Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2371 2023-07-02 17:26:28 |
2 format Meta information modification 2371 2023-07-03 04:33:00 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Alonso-Juarranz, M.; Mascaraque, M.; Carrasco, E.; Gracia-Cazaña, T.; De La Sen, O.; Gilaberte, Y.; Gonzalez, S.; Juarranz, �.; Falahat, F. Tumor Microenvironment of Squamous Cell Carcinomas. Encyclopedia. Available online: https://encyclopedia.pub/entry/46319 (accessed on 30 June 2024).
Alonso-Juarranz M, Mascaraque M, Carrasco E, Gracia-Cazaña T, De La Sen O, Gilaberte Y, et al. Tumor Microenvironment of Squamous Cell Carcinomas. Encyclopedia. Available at: https://encyclopedia.pub/entry/46319. Accessed June 30, 2024.
Alonso-Juarranz, Miguel, Marta Mascaraque, Elisa Carrasco, Tamara Gracia-Cazaña, Oscar De La Sen, Yolanda Gilaberte, Salvador Gonzalez, Ángeles Juarranz, Farzin Falahat. "Tumor Microenvironment of Squamous Cell Carcinomas" Encyclopedia, https://encyclopedia.pub/entry/46319 (accessed June 30, 2024).
Alonso-Juarranz, M., Mascaraque, M., Carrasco, E., Gracia-Cazaña, T., De La Sen, O., Gilaberte, Y., Gonzalez, S., Juarranz, �., & Falahat, F. (2023, July 02). Tumor Microenvironment of Squamous Cell Carcinomas. In Encyclopedia. https://encyclopedia.pub/entry/46319
Alonso-Juarranz, Miguel, et al. "Tumor Microenvironment of Squamous Cell Carcinomas." Encyclopedia. Web. 02 July, 2023.
Tumor Microenvironment of Squamous Cell Carcinomas
Edit

Squamous cell carcinomas arise from stratified squamous epithelia. Here, a comparative analysis based on recent studies defining the genetic alterations and composition of the stroma of oral and cutaneous squamous cell carcinomas (OSCC and CSCC, respectively) was performed. Both carcinomas share some but not all histological and genetic features. 

oral squamous cell carcinoma cutaneous squamous cell carcinoma tumor microenvironment cancer-associated fibroblasts

1. Introduction

SCC and its precursor lesions are complex systems in which neoplastic cells coexist with the other cell types and tissue components that comprise the tumor microenvironment (TME). The TME contains multiple different cell types, including cancer-associated fibroblasts (CAFs), neutrophils, macrophages, and regulatory T cells. Tumor cells and TME cell populations interact with each other via complex communication networks through the various secreted cytokines, chemokines, growth factors, and proteins of the extracellular matrix (ECM). The TME is known to be implicated in cancer cell survival, tumor progression, and the tumor response to therapy [1].

2. Extracellular Matrix

The extracellular matrix (ECM) is a non-cellular network of macromolecules (collagen, fibronectin, and laminin, etc.) that offers structural and biochemical support for cellular components, enabling it to influence cell communication, adhesion, and proliferation [2]. In cancer, the ECM is frequently deregulated and disorganized, which directly stimulates malignant cell transformation. Matrix metalloproteinases (MMPs) are critical molecules for the EMT process because they not only degrade cell adhesion molecules, favoring migration and metastasis, but also promote the initiation and proliferation of primary tumors [3]. MMPS are produced by tumor and stromal cells, such as fibroblast and inflammatory cells [2][3][4]. Alterations in the proteins of the ECM may impact the tumor progression in SCCs. Collagen is the major protein component of the ECM, which provides the cells with tensile strength and support for migration [2]. The loss of type IV collagen correlates with poorly differentiated OSCC [3] and CSCC [5]. Fibronectin is produced by fibroblasts and endothelial cells and mediates the cellular interaction with the ECM. In the development of cancer, increased levels of fibronectin have been associated with increased tumor progression, migration, and invasion, as well as an impaired response to treatment [6][7]. Additionally, the expression of the laminin receptor plays an important role in SCC progression [7][8], as reduced laminin expression has been correlated with an invasive phenotype of OSCC tumors [5].

