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 + 1530 word(s) 1530 2021-07-02 11:05:02

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.
Pertinhez, T. Oral Squamous Cell Carcinoma Biomarkers. Encyclopedia. Available online: https://encyclopedia.pub/entry/11737 (accessed on 29 March 2024).
Pertinhez T. Oral Squamous Cell Carcinoma Biomarkers. Encyclopedia. Available at: https://encyclopedia.pub/entry/11737. Accessed March 29, 2024.
Pertinhez, Thelma. "Oral Squamous Cell Carcinoma Biomarkers" Encyclopedia, https://encyclopedia.pub/entry/11737 (accessed March 29, 2024).
Pertinhez, T. (2021, July 06). Oral Squamous Cell Carcinoma Biomarkers. In Encyclopedia. https://encyclopedia.pub/entry/11737
Pertinhez, Thelma. "Oral Squamous Cell Carcinoma Biomarkers." Encyclopedia. Web. 06 July, 2021.
Oral Squamous Cell Carcinoma Biomarkers
Edit

Oral cancers are among the most common malignant tumors worldwide. More than 90% of all oral malignancies are oral squamous cell carcinomas (OSCC). While in Western countries, OSCC accounts for about 4% of all cancers; in India and Southeast Asia, it reaches up to 40%. The interaction between oral dysplastic/neoplastic cells and saliva makes this fluid a source of biomarkers, such as cytokines.

oral cancer oral squamous cell carcinoma biomarkers cytokines oral potentially malignant disorders

1. Introduction

The clinical course of OSCC is strictly related to the time of diagnosis [1]. According to the Surveillance, Epidemiology, and End Results (SEER) Program (based on the U. S. population), when OSCC is confined to the primary site, the 5-years relative survival rate is about 85.1%, decreasing to 66.8% and to 40.1% for tumors spread to regional lymph nodes and to distant sites, respectively [2].
Classical risk factors for OSCC development in Western countries are smoking and alcohol abuse. The role of some human papillomaviruses (HPV) infection has been clearly established for carcinomas of the oropharynx, but doubts still exist on the oncogenic potential of such viruses in the rest of the oral cavity [3].
As for the rest of the head and neck tumors, the pathogenesis of OSCC is still a matter of controversy. Most authors agree on a multi-step process in which the accumulation of genetic and epigenetic changes affect protein expression, thus altering a variety of signaling pathways [4]. Gene alterations may range from the gain or loss of single nucleotides, up to partial or complete deletion of chromosomes. The loss of genes has been described in significant conjunction with inactivating mutations of tumor suppressor genes [5].
In a recent review on the molecular landscape in head and neck cancer, Leemans et al. identified five cellular processes that may be dysregulated in OSCC pathogenesis: cell cycle, growth signal, survival, WNT signaling and epigenetic regulation [6].
Diagnosis of OSCC is based on clinical inspection followed by biopsy and histopathological evaluation of suspicious tissues. Vital staining (e.g., toluidine blue) and auto-fluorescence imaging may highlight tissues undergoing rapid cell division and represent complementary diagnostic aids, as they can differentiate normal from dysplastic or cancerous tissues and direct the biopsy to the target site [7]. Radiographic imaging is used to investigate the involvement of the surrounding tissue and structures, such as muscles, bone, and lymph nodes; in particular, computed tomography (CT) and magnetic resonance imaging (MRI) are essential for bone and neck nodes evaluation [8].
Some OSCCs are preceded by oral potentially malignant disorders (OPMDs), including leukoplakia, erythroplakia, lichen planus and oral submucous fibrosis [9][10], at the histological level.
Visual examination alone, however, may lead to ignoring subtle lesions and failing to differentiate malignant from benign oral conditions. As a result, OSCC is diagnosed at an already advanced stage, adversely affecting the survival rate. Many studies are, therefore, focused on the identification of easily accessible early diagnostic biomarkers.

