Next Article in Journal
Determination of Micromovements in Removable Prosthesis during Mastication: A Pilot Study with 3D Electromagnetic Articulography
Previous Article in Journal
Improving Generalizability of PET DL Algorithms: List-Mode Reconstructions Improve DOTATATE PET Hepatic Lesion Detection Performance
Previous Article in Special Issue
Gingival Fibroblasts Are Sensitive to Oral Cell Lysates Indicated by Their IL11 Expression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

State of the Art in the Diagnosis and Assessment of Oral Malignant and Potentially Malignant Disorders: Present Insights and Future Outlook—An Overview

by
Dardo Menditti
1,
Mario Santagata
1,
David Guida
1,*,
Roberta Magliulo
1,
Giovanni Maria D’Antonio
2,
Samuel Staglianò
1 and
Ciro Emiliano Boschetti
1
1
Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
2
Division of Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90128 Palermo, Italy
*
Author to whom correspondence should be addressed.
Bioengineering 2024, 11(3), 228; https://doi.org/10.3390/bioengineering11030228
Submission received: 25 January 2024 / Revised: 23 February 2024 / Accepted: 26 February 2024 / Published: 28 February 2024

Abstract

:
Oral Potentially Malignant Disorder (OPMD) is a significant concern for clinicians due to the risk of malignant transformation. Oral Squamous Cell Carcinoma (OSCC) is a common type of cancer with a low survival rate, causing over 200,000 new cases globally each year. Despite advancements in diagnosis and treatment, the five-year survival rate for OSCC patients remains under 50%. Early diagnosis can greatly improve the chances of survival. Therefore, understanding the development and transformation of OSCC and developing new diagnostic methods is crucial. The field of oral medicine has been advanced by technological and molecular innovations, leading to the integration of new medical technologies into dental practice. This study aims to outline the potential role of non-invasive imaging techniques and molecular signatures for the early detection of Oral Malignant and Potentially Malignant Disorders.

Graphical Abstract

1. Introduction

The oral cavity constitutes a complex biological system, both from a histological point of view and from a functional–biomechanical point of view [1]. The oral system is an organ able to perform certain functions such as chewing, swallowing, phonation, breathing, facial expressions; these functions are made possible mainly by a pair of joints: the tempo-mandibular joints and naturally by the related muscles [2]. These anatomical structures can be affected by various kinds of disorders, including genetic, congenital, inflammatory and traumatic events [3,4,5]. Moreover, the niche of the oral antrum represents a favorable domain for the invasion of external agents from the external environment [6]. The coexistence between external microbial agents and the host is guaranteed by a constant balance of the oral habitat, nowadays also known as oral microbiome [7]. An alteration of the microbial flora, due to various intrinsic and extrinsic factors, may determine the phenomenon of dysbiosis [8,9,10]. The mechanisms involved in oral dysbiosis have shown to lead to most of the dental and periodontal tissues of the oral cavity, which recognize microbiological agents—viral, bacterial and fungal—as the main nosological factors [11,12,13]. Oral mucosal soft tissues, instead, are mainly affected by mycotic superinfections, with a greater prevalence for frail patients, immunosuppressed subjects and prosthesis wearers [14,15,16]. Furthermore, a large group of viral agents is involved in the etiological mechanism of a wide spectrum of oral mucosa diseases, ranging from acute to chronic disorders [17,18,19,20,21].
Pathological cytoarchitectural changes in the tissue layers of the oral mucosa are seen in a wide range of disorders, ranging from benign [22] to potentially malignant and properly malignant conditions [23,24,25,26], up to the involvement of rare syndromic manifestations involving multiple areas of the body [27,28]. Benign disorders affecting the oral mucosa represent a large group of pathological manifestations, especially autoimmune, viral and neuropsychiatric diseases [29,30,31,32,33,34]. However, the oral cavity is linked to the possible occurrence of malignant lesions, and 90% of them are represented by Oral Squamous Cell Carcinoma (OSCC), the 16th most common worldwide cancer [35].
Furthermore, there are several pathological entities involving oral mucosa that may show an increased risk of malignant transformation, which are also known as Oral Potentially Malignant Disorders (OPMD), and they are represented by manifestations such as Oral Leukoplakia, Oral Eritroplakia and Proliferative Verrucous Leukoplakia [36,37].
The most demanding challenge for clinicians involves the early detection and treatment of the OPMD manifestations, in order to prevent malignancy transformation [38,39]. The strongest efforts, to date, are focused on all the possible strategies able to achieve a time-saving diagnosis, ranging from biomarkers to imaging techniques [40,41,42,43]. The purpose of this overview is to describe a broad picture of oral cancerous and precancerous entities, and to depict the current approaches and the future perspectives to achieve a quick diagnosis. The aim of this study was to examine the available knowledge in the literature on the potential use of technology and biomarkers or accurately identifying and categorizing malignant or potentially malignant oral lesions at an early stage.