2.2. Cancer Associated Fibroblasts

Fibroblasts are one of the main cell components of the connective tissue subjacent to the epithelia. The main functions of these cells are the synthesis of the ECM (collagen, laminin, and fibronectin, including those needed to form basal membranes), the regulation of epithelial differentiation, and the promotion of wound closure [9][10]. CAFs are activated fibroblasts with mesenchymal characteristics associated with cancer cells, which contribute to tumor-promoting inflammation and fibrosis. CAFs acquire specific characteristics, such as a distinct morphology (an elongated spindle-like shape), and express differential markers (α-sma—Alpha-Smooth Muscle Actin; FAP-1—Fibroblast Activation Protein-1; vimentin and S100A4) and a lack of lineage markers for epithelial cells, endothelial cells, and hematopoietic cells [11][12]. However, the precise origins and roles of the fibroblast populations within the tumor microenvironment remain poorly understood.
In the case of HNSCC, several populations of CAFs have been described that, according to specific markers, can be classified into three subgroups: classical CAFs, normal activated fibroblasts, and elastic fibroblasts. Classical CAFs are enriched for genes, encoding proteins such as FAP, PDGF (platelet-derived growth factor receptor), lysyl oxidase, and MMPs. Normal activated fibroblasts show a low expression of CAF markers and elastic fibroblasts are enriched for tropoelastin, fibrillin 1, and microfibril-associated protein 4. It seems that the presence of different CAFs can be related to overall patient prognoses [13].
CAFs are key in different stages of the development of OSSC and CSCC, stimulating the growth and progression of tumors and participating in the maintenance of a state of poor differentiation of the surrounding cells. They act synergistically with epithelial cells to promote carcinogenesis and influence the patterns of invasiveness and metastasis [13][14]. In this sense, CAFs could act in the initiation process of cancer, favoring the mutagenicity of epithelial cells, for example, by secreting ROS, which favor decreases in the pH and hypoxia in the TEM [15]. In tumor progression, they promote the migration and invasion of tumor cells by chronically maintaining the proinflammatory stimuli via the promotion of oxidative stress. They also contribute to this end by secreting a broad amount of cytokines and chemokines, such as transforming growth factor beta (TGFβ) and interleukins (IL-1 and IL-6), and a broad range of growth factors, such as EGF (epidermal growth factor), bFGF (basic fibroblast growth factor), VEGF (vascular endothelial growth factor), HGF (hepatocyte growth factor), tumor necrosis factor (TNF), interferon-(IFN), CXCL12, IL-6, galectin-1, sonic hedgehog protein (SHH), and bone morphogenetic protein (BMP), among others, which are tumor-promoting. In particular, HGF has been described to promote glycolysis in HNSCC cells [15]. All these molecules can influence tumor cell growth, angiogenesis, and the recruitment of immunosuppressive immune cells [16][17][18]. CAFs are also crucial producers of MMPs, playing an important role in modulating the TME through the remodeling and degradation of the ECM, which ultimately results in the promotion of the invasive phenotype of cancer cells [18][19][20].
In addition, they promote the EMT by secreting a variety of soluble activators that initiate the TGFβ cascade in epithelial tumor cells, leading to a change in morphology and response, acquiring mesenchymal characteristics [21][22]. As a major secreted factor of CAFs, TGFβ predominantly mediates the crosstalk between CAFs and cancer cells. Several in vitro studies have demonstrated that, in OSCC and CSCC, the TGFβ secreted by CAFs induces EMT and resistance to different therapies [23][24][25][26][27].

2.3. Immune Cells

The immune cell component of the TME is formed by tumor-infiltrating lymphocytes (TILs, including CD4+ and CD8+ T cells, B cells, natural killer T cells, and myeloid lineage cells (macrophages, neutrophils, monocytes, eosinophils, myeloid-derived suppressor cells or MDSCs, and mast cells or MCs)). In general, OSCC and CSCC tumors are highly infiltrated by immune cells, although the extent and composition of the immune cell infiltrate vary according to the anatomical subsite and etiological agent [28][29].
Tumor-infiltrating lymphocytes (TILs) are the major cell type in adaptive immunity that recognize specific antigens and produce specific immune responses. High levels of TILs generally correspond to better outcomes in OSCC, but this is dependent on the balance of cells with anti-tumor activity (effector T or Teff cells) versus those with immunosuppressive activity (regulatory T or Treg cells) in the TIL population [28][30].Teffs are related to high levels of cytotoxic CD8+, which produces IFN-ϒ [28]. On the contrary, high levels of CD4+Foxp3+ or Treg cells through IL-4 and IL-10 production have been correlated with immunosuppression and pro-tumor activity, triggering poor outcomes [31][32]. Likewise, UV radiation also causes an increase in Treg and a decrease in Teff cells in the skin, leading to a change in the T-cell balance and promoting the development of CSCC [33]. On the other hand, the presence of B lymphocytes is related to higher levels of CD8+ cell infiltration and, therefore, to a better prognosis in OSCC and CSCC [34][35].
Tertiary lymphoid structures (TLS) are crucial elements of the tumor immune microenvironment, corresponding to sites of lymphoid neogenesis with the potential of orchestrating anti-tumor responses. They correspond to ectopic lymphoid organs, emerging in the context of chronic inflammation such as the TME and even allowing for germinal center formation [36][37][38]. In OSCC patients, a high density of TLS has been associated with a better overall survival and identified as an independent positive prognostic factor [39][40]. In the case of CSCC, though not much work has been performed, clinically, the presence of TLS has been prominently associated with a better degree of histopathological grades and a higher level of sun exposure. Furthermore, the presence of intratumoral TLS has been associated with lower lymphovascular invasion. Therefore, TLSs are considered to be a positive prognostic factor for CSCC and will provide a theoretical basis for the future diagnostic and therapeutic value in this type of cancer [41]. The features and clinical significance of TLSs in SCC still remain unknown.
Tumor-associated macrophages (TAMs) interact, modulate, and influence tumor progression, invasion, and metastasis. Macrophages display a great plasticity, oscillating between M1 (antitumoral) and M2 (protumoral) phenotypes. M1 macrophages produce pro-inflammatory cytokines (IL-12 and IL-23), tumor necrosis factor-α (TNF-α), and chemokines (CCL-5, CXCL9, CXCL10, and CXCL5), which promote adaptive immunity. They also express high levels of major histocompatibility complex 2 (MHC-2) molecules, allowing for the presentation of tumor antigens [42][43]. In contrast, M2 macrophages play an immunoregulatory role and are involved in tissue remodeling, angiogenesis, and tumor progression. M2 macrophages act by releasing anti-inflammatory cytokines (IL-4, IL-13, IL-10, and TGFβ, etc.), overexpressing PD-L1 (Programmed death-ligand 1) and expressing comparatively lower levels of MHC-2 molecules [42][43][44]. Several studies have suggested a correlation between the level of TAM infiltration and a poor outcome in OSCC, which could be used as a potential prognostic marker [45][46].
Myeloid-derived suppressor cells (MDCSs) comprise a heterogeneous population of cells that play a crucial role in the negative regulation of the immune response in cancer by inhibiting both adaptive and innate immunity, establishing the premetastatic niche in different types of cancer [47][48]. In addition, MDSCs have also been linked to angiogenesis and the degradation of the ECM [49][50]. A high abundance of circulating MDSCs correlates with advanced stages of OSCC and is also known to promote CSCC development [50][51][52].
Mast cells (MCs) represent another important myeloid component of the immune system. MCs in the TME may have pro-tumoral functions, such as the promotion of angiogenesis (through VEGF production), ECM degradation (via MMPs production), and the induction of tumor cell proliferation (through tryptase and histamine) [53][54]. In OSCC and CSCC, the protective and pro-tumoral role of MCs has been described in several studies [55][56][57].

