Skip to main content
Erschienen in: Diagnostic Pathology 1/2017

Open Access 01.12.2017 | Review

Role of tumour-associated macrophages in oral squamous cells carcinoma progression: an update on current knowledge

verfasst von: Maria Noel Marzano Rodrigues Petruzzi, Karen Cherubini, Fernanda Gonçalves Salum, Maria Antonia Zancanaro de Figueiredo

Erschienen in: Diagnostic Pathology | Ausgabe 1/2017

Abstract

Background

Oral squamous cell carcinoma (OSCC) accounts over 90% of malignant neoplasms of the oral cavity. This pathological entity is associated to a high mortality rate that has remained unchanged over the past decades. Tumour-associated macrophages (TAMs) are believed to have potential involvement in OSCC progression. However, the molecular networks involved in communication between stroma and cancer cells have not yet been fully elucidated.

Main body

The role of M2 polarized cells in oral carcinogenesis is supported by a correlation between TAMs accumulation into OSCC stroma and poor clinical outcome. Signalling pathways such as the NF-κB and cytokines released in the tumour microenvironment promote a bidirectional cross-talk between M2 and OSCC cells. These interactions consequently result in an increased proliferation of malignant cells and enhances aggressiveness, thus reducing patients’ survival time.

Conclusions

Here, we present a comprehensive review of the role of interleukin (IL)-1, IL-4, IL-6, IL-8, IL-10 and the receptor tyrosine kinase Axl in macrophage polarization to an M2 phenotype and OSCC progression. Understanding the molecular basis of oral carcinogenesis and metastatic spread of OSCC would promote the development of targeted treatment contributing to a more favourable prognosis.
Abkürzungen
Akt
Protein kinase B
AMD3100
1,1′-[1,4-phenylenebis(methylene)]bis-1,4,8,11-tetraazacyclotetradecane octahydrochloride
Axl
Axl receptor tyrosine kinase
CaMK II
Ca(2+)/calmodulin-dependent kinase II
CB2
Cannabinoid receptor 2
CCL
Chemokine C-C motif ligand
CXCR
C-X-C motif chemokine receptors
EBV
Epstein-Barr virus
EGFR
Endothelial growth factor receptor
EMT
Epithelial-mesenchymal transition
ERK
Extracellular signal regulated MAP kinase
Gas-6
Growth arrest specific gene-6
GM-CSF
Granulocyte macrophage colony stimulating factor
HPV
Human papilloma virus
IKKβ
I-kappaB kinase beta
IL
Interleukin
IL-1R
Interleukin-1 receptor
INF-\( \gamma \)
Interferon-gamma
MAPK
Mitogen activated protein kinases
MMP
Matrix metalloproteinase
MyD88
Myeloid differentiation primary response gene 88
nAChR
Nicotine acetylcholine receptor
NF-κB
Nuclear factor kappa B
OSCC
Oral squamous cell carcinoma
PI3K
Phosphatidylinositol 3-kinase
S1P
Sphingosine-1-phosphate
SDF-1α
Stromal cell derived factor-1 alpha
SOCS3
Suppressor of cytokine signalling 3
STAT
Signal transducer and activator of transcription
TAMs
Tumour-associated macrophages
TGF-β1
Transforming growth factor beta 1
TH1
T helper 1
TH2
T helper 2
TLR
Toll-like receptors
TNF
Tumour necrosis factor
TRAF3
TNF receptor associated factors
TRIF
TIR-domain-containing adapter-inducing interferon-β
TRKA
Tyrosine kinase receptor A
uPA
Plasminogen activator urokinase
uPAR
Plasminogen activator urokinase receptor
VEGF
Vascular endothelial growth factor

Background

Cancer pathogenesis events take place in imbalanced microenvironments, where pathological states do not affect only neoplastic cells [13]. Instead, cancerous cells disrupt tissue homeostasis, disturbing different cell types and contributing to disease progression, through interactions with mediators of the immune system [46]. Oral squamous cell carcinoma (OSCC) is a solid tumour of epithelial origin that affects more than 400,000 individuals annually worldwide [7]. The mortality rate of this disease has remained largely unchanged for the last decades, with a 5-year survival under 50% [8]. There is compelling evidence that tumour-associated macrophages (TAMs) have potential involvement in the progression and metastatic spread of OSCC. The most important features associated with their presence in the lesion stroma include facilitation of angiogenesis, tumour cell invasion, augmentation of cell motility, persistent growth, and suppression of anti-tumour responses [913]. The signals involved in communication between tumour cells and macrophages have not yet been completely elucidated. However, the interaction among tumour and inflammatory cells seems to be bidirectional [14]. Here, we present the strategies by which tumour cells influence macrophage physiology to display a pro-tumour phenotype, and the contribution of TAMs to OSCC progression. Is given an overview of potential markers that could provide support for diagnostic, evaluation of clinical outcome, and be used as valuable antineoplastic targets.

Macrophage differentiation

In adults, inflammatory monocytes (CD64+/CD16- CCR2+ Ly6C+) constitutively originate tissue-resident macrophage populations [15, 16]. The exposure of these cells to microenvironmental stimuli results in complex phenotypic modifications in a time- and location-dependent manner [1719]. The activation of different regulatory mechanisms and transcription pathways result in a vast spectrum of macrophage subtypes, of which M1 and M2 represent the extreme polarization phenotypes [18, 19]. The M1 polarization state depends on microbial stimulus and a T helper type 1 (TH1) cytokine profile (classical activation pathway). Whereas M2 polarization depends on a T helper type 2 (TH2) cytokine profile (alternative activation pathway) [20]. Interferon-gamma (INF-\( \gamma \)) and interleukin (IL)-4 secretion sustain an M1 and an M2 phenotype commitment, respectively [21]. M1 are innate immune effector cells that fight intracellular microbial challenges by means of reactive oxygen species and nitrogen intermediates. Activation of signal transducer and activator of transcription (STAT)-1 in M1 macrophages is important for optimal TH1 responses [22], such as direct tumour cell death [23, 24]. M2 macrophages block TH1 and differentiate in the tumour stroma from blood monocytes, or resident macrophages in resting state, after making contact with neoplastic cells presenting aberrant production of certain cytokines [18]. Additionally, they promote cancer progression by STAT-3 activation, inducing and maintaining a pro-carcinogenic inflammatory microenvironment [25].

OSCC cells and TAM interactions

Histopathologically, OSCC presents as fibrous connective tissue with unusual amounts of extracellular matrix rich in fibroblasts, vascular vessels, and inflammatory cells [26]. Among the local milieu of OSCC stromal spaces, rich in perlecans and inflammatory cells, monocytes or resting macrophages are differentiated into LyC16high, CD163+, CD204+, and CD68+ expressing TAMs. These cells are considered of utmost biological importance for disease progression and correlate with increased dedifferentiation in primary tumour sites [2729]. Moreover, TAMs elicit tumour relapse and/or post-operative cervical lymph node metastasis via angiogenesis and suppression of anti-tumour immunity [9]. An increase in the number of CD163+ macrophages occurs in oral leukoplakia. However, they co-express phosphorylated STAT-1, suggesting that in premalignant lesions TAMs possess an M1 phenotype in a dominant TH1 microenvironment [30]. Polarization to an M2 TAM phenotype probably occurs gradually and early during the onset of cancer. It is suggested that several interleukins (IL-1, IL-4, IL-6, IL-8, and IL-10), and other factors, such as the receptor tyrosine kinase Axl, participate in promoting this phenomenon. In the next sections we propose a topic structured discussion of relevant findings that corroborate this theoretical assumption. Figure 1 briefly reviews the effect of interleukins on TAMs present in OSCC stroma.