2. Salivary Cytokines as Biomarkers for Oral Squamous Cell Carcinoma

It is well accepted that inflammation and cell-mediated immunity are active players in the control of oral carcinogenesis progression [11]. Cells’ evasion from immune surveillance has been described as a primary step in oncogenesis [12]. Pro-inflammatory cytokines (IL-6, IL-8, TNF-α, soluble interleukin-2 receptor (sIL-2R)) are key-molecules involved in the crosstalk between stromal and cancer cells, their expression being, to some extent, associated with tumor growth promotion or inhibition [13].
Serum levels of pro-inflammatory growth factors and cytokines in patients with OSCC or other oral potentially malignant disorders are still poorly investigated. Recent results would demonstrate that the serum level of IL-6, IL-8 and sIL-2R is significantly higher in patients with OSCC compared to healthy controls and to patients with OPMDs [11]. Thus, it appears feasible to accept that some salivary cytokines might be considered reliable biomarkers for OSCC diagnosis.
All the selected studies, investigating the role of specific salivary cytokines in the diagnosis of OSCC, identify the institutional care facility that enrolled the OSCC patients and, in most cases, mention the approval of an institutional ethical committee, thus presenting appropriate information about ethical protection.
As for saliva collection and processing, we ascertained, overall, a reasonable procedural consistency. In fact, most of the studies (1) analyze unstimulated whole saliva, obtained via passive drool; (2) include a preliminary step of centrifugation at 4 °C to separate cells and debris; (3) freeze the saliva supernatant, usually at −80 °C, until the analysis. Nevertheless, only in a few studies did the salivary protein content, including cytokine molecules, appear to be protected by the addition of protease inhibitors [14][15][16][17][18].
In all studies, an enzyme-linked immunosorbent assay (ELISA), based on antibodies against specific cytokines and a colorimetric revelation system, turned out to be the method of choice for cytokine quantitation. The method allows the accurate detection of the antigen, but measures only one cytokine in each sample, with consequent sample waste and high cost when the purpose is to measure multiple cytokines. In four included studies that investigated the potential variation in the concentration of several cytokines, a multiplex bead-based immunoassay was preferred [19][14][20][21]. Besides the high throughput multiplex analysis, the major advantage is its broader dynamic range of cytokine concentration measurable as compared to the ELISA test.
Since the possible presence of dental and periodontal infection may influence salivary cytokine concentrations [22], many of the selected studies evaluate the periodontal status of the study participants. While most of them simply exclude subjects with periodontitis from control and/or patient groups or include control subjects with periodontal status matched with the patient group, the studies of Brailo and co-workers [23] and Sato and co-workers [24][25] take into account the periodontal status of all the study participants. Thus, by using the community periodontal index, as described by the World Health Organization (WHO), they can demonstrate that the salivary concentrations of IL-6 and TNF-α are not affected by periodontal health.
In addition, except for a few cases [15][26][16][27], the authors agree on excluding subjects with any relevant comorbidity, for example, chronic or acute illnesses, autoimmune diseases, chronic inflammatory diseases, pathological dry mouth syndrome, or the inability to collect a sufficient saliva sample. We believe that the application of this exclusion criterion constitutes good practice, consistent with the diagnostic purposes of this topic area.