2.4. The Importance of the TME for the Treatment of OSCC and CSCC

OSCC and CSCC are generally treated with surgical resection and, depending on the disease state, this is accompanied by radiation or chemotherapy [58][59][60]. Traditional tumor treatment methods cannot solve the problems of tumor recurrence and metastasis, which are often associated with the TME, as mentioned in the previous section. Therefore, new single or combined strategies are being developed to address the TME, as described below.

2.4.1. CAF-Targeting Strategies

The importance of CAFs in tumor development and their role in therapy resistance have been demonstrated in several types of cancer, including OSCC and CSCC [16]. CAF-mediated resistance to cetuximab has been reported in OSCC [61][62]. Additionally, in CSCC, the presence of CAFS has been found to increase resistance to photodynamic therapy [23], suggesting that therapeutic CAF targeting could increase the response rates for a diverse range of treatments.
One of the main mediators related to these resistance effects is TGFβ. This cytokine modulates the tumor progression and therapy response through the CAF activation status, shape, and invasiveness [63]. Quan et al. [64] suggested that TGF-β1 induces EMT to increase the capacity of OSCC for invasion, and Gallego et al. [23] described that an increased secretion of CAF-derived TFGβ mediates resistance in CSCC. However, targeting TGF-β is potentially problematic for its dual role: in the early stages of tumorigenesis it can act as a tumor suppressor, while acting as a tumor promoter in later stages [65]. Even so, it has been described that a novel TGFβ inhibitor promoted anti-tumor immune responses in OSCC, alone and in combination with anti PD-L1 antibodies [66]. Although the signaling cascades involving TGFβ are the primary signaling pathways regulating CAF activation, there are other growth factors and signaling molecules also implicated in the differentiation process, including NOX4 (NADPH oxidase 4), FGF, IL-6, or TNF [67]. Hanley et al. [68] identified NOX4 as a critical regulator of CAF activation in OSCC, and its inhibitor Setanaxib suppressed CAF activation. In OSCCs and CSCCs, there have not been many more studies targeting CAFS in order to prevent resistance. However, in other cancers, more trials have been described, such as the depletion of FAP-expressing cells as an adjuvant to immunotherapy [69]. Additionally, a combination of paclitaxel (which suppresses the expression of α-SMA) with gemcitabine improved the overall survival in pancreatic cancer patients [70], suggesting a possible new therapeutic window for OSCC and CSCC.

