXCL1 and CXCL2 could decrease mo‐MDSC generation and improve host immunosurveillance. This article is protected by copyright. All rights reserved.
Article
Full-text available
Microbiota has been widely considered to play a critical role in human carcinogenesis. Human papilloma virus, hepatitis B and C virus, and Helicobacter pylori are implicated in the pathogenesis of cancer of uterine cervix, liver, and stomach, respectively. However, whether Porphyromonas gingivalis (P. gingivalis), a common Gram negative oral bacteria, is associated with oral carcinogenesis still remains unclear and its underlying mechanism needs to be addressed. Here, we established a combined experimental system of 4NQO-induced oral carcinoma model and chronic periodontitis model and investigated the effects of P. gingivalis infection on oral carcinogenesis and fatty acid metabolism during oral carcinogenesis. The data showed that in this animal model, P. gingivalis infection induced mice periodontitis, increased the tongue lesion size and multiplicity of each mouse and promoted oral cancer development. P. gingivalis treatment significantly increased the level of free fatty acids and altered the fatty acid profile in tongue tissues and the serum of mice. And P. gingivalis induced the formation of fatty liver of the mice. Besides, immunohistochemical analysis and qRT-PCR showed that the expression of fatty-acid synthase and acetyl-CoA carboxylase 1 were increased in the tongue and liver tissues of 4NQO-treated mice infected with P. gingivalis. These results showed that P. gingivalis promoted oral carcinogenesis and aggravated disturbance of fatty acid metabolism, indicating a close association among P. gingivalis, lipid metabolic and oral carcinogenesis.
Article
Full-text available
Background: There is a great interest in developing biomarkers to enhance early detection and clinical management of tongue squamous cell carcinoma (TSCC). However, the developmental path towards a clinically valid biomarker remains extremely challenging. Ideally, the initial key step in moving a newly discovered biomarker towards clinical implementation is independent replication. Therefore, the focus of this review is on biomarkers that consistently showed clinical relevance in two or more publications. Methods: We searched PubMed database for relevant papers across different TSCC sample sources, i.e., body fluids (saliva, serum/plasma) and tissues. No restriction regarding the date of publication was applied except for immunohistochemistry (IHC); only studies published between 2010 and June 2017 were included. Results: The search strategy identified 1429 abstracts, of which 96 papers, examining 150 biomarkers, were eventually included. Of these papers, 66% were exploratory studies evaluating single or a panel of biomarkers in one publication. Ultimately, based on studies that had undergone validation for their clinical relevance in at least two independent studies, we identified 10 promising candidates, consisting of different types of molecules (IL-6, IL-8, and Prolactin in liquid samples; HIF-1α, SOX2, E-cadherin, vimentin, MALAT1, TP53, and NOTCH1 in tissue biopsies) CONCLUSIONS: Although more exploratory research is needed with newer methods to identify biomarkers for TSCC, rigorous validation of biomarkers that have already shown unbiased assessment in at least two publications should be considered a high priority. Further research on these promising biomarkers or their combination in multi-institutional studies, could provide new possibilities to develop a specific panel for early diagnosis, prognosis, and individualized treatments.
Article
Full-text available
Betel quid and areca nut are known risk factors for many oral and oesophageal cancers, and their use is highly prevalent in the Asia-Pacific region. Additionally, betel quid and areca nut are associated with health effects on the cardiovascular, nervous, gastrointestinal, metabolic, respiratory, and reproductive systems. Unlike tobacco, for which the WHO Framework Convention on Tobacco Control provides evidence-based policies for reducing tobacco use, no global policy exists for the control of betel quid and areca nut use. Multidisciplinary research is needed to address this neglected global public health emergency and to mobilise efforts to control betel quid and areca nut use. In addition, future research is needed to advance our understanding of the basic biology, mechanisms, and epidemiology of betel quid and areca nut use, to advance possible prevention and cessation programmes for betel quid and areca nut users, and to design evidence-based screening and early diagnosis programmes to address the growing burden of cancers that are associated with use.
