Skip to main content
Erschienen in: BMC Cancer 1/2019

Open Access 01.12.2019 | Research article

Expression of toll-like receptors in non-endemic nasopharyngeal carcinoma

verfasst von: Miia Ruuskanen, Ilmo Leivo, Heikki Minn, Tero Vahlberg, Caj Haglund, Jaana Hagström, Heikki Irjala

Erschienen in: BMC Cancer | Ausgabe 1/2019

Abstract

Background

Nasopharyngeal carcinoma (NPC) is a malignant disease with an enigmatic etiology. NPC associates with Epstein-Barr virus (EBV) and human papillomaviruses (HPVs), while immunological factors also play a role in carcinogenesis. Toll-like receptors (TLRs) are pattern recognition receptors that participate in the immunological defence against pathogens, but their functions are also linked to cancer.

Methods

In our whole population-based study, we retrieved 150 Finnish NPC cases and studied their tumour samples for TLR1, TLR2, TLR4, TLR5, TLR7, and TLR9 expressions by immunohistochemistry, and for the presence of EBV and high-risk HPVs with EBV RNA and HPV E6/E7 mRNA in situ hybridizations. In addition, we analyzed the TLR expression patterns according to age, tumour histology, EBV/HPV status, and outcome.

Results

We found that all TLRs studied were highly expressed in NPC. Viral status of the tumours varied, and 62% of them were EBV-positive, 14% HPV-positive, and 24% virus-negative. The tumours with strong TLR2nucl or TLR5 expression were mostly virus-negative or HPV-positive keratinizing squamous cell carcinomas, and the patients with these tumours were significantly older than those with mild or negative TLR2nucl/TLR5 expression. In Kaplan-Meier analysis, the patients with strong TLR5 expression had worse survival compared to the patients with negative or mild TLR5 expression, but the results were linked to other patient and tumour characteristics. In multivariable-adjusted Cox regression analysis, the patients with positive TLR7 tumour expression had better overall survival than those with no TLR7 expression. The 5-year overall survival rates according to TLR7 expression were 66% (mild), 52% (moderate or strong), and 22% (negative).

Conclusions

TLRs are highly expressed in non-endemic NPC. Intensity of TLR2 and TLR5 expressions correlate with viral status, and TLR7 seems to be an independent prognostic factor of non-endemic NPC.
Hinweise
Jaana Hagström and Heikki Irjala contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CI
Confidence interval
DAB
Diaminobenzidine
DAMP
Damage-associated molecular pattern
DSS
Disease-specific survival
EBER
EBV-encoded RNA
EBV
Epstein-Barr virus
EDTA
Ethylenediaminetetra-acetic acid
FFPE
Formalin-fixed paraffin-embedded
Gy
Gray
HPV
Human papillomavirus
HR
Hazard ratio
ISH
In situ hybridization
KSCC
Keratinizing squamous cell carcinoma
NK-D
Non-keratinizing differentiated carcinoma
NK-U
Non-keratinizing undifferentiated carcinoma
NPC
Nasopharyngeal carcinoma
OPSCC
Oropharyngeal squamous cell carcinoma
OS
Overall survival
OTSCC
Oral tongue squamous cell carcinoma
PAMP
Pathogen-associated molecular pattern
TLR
Toll-like receptor
TMA
Tissue microarray
UICC
International union against cancer

Background

Toll-like receptors (TLRs) are a family of transmembrane receptors that play an important role in innate immune defence. TLRs recognize the receptor-specific pathogen-associated molecular patterns (PAMPs) of microbes, and respond by activating immune cells against them [1]. TLRs also recognize the endogenous damage-associated molecular patterns (DAMPs) released from injured tissues, and TLR pathways have been shown to maintain tissue homeostasis by regulating wound healing processes and apoptosis [24]. In humans, ten different TLRs (TLR1-10) have been characterized [1]. For TLR1-9, several specific ligands have been identified, whereas the ligand for TLR10 remains elusive [1, 5]. TLR1, TLR2, TLR4, TLR5, and TLR6 were originally characterized as exclusively expressed on the cell surface and TLR3, TLR7, TLR8, and TLR9 as almost exclusively expressed in intracellular compartments such as endosomes [1]. The subcellular localization of TLR10 has not yet been characterized [5]. Nevertheless, recent findings suggest that TLR localization may be altered across cell types and in response to stimulation or disease [5]. Many studies have shown changes in TLR expression with oncogenic transformation [6]. However, the actual function of cancer-associated TLR modulation remains controversial. TLR stimulation may have anti- or pro-tumoral effects depending on the TLR receptor and cancer type [6]. Over the past decade, interest in the role of TLRs in tumorigenesis has increased, and numerous preclinical and clinical trials are ongoing to develop TLR agonists for cancer therapy [7].
Nasopharyngeal carcinoma (NPC) is a highly invasive malignant tumour arising from the mucosal epithelium of the nasopharynx. NPC has marked geographical disparities in incidence, with the highest occurrence in Southeast Asia and lowest in Europe and North America [8]. NPC is subdivided into three major histological types: keratinizing squamous cell carcinoma (KSCC), non-keratinizing carcinoma, and basaloid squamous cell carcinoma [9]. Non-keratinizing carcinoma can be further subdivided into undifferentiated and differentiated types [9]. The etiology of NPC is poorly understood, but epidemiological studies indicate that both genetic and environmental factors contribute to its development [10]. In high-incidence endemic areas, more than 95% of NPC tumours show non-keratinizing histology with Epstein-Barr virus (EBV) association, while in low-incidence non-endemic areas, the histology and viral status are more diverse. In the latter areas, KSCCs and human papillomavirus (HPV)-positive tumours are additionally found [1113]. Although it is commonly accepted that EBV and HPV can contribute to carcinogenesis, these viruses alone seem insufficient for malignant transformation [1416]. Recent studies indicate that the host’s immunological responses to viruses and precancerous lesions have an important role in cancer development, and that tumour progression could partly be due to a failure in the innate immune response [16]. In HPV-positive oropharyngeal squamous cell carcinoma (OPSCC), strong expression of TLR5 and low expression of TLR7 has been associated with poor disease-specific survival [17]. Furthermore, the intensity of TLR4 expression has been significantly lower in laryngeal papillomas transforming into laryngeal squamous cell carcinoma than in papillomas without malignant transformation [18]. In NPC, the importance of TLRs in tumour immunity has been demonstrated in studies from endemic areas, where certain sequence variants in TLR genes were associated with increased NPC risk [1922]. However, data on TLR expression in NPC is scarce and limited to endemic cases.
The purpose of this whole population-based study on NPC was to characterize the expression of six TLRs of interest for malignant transformation and immune activation. We studied the expression of TLR1, TLR2, TLR4, TLR5, TLR7, and TLR9 in Finnish patients and related the findings to histopathological subtypes, viral status, and survival.