IL-1

Tumour released IL-1 cross-talks to TAMs and induces M2 polarization to an immunosuppressive phenotype via IL-1 receptor (IL-1R) and myeloid differentiation primary response gene 88 (MyD88), which requires I-kappaB kinase beta (IKKβ)-mediated nuclear factor kappa B (NF-κB) [31, 32]. Interleukin-1 beta (IL-1β) is a critical mediator of chronic inflammation and is implicated in OSCC during early and late stages of carcinogenesis. Pro-IL-1β is upregulated in tobacco and betel quid related oral cancer, and is secreted in an inflammasome-dependent manner [33], although it is absent in homeostatic conditions. In the presence of IL-1β TAMs suffer an upregulation of C-X-C motif chemokine receptors (CXCR), especially CXCR4, induced by the activation of extracellular signal regulated MAP kinase (ERK). Macrophages then become attracted by CXCR4 ligands, like stromal cell derived factor-1 alpha (SDF-1α) [34]. SDF-1α is highly inducible in hypoxic and proangiogenic niches, where it reinforces the autocrine/paracrine loop that contributes to an M2 phenotype [35]. Nevertheless, OSCC cells and TAMs together through IL-1β/IL-1R and CXCR4/ SDF-1α and via activation of the ERK signalling pathway produce tumour cell migration and invasion by inducing expression of matrix metalloproteinase (MMP) enzymes MMP-9 and MMP-13 [36]. These important angiogenic modulating enzymes promote the acquisition of vasculature for oxygenation, nutrition, and waste disposal, which are of fundamental importance for tumour growth [2]. Contrarily, the blockade of CXCR4 by the antagonist 1,1′-[1,4-phenylenebis(methylene)]bis-1,4,8,11-tetraazacyclotetradecane octahydrochloride (AMD3100) inhibited SDF-1 mediated lymph node metastasis [37]. Furthermore, a rich IL-1β microenvironment promotes CXCL1 production, and through CXCR2 this induces tyrosine phosphorylation of the endothelial growth factor receptor (EGFR) [33]. As a result, EGFR activates pathways leading to cell growth, DNA synthesis, and the expression of oncogenes like fos and jun [38]. IL-1α is found in tumour cell membranes and in intracellular locations, and is produced in larger amounts than IL-1β in highly metastatic tumours. Despite the lack of evidence that demonstrates its direct participation in OSCC progression through macrophage activation, IL-1α interacts with fibroblasts in the stroma. Therefore, IL-1α acts promoting cell proliferation and upregulating the secretion of IL-8, CXCL-1, and chemokine C-C motif ligand (CCL)-7 [39]. Coincidently, these cytokines are also commonly produced by TAMs, rising the hypothesis of a plausible interaction between OSCC and M2 cells by means of IL-1α.

IL-4

IL-4 is an anti-inflammatory and immunomodulatory cytokine that has been identified as a relevant factor for the activation of TAMs, as well as IL-1. Furthermore, an increased expression of IL-4 receptor alpha (IL-4Rα) correlates with increased OSCC recurrence [40]. Regarding this tumour entity, the interaction between malignant cells and TAMs occurs through the plasminogen activator urokinase (uPA) and its specific receptor uPAR, mainly through the activation of ERK1/2 and increase in the production of IL-4. In OSCC cells this receptor modifies several transduction pathways, affecting neoplastic cell behaviours and acts as a promoter of survival, proliferation, and metastasis [4143]. The high levels of IL-4 produced modifies the tumour microenvironment and facilitates an increase in arginase-1 levels, considered a biomarker of TAMs [43]. Similarly, this citokine induces cathepsin protease activity in TAMs, where they activate proteins including growth factors, transcription factors, and other proteases, such as MMPs [44]. Cathepsin B is considered a reliable marker for OSCC poor prognosis, correlating to higher tumour grade and lymph node metastasis [45].

IL-6

IL-6 expression in OSCC has been related to high lymph node metastatic rates and poor tumour differentiation, especially in male patients [46]. SDF-1alpha increases secretion of IL-6 in cultured human OSCC cells via CXCR4, ERK, and NF-κB pathways [47], in a similar manner to that seen for IL-1β/IL-1R. Moreover, the aberrant synthesis of IL-6 by neoplastic cells may be controlled by the CXCR4-specific inhibitor AMD3100 [47]. The calcium binding protein S100A9, associated with loss of differentiation and recurrence, tends to be deregulated in both tumour and stromal cells. The expression of S100A9 in monocytes exerts a tumour-promoting effect upon co-culture with oral cancer cells, in particular by releasing IL-6 and the activation of NF-κB or STAT-3 that is not achieved in tumour cell monoculture [48]. In response to apoptotic tumour cell supernatants, signalling patterns were identified that contributed to the TAMs phenotype. Two targets, IL-4Rα and cannabinoid receptor 2 (CB2), were validated and confirmed to regulate both IL-6 and IL-10 production in TAMs, contributing to autocrine/paracrine activation of STAT-3 in macrophages and tumour cells [49]. These findings emphasise the relevance of tumour cells and TAMs interactions for disease progression.

IL-8

IL-8 is a pro-angiogenic, pro-inflammatory mediator important for OSCC angiogenesis progression [50]. The mitogen activated protein kinases (MAPK) pathway is used by OSCC IL-8 to activate angiogenic activity in TAMs [51] augmenting, for example, vascular endothelial growth factor (VEGF) production. The receptors CXCR1 and CXCR2 have been detected in both oral normal keratinocytes and OSCC cells, where they exhibit higher expression. The presence of IL-8 CXC receptors in tumour cells increases ERK phosphorylation and MMP-7 and MMP-9 release, representing a tendency to proliferation, migration, and invasion [52]. Is important to consider that matrix metalloproteinase enzymes are essential for the achievement of a complete angiogenic potential of TAMs. At the same time, the progressive development of the tumour requires vast vasculature. Chronic periodontitis and tobacco consumption have both historically been associated to oral cancer. Then, recent published works propose the following interesting associations that also support the important role of IL-8 in OSCC progression. It is probable that Porphyromonas gingivalis contributes to OSCC progression, increasing IL-8 levels in the microenvironment and upregulating MMPs [53]. Nicotine also increases IL-8 release in OSCC, binding to the nicotine acetylcholine receptor (nAChR) and inducing calcium influx, that phosphorylates Ca(2+)/calmodulin-dependent kinase II (CaMK II) and NF-κB [54].