2. Oral Potentially Malignant Disorders

Oral Potentially Malignant Disorders (OPDMs) represent a wide scenario of pathological manifestations, which may not include the onset of oral cavity malignant neoplastic lesions; this nomenclature has been asserted because not all disorders described under this term may transform into cancer [38].

2.1. Oral Leukoplakia, Proliferative Verrucous Leukoplakia and Erythroplakia

One of the most frequently encountered lesions classified as an OPMD in the oral cavity is Oral Leukoplakia (OL) [44] (Figure 1). A smoking habit has shown to be recurrently associated with leukoplakia in more than 80% of cases; OL represents a non-scraping white lesion and it is defined by the World Health Organization (WHO) as a “white plaque of questionable risk having excluded (other) known diseases or disorders that carry no increased risk for cancer” [45]. From a clinical point of view, there are different varieties for this entity: homogeneous leukoplakia presents as a flat and uniform white plaque with well-defined margins (at least one). Non-homogeneous leukoplakia involves spots of erythema surrounded by areas of nodularity and verrucousity [46]. Oral Proliferative Verrucous Leukoplakia (PVL) is a distinguishing subset of non-homogenous leukoplakia more common in females and less associated with a smoking habit. PVLs may involve a single extensive area but are often multifocal and frequently occur on the gingiva, buccal mucosa, and tongue in both bordering and non-contiguous sites of the oral cavity [47,48]. The mechanisms underlying the transformation from hyperkeratosis or hyperplasia to various degrees of dysplasia and eventually carcinoma in situ and/or OSCC are still subject to ongoing debate. As a result, PVLs and leukoplakias necessitate multiple recurring biopsies at different sites. This ongoing need for biopsies is essential to continuously investigate the histotype and detect different grades of dysplasia or malignant transformation to OSCC [49,50,51].
Furthermore, the lesion called “Oral erythroplakia” (OE) has been defined by the WHO as “a fiery red patch that cannot be characterized clinically or pathologically as any other definable disease” and it has long been deemed as the oral mucosal lesion with the greatest potential for malignant transformation in the oral cavity [52]. OE does not occur on any traumatic, vascular, or inflammatory basis, but appears with comparable frequency in both sexes in middle-aged and elderly patients. A diagnostic biopsy is required to detect this potentially malignant lesion, more accurately analyze this histological subset and even prevent the risk of transformation [53].