Methods

Patients

We identified a total of 207 patients with primary NPC, which were diagnosed between 1990 and 2009, from the files of the nationwide Finnish Cancer Registry [23]. Clinical records for patient and disease characteristics, treatment and follow-up data were collected from eight major hospitals in Finland. All tumours were confirmed to originate from the nasopharyngeal epithelium. The clinical stage was determined according to the International Union Against Cancer (UICC) TNM staging system, 7th edition [24], and the dates and causes of death were acquired from the Finnish Cancer Registry and Statistics Finland. The study was approved by the Research Ethics Committee of the Hospital District of Southwest Finland, National Institute for Health and Welfare, and National Supervisory Authority for Welfare and Health.

Tissue microarray construction

Histological slides from NPC biopsies, taken at the time of diagnosis and stained with hematoxylin and eosin, were available for 168 patients. Tumour slides were reviewed and reclassified by an experienced head and neck pathologist (I.L.) according to the 4th edition of the World Health Organization (WHO) histological classification [9]. Formalin-fixed paraffin-embedded (FFPE) tissue blocks were available from 150 patients. Tissue microarrays (TMAs) were constructed using an automated tissue microarrayer (TMA Grand Master, 3D Histech Ltd., Budapest, Hungary) to create five new paraffin blocks from 1 mm core samples (n = 324) containing representative areas of NPC tumour tissue taken from the original FFPE blocks of tumours. Each patient with an available tumour block was represented in the array by at least one core, usually two. Scores from the duplicate cores were averaged to produce a single score. Five of the original tumour blocks represented neck metastases, while the remaining 145 were from the primary tumours. The TMAs also included as control tissues cores from normal liver and placenta. In addition, five samples from benign hyperplastic adenoid were stained separately for benign controls. In the final TMAs, representative tumour samples were lacking due to technical reasons in one patient for TLR1, in nine patients for TLR2 and TLR4, in seven patients for TLR5 and TLR7, and in one patient for TLR9.

Immunohistochemistry for TLRs

Immunohistochemical stains were performed on 3.5-μm thick TMA sections, which were mounted on glass slides, deparaffinized in xylene, and rehydrated in graded alcohol series. The sections were treated in a PreTreatment module (Lab Vision Corp, UK Ltd., Altrincham, UK) in Tris-HCl buffer (pH 8.5; TLR2, TLR4, and TLR7) or in Tris-EDTA buffer (pH 9.0; TLR1, TLR5, and TLR9) at 98 °C for 20 min. Endogenous peroxidase activity was blocked with 0.3% Dako REAL Peroxidase-Blocking Solution (Dako, Glostrup, Denmark) for 5 min. Immunostaining was performed in an Autostainer 480 (LabVision Corp, Fremont, CA, USA) incubating sections with primary antibodies against TLRs (Table 1) for 60 min (TLR9) or overnight (TLR1, TLR2, TLR4, TLR5, TLR7) followed by a 30 min incubation with Dako REAL EnVision Detection System (peroxidase/DAB+, rabbit/mouse). Between each step, slides were washed with PBS-0.04%-Tween20. Sections were counterstained with Dako Meyer’s hematoxylin and mounted in PERTEX (Histolab Product AB, Göteborg, Sweden). For each TLR, the five tumour TMAs were stained simultaneously in one and the same staining procedure, and five benign hyperplastic adenoid tissues were also stained for staining control comparison. Furthermore, the authors are experienced by previous use of these antibodies in various other tumour materials [17, 18].
Table 1
TLR antibodies used and TLR staining patterns in NPC and normal controls
Antigen
Primary antibody
Dilution
Staining pattern in NPC
Staining pattern in normal nasopharyngeal epithelial cells
TLR1
sc-30,000, polyclonal, rabbit, Santa Cruz Biotechnology Inc., CA, USA
1:100
Cytoplasmic
Cytoplasmic
TLR2
sc-10,739, polyclonal, rabbit, Santa Cruz Biotechnology Inc., CA, USA
1:200
Nuclear and cytoplasmic
Nuclear and cytoplasmic
TLR4
sc-10,741, polyclonal, rabbit, Santa Cruz Biotechnology Inc., CA, USA
1:300
Cytoplasmic
Nuclear and cytoplasmic
TLR5
NBP2–24787, monoclonal, mouse, Novus Biologicals, CO, USA
1:100
Nuclear membranous and cytoplasmic
Nuclear membranous and cytoplasmic
TLR7
IMG-581A, polyclonal, rabbit, Imgenex/Novus Biologicals, CO, USA
1:300
Nuclear membranous and nuclear
Nuclear and cytoplasmic
TLR9
sc-25,468, polyclonal, rabbit, Santa Cruz Biotechnology Inc., CA, USA
1:100
Cytoplasmic
Plasma membranous and cytoplasmic

Evaluation of TLR immunoreactivity in NPC

The interpretation of immunopositivity was performed by two head and neck pathologist (I.L. and J.H.), who were blinded to the clinical data (Fig. 1 and Table 1). TLR positivity was scored using a semi-quantitative scoring method where classification into negative, mild, and strong staining was based on the intensity of staining in cytoplasmic and nuclear areas. For a positive scoring, it was required that all or a large majority (> 80%) of tumour cells in the sample stained positively. Staining for all studied TLRs was scored in malignant NPC tissues. The scoring method has been documented previously [17], and representative examples are illustrated in Fig. 1. Using the scoring method, TLR1 positivity was classified as mild or strong granular cytoplasmic signals. TLR2 was expressed in both the cytoplasm and the nuclei, and the intensity of nuclear expression varied independently of cytoplasmic expression. Thus, cytoplasmic (TLR2cyto) and nuclear (TLR2nucl) positivities were scored separately as mild or strong. Nuclear expression was scored mild when the intensity of the staining was weaker or similar to cytoplasmic intensity, and strong when the staining was more intensive in the nuclei than in the cytoplasm. TLR4 positivity was scored mild or strong based on the intensity of cytoplasmic staining. TLR5 was expressed in the cytoplasm and on the nuclear membranes, and the two staining intensities were directly related: if cytoplasmic staining was strong, there was also strong staining on the nuclear membranes. Thus, the two were scored together as one TLR5 score: negative, mild (Fig. 1c) or strong (Fig. 1d). TLR7 staining was scored mild if some nuclear membranes were positive, moderate if all nuclear membranes and some nuclei were positive (Fig. 1e, f), and strong if nuclear membranes and nuclei stained substantially. TLR9 positivity was scored by the intensity of cytoplasmic staining as mild or strong.