2.4.2. Immunotherapy

Immunotherapy has outstanding application value in the field of tumor therapy, including antibody-based therapy, cytokine therapy, and gene therapy.
One of the main strategies for eliminating SCCs is based on the use of monoclonal antibodies (mAbs), which include immune checkpoint inhibitors or anti-angiogenesis mAbs.
There are several clinical trials focused on mAbs for OSCC and CSCC treatment.
PD-1 is a checkpoint protein belonging to a group of T cell receptors involved in T cell suppression. PD-1 is also expressed by B cells, monocytes, and natural killer and dendritic cells [71]. This transmembrane protein binds to PD-L1, which is present on the surface of tumor cells, and this interaction triggers a signal that inhibits the activated T cells and induces immunological exhaustion and T cell apoptosis [71][72]. Then, the PD-L1/PD-1 axis is a primary mechanism of cancer immune evasion and has thus become the main target for the development new drugs that have emerged in recent years. Targeting the immune checkpoint proteins with mAbs has yielded a net clinical benefit in cancer [73][74]. So far, several mAbs have been approved for PD-1/PD-L1 blockade in clinical studies for oral cancer treatment, including Cemiplimab, Nivolumab, Sintilimab, Toripalimab, Pembrolizumab, Aezolizumab, Avelumab, Camreluzimab, and Durvalamab. Likewise, mAbs blocking other immune checkpoint receptors such as CTLA-4 (Tremelimumab) are being studied [75]. Finally, in OSCC, the effect of inhibiting OX40, a costimulatory molecule that can enhance T cell immunity, is also being tested. Anti-human OX40 was used in a phase I clinical trial (NCT02274155) prior to surgery. The results demonstrated that anti-OX40 mAb could induce the activation and proliferation of T cells in hosts, suggesting its successful potential as a clinical strategy [76].
On the other hand, as angiogenesis plays an important role in tumor development and metastasis, mAbs are also being tested to inhibit these processes in both OSCC and CSCC. Cetuximab, an anti-EGFR mAb, and bevacizumab, an anti-VEGFR mAb, are being administered in clinical trials, either alone or in combination with other treatments.
Macrophages and fibroblasts, among other cell types belonging to the TME, produce different cytokines that can be pro- or anti-tumorigenic [25][42][43]. Several cytokines clinical trials have been completed in OSCC, although the results have not been published yet. Some of these trials are related to the administration of IL-2 (NCT00899821 and NCT00019331) or INFα (NCT00276523, NCT00054561, NCT00002506, and NCT00014261), in order to see if they promote anti-tumor responses in combination with other treatments.