IL-10

In more dedifferentiated tumour niches the microenvironment progressively acquires an immunosuppressive profile [14]. IL-10 is a cytokine that modulates immune responses, causing suppressive regulatory T cell differentiation that contributes to tumour cell proliferation [55]. Since persistent viral infection promotes IL-10 upregulation and impaired T-cell responses [56], it is believed that this cytokine plays a critical role in human papilloma virus (HPV)- and Epstein-Barr virus (EBV)-related OSCC progression [57, 58]. Moreover, IL-10 indicates poor outcomes in HPV-unrelated OSCC, especially when INF-γ secretion [59] and transforming growth factor beta 1 (TGF-β1) levels [60] are low. Receptors for IL-22, a member of the IL-10 family, are highly expressed in OSCC cells, including in metastatic sites, compared to healthy regions. It was observed that in the OSCC MISK81-5 cell line, IL-22 induced the translocation of phosphorylated STAT-3 and upregulated the expression of Bcl-xL, survivin, and c-Myc, all known anti-apoptotic genes, as well as suppressor of cytokine signalling 3 (SOCS3) [61]. In this context, diverse pathways for IL-10 production by TAMs have been described, highlighting their contribution to an immunosuppressive state in the tumour stroma. TAMs present a defective TLR response caused by tumour-selective disruption of the MyD88 signalling cascade, and affect the TIR-domain-containing adapter-inducing interferon-β (TRIF)/TNF receptor associated factors (TRAF3)-dependent pathway in their own favour, leading to favourable transcription at the IL-10 promoter region [62]. In the presence of apoptotic tumour cell-factors like sphingosine-1-phosphate (S1P), TAMs use tyrosine kinase receptor A (TRKA), phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt), and MAPK signalling to induce IL-10 [63].

Gas6/Axl

TAMs acquire, possibly by cancer-derived factors like IL-10, the capacity to produce high levels of Gas-6 that promotes tumour development [64]. At the same time, in a bidirectional interaction, OSCC cells, that also produce Gas-6, polarize TAM toward a tumour-promoter phenotype. In OSCC, Gas-6 cooperates with Axl and achieves biological and clinical relevance by triggering the signalling pathways of PI3/Akt and NF-κB [65]. TAMs and OSCC interact in Gas-6/Axl axis-modulated epithelial-mesenchymal transition by upregulating cadherin, n-cadherin, and vimentin expression, and promoting cell invasion and migration. It was found that Axl expression correlates with clinical stage and lymph node status in OSCC patients. Moreover, TAMs count was associated with phosphorylated Axl immunoactivity in OSCC tissues [13, 65, 66]. Gas-6/Axl and NF-κB may be interesting targets for therapeutic intervention, since NF-κB promotes cancer resistance to apoptosis and production of growth factors in the stroma, which stimulates tumour progression [31].

Role of TAMs in OSCC histopathological diagnosis

Although further clinicopathological studies are needed before interactions between stromal cells and malignant cells can be defined as a key process for OSCC progression, evidence suggests that TAMs play several tumour-promoter roles during carcinogenesis [10, 28]. The presence of these polarized cells should be used as a potential marker to distinguish incipient OSCC from invasive lesions, avoiding underdiagnoses. As indicated by Matwaly et al. [67], the oral mucosa lacks an objective, standard-like structure that is found in other anatomical regions like the oesophagus, which makes the detection of invasiveness in oral cancer demanding. For a better understanding of TAMs in OSCC, more studies are necessary to define, by means of gene profiling, macrophage subpopulations with different tumour promoting abilities. A better indicator of the dynamic regulation of macrophage phenotype may be cellular cytokines, evaluated by means of tests conducted over multiple time points [67]. However, this methodology is time and cost demanding and probably unfeasible in clinical situations, especially in less developed countries where the prevalence of OSCC is higher. However, from the available evidence, it is possible to suggest that screening for TAM markers in oral biopsies certainly may contribute to accurate assessment of OSCC behaviour, being a valuable tool for the estimation of prognosis in cases related and unrelated to viral infection [5760, 68].

New diagnostic alternatives

Weber et al. [27] propose that even trauma from incisional biopsies might influence tumour biology leading to a worse prognosis and increased risk of developing lymph node metastases in OSCC patients. A wound-healing reaction consecutive to tissue trauma probably provides a microenvironmental stimulus that affects macrophage polarisation [69]. Until the present, diagnostic procedures and therapeutic planning for OSCC have been supported mainly by histopathological findings. Despite being inviable at present, mostly due to the lack of standardized techniques, interpretation, and validation of parameters, the development of new minimal invasive diagnostic strategies should consider the screening of salivary and serum markers that reflect tumour behaviour, associated or not with the improvement of classical techniques like exfoliative cytology. Several studies have demonstrated valuable associations among OSCC clinical stages and prognosis, and salivary or serum markers associated with TAM’s dynamic participation in the tumour stroma [58, 68, 7073]. Although salivary markers associated to TAM polarization are not yet used as parameters for definitive diagnoses, they should be taken into consideration to evaluate patients with potent malignant disorders, like proliferative verrucous leukoplakia [74], as well as for recurrence in OSCC treated patients.

Targeting TAMs in OSCC therapeutics

TAMs are potential targets for combination therapy in cancer treatment [75]. As we move forward, comprehension of the role of stromal cells in OSCC progression, suggest that therapies that only target TAMs may be possible, leading to an imbalance in tumour growth and invasiveness [75, 76]. However, despite its conceivable relevance, essentially mostly from positive clinical implications, the research in this field is incipient among cancer researchers. Recently a few studies have proposed targeting TAMs pathways to block cancer development [77, 78]. Signalling pathways such as the NF-κB and cytokines released in the tumour microenvironment through OSCC cells and TAMs interactions are attractive targets [79]. Inhibitors of cytokines involved in tumour signalling present potential for use to combat cancer, specially those implicated in promoting a malignancy cycle between OSCC cells and TAMs. Considering that chirurgical approaches are gold standard procedures for OSCC treatment, chemical interventions would be considered of lesser importance. However, it is relevant to underscore that during the last 30 years the disease-free survival and overall survival rates of OSCC patients have remained unchanged, perhaps due to limited care access or professional failures in performing early diagnoses, which is of the utmost relevance for prognosis. For these cases in particular, new therapeutic options are urgently needed.

Conclusions

Impaired tumour-preventive responses in OSCC are promoted by malignant cells and by soluble factors of the microenvironment that attract and polarize macrophages to a tumour-promoting state. Besides, macrophages reinforce the loop that promotes cancer growth and metastasis. This link between inflammation and cancer regulate OSCC progression and signalling pathways that provide a cross-talk between cancer cells and TAMs should be taken into consideration as valuable antineoplastic targets.

Acknowledgement

Not applicable.

Funding

Not applicable.

Availability of data and material

Not applicable.