In situ hybridization for EBV-encoded RNA and HPV E6/E7 mRNA

In situ hybridization (ISH) for EBV-encoded RNA (EBER) and for high risk HPV E6/E7 mRNA were performed on 3.5-μm thick TMA sections as described earlier [13]. This high risk HPV cocktail probe (RNAscope, Advanced Cell Diagnostics, Inc., Hayward, CA, USA) detects HPV genotypes 16, 18, 26, 31, 33, 35, 39, 45, 51, 52, 53, 56, 58, 59, 66, 68, 73 and 82. Positive hybridization for EBER was defined as strong diffuse signals in the nucleus of nearly all (> 90%) tumour cells. A positive HPV E6/E7 mRNA staining was defined as punctate staining that co-localised to the cytoplasm and/or nucleus of any of the malignant cells. The positivity was graded mild when at least 10 cells were intensively positive or less than 50% of the cells were positive with low intensity, moderate when over 50 to 70% of the tumour cells were positive, and strong when strong intensity of dot-like nuclear and cytoplasmic signals were present in over 70% of the cells or nearly all tumour cells were positive with low intensity.

Immunohistochemistry for p16

Immunohistochemical stains were performed on 3.5-μm thick TMA sections as described earlier, and p16 expression was defined positive when there were strong diffuse nuclear and cytoplasmic staining in 75% or more of tumour cells [13].

Treatment

Patients were treated with radiotherapy with or without concurrent chemotherapy as described earlier [13]. Typically patients received 2 Gy daily fractions five times per week, and the median dose was 70 Gy in the nasopharyngeal tumour area, 60 Gy in the involved lymph nodes, and 50 Gy in the elective neck area. The median treatment time was 7 weeks. In total, 76 patients (51%) received concurrent chemotherapy usually (87%) with platinum based cytostatic drugs. Seven patients (4%) with a compromised general condition and/or distant metastases received only palliative treatment, and were omitted from the survival analyses.

Statistical analysis

Statistical analyses were carried out using the SAS System for Windows, release 9.4 (SAS Institute Inc., Cary, NC). Mean ages in the TLR groups were compared with 1-way analysis of variance using the Tukey’s method for pairwise comparisons. The associations of categorical variables with different TLR statuses were compared with Pearson chi-squared test or Fisher’s exact test. Survival rates were calculated using the Kaplan-Meier method. Follow-up time was calculated from the end of the primary treatment, usually from the last day of radiotherapy, to the end of follow up or the date of death. Log-rank test was used to compare Kaplan-Meier survival curves. Age-adjusted and multivariable-adjusted Cox regression was used to test the association of TLR status with disease-specific survival (DSS) and overall survival (OS). The multivariable Cox regression analysis was adjusted for the potential confounding factors age, gender, stage and viral status. To avoid multicollinearity problems histology was excluded from the multivariable model due to the high correlation with viral status. The results are expressed using hazard ratios (HRs) with 95% confidence intervals (CIs). P-values of less than 0.05 were considered as statistically significant.

Results

Overall characterization of the patients

We analyzed the clinical data of 150 patients who had appropriate tissue specimens in TMA (Table 2). The mean age (SD) of the patients was 57.0 [15] years, and 101/150 (67%) were men. Almost all (145/150, 97%) patients were Caucasians of Finnish ethnic background. The majority (63%) presented with stage III/IV disease at the time of the diagnosis. Thirty-three patients (22%) had KSCC, 25 patients (17%) had non-keratinizing differentiated carcinoma (NK-D), and 92 patients (61%) had non-keratinizing undifferentiated carcinoma (NK-U). There were no basaloid SCCs. More detailed information on the patients has been reported in our earlier study [13].
Table 2
Patient characteristics
Characteristic
n (%)
All patients
150 (100)
Gender
 Male
101 (67)
 Female
49 (33)
Mean age at diagnosis
 Years (SD)
57.0 (15)
 Range
12–85
Ethnicity
 Finnish
145 (97)
 Othera
5 (3)
Smoking
 Smoker or ex-smoker
71 (47)
 Non-smoker
36 (24)
 Not known
43 (29)
T class
 T1
54 (36)
 T2
40 (27)
 T3
26 (17)
 T4
30 (20)
N class
 N0
53 (35)
 N1
33 (22)
 N2
51 (34)
 N3a
10 (7)
 N3b
3 (2)
Overall stage
 I
19 (12)
 II
37 (25)
 III
52 (35)
 IV
42 (28)
Histology
 Keratinizing squamous cell carcinoma
33 (22)
 Non-keratinizing differentiated carcinoma
25 (17)
 Non-keratinizing undifferentiated carcinoma
92 (61)
Virus status
 EBV-positive
93 (62)
 HPV-positive
21 (14)
 EBV/HPV-negative
36 (24)
Treatment
 Radiotherapy
67 (45)
 Chemoradiotherapy
76 (51)
 Palliative
7 (4)
Irradiation technique
 2-dimensional radiotherapy (2D)
15 (10)
 3-dimensional radiotherapy (3D)
88 (60)
 Intensity modulated radiotherapy (IMRT)
44 (30)
a Three from South-East Asia, one from Africa, one from Eastern Europe

EBV and HPV status

Of the total of 150 tumours, 93 (62%) were positive in EBER ISH and thus regarded as EBV-positive. HPV positivity was examined with p16 immunohistochemistry and high risk HPV E6/E7 mRNA ISH, and 21/150 (14%) tumours were positive with both methods forming the HPV-positive group. It is worthy to note that all p16-positive tumours showed positivity in HPV mRNA ISH; the concordance between these two methods was 100%. There were no co-infections with EBV and HPV. In 36/150 (24%) cases the tumours were negative for both EBV and HPV (EBV/HPV-negative group) [13].

Overall TLR expression in NPC and in benign adenoid tissues

TLR1, TLR2, TLR4, TLR7, and TLR9 were very highly expressed in NPC (Table 3). In TLR5 staining only, the proportion of negative samples was as high as 39%. TLR1 was found in the cytoplasm of cancer cells in 146/149 (98%) samples, and TLR1 positivity was strong in 82% of all samples. TLR2 was expressed in the cytoplasm and in the nuclei in 133/141 (94%) cases. In most of the samples (66%), nuclear staining was similar or milder than cytoplasmic staining, while in 28% of the samples it was stronger. TLR4 was positive in the cytoplasm of 130/141 (92%) specimens, and the percentages for mild and strong expression were 35 and 57%, respectively. Also in TLR5 staining, intensities in nuclear membranous and cytoplasmic staining were divisible into mild (53/143, 37%) and strong (32/143, 22%). TLR7 expression was found on the nuclear membranes and partly in the nuclei in 134/143 (94%) samples. Occasional large cancer cells also had cytoplasmic TLR7 staining. Mild TLR7 expression was found in 45/143 (31%) and moderate or strong expression in 89/143 (62%) cases. We united the groups with moderate and strong TLR7 expression, as these categories gave very similar results. TLR9 was expressed in the cytoplasm of 145/149 (97%) samples. Five controls of benign adenoid tissues expressed TLR1 and TLR2 in similar locations as in NPC tissues (Table 1). However, TLR4, TLR5, TLR7, and TLR9 were expressed differently in benign samples. TLR4 expression was also found in the nuclei of the benign cells. TLR5 was expressed on the nuclear membranes and in the cytoplasm in both malignant NPC and benign adenoid tissues, but the benign nasopharyngeal epithelium expressed TLR5 in the basal cells only (Fig. 1g), while the expression was ubiquitous in NPC (Fig. 1c, d). In benign adenoid tissues, TLR7 was expressed in the cytoplasm, while the nuclear membranes, positive in NPC, were not stained (Fig. 1h). TLR9 was found both in the cytoplasm and on the plasma membranes in the adenoid samples. The scorings were repeated with blinding of the data, and similar results were obtained.
Table 3
Expression of different TLRs in NPC in Finland
TLR type
Intensity of TLR expression, n (%)
Negative
Mild
Strong
Total
TLR1
3 (2)
24 (16)
122 (82)
149
TLR2nucl
8 (6)
93 (66)
40 (28)
141
TLR2cyto
8 (6)
59 (42)
74 (52)
141
TLR4
11 (8)
50 (35)
80 (57)
141
TLR5
58 (39)
53 (37)
32 (22)
143
TLR7
9 (7)
45 (31)
89 (62)
143
TLR9
4 (3)
61 (41)
84 (56)
149