The interfering behavioral habits considered are mainly alcohol consumption and tobacco chewing or smoking. The use of these substances is frequent in OSCC patients, although preferential habits appear to be associated with the specific geographical origin of the patients. In only nine studies [19][14][23][28][27][29][30][31][25], the authors tested whether the salivary cytokine level was affected by social habits. As far as IL-1α, IL-1β, IL-6, TNF-α and IL-8 are individually concerned, their salivary concentrations did not appear to be affected by (or correlated to) specific habits.
In thirteen articles, salivary cytokine level is also assessed in the OPMD patient group and compared with OSCC and control groups. OPMD patients suffer prevalently from leucoplakia, but also from oral sub mucous fibrosis or oral lichen planus, while erythroplakia did not appear in any study, possibly because of its rare occurrence. In twelve studies, the salivary levels of IL-6, IL-8, and TNF-α of OPMD patients show a statistically significant higher concentration when compared with the control, but lower when compared to OSCC patients [19][32][29][33][34][30][35][36]. Likewise, comparable findings are obtained with IL-6 and TNF-α salivary concentrations [23][28] when compared to control subjects. In these cases, the salivary cytokines concentration allows us to differentiate between OPMD, OSCC and healthy subjects, thus reflecting a multi-diagnostic potential and corroborating the use of selected cytokines as biomarkers.
Among the studies including OPMD subjects, six articles investigated the diagnostic utility of the salivary cytokine level by ROC curve analysis and compared the ability of selected cytokines (IL-6, IL-8, TNF-α) to differentiate between patients with OSCC and OPMD [19][17][29][33][34][30]. The significant area under the curve (AUC) ranges from 0.70 to 0.99, reflecting an effective power of discrimination.
As observed in some studies [19][17][31], the combination of different salivary biomarkers is of great value for OSCC detection. A predictive model based on six cytokines and used for distinguishing OSCC from control subjects yielded a sensible increase in AUC as compared to individual cytokine analysis [19]. Additionally, the combination of cytokine proteomic and transcriptomic biomarkers generated an increased discriminatory effect between OSSC and control subjects [17][31], although further inclusion of a risk factor exposure (areca nut, smoking) provided the best panel of variables useful for OSCC detection [17].
Unfortunately, in OSCC patients, treatment outcome and prognosis are seriously affected not only by late diagnosis but also by frequent loco regional recurrences. Sato and co-workers [24][25] perceived the potential of IL-6 in terms of recurrence prediction and demonstrated that the sequential analysis of salivary cytokines post-treatment could be a useful marker for the diagnosis of early and late loco regional recurrence, a common cause of mortality in patients with OSCC. To give one example, they measured an increased postoperative IL-6 concentration in subjects with early recurrence as compared to subjects without recurrence. Apparently, if validated with a larger number of patients enrolled prospectively, the analysis of the dynamic behavior of that cytokine level in the post-treatment phase might be an effective tool for the early identification/prediction of recurrence.
According to these observations, we also expect an appropriate combination of biomarkers to be validated in longitudinal studies, and their reliability to be confirmed with respect to potential confounding factors such as behavioral habits and periodontitis.