Authors’ contributions

MNP, KC, FS and MAF were major contributors in writing the manuscript. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
Not applicable.
Not applicable.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis 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.
Literatur
2.
Zurück zum Zitat Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2010;144(5):646–74.CrossRef Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2010;144(5):646–74.CrossRef
4.
Zurück zum Zitat Landskron G, de la Fuente M, Thuwajit P, et al. Chronic inflammation and cytokines in the tumor microenvironment. J Immunol Res. 2014;2014:149185.CrossRefPubMedPubMedCentral Landskron G, de la Fuente M, Thuwajit P, et al. Chronic inflammation and cytokines in the tumor microenvironment. J Immunol Res. 2014;2014:149185.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Christopher AF, Gupta M, Bansal P.Micronome revealed miR-19a/b as key regulator of SOCS3 during cancer related inflammation of oral squamous cell carcinoma. Gene. 2016;594(1):30-40. doi:10.1016/j.gene.2016.08.044. Epub 2016 Aug 28. Christopher AF, Gupta M, Bansal P.Micronome revealed miR-19a/b as key regulator of SOCS3 during cancer related inflammation of oral squamous cell carcinoma. Gene. 2016;594(1):30-40. doi:10.​1016/​j.​gene.​2016.​08.​044. Epub 2016 Aug 28.
6.
Zurück zum Zitat Singh PK, Chandra G, Bogra J, et al. Association of interleukin-6 genetic polymorphisms with risk of OSCC in Indian population. Meta Gene. 2015;4:142–51.CrossRefPubMedPubMedCentral Singh PK, Chandra G, Bogra J, et al. Association of interleukin-6 genetic polymorphisms with risk of OSCC in Indian population. Meta Gene. 2015;4:142–51.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Wehrhan F, Büttner-Herold M, Hyckel P, et al. Increased malignancy of oral squamous cell carcinomas (OSCC) is associated with macrophage polarization in regional lymph nodes – an immunohistochemical study. BMC Cancer. 2014;14:522.CrossRefPubMedPubMedCentral Wehrhan F, Büttner-Herold M, Hyckel P, et al. Increased malignancy of oral squamous cell carcinomas (OSCC) is associated with macrophage polarization in regional lymph nodes – an immunohistochemical study. BMC Cancer. 2014;14:522.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Chiu KC, Lee CH, Liu SY, et al. Polarization of tumor-associated macrophages and Gas6/Axl signaling in oral squamous cell carcinoma. Oral Oncol. 2015;51(7):683–9.CrossRefPubMed Chiu KC, Lee CH, Liu SY, et al. Polarization of tumor-associated macrophages and Gas6/Axl signaling in oral squamous cell carcinoma. Oral Oncol. 2015;51(7):683–9.CrossRefPubMed
11.
Zurück zum Zitat He KF, Zhang L, Huang CF, et al. CD163+ tumor-associated macrophages correlated with poor prognosis and cancer stem cells in oral squamous cell carcinoma. Biomed Res Int. 2014;2014:838632.PubMedPubMedCentral He KF, Zhang L, Huang CF, et al. CD163+ tumor-associated macrophages correlated with poor prognosis and cancer stem cells in oral squamous cell carcinoma. Biomed Res Int. 2014;2014:838632.PubMedPubMedCentral
12.
Zurück zum Zitat Ni YH, Ding L, Huang XF, et al. Microlocalization of CD68+ tumor-associated macrophages in tumor stroma correlated with poor clinical outcomes in oral squamous cell carcinoma patients. Tumour Biol. 2015;36(7):5291–8.CrossRefPubMed Ni YH, Ding L, Huang XF, et al. Microlocalization of CD68+ tumor-associated macrophages in tumor stroma correlated with poor clinical outcomes in oral squamous cell carcinoma patients. Tumour Biol. 2015;36(7):5291–8.CrossRefPubMed
13.
Zurück zum Zitat Lee CH, Liu SY, Chou KC, Yeh CT, et al. Tumor-associated macrophages promote oral cancer progression through activation of the Axl signaling pathway. Ann Surg Oncol. 2014;21(3):1031–7.CrossRefPubMed Lee CH, Liu SY, Chou KC, Yeh CT, et al. Tumor-associated macrophages promote oral cancer progression through activation of the Axl signaling pathway. Ann Surg Oncol. 2014;21(3):1031–7.CrossRefPubMed
14.
Zurück zum Zitat Joyce JA, Pollard JW. Microenvironmental regulation of metastasis. Nature Rev Cancer. 2009;9:239–52.CrossRef Joyce JA, Pollard JW. Microenvironmental regulation of metastasis. Nature Rev Cancer. 2009;9:239–52.CrossRef
15.
Zurück zum Zitat Grage-Griebenow E, Flad HD, Ernst M. Heterogeneity of human peripheral blood monocyte subsets. J Leukoc Biol. 2001;69(1):11–20.PubMed Grage-Griebenow E, Flad HD, Ernst M. Heterogeneity of human peripheral blood monocyte subsets. J Leukoc Biol. 2001;69(1):11–20.PubMed
16.
Zurück zum Zitat Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005;5(12):953–64.CrossRefPubMed Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005;5(12):953–64.CrossRefPubMed
17.
Zurück zum Zitat McWhorter FY, Davis CT, Liu WF. Physical and mechanical regulation of macrophage phenotype and function. Cell Mol Life Sci. 2015;72(7):1303–16.CrossRefPubMed McWhorter FY, Davis CT, Liu WF. Physical and mechanical regulation of macrophage phenotype and function. Cell Mol Life Sci. 2015;72(7):1303–16.CrossRefPubMed
19.
Zurück zum Zitat Melton DW, McManus LM, Gelfond JAL, et al. Temporal phenotypic features distinguish polarized macrophages in vitro. Autoimmunity. 2015;48(3):161–76.CrossRefPubMedPubMedCentral Melton DW, McManus LM, Gelfond JAL, et al. Temporal phenotypic features distinguish polarized macrophages in vitro. Autoimmunity. 2015;48(3):161–76.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Barros MHM, Hauck F, Dreyer JH, et al. Macrophage polarisation: an immunohistochemical approach for identifying M1 and M2 macrophages. PLoS One. 2013;8(11):e80908.CrossRefPubMedPubMedCentral Barros MHM, Hauck F, Dreyer JH, et al. Macrophage polarisation: an immunohistochemical approach for identifying M1 and M2 macrophages. PLoS One. 2013;8(11):e80908.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Wang N, Liang H, Zen K. Molecular mechanisms that influence the macrophage M1–M2 polarization balance. Front Immunol. 2014;5:614.PubMedPubMedCentral Wang N, Liang H, Zen K. Molecular mechanisms that influence the macrophage M1–M2 polarization balance. Front Immunol. 2014;5:614.PubMedPubMedCentral