Association of TLR expression with patient characteristics and tumour histology

TLR2 and TLR5 expression associated with age. The patients with mild TLR2nucl expression were significantly younger than the patients with strong TLR2nucl expression with mean (SD) ages of 54.7 [16] and 61.9 [13], respectively (p = 0.036). In addition, the patients with negative or mild TLR5 expression were younger than those with strong TLR5 expression with mean ages of 54.4 [15], 55.5 [16], and 63.4 [13], respectively (p = 0.021 and p = 0.053). There were significantly more women in the group with strong TLR5 expression (50%) compared to the group with negative TLR5 expression (24%, p = 0.013). Furthermore, TLR2nucl and TLR5 expressions correlated with histology (p = 0.006 and p < 0.0001, respectively): keratinizing tumours had often strong TLR2nucl (18/31, 58%) and strong TLR5 (19/32, 59%) expressions, while undifferentiated tumours presented mostly with mild TLR2nucl (65/87, 75%) and negative TLR5 (48/88, 55%) expressions. No statistically significant associations were found between any TLR and smoking, TNM classification, or overall stage.

Association between TLR expression and viral status

Cases were classified into three groups according to their viral status as follows: EBV-positive, HPV-positive, and EBV/HPV-negative. We found that TLR1, TLR4, TLR7, and TLR9 expressions were not related to viral status, while the opposite was true for the expressions of TLR2 and TLR5 (p < 0.0001 in both). The expression of TLR2nucl was stronger in the HPV-positive group and in the EBV/HPV-negative group than in the EBV-positive group. Accordingly, strong TLR2nucl positivity was seen in 57 and 53% versus 12% of the cases in each viral status group, respectively. The same tendency was seen in TLR5 expression, which was strong in 58% of the EBV/HPV-negative tumours, strong (29%) or mild (52%) in the HPV-positive tumours, and negative in 55% of the EBV-positive cases. Figure 2 shows the relationship between viral status and TLR2nucl/TLR5 expressions.

Association of TLR expression with clinical outcome

The median follow-up time was 63 months for patients treated with curative intent (n = 143) and for most patients (97%) it was 5 years or more. In Kaplan-Meier analysis, the patients with strong TLR5 expression had worse OS than those with mild or negative TLR5 expression (Fig. 3a; p = 0.019). The 5-year OS rates according to the TLR5 expression categories were 65% (mild), 57% (negative), and 40% (strong). DSS was nearly significantly worse in the patients with strong TLR5 expression compared to the patients with mild or negative expression (Fig. 3b; p = 0.062). The 5-year DSS rates were 69% (mild), 63% (negative), and 48% (strong). In contrast, the patients with no TLR7 expression had worse OS and DSS than the patients with positive TLR7 staining (Fig. 3c and d; p = 0.008 and p = 0.019, respectively). The 5-year OS rates according to TLR7 expression were 66% (mild), 52% (moderate or strong), and 22% (negative), and the corresponding DSS rates were 70% (mild), 59% (moderate or strong), and 22% (negative). The patients with negative TLR9 expression had worse OS compared to the patients with positive TLR9 expression, but the significance of this is limited by the presence of only four TLR9-negative cases. The 5-year OS rates according to TLR9 expression were 63% (mild), 52% (strong), and 25% (negative). In Kaplan-Meier analysis, we did not find survival differences related to TLR1, TLR2, and TLR4 expression.

TLR7 and TLR9 as prognostic factors in NPC

In multivariable-adjusted Cox regression analysis, positive TLR7 expression (mild, p = 0.018; moderate or strong, p = 0.038) compared to negative TLR7 expression was a significant prognostic factor for a better OS (Table 4). When analyzing DSS in multivariable-adjusted Cox regression analysis, only mild TLR7 expression (p = 0.046) compared to negative TLR7 expression remained as a significant prognostic factor for a better DSS. In age-adjusted Cox regression analysis, positive TLR9 expression (mild, p = 0.004; strong, p = 0.034) compared to no expression was associated with a better OS. However, when adjusted for gender, stage, and viral status TLR9 expression did not remain as a significant prognostic factor.
Table 4
Age-adjusted and multivariable Cox regression analysis of 143 patients for overall survival (a) and disease-specific survival (b)
 
Age-adjusted
Multivariablea
HR (95% CI)
p-value
HR (95% CI)
p-value
a) Overall survival (OS)
 Stage
  II vs. I
2.13 (0.96–4.73)
0.064 NS
  
  III vs. I
2.79 (1.30–5.95)
0.008
  
  IV vs. I
4.52 (2.12–9.64)
< 0.001
  
 Viral status
  HPV-pos vs. EBV-pos
1.62 (0.88–3.00)
0.123 NS
  
  HPV/EBV-neg vs. EBV-pos
2.67 (1.64–4.34)
< 0.001
  
 TLR1
  Mild vs. neg
0.30 (0.09–1.04)
0.057 NS
0.69 (0.18–2.70)
0.595 NS
  Strong vs. neg
0.24 (0.08–0.79)
0.018
0.48 (0.14–1.70)
0.256 NS
 TLR2 nuclear
  Mild vs. neg
0.56 (0.25–1.24)
0.151 NS
0.53 (0.23–1.19)
0.125 NS
  Strong vs. neg
0.62 (0.27–1.45)
0.271 NS
0.52 (0.21–1.30)
0.162 NS
 TLR2 cytoplasmic
  Mild vs. neg
0.62 (0.27–1.41)
0.256 NS
0.49 (0.20–1.17)
0.109 NS
  Strong vs. neg
0.70 (0.31–1.55)
0.373 NS
0.68 (0.30–1.56)
0.363 NS
 TLR4
  Mild vs. neg
1.16 (0.51–2.65)
0.727 NS
1.07 (0.46–2.50)
0.872 NS
  Moderate/strong vs. neg
1.15 (0.52–2.55)
0.731 NS
1.13 (0.50–2.56)
0.775 NS
 TLR5
  Mild vs. neg
0.75 (0.45–1.24)
0.259 NS
0.65 (0.38–1.10)
0.108 NS
  Strong vs. neg
1.37 (0.80–2.33)
0.250 NS
0.88 (0.47–1.61)
0.666 NS
 TLR7
  Mild vs. neg
0.28 (0.12–0.64)
0.003
0.37 (0.16–0.84)
0.018
  Moderate/strong vs. neg
0.49 (0.23–1.04)
0.062 NS
0.44 (0.20–0.95)
0.038
 TLR9
  Mild vs. neg
0.21 (0.07–0.60)
0.004
0.37 (0.12–1.16)
0.087 NS
  Strong vs. neg
0.32 (0.12–0.92)
0.034
0.49 (0.17–1.46)
0.199 NS
b) Disease-specific survival (DSS)
 Stage
  II vs. I
2.50 (0.68–9.14)
0.166 NS
  