2.2. Oral Lichen Planus

To date, experts adhere to the distinction between precancerous lesions and precancerous conditions for the main reason that the onset of a malignancy in the mouth of a patient known to have a precancerous lesion would correspond with the site of precancer, while in precancerous conditions, malignancy may originate in any anatomical site of the oral cavity [45].
One of the most recognized oral potentially malignant conditions is represented by Oral Lichen Planus (OLP), a chronic immunological inflammatory disease that usually affects middle-aged patients, particularly women [54]. This disorder may involve the skin and mucosal membranes; moreover, OLP is usually linked to various patterns and the subtypes can be categorized as follows: reticular form, which is the most common subtype, distinguished by Wickham striae and hyperkeratotic plaques or papules; atrophic–erosive form, with areas of ulceration usually associated with keratotic white striae, and also ulcerations [55]. The plaque form of Oral Lichen Planus can mimic Oral Leukoplakias, which highlights the importance of biopsy [56]. OLP generally involves the buccal mucosa, followed by the gingiva and tongue, usually with multiple, bilateral lesions. Erosive and atrophic OLP can provoke intense discomfort and inhibit speech and swallowing [55]. It is still debated whether Oral Lichen Planus can progress into Oral Squamous Cell Carcinoma: data concerning the transformation rates range between 0% and 12.5%, and no clinical or histopathological suggestion can support the prognosis of potential malignant transformation [57,58]. The erosive form bears the highest risk rate of malignant transformation, followed by atrophic OLP, while the reticular white type has the lowest risk rate [59,60].

2.3. Submucosal Fibrosis

Another characteristic oral potentially malignant condition is represented by Submucosal Fibrosis [61]. This condition presents as a chronic fibrotic lesion of the oral mucosa, possibly as the expression of a large wound overhealing, based on a chronic mechanical or chemical insult [62]. With a predominance in males, the most commonly affected sites are the buccal mucosa, the lingual margins, the lip, the palate, and the gums [63]. Clinically, Submucosal Fibrosis is characterized by a loss of elasticity in the affected tissues, with palpable fibrous bands that involve the mobility of the tongue and restrict mouth opening [64].
To date, Submucosal Fibrosis has been widely recognized to carry a malignant potential; an estimated transformation rate of 8% has been reported [65].

2.4. Actinic Keratosis

Actinic Keratosis (AK) is a common precancerous skin lesion that may also affect oral and peri-oral tissues, caused by long-term sun exposure. It appears as a rough, scaly patch on sun-exposed areas of the skin, such as the face, neck, arms, and hands. If left untreated, AK can develop into squamous cell cancer. Early detection and treatment, such as cryotherapy or topical medications, can prevent the progression of AK to malignant transformation. Regular use of sun protection, such as sunscreen and protective clothing, can also reduce the risk of developing AK. It is important to seek the advice of a healthcare professional for proper diagnosis and management of actinic keratosis [66].

2.5. Graft versus Host Disease

According to the WHO Classification of Tumours, 4th Edition (2017) [67], Oral Graft versus Host Disease (GVHD) is considered as a potentially malignant disorder, and it describes a condition that can occur after a bone marrow or stem cell transplant, when the transplanted cells attack the recipient’s body. In Oral GVHD, the mouth, including the lips, gums, tongue, and inside of the cheeks, is affected [68]. Symptoms may include redness, swelling, sores, and painful or difficulty in swallowing. Oral GVHD can be a severe and life-threatening condition, and early detection and prompt treatment are important for the best outcome. Treatment may include topical or systemic medications, and in severe cases, additional interventions such as surgery may be necessary. Regular monitoring by a healthcare professional is important for the management of Oral GVHD. Nevertheless, no Oral Potentially Malignant Disorder is considered as a compulsory forerunner to cancer; likewise, most Oral Potentially Malignant Disorders do not lead to cancer [69]. Hence, the most challenging struggle involves determining lesions that display a higher risk of transformation from those carrying a lower risk. Dysplastic features are supposed to be the most valuable indicator of potential malignant progression [70,71].
Nonetheless, a lesion with no sign of dysplasia carries the potential to transform into cancer, and there are no specific clinical signs directly associated with dysplastic features; therefore, a biopsy is invariably mandatory to assess whether dysplasia is present [72].