22.
Zurück zum Zitat Kim HS, Kim DC, Kim H-M, et al. STAT1 deficiency redirects IFN signalling toward suppression of TLR response through a feedback activation of STAT3. Scientific Reports. 2015;5:13414.CrossRefPubMedPubMedCentral Kim HS, Kim DC, Kim H-M, et al. STAT1 deficiency redirects IFN signalling toward suppression of TLR response through a feedback activation of STAT3. Scientific Reports. 2015;5:13414.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Ma J, Liu L, Che G, et al. The M1 form of tumor-associated macrophages in non-small cell lung cancer is positively associated with survival time. BMC Cancer. 2010;10:112.CrossRefPubMedPubMedCentral Ma J, Liu L, Che G, et al. The M1 form of tumor-associated macrophages in non-small cell lung cancer is positively associated with survival time. BMC Cancer. 2010;10:112.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Mori K, Haraguchi S, Hiori M, et al. Tumor-associated macrophages in oral premalignant lesions coexpress CD163 and STAT1 in a Th1-dominated microenvironment. BMC Cancer. 2015;15:573.CrossRefPubMedPubMedCentral Mori K, Haraguchi S, Hiori M, et al. Tumor-associated macrophages in oral premalignant lesions coexpress CD163 and STAT1 in a Th1-dominated microenvironment. BMC Cancer. 2015;15:573.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Metwaly H, Maruyama S, Yamazaki M, et al. Parenchymal-stromal switching for extracellular matrix production on invasion of oral squamous cell carcinoma. Hum Pathol. 2012;43(11):1973–81.CrossRefPubMed Metwaly H, Maruyama S, Yamazaki M, et al. Parenchymal-stromal switching for extracellular matrix production on invasion of oral squamous cell carcinoma. Hum Pathol. 2012;43(11):1973–81.CrossRefPubMed
27.
Zurück zum Zitat Weber M, Moebius P, Büttner-Herold M, et al. Macrophage polarisation changes within the time between diagnostic biopsy and tumour resection in oral squamous cell carcinomas—an immunohistochemical study. Br J Cancer. 2015;113(3):510–9.CrossRefPubMedPubMedCentral Weber M, Moebius P, Büttner-Herold M, et al. Macrophage polarisation changes within the time between diagnostic biopsy and tumour resection in oral squamous cell carcinomas—an immunohistochemical study. Br J Cancer. 2015;113(3):510–9.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Weber M, Iliopoulos C, Moebius P, et al. Prognostic significance of macrophage polarization in early stage oral squamous cell carcinomas. Oral Oncol. 2016;52:75–84.CrossRefPubMed Weber M, Iliopoulos C, Moebius P, et al. Prognostic significance of macrophage polarization in early stage oral squamous cell carcinomas. Oral Oncol. 2016;52:75–84.CrossRefPubMed
30.
Zurück zum Zitat Bôas DS, Takiya CM, Gurgel CA, Cabral MG, Santos JN. Tumor-infiltrating macrophage and microvessel density in oral squamous cell carcinoma. Braz Dent J. 2013;24(3):194-9. doi:10.1590/0103-6440201302049. PMID:23969905. Bôas DS, Takiya CM, Gurgel CA, Cabral MG, Santos JN. Tumor-infiltrating macrophage and microvessel density in oral squamous cell carcinoma. Braz Dent J. 2013;24(3):194-9. doi:10.​1590/​0103-6440201302049. PMID:23969905.
31.
32.
Zurück zum Zitat Watari K, Shibata T, Kawahara A, et al. Tumor-derived interleukin-1 promotes lymphangiogenesis and lymph node metastasis through M2-type macrophages. PLoS One. 2014;9(6):e99568.CrossRefPubMedPubMedCentral Watari K, Shibata T, Kawahara A, et al. Tumor-derived interleukin-1 promotes lymphangiogenesis and lymph node metastasis through M2-type macrophages. PLoS One. 2014;9(6):e99568.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Lee CH, Chang JS, Syu SH, et al. IL-1β promotes malignant transformation and tumor aggressiveness in oral cancer. J Cell Physiol. 2015;230(4):875–84.CrossRefPubMed Lee CH, Chang JS, Syu SH, et al. IL-1β promotes malignant transformation and tumor aggressiveness in oral cancer. J Cell Physiol. 2015;230(4):875–84.CrossRefPubMed
34.
Zurück zum Zitat Sun Y, Zhu D, Wang G, et al. Pro-inflammatory cytokine IL-1β up-regulates CXC chemokine receptor 4 via notch and ERK signaling pathways in tongue squamous cell carcinoma. PLoS One. 2015;10(7):e0132677.CrossRefPubMedPubMedCentral Sun Y, Zhu D, Wang G, et al. Pro-inflammatory cytokine IL-1β up-regulates CXC chemokine receptor 4 via notch and ERK signaling pathways in tongue squamous cell carcinoma. PLoS One. 2015;10(7):e0132677.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Sánchez-Martín L, Estecha A, Samaniego R, et al. The chemokine CXCL12 regulates monocyte-macrophage differentiation and RUNX3 expression. Blood. 2011;117(1):88–97.CrossRefPubMed Sánchez-Martín L, Estecha A, Samaniego R, et al. The chemokine CXCL12 regulates monocyte-macrophage differentiation and RUNX3 expression. Blood. 2011;117(1):88–97.CrossRefPubMed
36.
Zurück zum Zitat Yu T, Wu Y, Helman JI, Wen Y, Wang C, Li L. CXCR4 promotes oral squamous cell carcinoma migration and invasion through inducing expression of MMP-9 and MMP-13 via the ERK signaling pathway. Mol Cancer Res. 2011;9(2):161–72.CrossRefPubMed Yu T, Wu Y, Helman JI, Wen Y, Wang C, Li L. CXCR4 promotes oral squamous cell carcinoma migration and invasion through inducing expression of MMP-9 and MMP-13 via the ERK signaling pathway. Mol Cancer Res. 2011;9(2):161–72.CrossRefPubMed
37.
Zurück zum Zitat Uchida D, Onoue T, Kuribayashi N, et al. Blockade of CXCR4 in oral squamous cell carcinoma inhibits lymph node metastases. Eur J Cancer. 2011;47(3):452–9.CrossRefPubMed Uchida D, Onoue T, Kuribayashi N, et al. Blockade of CXCR4 in oral squamous cell carcinoma inhibits lymph node metastases. Eur J Cancer. 2011;47(3):452–9.CrossRefPubMed
38.