  III vs. I
4.72 (1.40–15.95)
0.013
  
  IV vs. I
8.63 (2.59–28.80)
< 0.001
  
 Viral status
  HPV-pos vs. EBV-pos
1.10 (0.49–2.47)
0.822 NS
  
  HPV/EBV-neg vs. EBV-pos
2.22 (1.25–3.96)
0.007
  
 TLR1
  Mild vs. neg
0.50 (0.11–2.25)
0.369 NS
1.30 (0.26–6.57)
0.748 NS
  Strong vs. neg
0.32 (0.08–1.32)
0.113 NS
0.76 (0.17–3.43)
0.720 NS
 TLR2 nuclear
  Mild vs. neg
0.75 (0.26–2.10)
0.578 NS
0.88 (0.31–2.52)
0.811 NS
  Strong vs. neg
0.84 (0.28–2.54)
0.757 NS
1.05 (0.33–3.34)
0.937 NS
 TLR2 cytoplasmic
  Mild vs. neg
0.62 (0.23–1.65)
0.336 NS
0.69 (0.25–1.90)
0.468 NS
  Strong vs. neg
0.71 (0.28–1.84)
0.481 NS
0.89 (0.33–2.34)
0.806 NS
 TLR4
  Mild vs. neg
1.52 (0.54–4.41)
0.441 NS
1.65 (0.56–4.88)
0.368 NS
  Moderate/strong vs. neg
1.12 (0.40–3.20)
0.827 NS
1.28 (0.44–3.73)
0.652 NS
 TLR5
  Mild vs. neg
0.87 (0.47–1.61)
0.665 NS
0.81 (0.43–1.53)
0.518 NS
  Strong vs. neg
1.62 (0.85–3.06)
0.141 NS
1.34 (0.64–2.80)
0.442 NS
 TLR7
  Mild vs. neg
0.28 (0.11–0.70)
0.006
0.39 (0.15–0.98)
0.046
  Moderate/strong vs. neg
0.47 (0.21–1.05)
0.067 NS
0.47 (0.21–1.09)
0.079 NS
 TLR9
  Mild vs. neg
0.27 (0.08–0.93)
0.038
0.54 (0.15–1.98)
0.351 NS
  Strong vs. neg
0.39 (0.12–1.29)
0.123 NS
0.70 (0.20–2.43)
0.576 NS
a Adjusted for age, gender, stage, and viral status

Discussion

TLR signalling has been associated with tumour development [25]. The expression pattern and the function of TLRs in tumour progression seem to be cell-type specific and relate to different conditions, such as infections [5]. In our whole population-based study of NPC in Finland, we examined the expression of TLR1, TLR2, TLR4, TLR5, TLR7, and TLR9 in 150 tumours and analysed their pattern of expression according to EBV and HPV status, and clinical outcome. To our knowledge, this is the first study on TLRs in non-endemic NPC published to date. A few studies, conducted in high-incidence areas, have reported that genetic polymorphisms of TLR3, TLR4, TLR9, and TLR10 are associated with a risk of developing NPC in endemic populations [1922], but outcome results related to TLR expression are lacking worldwide.
The present study demonstrated that the expression patterns of TLR2 and TLR5 were related to the viral status while both TLRs were expressed significantly less in EBV-positive than in HPV-positive or EBV/HPV-negative NPC. These expressions were also related to well-established prognostic factors such as age and histology [23], which tend to dilute the independent prognostic significance of TLR2 and TLR5 in multivariable Cox regression analysis. However, in Kaplan-Meier analysis, the patients with strong TLR5 expression had worse survival compared to those with mild or negative expression (Fig. 3). This is in line with clinical studies on OPSCC and oral tongue squamous cell carcinoma (OTSCC), which report the association of poor DSS with strong expression of TLR5 [17, 26]. The exact effects of TLR-mediation on tumour growth are not known, but several in vitro studies on other types of carcinomas have shown that activation of TLR5 can promote tumorigenesis. For example, such activation has enhanced the proliferation of gastric cancer cells, and the migration and invasion of salivary gland adenocarcinoma [27, 28].
In contrast to TLR5, the patients with positive TLR7 expression had better DSS and OS than the patients with no TLR7 expression, and TLR7 was found to be a significant prognostic factor in multivariable Cox regression analysis. The finding that the patients with mild TLR7 expression had slightly better 5-year survival than the patients with strong TLR7 expression was not expected, but a similar result has been reported in patients with oral squamous cell carcinoma [29]. However, that report did not mention the survival rates of the patients with TR7-negative tumours. Nevertheless, it described variable TLR7 expression patterns when comparing between normal, dysplastic, and carcinoma tissues [29]. In our study, TLR7 expression in NPC samples was prominent on nuclear membranes, while in benign tissues TLR7 was expressed in the cytoplasm and in the nuclei. Another example of a defensive marker with derangement of function, in both over- and underexpression, is matrix metalloproteinase-8 [30].
Our findings suggest that viruses, or tumours related to specific viruses, or simply malignant neoplasms evoke distinct immunological reactions, but the hosts’ responses vary because of yet unknown causes. This might partly explain the differences in the effectiveness of oncological treatments. However, according to the hypothesis proposed by Wee et al. [31], causation may also be the reverse. They suggested that genetic TLR polymorphisms, especially in X-chromosome-linked TLR8, affect the innate immune response and make certain populations and individuals more vulnerable to infection-related cancers [31]. In NPC, altered TLR function could allow the virus to enter the nasopharyngeal mucosa and cause persistent infection finally resulting in a carcinogenic process [31]. We did not evaluate TLR8 expression in our tumour cells, but we found the usual but unexplained male preponderance in EBV- and HPV-positive patients (74 and 76% were males, respectively). In line with this, there were significantly more women in the virus-negative group (56%) compared to the EBV-positive (p = 0.001) and the HPV-positive (p = 0.020) groups. This suggests that men are more susceptible to virus-related NPC than women even in a non-endemic low-incidence population.
We studied TLR expression by immunohistochemistry to determine the expression sites of TLRs in carcinoma cells. As stated in a recent review article by Hamonic et al. [5], comprehensive understanding of the changing localizations of TLRs could aid us in understanding the basis of cancer immunology and possibly in developing new treatment modalities. We found that both in the benign adenoid control tissues and in the NPC samples, TLR1, TLR2, TLR4, and TLR5 were expressed diffusely in the cytoplasm instead of the cell surface, where they have been reported to localize when activated by pathogen-associated molecular patterns [32]. Interestingly, TLR4 expression was detected in the nuclei of benign controls but not in the nuclei of NPC cells. In benign samples, TLR5 was expressed exclusively in the basal layer of the nasopharyngeal epithelium as reported also for the normal oral mucosa [33], while in NPC the expression was diffuse. Similar to NPC, Pimentel-Nunes et al. have reported changing TLR localizations in gastric carcinogenesis [34]. They found that the normal gastric mucosa expressed TLR2, TLR4, and TLR5 in a polarized manner in the apical and particularly the basolateral membrane. By contrast, in metaplasia, dysplasia, and adenocarcinoma, these TLRs were expressed throughout the cytoplasm with no apparent polarization [34]. Also in previous studies on OPSCC and OTSCC, TLR5 has been localized in the cytoplasm rather than the membranes of the neoplastic cells [26, 35]. These findings suggest that activation of TLRs in abnormal locations may be related to carcinogenetic processes. We can only speculate whether TLRs have different functions in benign and malignant tissues, or if they become activated in different cellular compartments by PAMPs or DAMPs. Further research is needed to describe the mechanisms and causal connections between these phenomena.
In the present study, the retrospective nature of patient data carries limitations regarding the history of smoking habits and alcohol consumption. This limited our possibilities to evaluate the impact of these known carcinogens on the differences in expression of the studied TLRs. However, the uniform nationwide health care system in Finland enabled us to collect complete treatment and follow-up data, and a high proportion of diagnostic histopathological samples for TMA. In fact, histopathology was available for 150 (72%) of a total of 207 patients. While the size of the Finnish population limited the number of cases, we can nevertheless attest that the present cases represent a truly whole population-based material for the NPC patients treated during this 20-year period [36].