3. Oral Malignancies

Oral malignant neoplasms belong to the variety of head and neck cancers (H&N cancers), which represent a wide group of epithelial malignant tumors involving the lining mucosa of the nasal cavity and paranasal sinuses, nasopharynx, hypopharynx, larynx and trachea, oropharynx, oral cavity and salivary glands [73]. Although there is a large group of mostly rare malignant neoplasms affecting the oromaxilofacial area [74,75,76], Oral Squamous Cell Carcinoma (OSCC) represents a malignant neoplasm involving the oral cavity, leading to more than 90% of malignant tumors of the head and neck area, ranking as the 16th most common worldwide cancer [77], and it stands as the most common malignancy in South-East Asia (India, Sri Lanka, Pakistan, Bangladesh, and Taiwan) and the Pacific regions (Papua New Guinea and Melanesia) due to the betel chewing habit [78] (Figure 2). Tobacco and alcohol consumption represent two of the major risk factors for the development of oral cancer [79]; nevertheless, many other elements have been reported to increase the risk of OSCC, such as micronutrient depletion, hormonal, protein and enzyme imbalances [80,81,82,83,84], poor oral hygiene, chronic traumatism, and viruses [85,86]. Poor oral hygiene has been always associated with the risk of the development of periodontal disease, so current investigations are focusing on the possible association between the periodontal status of health and oral cancer risk [87]. The potential role of human papillomavirus (HPV) infection and its relationship with cancerogenesis are the most explored but still debated topics among all human viruses. Human papillomavirus infection has a potential role in carcinogenesis. To date, more than 100 genotypes of HPV are known, divided into low risk and high risk. The viral proteins E6 and E7 play a key role in pathogenesis, particularly by inhibiting the oncosuppressors p53 and Rb, with overexpression of the p16 protein. Data in the literature show that HPV16 represents about 86% of HPV+ oropharyngeal squamous cell carcinomas (OPSCC) cases, while the association between OSCC and human papillomavirus infection is still debated [88,89,90,91]. From a purely clinical point of view, various forms have been reported, including the endophytic form, infiltrative exophytic form, vegetative or papillary, ulcerative form and mixed form [92]. The recurrence and prognosis of OSCC have been deeply investigated in different studies and a close relationship with histological malignancy grading and different clinical parameters has been found [35,93,94]. Besides the recurrence of OSCC, survival and mortality rely on multiple and additional biological, histological, macroscopic, and microscopic aspects that have been analyzed in order to determine the causalities, support early diagnosis and develop suitable remedies for the different characteristics and grades of Oral Squamous Cell Carcinomas [95,96]. Site, size and thickness—or the depth of invasion—of the cancer have been the most examined prognostic factors for achieving the current survival rates. The closer the tumor origin is to the inner sites of the mouth, the lower the survival rates [97]; furthermore, the survival rate decreases in relation to the advanced involvement of regional lymph nodes [98]. To date, approximately 60% of OSCC lesions are detected in locally advanced stages, when the 5-year survival rate is <50–60% [99]. In addition, conventional histopathological data seem to be unsatisfactory in accurately foretelling the clinical evolution of OSCC [100]; consequently, the need for a label of molecular markers that can predictively demarcate which cancer will display an aggressive behavior and poorer prognosis should be highlighted [101,102,103]. Treatment options include surgery, radiation therapy, chemotherapy, and targeted therapy [93]. The type and extent of treatment will depend on the size and location of the tumor and the overall health of the patient. It is important to work with a healthcare team to develop an individualized treatment plan. Regular dental check-ups and oral cavity screenings can help detect OSCC in its early stages and increase the chances of successful treatment [67].
The early diagnosis of the asymptomatic first stages of oral cancer is still the pivotal and most demanding step to achieving an adequate clinical outcome in most patients.