Zurück zum Zitat Voldborg BR, Damstrup L, Spang-Thomsen M, Poulsen HS. Epidermal growth factor receptor (EGFR) and EGFR mutations, function and possible role in clinical trials. Ann Oncol. 1997;8(12):1197–206.CrossRefPubMed Voldborg BR, Damstrup L, Spang-Thomsen M, Poulsen HS. Epidermal growth factor receptor (EGFR) and EGFR mutations, function and possible role in clinical trials. Ann Oncol. 1997;8(12):1197–206.CrossRefPubMed
39.
Zurück zum Zitat Bae JY, Kim EK, Yang DH, et al. Reciprocal Interaction between Carcinoma-Associated Fibroblasts and Squamous Carcinoma Cells through Interleukin-1α Induces Cancer Progression. Neoplasia (New York, NY). 2014;16(11):928–38.CrossRef Bae JY, Kim EK, Yang DH, et al. Reciprocal Interaction between Carcinoma-Associated Fibroblasts and Squamous Carcinoma Cells through Interleukin-1α Induces Cancer Progression. Neoplasia (New York, NY). 2014;16(11):928–38.CrossRef
40.
Zurück zum Zitat Kwon M, Kim JW, Roh JL, Park Y, Cho KJ, Choi SH, Nam SY, Kim SY, Lee BH. Recurrence and cancer-specific survival according to the expression of IL-4Rα and IL-13Rα1 in patients with oral cavity cancer. Eur J Cancer. 2015;51(2):177–85.CrossRefPubMed Kwon M, Kim JW, Roh JL, Park Y, Cho KJ, Choi SH, Nam SY, Kim SY, Lee BH. Recurrence and cancer-specific survival according to the expression of IL-4Rα and IL-13Rα1 in patients with oral cavity cancer. Eur J Cancer. 2015;51(2):177–85.CrossRefPubMed
41.
Zurück zum Zitat Shi Z, Stack MS. Urinary-type plasminogen activator (uPA) and its receptor (uPAR) in squamous cell carcinoma of the oral cavity. Biochem J. 2007;407(2):153–9.CrossRefPubMed Shi Z, Stack MS. Urinary-type plasminogen activator (uPA) and its receptor (uPAR) in squamous cell carcinoma of the oral cavity. Biochem J. 2007;407(2):153–9.CrossRefPubMed
42.
Zurück zum Zitat Yoshizawa K, Nozaki S, Kitahara H, et al. Expression of urokinase-type plasminogen activator/urokinase-type plasminogen activator receptor and maspin in oral squamous cell carcinoma: Association with mode of invasion and clinicopathological factors. Oncol Rep. 2011;26(6):1555–60.PubMed Yoshizawa K, Nozaki S, Kitahara H, et al. Expression of urokinase-type plasminogen activator/urokinase-type plasminogen activator receptor and maspin in oral squamous cell carcinoma: Association with mode of invasion and clinicopathological factors. Oncol Rep. 2011;26(6):1555–60.PubMed
43.
Zurück zum Zitat Hu J, Jo M, Eastman BM, et al. uPAR Induces Expression of Transforming Growth Factor β and Interleukin-4 in Cancer Cells to Promote Tumor-Permissive Conditioning of Macrophages. Am J Pathol. 2014;184(12):3384–93.CrossRefPubMedPubMedCentral Hu J, Jo M, Eastman BM, et al. uPAR Induces Expression of Transforming Growth Factor β and Interleukin-4 in Cancer Cells to Promote Tumor-Permissive Conditioning of Macrophages. Am J Pathol. 2014;184(12):3384–93.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Gocheva V, Wang H-W, Gadea BB, et al. IL-4 induces cathepsin protease activity in tumor-associated macrophages to promote cancer growth and invasion. Genes Dev. 2010;24(3):241–55.CrossRefPubMedPubMedCentral Gocheva V, Wang H-W, Gadea BB, et al. IL-4 induces cathepsin protease activity in tumor-associated macrophages to promote cancer growth and invasion. Genes Dev. 2010;24(3):241–55.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Yang W-E, Ho C-C, Yang S-F, et al. Cathepsin B Expression and the correlation with clinical aspects of oral squamous cell carcinoma. PLoS One. 2016;11(3):e0152165.CrossRefPubMedPubMedCentral Yang W-E, Ho C-C, Yang S-F, et al. Cathepsin B Expression and the correlation with clinical aspects of oral squamous cell carcinoma. PLoS One. 2016;11(3):e0152165.CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Chen C-J, Sung W-W, Lin Y-M, et al. Gender Difference in the Prognostic Role of Interleukin 6 in Oral Squamous Cell Carcinoma. PLoS One. 2012;7(11):e50104.CrossRefPubMedPubMedCentral Chen C-J, Sung W-W, Lin Y-M, et al. Gender Difference in the Prognostic Role of Interleukin 6 in Oral Squamous Cell Carcinoma. PLoS One. 2012;7(11):e50104.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Tang C-H, Chuang J-Y, Fong Y-C, Maa M-C, Way T-D, Hung C-H. Bone-derived SDF-1 stimulates IL-6 release via CXCR4, ERK and NF-κB pathways and promotes osteoclastogenesis in human oral cancer cells. Carcinogenesis. 2008;29(8):1483–92.CrossRefPubMedPubMedCentral Tang C-H, Chuang J-Y, Fong Y-C, Maa M-C, Way T-D, Hung C-H. Bone-derived SDF-1 stimulates IL-6 release via CXCR4, ERK and NF-κB pathways and promotes osteoclastogenesis in human oral cancer cells. Carcinogenesis. 2008;29(8):1483–92.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Fang W-Y, Chen Y-W, Hsiao J-R, et al. Elevated S100A9 expression in tumor stroma functions as an early recurrence marker for early-stage oral cancer patients through increased tumor cell invasion, angiogenesis, macrophage recruitment and interleukin-6 production. Oncotarget. 2015;6(29):28401–24.CrossRefPubMedPubMedCentral Fang W-Y, Chen Y-W, Hsiao J-R, et al. Elevated S100A9 expression in tumor stroma functions as an early recurrence marker for early-stage oral cancer patients through increased tumor cell invasion, angiogenesis, macrophage recruitment and interleukin-6 production. Oncotarget. 2015;6(29):28401–24.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Ley S, Weigert A, Hériché JK, et al. RNAi screen in apoptotic cancer cell-stimulated human macrophages reveals co-regulation of IL-6/IL-10 expression. Immunobiology. 2013;218(1):40–51.CrossRefPubMed Ley S, Weigert A, Hériché JK, et al. RNAi screen in apoptotic cancer cell-stimulated human macrophages reveals co-regulation of IL-6/IL-10 expression. Immunobiology. 2013;218(1):40–51.CrossRefPubMed
50.
Zurück zum Zitat Punyani SR, Sathawane RS. Salivary level of interleukin-8 in oral precancer and oral squamous cell carcinoma. Clin Oral Investig. 2013;17(2):517–24.CrossRefPubMed Punyani SR, Sathawane RS. Salivary level of interleukin-8 in oral precancer and oral squamous cell carcinoma. Clin Oral Investig. 2013;17(2):517–24.CrossRefPubMed