Conclusions

Our study is the first to evaluate TLR expressions in NPC of a non-endemic area. We found that TLRs were highly expressed in NPC, and intensity of TLR2 and TLR5 expressions were related to viral status. Expressions of TLR2 and TLR5 were strong in virus-negative cases, which will logically lead us to further studies on other possible factors causing NPC. In addition, TLR7 seems to be an independent prognostic factor of non-endemic NPC.

Acknowledgements

We thank Stina Syrjänen and Reija Randen-Brady for HPV evaluations. We are also grateful to Päivi Peltokangas, Sinikka Collanus, Heikki Peuravuori, and Auria Biobank for their technical assistance.
The study protocol was approved by the Research Ethics Committee of the Hospital District of Southwest Finland, and formal consent was deemed unnecessary by this ethical committee. This article does not contain any studies with human participants or animals performed by any of the authors.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Takeda K, Kaisho T, Akira S. Toll-like receptors. Annu Rev Immunol. 2003;21(1):335.CrossRef Takeda K, Kaisho T, Akira S. Toll-like receptors. Annu Rev Immunol. 2003;21(1):335.CrossRef
2.
Zurück zum Zitat Tsan M. Toll-like receptors, inflammation and cancer. Semin Cancer Biol. 2006;16(1):32–7.CrossRef Tsan M. Toll-like receptors, inflammation and cancer. Semin Cancer Biol. 2006;16(1):32–7.CrossRef
3.
Zurück zum Zitat Macedo L, Pinhal-Enfield G, Alshits V, Elson G, Cronstein BN, Leibovich SJ. Wound healing is impaired in MyD88-deficient mice: a role for MyD88 in the regulation of wound healing by adenosine A2A receptors. Am J Pathol. 2007;171(6):1774–88.CrossRef Macedo L, Pinhal-Enfield G, Alshits V, Elson G, Cronstein BN, Leibovich SJ. Wound healing is impaired in MyD88-deficient mice: a role for MyD88 in the regulation of wound healing by adenosine A2A receptors. Am J Pathol. 2007;171(6):1774–88.CrossRef
4.
Zurück zum Zitat Salaun B, Coste I, Rissoan M, Lebecque SJ, Renno T. TLR3 can directly trigger apoptosis in human cancer cells. J Immunol (Baltimore, Md: 1950). 2006;176(8):4894.CrossRef Salaun B, Coste I, Rissoan M, Lebecque SJ, Renno T. TLR3 can directly trigger apoptosis in human cancer cells. J Immunol (Baltimore, Md: 1950). 2006;176(8):4894.CrossRef
5.
Zurück zum Zitat Hamonic G, Paternak JA, Wilson HL. Recognizing conserved non-canonical localization patterns of toll-like receptors in tissues and across species. Cell Tissue Res. 2018;372(1):1–11.CrossRef Hamonic G, Paternak JA, Wilson HL. Recognizing conserved non-canonical localization patterns of toll-like receptors in tissues and across species. Cell Tissue Res. 2018;372(1):1–11.CrossRef
6.
Zurück zum Zitat Dajon M, Iribarren K, Cremer I. Toll-like receptor stimulation in cancer: a pro- and anti-tumor double-edged sword. Immunobiology. 2017;222(1):89.CrossRef Dajon M, Iribarren K, Cremer I. Toll-like receptor stimulation in cancer: a pro- and anti-tumor double-edged sword. Immunobiology. 2017;222(1):89.CrossRef
7.
Zurück zum Zitat Iribarren K, Bloy N, Buqué A, Cremer I, Eggermont A, Fridman WH, et al. Trial watch: Immunostimulation with toll-like receptor agonists in cancer therapy. Oncoimmunology. 2016;5(3):e1088631.CrossRef Iribarren K, Bloy N, Buqué A, Cremer I, Eggermont A, Fridman WH, et al. Trial watch: Immunostimulation with toll-like receptor agonists in cancer therapy. Oncoimmunology. 2016;5(3):e1088631.CrossRef
8.
Zurück zum Zitat Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61(2):69.CrossRef Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61(2):69.CrossRef
9.
Zurück zum Zitat El-Naggar AK, Chan JKC, Grandis JR, Takata T, Slootweg PJ. WHO classification of head and neck tumours. 4th ed. Lyon: International Agency for Research on Cancer; 2017. El-Naggar AK, Chan JKC, Grandis JR, Takata T, Slootweg PJ. WHO classification of head and neck tumours. 4th ed. Lyon: International Agency for Research on Cancer; 2017.
10.
Zurück zum Zitat Yu WM, Hussain SSM. Incidence of nasopharyngeal carcinoma in Chinese immigrants, compared with Chinese in China and South East Asia: review. J Laryngol Otol. 2009;123(10):1067–74.CrossRef Yu WM, Hussain SSM. Incidence of nasopharyngeal carcinoma in Chinese immigrants, compared with Chinese in China and South East Asia: review. J Laryngol Otol. 2009;123(10):1067–74.CrossRef
11.
Zurück zum Zitat Chua MLK, Wee JTS, Hui EP, Chan ATC. Nasopharyngeal carcinoma. Lancet. 2016;387(10022):1012–24.CrossRef Chua MLK, Wee JTS, Hui EP, Chan ATC. Nasopharyngeal carcinoma. Lancet. 2016;387(10022):1012–24.CrossRef
12.
Zurück zum Zitat Stenmark MH, Mchugh JB, Schipper M, Walline HM, Komarck C, Feng FY, et al. Nonendemic HPV-positive nasopharyngeal carcinoma: association with poor prognosis. Int J Radiat Oncol Biol Phys. 2014;88(3):580–8.CrossRef Stenmark MH, Mchugh JB, Schipper M, Walline HM, Komarck C, Feng FY, et al. Nonendemic HPV-positive nasopharyngeal carcinoma: association with poor prognosis. Int J Radiat Oncol Biol Phys. 2014;88(3):580–8.CrossRef
13.