References

  1. Anderson, N.M.; Simón, M.C. The tumor microenvironment. Curr. Biol. 2020, 30, R921–R925.
  2. Walker, C.; Mojares, E.; del Río Hernández, A. Role of Extracellular Matrix in Development and Cancer Progression. Int. J. Mol. Sci. 2018, 19, 3028.
  3. Najafi, M.; Farhood, B.; Mortezaee, K. Extracellular matrix (ECM) stiffness and degradation as cancer drivers. J. Cell. Biochem. 2019, 120, 2782–2790.
  4. Thomson, J.; Bewicke-Copley, F.; Anene, C.A.; Gulati, A.; Nagano, A.; Purdie, K.; Inman, G.J.; Proby, C.M.; Leigh, I.M.; Harwood, C.A.; et al. The Genomic Landscape of Actinic Keratosis. J. Investig. Dermatol. 2021, 141, 1664–1674.e7.
  5. Harada, T.; Shinohara, M.; Nakamura, S.; Oka, M. An immunohistochemical study of the extracellular matrix in oral squamous cell carcinoma and its association with invasive and metastatic potential. Virchows Archiv. 1994, 424, 257–266.
  6. Knowles, L.M.; Gurski, L.A.; Engel, C.; Gnarra, J.R.; Maranchie, J.K.; Pilch, J. Integrin αvβ3 and Fibronectin Upregulate Slug in Cancer Cells to Promote Clot Invasion and Metastasis. Cancer Res. 2013, 73, 6175–6184.
  7. Scanlon, C.S.; Van Tubergen, E.A.; Inglehart, R.C.; D’Silva, N.J. Biomarkers of Epithelial-Mesenchymal Transition in Squamous Cell Carcinoma. J. Dent. Res. 2013, 92, 114–121.
  8. Janes, S.M.; Watt, F.M. New roles for integrins in squamous-cell carcinoma. Nat. Rev. Cancer 2006, 6, 175–183.
  9. Thulabandu, V.; Chen, D. Atit RP. Dermal fibroblast in cutaneous development and healing. WIREs Dev. Biol. 2018, 7, 307.
  10. Driskell, R.R.; Watt, F.M. Understanding fibroblast heterogeneity in the skin. Trends Cell Biol. 2015, 25, 92–99.
  11. Teichgräber, V.; Monasterio, C.; Chaitanya, K.; Boger, R.; Gordon, K.; Dieterle, T.; Jäger, D.; Bauer, S. Specific inhibition of fibroblast activation protein (FAP)-alpha prevents tumor progression in vitro. Adv. Med. Sci. 2015, 60, 264–272.
  12. Nurmik, M.; Ullmann, P.; Rodriguez, F.; Haan, S.; Letellier, E. In search of definitions: Cancer-associated fibroblasts and their markers. Int. J. Cancer 2020, 146, 895–905.
  13. Sasaki, K.; Sugai, T.; Ishida, K.; Osakabe, M.; Amano, H.; Kimura, H.; Sakuraba, M.; Kashiwa, K.; Kobayashi, S. Analysis of cancer-associated fibroblasts and the epithelial-mesenchymal transition in cutaneous basal cell carcinoma, squamous cell carcinoma, and malignant melanoma. Hum. Pathol. 2018, 79, 1–8.
  14. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598.
  15. Kumar, D.; New, J.; Vishwakarma, V.; Joshi, R.; Enders, J.; Lin, F.; Dasari, S.; Gutierrez, W.R.; Leef, G.; Ponnurangam, S.; et al. Cancer-Associated Fibroblasts Drive Glycolysis in a Targetable Signaling Loop Implicated in Head and Neck Squamous Cell Carcinoma Progression. Cancer Res 2018, 78, 3769–3782.
  16. Kuzet, S.E.; Gaggioli, C. Fibroblast activation in cancer: When seed fertilizes soil. Cell Tissue Res. 2016, 365, 607–619.
  17. Smithmyer, M.E.; Spohn, J.B.; Kloxin, A.M. Probing Fibroblast Activation in Response to Extracellular Cues with Whole Protein- or Peptide-Functionalized Step-Growth Hydrogels. ACS Biomater. Sci. Eng. 2018, 4, 3304–3316.
  18. Joshi, R.S.; Kanugula, S.S.; Sudhir, S.; Pereira, M.P.; Jain, S.; Aghi, M.K. The Role of Cancer-Associated Fibroblasts in Tumor Progression. Cancers 2021, 13, 1399.
  19. Eke, I.; Storch, K.; Krause, M.; Cordes, N. Cetuximab Attenuates Its Cytotoxic and Radiosensitizing Potential by Inducing Fibronectin Biosynthesis. Cancer Res. 2013, 73, 5869–5879.
  20. Grauel, A.L.; Nguyen, B.; Ruddy, D.; Laszewski, T.; Schwartz, S.; Chang, J.; Chen, J.; Piquet, M.; Pelletier, M.; Yan, Z.; et al. TGFβ-blockade uncovers stromal plasticity in tumors by revealing the existence of a subset of interferon-licensed fibroblasts. Nat. Commun. 2020, 11, 6315.
  21. Ghahremanifard, P.; Chanda, A.; Bonni, S.; Bose, P. TGF-β Mediated Immune Evasion in Cancer—Spotlight on Cancer-Associated Fibroblasts. Cancers 2020, 12, 3650.
  22. Xiao, L.; Zhu, H.; Shu, J.; Gong, D.; Zheng, D.; Gao, J. Overexpression of TGF-β1 and SDF-1 in cervical cancer-associated fibroblasts promotes cell growth, invasion and migration. Arch. Gynecol. Obstet. 2022, 305, 179–192.