51.
Zurück zum Zitat Nishio Y, Gojoubori T, Kaneko Y, Shimizu N, Asano M. Cancer cell-derived IL-8 induces monocytic THP1 cells to secrete IL-8 via the mitogen-activated protein kinase pathway. Tumour Biol. 2015;36(12):9171–7.CrossRefPubMed Nishio Y, Gojoubori T, Kaneko Y, Shimizu N, Asano M. Cancer cell-derived IL-8 induces monocytic THP1 cells to secrete IL-8 via the mitogen-activated protein kinase pathway. Tumour Biol. 2015;36(12):9171–7.CrossRefPubMed
52.
Zurück zum Zitat Khurram SA, Bingle L, McCabe BM, Farthing PM, Whawell SA. The chemokine receptors CXCR1 and CXCR2 regulate oral cancer cell behaviour. J Oral Pathol Med. 2014;43(9):667–74.CrossRefPubMed Khurram SA, Bingle L, McCabe BM, Farthing PM, Whawell SA. The chemokine receptors CXCR1 and CXCR2 regulate oral cancer cell behaviour. J Oral Pathol Med. 2014;43(9):667–74.CrossRefPubMed
53.
Zurück zum Zitat Ha NH, Park DG, Woo BH, da Kim J, Choi JI, Park BS, Kim YD, Lee JH, Park HR. Porphyromonas gingivalis increases the invasiveness of oral cancer cells by upregulating IL-8 and MMPs. Cytokine. 2016;86:64–72.CrossRefPubMed Ha NH, Park DG, Woo BH, da Kim J, Choi JI, Park BS, Kim YD, Lee JH, Park HR. Porphyromonas gingivalis increases the invasiveness of oral cancer cells by upregulating IL-8 and MMPs. Cytokine. 2016;86:64–72.CrossRefPubMed
54.
Zurück zum Zitat Tsunoda K, Tsujino I, Koshi R, et al. Nicotine-Mediated Ca(2+)-Influx Induces IL-8 Secretion in Oral Squamous Cell Carcinoma Cell. J Cell Biochem. 2016;117(4):1009–15.CrossRefPubMed Tsunoda K, Tsujino I, Koshi R, et al. Nicotine-Mediated Ca(2+)-Influx Induces IL-8 Secretion in Oral Squamous Cell Carcinoma Cell. J Cell Biochem. 2016;117(4):1009–15.CrossRefPubMed
55.
Zurück zum Zitat Gasparoto TH, de Souza Malaspina TS, Benevides L, et al. Patients with oral squamous cell carcinoma are characterized by increased frequency of suppressive regulatory T cells in the blood and tumor microenvironment. Cancer Immunol Immunother. 2010;59(6):819–28.CrossRefPubMed Gasparoto TH, de Souza Malaspina TS, Benevides L, et al. Patients with oral squamous cell carcinoma are characterized by increased frequency of suppressive regulatory T cells in the blood and tumor microenvironment. Cancer Immunol Immunother. 2010;59(6):819–28.CrossRefPubMed
56.
57.
Zurück zum Zitat Chuang C-Y, Sung W-W, Wang L, et al. Differential impact of IL-10 expression on survival and relapse between HPV16-positive and -negative oral squamous cell carcinomas. PLoS One. 2012;7(10):e47541.CrossRefPubMedPubMedCentral Chuang C-Y, Sung W-W, Wang L, et al. Differential impact of IL-10 expression on survival and relapse between HPV16-positive and -negative oral squamous cell carcinomas. PLoS One. 2012;7(10):e47541.CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Polz-Dacewicz M, Strycharz-Dudziak M, Dworzański J, Stec A, Kocot J. Salivary and serum IL-10, TNF-α, TGF-β, VEGF levels in oropharyngeal squamous cell carcinoma and correlation with HPV and EBV infections. Infect Agents Cancer. 2016;11:45.CrossRefPubMedPubMedCentral Polz-Dacewicz M, Strycharz-Dudziak M, Dworzański J, Stec A, Kocot J. Salivary and serum IL-10, TNF-α, TGF-β, VEGF levels in oropharyngeal squamous cell carcinoma and correlation with HPV and EBV infections. Infect Agents Cancer. 2016;11:45.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Wang S, Sun M, Gu C, et al. Expression of CD163, interleukin-10, and interferon-gamma in oral squamous cell carcinoma: mutual relationships and prognostic implications. Eur J Oral Sci. 2014;122(3):202–9.CrossRefPubMed Wang S, Sun M, Gu C, et al. Expression of CD163, interleukin-10, and interferon-gamma in oral squamous cell carcinoma: mutual relationships and prognostic implications. Eur J Oral Sci. 2014;122(3):202–9.CrossRefPubMed
60.
Zurück zum Zitat Gonçalves AS, Arantes DA, Bernardes VF, et al. Immunosuppressive mediators of oral squamous cell carcinoma in tumour samples and saliva. Hum Immunol. 2015;76(1):52–8.CrossRefPubMed Gonçalves AS, Arantes DA, Bernardes VF, et al. Immunosuppressive mediators of oral squamous cell carcinoma in tumour samples and saliva. Hum Immunol. 2015;76(1):52–8.CrossRefPubMed
61.
Zurück zum Zitat Naher L, Kiyoshima T, Kobayashi I, et al. STAT3 signal transduction through interleukin-22 in oral squamous cell carcinoma. Int J Oncol. 2012;41(5):1577–86.PubMedPubMedCentral Naher L, Kiyoshima T, Kobayashi I, et al. STAT3 signal transduction through interleukin-22 in oral squamous cell carcinoma. Int J Oncol. 2012;41(5):1577–86.PubMedPubMedCentral
62.
Zurück zum Zitat Banerjee S, Halder K, Bose A, et al. TLR signaling-mediated differential histone modification at IL-10 and IL-12 promoter region leads to functional impairments in tumor-associated macrophages. Carcinogenesis. 2011;32(12):1789–97.CrossRefPubMed Banerjee S, Halder K, Bose A, et al. TLR signaling-mediated differential histone modification at IL-10 and IL-12 promoter region leads to functional impairments in tumor-associated macrophages. Carcinogenesis. 2011;32(12):1789–97.CrossRefPubMed
63.
Zurück zum Zitat Ley S, Weigert A, Weichand B, Henke N, Mille-Baker B, Janssen RA, Brüne B. The role of TRKA signaling in IL-10 production by apoptotic tumor cell-activated macrophages. Oncogene. 2013;32(5):631–40.CrossRefPubMed Ley S, Weigert A, Weichand B, Henke N, Mille-Baker B, Janssen RA, Brüne B. The role of TRKA signaling in IL-10 production by apoptotic tumor cell-activated macrophages. Oncogene. 2013;32(5):631–40.CrossRefPubMed
65.
Zurück zum Zitat Wu G, Ma Z, Hu W, Wang D, Gong B, Fan C, Jiang S, Li T, Gao J, Yang Y. Molecular insights of Gas6/TAM in cancer development and therapy. Cell Death Dis. 2017;8(3):e2700. doi: 10.1038/cddis.2017.113. Review. PMID: 28333143. Wu G, Ma Z, Hu W, Wang D, Gong B, Fan C, Jiang S, Li T, Gao J, Yang Y. Molecular insights of Gas6/TAM in cancer development and therapy. Cell Death Dis. 2017;8(3):e2700. doi: 10.​1038/​cddis.​2017.​113. Review. PMID: 28333143.