Zurück zum Zitat Ruuskanen M, Irjala H, Minn H, Vahlberg T, Randen-Brady R, Hagström J, et al. Epstein-Barr virus and human papillomaviruses as favorable prognostic factors in nasopharyngeal carcinoma – a nationwide study in Finland. Head Neck. 2019;57:251–6. Ruuskanen M, Irjala H, Minn H, Vahlberg T, Randen-Brady R, Hagström J, et al. Epstein-Barr virus and human papillomaviruses as favorable prognostic factors in nasopharyngeal carcinoma – a nationwide study in Finland. Head Neck. 2019;57:251–6.
14.
Zurück zum Zitat Rickinson AB. Co-infections, inflammation and oncogenesis: future directions for EBV research. Semin Cancer Biol. 2014;26:99–115.CrossRef Rickinson AB. Co-infections, inflammation and oncogenesis: future directions for EBV research. Semin Cancer Biol. 2014;26:99–115.CrossRef
15.
Zurück zum Zitat Stanley MA. Epithelial cell responses to infection with human papillomavirus. Clin Microbiol Rev. 2012;25(2):215–22.CrossRef Stanley MA. Epithelial cell responses to infection with human papillomavirus. Clin Microbiol Rev. 2012;25(2):215–22.CrossRef
16.
Zurück zum Zitat Chow MT, Möller A, Smyth MJ. Inflammation and immune surveillance in cancer. Semin Cancer Biol. 2012;22(1):23–32.CrossRef Chow MT, Möller A, Smyth MJ. Inflammation and immune surveillance in cancer. Semin Cancer Biol. 2012;22(1):23–32.CrossRef
17.
Zurück zum Zitat Jouhi L, Mohamed H, Mäkitie A, Remes S, Haglund C, Atula T, et al. Toll-like receptor 5 and 7 expression may impact prognosis of HPV-positive oropharyngeal squamous cell carcinoma patients. Cancer Immunol Immunother. 2017;66(12):1619–29.CrossRef Jouhi L, Mohamed H, Mäkitie A, Remes S, Haglund C, Atula T, et al. Toll-like receptor 5 and 7 expression may impact prognosis of HPV-positive oropharyngeal squamous cell carcinoma patients. Cancer Immunol Immunother. 2017;66(12):1619–29.CrossRef
18.
Zurück zum Zitat Ilmarinen T, Hagström J, Haglund C, Auvinen E, Leivo I, Pitkäranta A, et al. Low expression of nuclear toll-like receptor 4 in laryngeal Papillomas transforming into squamous cell carcinoma. Otolaryngol Head Neck Surg. 2014;151(5):785–90.CrossRef Ilmarinen T, Hagström J, Haglund C, Auvinen E, Leivo I, Pitkäranta A, et al. Low expression of nuclear toll-like receptor 4 in laryngeal Papillomas transforming into squamous cell carcinoma. Otolaryngol Head Neck Surg. 2014;151(5):785–90.CrossRef
19.
Zurück zum Zitat He J-F, Jia W-H, Fan Q, Zhou X-X, Qin H-D, Shugart YY, et al. Genetic polymorphisms of TLR3 are associated with nasopharyngeal carcinoma risk in Cantonese population. BMC Cancer. 2007;7:194–200.CrossRef He J-F, Jia W-H, Fan Q, Zhou X-X, Qin H-D, Shugart YY, et al. Genetic polymorphisms of TLR3 are associated with nasopharyngeal carcinoma risk in Cantonese population. BMC Cancer. 2007;7:194–200.CrossRef
20.
Zurück zum Zitat Song C, Chen L, Zhang R, Yu X, Zeng Y. Functional variant in the 3′-untranslated region of toll-like receptor 4 is associated with nasopharyngeal carcinoma risk. Cancer Biol Ther. 2006;5(10):1285–91.CrossRef Song C, Chen L, Zhang R, Yu X, Zeng Y. Functional variant in the 3′-untranslated region of toll-like receptor 4 is associated with nasopharyngeal carcinoma risk. Cancer Biol Ther. 2006;5(10):1285–91.CrossRef
21.
Zurück zum Zitat Dai Q, Li XP, Chai L, Long HA, Yang ZH. Polymorphisms of toll-like receptor 9 are associated with nasopharyngeal carcinoma susceptibility. Tumor Biol. 2014;35(4):3247–53.CrossRef Dai Q, Li XP, Chai L, Long HA, Yang ZH. Polymorphisms of toll-like receptor 9 are associated with nasopharyngeal carcinoma susceptibility. Tumor Biol. 2014;35(4):3247–53.CrossRef
22.
Zurück zum Zitat Zhou X, Jia W, Shen G, Qin H, Yu X, Chen L, et al. Sequence variants in toll-like receptor 10 are associated with nasopharyngeal carcinoma risk. Cancer Epidemiol Biomark Prev. 2006;15(5):862.CrossRef Zhou X, Jia W, Shen G, Qin H, Yu X, Chen L, et al. Sequence variants in toll-like receptor 10 are associated with nasopharyngeal carcinoma risk. Cancer Epidemiol Biomark Prev. 2006;15(5):862.CrossRef
23.
Zurück zum Zitat Ruuskanen M, Grenman R, Leivo I, Vahlberg T, Mäkitie A, Saarilahti K, et al. Outcome of nasopharyngeal carcinoma in Finland: a nationwide study. Acta Oncol. 2018;57(2):251–6.CrossRef Ruuskanen M, Grenman R, Leivo I, Vahlberg T, Mäkitie A, Saarilahti K, et al. Outcome of nasopharyngeal carcinoma in Finland: a nationwide study. Acta Oncol. 2018;57(2):251–6.CrossRef
24.
Zurück zum Zitat Sobin LH, Gospodarowicz MK, Wittekind CTNM. Classification of Malignant Tumours. 7th ed. Hoboken: John Wiley & Sons, Incorporated; 2009. Sobin LH, Gospodarowicz MK, Wittekind CTNM. Classification of Malignant Tumours. 7th ed. Hoboken: John Wiley & Sons, Incorporated; 2009.
25.
Zurück zum Zitat Huang B, Zhao J, Li H, He K, Chen Y, Mayer L, et al. Toll-like receptors on tumor cells facilitate evasion of immune surveillance. Cancer Res. 2005;65(12):5009–14.CrossRef Huang B, Zhao J, Li H, He K, Chen Y, Mayer L, et al. Toll-like receptors on tumor cells facilitate evasion of immune surveillance. Cancer Res. 2005;65(12):5009–14.CrossRef
26.