4. Future Perspectives

Early diagnosis of oral malignant and potentially malignant lesions is crucial for improving the chances of successful treatment. Prompt detection of oral cancer can be aided through routine dental exams and screenings for the disease [78]. During a screening, a dentist or physician will examine the mouth, neck, face, and lymph nodes for signs of abnormalities. They may also feel for lumps or thick patches in the tissues and check for any changes in the way the teeth fit together when the mouth is closed [73]. In some cases, clinicians should be assisted by new technological instruments, such as a bright light or magnifying glass, in order to take a closer look at the tissues. If an abnormality is detected, the individual may be referred for further testing, such as a biopsy, to determine if it is cancerous [104]. Within this context, AJCC/UICC suggest performing HPV tests using techniques such as DNA/RNA HIS (in situ hybridization), DNA-PCR, p16 immunostaining or the more recent circulating tumor tissue-modified viral (TTMV)-HPV DNA plasma testing (still being studied). Indeed, the new TNM classification for head and neck cancers (8th edition) distinguishes between HPV-negative oropharyngeal cancers and HPV-positive oropharyngeal cancers (OPC) to improve patient communication, clinical management, risk stratification and disease monitoring [105,106]. Therefore, it is important for individuals to be aware of any changes in the oral tissues and seek medical attention if necessary.
The main works retrieved and included in this scoping review have been summarized and synthetized and are presented in Table 1.
The need to intercept premalignant lesions, and to identify them before they give rise to a malignant neoplasm, has directed efforts towards the search for increasingly effective and reliable innovative methodologies [104]. Following this pursuit, in the last few decades, the advancement of scientific technologies and molecular research have allowed us to explore markers and genetic and epigenetic factors and to define their link to early diagnosis and OSCC progression and prognosis, thus treating them as essential in future individual and tailor-made approaches [107,108,109]. In the future, advancements in technology and medical research may lead to improved methods for diagnosing oral cancer. For example, researchers are exploring the use of biomarkers, such as saliva and blood tests, to detect oral cancer at its earliest stages [110]. Artificial intelligence and machine learning may also play a role in oral cancer diagnosis by helping healthcare providers to identify and analyze patterns in large amounts of data [111]. Additionally, there is ongoing research into the use of devices such as handheld spectroscopy tools and portable imaging devices, which may make it easier for healthcare providers to perform oral cancer screenings in various settings. The future of oral cancer diagnosis is likely to involve a combination of these and other innovative technologies, with the goal of improving accuracy and early detection rates.

4.1. Biomarkers

Biomarkers for oral malignant and premalignant lesions refer to substances or characteristics in the body that indicate the presence of a disease or condition. These biomarkers represent a strategy target that may be involved in diagnosing and monitoring the evolution of these kinds of lesions in their early stages or to monitor the effectiveness of treatment [112]. Examples of oral cancer biomarkers include certain proteins, genes, or microRNA.
One of the major targets of interest has been the down-regulation of cell adhesion proteins, which has proven to contribute to OSCC progression [113]. Several authors have investigated E-cadherin and P-cadherin expression and reported their role as negative prognostic factors of OSCC, due to their aggressive biological behavior with a tendency to infiltrate and metastasize [70,80]. Likewise, several studies have been conducted focusing on the prognostic role of CD44 in oral carcinoma progression; according to Carinci et al., an under-expressed level of CD44 is related to a reduced survival rate and contrariwise, the over-expression of CD44 in primary tumors was compatible with a more extended survival outcome [114]. Following this path, the advancements of molecular analyses have led to the exploration of potential markers and genetic and epigenetic factors in an effort to elucidate their link to early diagnosis and OSCC progression and prognosis and use them as targets in future studies [83,85].
The identification of molecular markers that can accurately characterize lesions that will display aggressive behavior and worse prognosis has been enhanced with the improvement of imaging techniques, which have been introduced in oral medicine to evaluate their potential role in early diagnosing tumoral signs of alteration, thus lessening the need for biopsy [115,116,117,118].
It is important to note that biomarker research is still in its early stages and more research is needed to fully understand their role in oral cancer diagnosis and treatment.
Therefore, due to the lack of large-scale studies and insufficient data, the use of biomarkers alone is not enough to diagnose and follow up these lesions and biopsy remains mandatory [119].