  23. Gallego-Rentero, M.; Gutiérrez-Pérez, M.; Fernández-Guarino, M.; Mascaraque, M.; Portillo-Esnaola, M.; Gilaberte, Y.; Carrasco, E.; Juarranz, Á. TGFβ1 Secreted by Cancer-Associated Fibroblasts as an Inductor of Resistance to Photodynamic Therapy in Squamous Cell Carcinoma Cells. Cancers 2021, 13, 5613.
  24. Guo, S.; Deng, C.X. Effect of Stromal Cells in Tumor Microenvironment on Metastasis Initiation. Int. J. Biol. Sci. 2018, 14, 2083–2093.
  25. Wheeler, S.E.; Shi, H.; Lin, F.; Dasari, S.; Bednash, J.; Thorne, S.; Watkins, S.; Joshi, R.; Thomas, S.M. Enhancement of head and neck squamous cell carcinoma proliferation, invasion, and metastasis by tumor-associated fibroblasts in preclinical models. Head Neck 2013, 36, 385–392.
  26. Hwang, Y.S.; Xianglan, Z.; Park, K.K.; Chung, W.Y. Functional invadopodia formation through stabilization of the PDPN transcript by IMP-3 and cancer-stromal crosstalk for PDPN expression. Carcinogenesis 2012, 33, 2135–2146.
  27. Ruffin, A.T.; Li, H.; Vujanovic, L.; Zandberg, D.P.; Ferris, R.L.; Bruno, T.C. Improving head and neck cancer therapies by immunomodulation of the tumour microenvironment. Nat. Rev. Cancer 2023, 23, 173–188.
  28. Ansary, T.M.; Hossain, M.R.; Komine, M.; Ohtsuki, M. Immunotherapy for the Treatment of Squamous Cell Carcinoma: Potential Benefits and Challenges. Int. J. Mol. Sci. 2022, 23, 8530.
  29. Fang, J.; Li, X.; Da Ma, D.; Liu, X.; Chen, Y.; Wang, Y.; Lui, V.W.Y.; Xia, J.; Bin Cheng, B.; Wang, Z. Prognostic significance of tumor infiltrating immune cells in oral squamous cell carcinoma. BMC Cancer 2017, 17, 375.
  30. Zhang, X.; Zeng, Y.; Qu, Q.; Zhu, J.; Liu, Z.; Ning, W.; Zeng, H.; Zhang, N.; Du, W.; Chen, C.; et al. PD-L1 induced by IFN-γ from tumor-associated macrophages via the JAK/STAT3 and PI3K/AKT signaling pathways promoted progression of lung cancer. Int. J. Clin. Oncol. 2017, 22, 1026–1033.
  31. Woolaver, R.; Wang, X.; Krinsky, A.L.; Waschke, B.C.; Chen, S.M.Y.; Popolizio, V.; Nicklawsky, A.G.; Gao, D.; Chen, Z.; Jimeno, A.; et al. Differences in TCR repertoire and T cell activation underlie the divergent outcomes of antitumor immune responses in tumor-eradicating versus tumor-progressing hosts. J. Immunother. Cancer 2021, 9, e001615.
  32. Gurin, D.; Slavik, M.; Hermanova, M.; Selingerova, I.; Kazda, T.; Hendrych, M.; Shatokhina, T.; Vesela, M. The tumor immune microenvironment and its implications for clinical outcome in patients with oropharyngeal squamous cell carcinoma. J. Oral Pathol. Med. 2020, 49, 886–896.
  33. Nasti, T.H.; Iqbal, O.; Tamimi, I.A.; Geise, J.T.; Katiyar, S.K.; Yusuf, N. Differential Roles of T-cell Subsets in Regulation of Ultraviolet Radiation Induced Cutaneous Photocarcinogenesis. Photochem. Photobiol. 2011, 87, 387–398.
  34. Hladíková, K.; Koucký, V.; Bouček, J.; Laco, J.; Grega, M.; Hodek, M.; Zábrodský, M.; Vošmik, M.; Rozkošová, K.; Vošmiková, H.; et al. Tumor-infiltrating B cells affect the progression of oropharyngeal squamous cell carcinoma via cell-to-cell interactions with CD8+ T cells. J. Immunother. Cancer 2019, 7, 261.
  35. Fogarty, G.B.; Bayne, M.; Bedford, P.; Bond, R.; Kannourakis, G. Three Cases of Activation of Cutaneous Squamous-cell Carcinoma During Treatment with Prolonged Administration of Rituximab. Clin. Oncol. 2006, 18, 155–156.
  36. Sautès-Fridman, C.; Petitprez, F.; Calderaro, J.; Fridman, W.H. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat. Rev. Cancer 2019, 19, 307–325.
  37. Schumacher, T.N.; Thommen, D.S. Tertiary lymphoid structures in cancer. Science 2022, 375, eabf9419.
  38. Zhao, Z.; Ding, H.; Lin, Z.-B.; Qiu, S.-H.; Zhang, Y.-R.; Guo, Y.-G.; Chu, X.-D.; I Sam, L.; Pan, J.-H.; Pan, Y.-L. Relationship between Tertiary Lymphoid Structure and the Prognosis and Clinicopathologic Characteristics in Solid Tumors. Int. J. Med. Sci. 2021, 18, 2327–2338.
  39. Domblides, C.; Rochefort, J.; Riffard, C.; Panouillot, M.; Lescaille, G.; Teillaud, J.-L.; Mateo, V.; Dieu-Nosjean, M.-C. Tumor-Associated Tertiary Lymphoid Structures: From Basic and Clinical Knowledge to Therapeutic Manipulation. Front. Immunol. 2021, 12, 698604.
  40. Wu, Y.; Wu, F.; Yan, G.; Zeng, Q.; Jia, N.; Zheng, Z.; Fang, S.; Liu, Y.; Zhang, G.; Wang, X. Features and clinical significance of tertiary lymphoid structure in cutaneous squamous cell carcinoma. J. Eur. Acad. Dermatol. Venereol. 2022, 36, 2043–2050.