Zurück zum Zitat Kauppila JH, Mattila AE, Karttunen TJ, Salo T. Toll-like receptor 5 (TLR5) expression is a novel predictive marker for recurrence and survival in squamous cell carcinoma of the tongue. Br J Cancer. 2013;108(3):638–43.CrossRef Kauppila JH, Mattila AE, Karttunen TJ, Salo T. Toll-like receptor 5 (TLR5) expression is a novel predictive marker for recurrence and survival in squamous cell carcinoma of the tongue. Br J Cancer. 2013;108(3):638–43.CrossRef
27.
Zurück zum Zitat Song E, Kang M, Kim Y, Kim S, Lee S, Kim C, et al. Flagellin promotes the proliferation of gastric cancer cells via the toll-like receptor 5. Int J Mol Med. 2011;28(1):115–9.PubMed Song E, Kang M, Kim Y, Kim S, Lee S, Kim C, et al. Flagellin promotes the proliferation of gastric cancer cells via the toll-like receptor 5. Int J Mol Med. 2011;28(1):115–9.PubMed
28.
Zurück zum Zitat Park J, Yoon H, Kim D, Kim S, Ahn S, Yoon J. Toll-like receptor 5 activation promotes migration and invasion of salivary gland adenocarcinoma. J Oral Pathol Med. 2011;40(2):187–93.CrossRef Park J, Yoon H, Kim D, Kim S, Ahn S, Yoon J. Toll-like receptor 5 activation promotes migration and invasion of salivary gland adenocarcinoma. J Oral Pathol Med. 2011;40(2):187–93.CrossRef
29.
Zurück zum Zitat Ni YH, Ding L, Zhang DY, Hou YY, Huang X, Hu Q. Distinct expression patterns of toll-like receptor 7 in tumour cells and fibroblast-like cells in oral squamous cell carcinoma. Histopathology. 2015;67(5):730–9.CrossRef Ni YH, Ding L, Zhang DY, Hou YY, Huang X, Hu Q. Distinct expression patterns of toll-like receptor 7 in tumour cells and fibroblast-like cells in oral squamous cell carcinoma. Histopathology. 2015;67(5):730–9.CrossRef
30.
Zurück zum Zitat Mäkinen LK, Häyry V, Atula T, Haglund C, Keski-Säntti H, Leivo I, et al. Prognostic significance of matrix metalloproteinase-2, −8, −9, and −13 in oral tongue cancer. J Oral Pathol Med. 2012;41(5):394–9.CrossRef Mäkinen LK, Häyry V, Atula T, Haglund C, Keski-Säntti H, Leivo I, et al. Prognostic significance of matrix metalloproteinase-2, −8, −9, and −13 in oral tongue cancer. J Oral Pathol Med. 2012;41(5):394–9.CrossRef
31.
Zurück zum Zitat Wee J, Nei WL, Yeoh KW, Yeo RM, Loong SL, Qian C. Why are east Asians more susceptible to several infection-associated cancers (carcinomas of the nasopharynx, stomach, liver, adenocarcinoma of the lung, nasal NK/T-cell lymphomas)? Med Hypotheses. 2012;79(6):833–42.CrossRef Wee J, Nei WL, Yeoh KW, Yeo RM, Loong SL, Qian C. Why are east Asians more susceptible to several infection-associated cancers (carcinomas of the nasopharynx, stomach, liver, adenocarcinoma of the lung, nasal NK/T-cell lymphomas)? Med Hypotheses. 2012;79(6):833–42.CrossRef
32.
Zurück zum Zitat Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124(4):783–801.CrossRef Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124(4):783–801.CrossRef
33.
Zurück zum Zitat Ahmed Haji Omar A, Korvala J, Haglund C, Virolainen S, Häyry V, Atula T, et al. Toll-like receptors -4 and −5 in oral and cutaneous squamous cell carcinomas. J Oral Pathol Med. 2015;44(4):258–65.CrossRef Ahmed Haji Omar A, Korvala J, Haglund C, Virolainen S, Häyry V, Atula T, et al. Toll-like receptors -4 and −5 in oral and cutaneous squamous cell carcinomas. J Oral Pathol Med. 2015;44(4):258–65.CrossRef
34.
Zurück zum Zitat Pimentel-Nunes P, Gonçalves N, Boal-Carvalho I, Afonso L, Lopes P, Roncon-Albuquerque R, et al. Helicobacter pylori induces increased expression of toll-like receptors and decreased toll-interacting protein in gastric mucosa that persists throughout gastric carcinogenesis. Helicobacter. 2013;18(1):22–32.CrossRef Pimentel-Nunes P, Gonçalves N, Boal-Carvalho I, Afonso L, Lopes P, Roncon-Albuquerque R, et al. Helicobacter pylori induces increased expression of toll-like receptors and decreased toll-interacting protein in gastric mucosa that persists throughout gastric carcinogenesis. Helicobacter. 2013;18(1):22–32.CrossRef
35.
Zurück zum Zitat Jouhi L, Datta N, Renkonen S, Atula T, Mäkitie A, Haglund C, et al. Expression of toll-like receptors in HPV-positive and HPV-negative oropharyngeal squamous cell carcinoma - an in vivo and in vitro study. Tumor Biol. 2015;36(10):7755–64.CrossRef Jouhi L, Datta N, Renkonen S, Atula T, Mäkitie A, Haglund C, et al. Expression of toll-like receptors in HPV-positive and HPV-negative oropharyngeal squamous cell carcinoma - an in vivo and in vitro study. Tumor Biol. 2015;36(10):7755–64.CrossRef
36.
Zurück zum Zitat Teppo L, Pukkala E, Lehtonen M. Data quality and quality control of a population-based cancer registry. Experience in Finland. Acta Oncol. 1994;33(4):365–9.CrossRef Teppo L, Pukkala E, Lehtonen M. Data quality and quality control of a population-based cancer registry. Experience in Finland. Acta Oncol. 1994;33(4):365–9.CrossRef
Metadaten
Titel
Expression of toll-like receptors in non-endemic nasopharyngeal carcinoma
verfasst von
Miia Ruuskanen
Ilmo Leivo
Heikki Minn
Tero Vahlberg
Caj Haglund
Jaana Hagström
Heikki Irjala
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2019
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-019-5816-9

Weitere Artikel der Ausgabe 1/2019

BMC Cancer 1/2019 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.