4.2. Reflectance Confocal Microscopy (RCM)

The in vivo and real-time visualization of oral cavity tissue layers demonstrated its usefulness during the last few decades in the diagnosis of suspicious lesions, showing a considerable sensitivity both for OSCC and Oral Potentially Malignant Disorders [120,121]. A critical and deep investigation of the imaging technology’s applicability to potentially malignant disorders has been carried out. Reflectance Confocal Microscopy (RCM) represents one of the most interesting devices, based on a system composed by a diode laser at 830 nm and combined with a water immersion objective lens [122]. RCM offers optical scanning by horizontal planes, layer by layer, with the ability to deeply penetrate both ex vivo samples and in vivo tissues [123]. Interesting confocal images of dysplastic and neoplastic oral tissues were documented by Maitland et al., proving a strong correlation between RCM and histological sections [124]. Furthermore, Agozzino et al. exhibited remarkable data from their RCM experience with in vivo imaging of Oral Malignant Disorders; the authors reported confocal images of epithelial layers with atypia and polymorphic keratinocytes in Oral Squamous Cell Carcinoma, before its histological confirmation [125].

4.3. Optical Coherence Tomography (OCT)

Similarly, based on interferometer technology, Optical Coherence Tomography (OCT) has proven to be a notable fit for oral cavity exploration [126]. OCT allows us to produce real-time cross-sectional sub-surface tissue images, conventionally explained as tomographic images, and generate real-time depth-resolved two-dimensional (2D) and three-dimensional (3D) images at superior spatial resolution (10–20 μm) and a satisfactory depth of penetration into the tissues [127]. In oral medicine, OCT has proven to provide several advantages over other imaging techniques, including its non-invasive nature, fast imaging speed, and high level of reproducibility [128]. These characteristics make OCT an important tool for ongoing monitoring of oral health and for guiding therapeutic interventions [118]. Additionally, OCT has the potential to aid in the early detection and diagnosis of oral diseases, potentially leading to improved outcomes for patients [129]. OCT technology has emphasized its useful diagnostic aid potential in oral disorders; a study by Wilder-Smith et al. showed 50 white or red potentially malignant lesions, explored by OCT, thus revealing a decisive correlation between OCT-based diagnostic pictures and the histological outcomes [130]. In addition, Heidari’s group highlighted the potential diagnostic accuracy of OCT scanning, through a diagnostic algorithm compared to clinical observations. The authors reported that OCT-based diagnosis was a possible resource to enhance the sensitivity and specificity of current procedures [131]. Furthermore, in a study by Sunny et al., they reported 125 OCT images of OSCCs. The OCT-based images highly correlated with the histological results, further revealing the ability to differentiate OSCC from non-dysplastic and dysplastic lesions and to differentiate oral epithelial dysplasia (OED) from non-dysplastic oral lesions [128].