References

  1. Speight, P.M. Update on Oral Epithelial Dysplasia and Progression to Cancer. Head Neck Pathol. 2007, 1, 61–66.
  2. Cancer Stat Facts: Oral Cavity and Pharynx Cancer. Available online: (accessed on 25 February 2021).
  3. Castellsague, X.; Alemany, L.; Quer, M.; Halec, G.; Quirós, B.; Tous, S.; Clavero, O.; Alos, L.; Biegner, T.; Szafarowski, T.; et al. HPV involvement in head and neck cancers: Comprehensive assessment of biomarkers in 3680 patients. J. Natl. Cancer Inst. 2016, 108, djv403.
  4. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674.
  5. Leemans, C.R.; Braakhuis, B.J.M.; Brakenhoff, R.H. The molecular biology of head and neck cancer. Nat. Rev. Cancer 2011, 11, 9–22.
  6. Leemans, C.R.; Snijders, P.J.F.; Brakenhoff, R.H. The molecular landscape of head and neck cancer. Nat. Rev. Cancer 2018, 18, 269–282.
  7. Giovannacci, I.; Vescovi, P.; Manfredi, M.; Meleti, M. Non-invasive visual tools for diagnosis of oral cancer and dysplasia: A systematic review. Med. Oral Patol. Oral Cir. Bucal 2016, 21, e305–e315.
  8. Montero, P.H.; Patel, S.G. Cancer of the oral cavity. Surg. Oncol. Clin. N. Am. 2015, 24, 491–508.
  9. Van der Waal, I. Potentially malignant disorders of the oral and oropharyngeal mucosa; terminology, classification and present concepts of management. Oral Oncol. 2009, 45, 317–323.
  10. Carvalho, M.F.M.S.d.; Cavalieri, D.; Do Nascimento, S.; Lourenço, T.G.B.; Ramos, D.V.R.; Pasqualin, D.C.; Martins, L.A.L.; Rocha, F.A.; Heller, D.; Marti, L. Cytokines levels and salivary microbiome play a potential role in oral lichen planus diagnosis. Sci. Rep. 2019, 9, 18137.
  11. Schiegnitz, E.; Kämmerer, P.W.; Schön, H.; Blatt, S.; Berres, M.; Sagheb, K.; Al-Nawas, B. Proinflammatory cytokines as serum biomarker in oral carcinoma-A prospective multi-biomarker approach. J. Oral Pathol. Med. 2018, 47, 268–274.
  12. Arduino, P.G.; Menegatti, E.; Cappello, N.; Martina, E.; Gardino, N.; Tanteri, C.; Cavallo, F.; Scully, C.; Broccoletti, R. Possible role for interleukins as biomarkers for mortality and recurrence in oral cancer. Int. J. Biol. Markers 2015, 30, e262–e266.
  13. Nibali, L.; Fedele, S.; D’Aiuto, F.; Donos, N. Interleukin-6 in oral diseases: A review. Oral Dis. 2012, 18, 236–243.
  14. Aziz, S.; Ahmed, S.S.; Ali, A.; Khan, F.A.; Zulfiqar, G.; Iqbal, J.; Khan, A.A.; Shoaib, M. Salivary immunosuppressive cytokines IL-10 and IL-13 are significantly elevated in oral squamous cell carcinoma patients. Cancer Investig. 2015, 33, 318–328.
  15. Katakura, A.; Kamiyama, I.; Takano, N.; Shibahara, T.; Muramatsu, T.; Ishihara, K.; Takagi, R.; Shouno, T. Comparison of salivary cytokine levels in oral cancer patients and healthy subjects. Bull. Tokyo Dent. Coll. 2007, 48, 199–203.
  16. Korostoff, A.; Reder, L.; Masood, R.; Sinha, U.K. The role of salivary cytokine biomarkers in tongue cancer invasion and mortality. Oral Oncol. 2011, 47, 282–287.
  17. Gleber-Netto, F.O.; Yakob, M.; Li, F.; Feng, Z.; Dai, J.; Kao, H.K.; Chang, Y.L.; Chang, K.P.; Wong, D.T. Salivary Biomarkers for Detection of Oral Squamous Cell Carcinoma in a Taiwanese Population. Clin. Cancer Res. 2016, 22, 3340–3347.
  18. Cheng, Y.S.; Jordan, L.; Gorugantula, L.M.; Schneiderman, E.; Chen, H.S.; Rees, T. Salivary interleukin-6 and -8 in patients with oral cancer and patients with chronic oral inflammatory diseases. J. Periodontol. 2014, 85, 956–965.
  19. Dikova, V.; Jantus-Lewintre, E.; Bagan, J. Potential non-invasive biomarkers for early diagnosis of oral squamous cell carcinoma. J. Clin. Med. 2021, 10, 1658.
  20. Lee, L.T.; Wong, Y.K.; Hsiao, H.Y.; Wang, Y.W.; Chan, M.Y.; Chang, K.W. Evaluation of saliva and plasma cytokine biomarkers in patients with oral squamous cell carcinoma. Int. J. Oral Maxillofac. Surg. 2018, 47, 699–707.