66.
Zurück zum Zitat Lee CH, Yen CY, Liu SY, Chen CK, Chiang CF, Shiah SG, Chen PH, Shieh YS. Axl is a prognostic marker in oral squamous cell carcinoma. Ann Surg Oncol. 2012;19 Suppl 3:S500–8.CrossRefPubMed Lee CH, Yen CY, Liu SY, Chen CK, Chiang CF, Shiah SG, Chen PH, Shieh YS. Axl is a prognostic marker in oral squamous cell carcinoma. Ann Surg Oncol. 2012;19 Suppl 3:S500–8.CrossRefPubMed
67.
Zurück zum Zitat Patankar SR, Wankhedkar DP, Tripathi NS, Bhatia SN, Sridharan G. Extracellular matrix in oral squamous cell carcinoma: Friend or foe? Indian J Dent Res. 2016;27(2):184-9. doi:10.4103/0970-9290.183125. Patankar SR, Wankhedkar DP, Tripathi NS, Bhatia SN, Sridharan G. Extracellular matrix in oral squamous cell carcinoma: Friend or foe? Indian J Dent Res. 2016;27(2):184-9. doi:10.​4103/​0970-9290.​183125.
68.
Zurück zum Zitat Jiang T, Liu G, Wang L, Liu H. Elevated Serum Gas6 Is a Novel Prognostic Biomarker in Patients with Oral Squamous Cell Carcinoma. PLoS One. 2015;10(7):e0133940.CrossRefPubMedPubMedCentral Jiang T, Liu G, Wang L, Liu H. Elevated Serum Gas6 Is a Novel Prognostic Biomarker in Patients with Oral Squamous Cell Carcinoma. PLoS One. 2015;10(7):e0133940.CrossRefPubMedPubMedCentral
69.
Zurück zum Zitat Kumar V, Gabrilovich DI. Hypoxia-inducible factors in regulation of immune responses in tumour microenvironment. Immunology. 2014;143(4):512–9.CrossRefPubMedPubMedCentral Kumar V, Gabrilovich DI. Hypoxia-inducible factors in regulation of immune responses in tumour microenvironment. Immunology. 2014;143(4):512–9.CrossRefPubMedPubMedCentral
70.
Zurück zum Zitat Cheng Y-SL, Rees T, Wright J. A review of research on salivary biomarkers for oral cancer detection. Clin Trans Med. 2014;3:3.CrossRef Cheng Y-SL, Rees T, Wright J. A review of research on salivary biomarkers for oral cancer detection. Clin Trans Med. 2014;3:3.CrossRef
71.
Zurück zum Zitat Rajkumar K, Nandhini G, Ramya R, Rajashree P, Kumar AR, Anandan SN. Validation of the diagnostic utility of salivary interleukin 8 in the differentiation of potentially malignant oral lesions and oral squamous cell carcinoma in a region with high endemicity. Oral Surg Oral Med Oral Pathol Oral Radiol. 2014;118(3):309–19.CrossRefPubMed Rajkumar K, Nandhini G, Ramya R, Rajashree P, Kumar AR, Anandan SN. Validation of the diagnostic utility of salivary interleukin 8 in the differentiation of potentially malignant oral lesions and oral squamous cell carcinoma in a region with high endemicity. Oral Surg Oral Med Oral Pathol Oral Radiol. 2014;118(3):309–19.CrossRefPubMed
72.
Zurück zum Zitat Aziz S, Ahmed SS, Ali A, et al. Salivary Immunosuppressive Cytokines IL-10 and IL-13 Are Significantly Elevated in Oral Squamous Cell Carcinoma Patients. Cancer Invest. 2015;33(7):318–28.CrossRefPubMed Aziz S, Ahmed SS, Ali A, et al. Salivary Immunosuppressive Cytokines IL-10 and IL-13 Are Significantly Elevated in Oral Squamous Cell Carcinoma Patients. Cancer Invest. 2015;33(7):318–28.CrossRefPubMed
73.
Zurück zum Zitat Panneer Selvam N, Sadaksharam J. Salivary interleukin-6 in the detection of oral cancer and precancer. Asia Pac J Clin Oncol. 2015;11(3):236–41.CrossRefPubMed Panneer Selvam N, Sadaksharam J. Salivary interleukin-6 in the detection of oral cancer and precancer. Asia Pac J Clin Oncol. 2015;11(3):236–41.CrossRefPubMed
74.
Zurück zum Zitat Bagan L, Sáez GT, Tormos MC, et al. Salivary and serum interleukin-6 levels in proliferative verrucous leukoplakia. Clin Oral Investig. 2016;20(4):737–43.CrossRefPubMed Bagan L, Sáez GT, Tormos MC, et al. Salivary and serum interleukin-6 levels in proliferative verrucous leukoplakia. Clin Oral Investig. 2016;20(4):737–43.CrossRefPubMed
76.
Zurück zum Zitat Modi BG, Neustadter J, Binda E, et al. Langerhans Cells Facilitate Epithelial DNA Damage and Squamous Cell Carcinoma. Science (New York, NY). 2012;335(6064):104–8.CrossRef Modi BG, Neustadter J, Binda E, et al. Langerhans Cells Facilitate Epithelial DNA Damage and Squamous Cell Carcinoma. Science (New York, NY). 2012;335(6064):104–8.CrossRef
77.
Zurück zum Zitat Hayashi N, Kataoka H, Yano S, Tanaka M, Moriwaki K, Akashi H, Suzuki S, Mori Y, Kubota E, Tanida S, Takahashi S, Joh T. A novel photodynamic therapy targeting cancer cells and tumor-associated macrophages. Mol Cancer Ther. 2015;14(2):452–60.CrossRefPubMed Hayashi N, Kataoka H, Yano S, Tanaka M, Moriwaki K, Akashi H, Suzuki S, Mori Y, Kubota E, Tanida S, Takahashi S, Joh T. A novel photodynamic therapy targeting cancer cells and tumor-associated macrophages. Mol Cancer Ther. 2015;14(2):452–60.CrossRefPubMed
78.
Zurück zum Zitat Junankar S, Shay G, Jurczyluk J, et al. Real-time intravital imaging establishes tumour-associated macrophages as the extraskeletal target of bisphosphonate action in cancer. Cancer Discov. 2015;5(1):35–42.CrossRefPubMed Junankar S, Shay G, Jurczyluk J, et al. Real-time intravital imaging establishes tumour-associated macrophages as the extraskeletal target of bisphosphonate action in cancer. Cancer Discov. 2015;5(1):35–42.CrossRefPubMed
79.
Zurück zum Zitat Mancino A, Lawrence T. Nuclear factor-kappaB and tumor-associated macrophages. Clin Cancer Res. 2010;16(3):784–9.CrossRefPubMed Mancino A, Lawrence T. Nuclear factor-kappaB and tumor-associated macrophages. Clin Cancer Res. 2010;16(3):784–9.CrossRefPubMed
Metadaten
Titel
Role of tumour-associated macrophages in oral squamous cells carcinoma progression: an update on current knowledge
verfasst von
Maria Noel Marzano Rodrigues Petruzzi
Karen Cherubini
Fernanda Gonçalves Salum
Maria Antonia Zancanaro de Figueiredo
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Diagnostic Pathology / Ausgabe 1/2017
Elektronische ISSN: 1746-1596
DOI
https://doi.org/10.1186/s13000-017-0623-6

Weitere Artikel der Ausgabe 1/2017

Diagnostic Pathology 1/2017 Zur Ausgabe

Neu im Fachgebiet Pathologie