4.4. High-Frequency Ultrasound

On the other hand, ultrasonography represents a routine support technique in various branches of medicine, such as gynecology, gastroenterology, cardiology, angiology, and in the head and neck region, to detect structural abnormalities and/or tumors of the salivary glands [132].
This technique for oral cavity exploration is not widely used, but various applications have been reported in the literature: ranging from the study of periodontal and peri-implant tissues to the clinical and surgical characterization of benign and malignant lesions of the oral mucosa [133,134]. Thanks to the advancement of technology, some recent ex vivo and in vivo studies suggest that high-frequency ultrasound holds the ability to distinguish between tissue layers based on image contrast [135]. This contrast represents the result of acoustic backscatter, which is directly proportional to the change in density found at the boundaries between tissue layers. As a result, the layers of less dense adipose tissue appear hypoechoic (black), while denser tissue layers such as muscle and connective tissue appear hyperechoic (white) on images [136]. Intraoral ultrasonography is a diagnostic imaging technique that uses high-frequency sound waves to produce images of structures within the oral cavity [133]. This type of ultrasonography has shown feasibility to be adopted in oral medicine to evaluate the size and position of various structures such as teeth, jaw bones, and soft tissues, as well as to identify pathological conditions such as neoformation, cysts, and abscesses. The procedure constitutes a non-invasive method and does not involve radiation exposure, making it a safe and effective tool for routine dental evaluations [53].
As shown in a study by Iida et al., ultrasonography represents a reproducible and valid technique for the evaluation of a tumor’s Depth of Invasion (DOI), confirming the accuracy of a preoperative measurement of DOI in 56 patients affected by tongue squamous cell carcinoma (TSCC) [137]. Di Stasio et al. presented striking images of squamous cell carcinoma of the tongue, which ultrasonographically presents as a rather defined hypoechoic lesion in relation to the surrounding tissues with homogeneous or relatively hyperechoic echogenicity, emphasizing the great potential for diagnostic support in this type of neoplastic lesions [134].