  21. Val, M.; Sidoti Pinto, G.A.; Manini, L.; Gandolfo, S.; Pentenero, M. Variations of salivary concentration of cytokines and chemokines in presence of oral squamous cell carcinoma. A case-crossover longitudinal prospective study. Cytokine 2019, 120, 62–65.
  22. Melguizo-Rodríguez, L.; Costela-Ruiz, V.J.; Manzano-Moreno, F.J.; Ruiz, C.; Illescas-Montes, R. Salivary biomarkers and their application in the diagnosis and monitoring of the most common oral pathologies. Int. J. Mol. Sci. 2020, 21, 5173.
  23. Brailo, V.; Vucicevic-Boras, V.; Lukac, J.; Biocina-Lukenda, D.; Zilic-Alajbeg, I.; Milenovic, A.; Balija, M. Salivary and serum interleukin 1 beta, interleukin 6 and tumor necrosis factor alpha in patients with leukoplakia and oral cancer. Med. Oral Patol. Oral Cir. Bucal 2012, 17, e10–e15.
  24. Sato, J.; Ohuchi, M.; Abe, K.; Satoh, T.; Abe, T.; Yamazaki, Y.; Satoh, A.; Notani, K.I.; Kitagawa, Y. Correlation between salivary interleukin-6 levels and early locoregional recurrence in patients with oral squamous cell carcinoma: Preliminary study. Head Neck 2013, 35, 889–894.
  25. Sato, J.; Ohuchi, M.; Wada, M.; Ohga, N.; Asaka, T.; Yoshikawa, K.; Miyakoshi, M.; Hata, H.; Satoh, A.; Kitagawa, Y. Differences in sequential posttreatment salivary IL-6 levels between patients with and patients without locoregional recurrences of oral squamous cell carcinoma: Part III of a cohort study. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2015, 120, 751–760.
  26. Rhodus, N.L.; Ho, V.; Miller, C.S.; Myers, S.; Ondrey, F. NF-κB dependent cytokine levels in saliva of patients with oral preneoplastic lesions and oral squamous cell carcinoma. Cancer Detect. Prev. 2005, 29, 42–45.
  27. 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.
  28. Juretić, M.; Cerović, R.; Belušić-Gobić, M.; Brekalo Pršo, I.; Kqiku, L.; Špalj, S.; Pezelj-Ribarić, S. Salivary levels of TNF-α and IL-6 in patients with oral premalignant and malignant lesions. Folia Biol. 2013, 59, 99–102.
  29. Dineshkumar, T.; Ashwini, B.K.; Rameshkumar, A.; Rajashree, P.; Ramya, R.; Rajkumar, K. Salivary and serum interleukin-6 levels in oral premalignant disorders and squamous cell carcinoma: Diagnostic value and clinicopathologic correlations. Asian Pac. J. Cancer Prev. 2016, 17, 4899–4906.
  30. Krishnan, R.; Thayalan, D.K.; Padmanaban, R.; Ramadas, R.; Annasamy, R.K.; Anandan, N. Association of serum and salivary tumor necrosis factor-α with histological grading in oral cancer and its role in differentiating premalignant and malignant oral disease. Asian Pac. J. Cancer Prev. 2014, 15, 7141–7148.
  31. Brinkmann, O.; Kastratovic, D.A.; Dimitrijevic, M.V.; Konstantinovic, V.S.; Jelovac, D.B.; Antic, J.; Nesic, V.S.; Markovic, S.Z.; Martinovic, Z.R.; Akin, D.; et al. Oral squamous cell carcinoma detection by salivary biomarkers in a serbian population. Oral Oncol. 2011, 47, 51–55.
  32. Punyani, S.R.; Sathawane, R.S. Salivary level of interleukin-8 in oral precancer and oral squamous cell carcinoma. Clin. Oral Investig. 2013, 17, 517–524.
  33. Rajkumar, K.; Nandhini, G.; Ramya, R.; Rajashree, P.; Kumar, A.R.; Anandan, S.N. 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, 309–319.
  34. Ameena, M.; Rathy, R. Evaluation of tumor necrosis factor alpha in the saliva of oral cancer, leukoplakia, and healthy controls—A comparative study. J. Int. Oral Health 2019, 11, 92–99.
  35. Deepthi, G.; Nandan, S.R.K.; Kulkarni, P.G. Salivary tumor necrosis factor-α as a biomarker in oral leukoplakia and oral squamous cell carcinoma. Asian Pac. J. Cancer Prev. 2019, 20, 2087–2093.
  36. Selvam, N.P.; Sadaksharam, J. Salivary interleukin-6 in the detection of oral cancer and precancer. Asia Pac. J. Clin. Oncol. 2015, 11, 236–241.
More
Information
Subjects: Oncology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 323
Revision: 1 time (View History)
Update Date: 29 Mar 2022
1000/1000