Article
In the last decade, human microbiome research is rapidly growing involving several fields of clinical medicine and population health. Although the microbiome seems to be linked to all sorts of diseases, cancer has the biggest potential to be investigated. Following the publication of the National Institute of Health - Human Microbiome Project (NIH-HMP), the link between Head and Neck Cancer (HNC) and microbiome seems to be a fast-moving field in research area. However, robust evidence-based literature is still quite scarce. Nevertheless the relationship between oral microbiome and HNC could have important consequences for prevention and early detection of this type of tumors. The aims of the present review are: (i) to discuss current pre-clinical evidence of a role of oral microbiome in HNC; (ii) to report recent developments in understanding the human microbiome's relationship with HNC oncogenesis; (iii) to explore the issue of treatment response and treatment toxicity; (iv) to describe the role of microbiota as potentially modifiable factor suitable for targeting by therapeutics. Further studies are needed to better establish the causal relationship between oral microbiome and HNC oncogenesis. Future trials should continue to explore oral microbiome in order to build the scientific and clinical rationale of HNC preventative and ameliorate treatment outcome.
Article
This article provides a status report on the global burden of cancer worldwide using the GLOBOCAN 2018 estimates of cancer incidence and mortality produced by the International Agency for Research on Cancer, with a focus on geographic variability across 20 world regions. There will be an estimated 18.1 million new cancer cases (17.0 million excluding nonmelanoma skin cancer) and 9.6 million cancer deaths (9.5 million excluding nonmelanoma skin cancer) in 2018. In both sexes combined, lung cancer is the most commonly diagnosed cancer (11.6% of the total cases) and the leading cause of cancer death (18.4% of the total cancer deaths), closely followed by female breast cancer (11.6%), prostate cancer (7.1%), and colorectal cancer (6.1%) for incidence and colorectal cancer (9.2%), stomach cancer (8.2%), and liver cancer (8.2%) for mortality. Lung cancer is the most frequent cancer and the leading cause of cancer death among males, followed by prostate and colorectal cancer (for incidence) and liver and stomach cancer (for mortality). Among females, breast cancer is the most commonly diagnosed cancer and the leading cause of cancer death, followed by colorectal and lung cancer (for incidence), and vice versa (for mortality); cervical cancer ranks fourth for both incidence and mortality. The most frequently diagnosed cancer and the leading cause of cancer death, however, substantially vary across countries and within each country depending on the degree of economic development and associated social and life style factors. It is noteworthy that high‐quality cancer registry data, the basis for planning and implementing evidence‐based cancer control programs, are not available in most low‐ and middle‐income countries. The Global Initiative for Cancer Registry Development is an international partnership that supports better estimation, as well as the collection and use of local data, to prioritize and evaluate national cancer control efforts. CA: A Cancer Journal for Clinicians 2018;0:1‐31. © 2018 American Cancer Society
Article
p>Bacteria may play a role in esophageal adenocarcinoma (EAC) and esophageal squamous cell carcinoma (ESCC), although evidence is limited to cross-sectional studies. In this study, we examined the relationship of oral microbiota with EAC and ESCC risk in a prospective study nested in two cohorts. Oral bacteria were assessed using 16S rRNA gene sequencing in prediagnostic mouthwash samples from n = 81/160 EAC and n = 25/50 ESCC cases/matched controls. Findings were largely consistent across both cohorts. Metagenome content was predicted using PiCRUST. We examined associations between centered log-ratio transformed taxon or functional pathway abundances and risk using conditional logistic regression adjusting for BMI, smoking, and alcohol. We found the periodontal pathogen Tannerella forsythia to be associated with higher risk of EAC. Furthermore, we found that depletion of the commensal genus Neisseria and the species Streptococcus pneumoniae was associated with lower EAC risk. Bacterial biosynthesis of carotenoids was also associated with protection against EAC. Finally, the abundance of the periodontal pathogen Porphyromonas gingivalis trended with higher risk of ESCC. Overall, our findings have potential implications for the early detection and prevention of EAC and ESCC. Cancer Res; 77(23); 6777–87. ©2017 AACR .</p