  41. Lerman, I.; Mitchell, D.C.; Richardson, C.T. Human cutaneous B cells: What do we really know? Ann. Transl. Med. 2021, 9, 440–449.
  42. Anfray, C.; Ummarino, A.; Torres-Andón, F.; Allavena, P. Current Strategies to Target Tumor-Associated-Macrophages to Improve Anti-Tumor Immune Responses. Cells 2019, 9, 46.
  43. Mantovani, A.; Biswas, S.K.; Galdiero, M.R.; Sica, A.; Locati, M. Macrophage plasticity and polarization in tissue repair and remodelling. J. Pathol. 2013, 229, 176–185.
  44. Makita, N.; Hizukuri, Y.; Yamashiro, K.; Murakawa, M.; Hayashi, Y. IL-10 enhances the phenotype of M2 macrophages induced by IL-4 and confers the ability to increase eosinophil migration. Int. Immunol. 2015, 27, 131–141.
  45. Hu, Y.; He, M.-Y.; Zhu, L.-F.; Yang, C.-C.; Zhou, M.-L.; Wang, Q.; Zhang, W.; Zheng, Y.-Y.; Wang, D.-M.; Xu, Z.-Q.; et al. Tumor-associated macrophages correlate with the clinicopathological features and poor outcomes via inducing epithelial to mesenchymal transition in oral squamous cell carcinoma. J. Exp. Clin. Cancer Res. 2016, 35, 12.
  46. Seminerio, I.; Kindt, N.; Descamps, G.; Bellier, J.; Lechien, J.R.; Mat, Q.; Pottier, C.; Journé, F.; Saussez, S. High infiltration of CD68+ macrophages is associated with poor prognoses of head and neck squamous cell carcinoma patients and is influenced by human papillomavirus. Oncotarget 2018, 9, 11046–11059.
  47. Pang, X.; Fan, H.-Y.; Tang, Y.-L.; Wang, S.-S.; Cao, M.-X.; Wang, H.-F.; Dai, L.-L.; Wang, K.; Yu, X.-H.; Wu, J.-B.; et al. Myeloid derived suppressor cells contribute to the malignant progression of oral squamous cell carcinoma. PLoS ONE 2020, 15, e0229089.
  48. Youn, J.I.; Nagaraj, S.; Collazo, M.; Gabrilovich, D.I. Subsets of Myeloid-Derived Suppressor Cells in Tumor-Bearing Mice. J. Immunol. 2008, 181, 5791–5802.
  49. Bruno, A.; Mortara, L.; Baci, D.; Noonan, D.M.; Albini, A. Myeloid Derived Suppressor Cells Interactions With Natural Killer Cells and Pro-angiogenic Activities: Roles in Tumor Progression. Front. Immunol. 2019, 10, 771.
  50. Vasquez-Dunddel, D.; Pan, F.; Zeng, Q.; Gorbounov, M.; Albesiano, E.; Fu, J.; Blosser, R.L.; Tam, A.J.; Bruno, T.; Zhang, H.; et al. STAT3 regulates arginase-I in myeloid-derived suppressor cells from cancer patients. J. Clin. Investig. 2013, 123, 1580–1589.
  51. Bottomley, M.J.; Thomson, J.; Harwood, C.; Leigh, I. The Role of the Immune System in Cutaneous Squamous Cell Carcinoma. Int. J. Mol. Sci. 2019, 20, 2009.
  52. Seddon, A.; Hock, B.; Miller, A.; Frei, L.; Pearson, J.; McKenzie, J.; Simcock, J.; Currie, M. Cutaneous squamous cell carcinomas with markers of increased metastatic risk are associated with elevated numbers of neutrophils and/or granulocytic myeloid derived suppressor cells. J. Dermatol. Sci. 2016, 83, 124–130.
  53. Du, R.; Lu, K.V.; Petritsch, C.; Liu, P.; Ganss, R.; Passegué, E.; Song, H.; VandenBerg, S.; Johnson, R.S.; Werb, Z.; et al. HIF1α Induces the Recruitment of Bone Marrow-Derived Vascular Modulatory Cells to Regulate Tumor Angiogenesis and Invasion. Cancer Cell 2008, 13, 206–220.
  54. Komi, D.E.A.; Redegeld, F.A. Role of Mast Cells in Shaping the Tumor Microenvironment. Clin. Rev. Allergy. Immunol. 2020, 58, 313–325.
  55. Lätti, S.; Leskinen, M.; Shiota, N.; Wang, Y.; Kovanen, P.T.; Lindstedt, K.A. Mast cell-mediated apoptosis of endothelial cells in vitro: A paracrine mechanism involving TNF-α-mediated down-regulation of bcl-2 expression. J. Cell Physiol. 2003, 195, 130–138.
  56. Gudiseva, S.; Santosh, A.B.R.; Chitturi, R.; Anumula, V.; Poosarla, C.; Baddam, V.R.R. The role of mast cells in oral squamous cell carcinoma. Contemp. Oncol. 2017, 1, 21–29.
  57. Shrestha, A.; Keshwar, S.; Raut, T. Evaluation of Mast Cells in Oral Potentially Malignant Disorders and Oral Squamous Cell Carcinoma. Int. J. Dent. 2021, 2021, 5609563.
  58. Koyfman, S.A.; Ismaila, N.; Crook, D.; D’Cruz, A.; Rodriguez, C.P.; Sher, D.J.; Silbermins, D.; Sturgis, E.M.; Tsue, T.T.; Weiss, J.; et al. Management of the Neck in Squamous Cell Carcinoma of the Oral Cavity and Oropharynx: ASCO Clinical Practice Guideline. J. Clin. Oncol. 2019, 37, 1753–1774.
  59. Cramer, J.D.; Burtness, B.; Le, Q.T.; Ferris, R.L. The changing therapeutic landscape of head and neck cancer. Nat. Rev. Clin. Oncol. 2019, 16, 669–683.
  60. Shelton, M.E.; Adamson, A.S. Review and Update on Evidence-Based Surgical Treatment Recommendations for Nonmelanoma Skin Cancer. Dermatol. Clin. 2019, 37, 425–433.
  61. Yegodayev, K.M.; Novoplansky, O.; Golden, A.; Prasad, M.; Levin, L.; Jagadeeshan, S.; Zorea, J.; Dimitstein, O.; Joshua, B.-Z.; Cohen, L.; et al. TGF-Beta-Activated Cancer-Associated Fibroblasts Limit Cetuximab Efficacy in Preclinical Models of Head and Neck Cancer. Cancers 2020, 12, 339.
  62. Johansson, A.-C.; Ansell, A.; Jerhammar, F.; Lindh, M.B.; Grénman, R.; Munck-Wikland, E.; Östman, A.; Roberg, K. Cancer-Associated Fibroblasts Induce Matrix Metalloproteinase–Mediated Cetuximab Resistance in Head and Neck Squamous Cell Carcinoma Cells. Mol. Cancer Res. 2012, 10, 1158–1168.
  63. Stylianou, A.; Gkretsi, V.; Stylianopoulos, T. Transforming growth factor-β modulates pancreatic cancer associated fibroblasts cell shape, stiffness and invasion. Biochim. Biophys. Acta Gen. Subj. 2018, 1862, 1537–1546.
  64. Quan, J.; Elhousiny, M.; Johnson, N.W.; Gao, J. Transforming growth factor-β1 treatment of oral cancer induces epithelial-mesenchymal transition and promotes bone invasion via enhanced activity of osteoclasts. Clin. Exp. Metastasis 2013, 30, 659–670.
  65. Lebrun, J.J. The Dual Role of TGFβ in Human Cancer: From Tumor Suppression to Cancer Metastasis. ISRN Mol. Biol. 2012, 2012, 381428.
  66. Ludwig, N.; Wieteska, Ł.; Hinck, C.S.; Yerneni, S.S.; Azambuja, J.H.; Bauer, R.J.; Reichert, T.E.; Hinck, A.P.; Whiteside, T.L. Novel TGFβ Inhibitors Ameliorate Oral Squamous Cell Carcinoma Progression and Improve the Antitumor Immune Response of Anti–PD-L1 Immunotherapy. Mol. Cancer Ther. 2021, 20, 1102–1111.
  67. Mao, X.; Xu, J.; Wang, W.; Liang, C.; Hua, J.; Liu, J.; Zhang, B.; Meng, Q.; Yu, X.; Shi, S. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: New findings and future perspectives. Mol. Cancer 2021, 20, 1–30.
  68. Hanley, C.J.; Mellone, M.; Ford, K.; Thirdborough, S.M.; Mellows, T.; Frampton, S.J.; Smith, D.M.; Harden, E.; Szyndralewiez, C.; Bullock, M.; et al. Targeting the Myofibroblastic Cancer-Associated Fibroblast Phenotype Through Inhibition of NOX4. J. Natl. Cancer Inst. 2018, 110, 109–120.
  69. Feig, C.; Jones, J.O.; Kraman, M.; Wells, R.J.; Deonarine, A.; Chan, D.S.; Connell, C.M.; Roberts, E.W.; Zhao, Q.; Caballero, O.L.; et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 20212–20217.
  70. Von Hoff, D.D.; Ervin, T.; Arena, F.P.; Chiorean, E.G.; Infante, J.; Moore, M.; Seay, T.; Tjulandin, S.A.; Ma, W.W.; Saleh, M.N.; et al. Increased Survival in Pancreatic Cancer with nab-Paclitaxel plus Gemcitabine. N. Engl. J. Med. 2013, 369, 1691–1703.
  71. Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and Its Ligands in Tolerance and Immunity. Annu. Rev. Immunol. 2008, 26, 677–704.
  72. Flies, D.B.; Sandler, B.J.; Sznol, M.; Chen, L. Blockade of the B7-H1/PD-1 pathway for cancer immunotherapy. Yale J. Biol. Med. 2011, 84, 409–421.
  73. Fisher, J.; Zeitouni, N.; Fan, W.; Samie, F.H. Immune checkpoint inhibitor therapy in solid organ transplant recipients: A patient-centered systematic review. J. Am. Acad. Dermatol. 2020, 82, 1490–1500.
  74. Long, J.; Qi, Z.; Rongxin, Z. PD-1/PD-L1 pathway blockade works as an effective and practical therapy for cancer immunotherapy. Cancer Biol. Med. 2018, 15, 116.
  75. Ferrarotto, R.; Bell, D.; Rubin, M.L.; Hutcheson, K.A.; Johnson, J.M.; Goepfert, R.P.; Phan, J.; Elamin, Y.Y.; Torman, D.K.; Warneke, C.L.; et al. Impact of Neoadjuvant Durvalumab with or without Tremelimumab on CD8+ Tumor Lymphocyte Density, Safety, and Efficacy in Patients with Ovropharynx Cancer: CIAO Trial Results. Clin. Cancer Res. 2020, 26, 3211–3219.
  76. Duhen, R.; Ballesteros-Merino, C.; Frye, A.K.; Tran, E.; Rajamanickam, V.; Chang, S.-C.; Koguchi, Y.; Bifulco, C.B.; Bernard, B.; Leidner, R.S.; et al. Neoadjuvant anti-OX40 (MEDI6469) therapy in patients with head and neck squamous cell carcinoma activates and expands antigen-specific tumor-infiltrating T cells. Nat. Commun. 2021, 12, 1–14.
More
Information
Subjects: Cell Biology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , ,
View Times: 198
Revisions: 2 times (View History)
Update Date: 03 Jul 2023
1000/1000
Video Production Service