4.5. Photodiagnosis and Photodynamic Therapy

An additional method for early diagnosis and treatment of malignant and potentially malignant lesions of the oral cavity involves the use of photosensitizing substances, irradiated by light through laser and nonlaser devices; the most commonly used device is the diode laser because it is small and inexpensive [138,139]. Siddiqui et al. recently proposed a portable and easy-to-use fiber-coupled LED system that uses a smartphone fluorescence imaging device [140]. Regarding photosensitizing agents, the use of Photofrin®, administered intravenously, has been suggested for the treatment of oral cancer at any stage [141]. On the other hand, 5-aminolevulinic acid (5-ALA), administered both topically and systemically, is unable to penetrate tissue so it is more suitable for use in the treatment of premalignant lesions [142].
Table 1. Summary data of the included studies.
Table 1. Summary data of the included studies.
Cit. Num.First AuthorYear of PublicationTitleMethodologyType of Neoplastic LesionAnalysed Outcome
[87]Lucchese A2005Proteomic Scan for Tyrosinase Peptide Antigenic Pattern in Vitiligo and Melanoma: Role of Sequence Similarity and HLA-DR1 Affinity1biomarkersvitiligo, melanomathe tyrosinase autoepitope profile of melanoma/vitiligo patient sera
[88]Tiwari R2004Computational peptide dissection of Melan-a/MART-1 oncoprotein antigenicity biomarkersmelanomahuman Melan-A/MART-1 sequence
[89]Willers J2005Definition of Anti-Tyrosinase MAb T311 Linear Determinant by Proteome-Based Similarity Analysisbiomarkersvitiligo, melanomathe human tyrosinase protein sequence
[91]Gupta A2018Role of E-Cadherin in Progression of Oral Squamous Cell Carcinoma: A Retrospective Immunohistochemical StudybiomarkersOED, OSCCE-cadherin expression in oral carcinogenesis
[66]Lo Muzio L2005P-Cadherin Expression and Survival Rate in Oral Squamous Cell Carcinoma: An Immunohistochemical StudybiomarkersOSCCprevalence of P-cadherin expression in oral squamous cell carcinoma
[92]Carinci F2002CD44 as Prognostic Factor in Oral and Oropharyngeal Squamous Cell CarcinomabiomarkersOSCCCD44 expression
[69]Pannone G2007CYCLOOXYGENASE ISOZYMES IN ORAL SQUAMOUS CELL CARCINOMA: A REAL-TIME RT-PCR STUDY WITH CLINIC P A THOLOGICAL CORRELA TIONSbiomarkersOSCCCOX-l and COX-2 m-RNA expression
[71]Aquino G2012pEGFR-Tyr 845 expression as prognostic factors in oral squamous cell carcinomabiomarkersOSCCEGFR expression
[97]Lucchese A2016The Potential Role of in Vivo Reflectance Confocal Microscopy for Evaluating Oral Cavity Lesions: A Systematic Review.in-vivo imagingOED, OSCCreflectance confocal microscopy (RCM) for evaluating oral cavity lesions
[100]Maitland KC2008In Vivo Imaging of Oral Neoplasia Using a Miniaturized Fiber Optic Confocal Reflectance Microscopein-vivo imagingOSCCreflectance confocal appearance for evaluating oral cavity lesions
[101]Agozzino M2014Noninvasive, in Vivo Assessment of Oral Squamous Cell Carcinomain-vivo imagingOSCCreflectance confocal appearance for evaluating oral cavity lesions
[103]Gentile E2017The Potential Role of in Vivo Optical Coherence Tomography for Evaluating Oral Soft Tissue: A Systematic Reviewin-vivo imagingOED, OSCCoptical coherence tomography (OCT) for evaluating oral cavity lesions
[104]Wilder-Smith P 2009In Vivo Diagnosis of Oral Dysplasia and Malignancy Using Optical Coherence Tomography: Preliminary Studies in 50 Patientsin-vivo imagingOED, OSCCoptical coherence tomography (OCT) for evaluating oral cavity lesions
[105]Heidari AE2019Optical Coherence Tomography as an Oral Cancer Screening Adjunct in a Low Resource Settingsin-vivo imagingOED, OSCCoptical coherence tomography (OCT) for evaluating oral cavity lesions
[106]Sunny SP2019Intra-Operative Point-of-Procedure Delineation of Oral Cancer Margins Using Optical Coherence Tomographyin-vivo imagingOED, OSCCoptical coherence tomography (OCT) for evaluating oral cavity malignant lesions
[107]Sengupta S2018Review on the Use of Magnetic Fields and Ultrasound for Non-Invasive Cancer Treatmentin-vivo imagingcancerseffects of magnetic fields and ultrasound on cancer cells and their application for cancer treatment
[108]Romano A2022An Overview of High-Definition Ultrasounds Applied in Oral Diseasesin-vivo imagingOSCCHigh-Definition Ultrasound for evaluating oral cavity lesions
[109]Di Stasio D2022High-Definition Ultrasound Characterization of Squamous Carcinoma of the Tongue: A Descriptive Observational Studyin-vivo imaging squamous carcinoma of the tongueHigh-Definition Ultrasound for evaluating oral cavity lesions
[112]Iida, Y2018Depth of Invasion in Superficial Oral Tongue Carcinoma Quantified Using Intraoral Ultrasonographyin-vivo imagingoral tongue carcinomaHigh-Definition Ultrasound for evaluating Depth of invasion (DOI) in oral carcinoma
[140]Siddiqui SA2022Clinical evaluation of a mobile, low-cost system for fluorescence guided photodynamic therapy of early oral cancer in India. in-vivo imagingOED, OSCCPhotodiagnosis (PD) and Photodynamic therapy (PDT) for detection and treatment of early oral cancer
OED = oral epithelial dysplasia, OSCC = oral squamous cell carcinoma.
Protoporphyrin IX (the actual photosensitizer) is produced precisely from 5-ALA in cells capable of heme synthesis and there is a negative feedback mechanism involving ALA synthase, thus preventing overproduction. Unlike other sensitizing substances that induce photosensitivity for several weeks, this agent peaks in 4–6 h and is cleared in about 24 h, without significant collateral effects, making it possible to repeat treatment even at short intervals [143]. On the other hand, the latest generation of photosensitizing agents is conjugated with antibodies, protein/receptor complexes or liposomes, thus presenting a greater affinity for the target tissue [144]. After dosing, the extent of the lesion is first assessed using a blue light that activates red fluorescence (photodiagnosis, PD), with higher intensity in tumor cells due to their ability to produce more protoporphyrin than the surrounding cells.
Photodynamic therapy exploits the same agent illuminated by red light; this light activation triggers a photochemical reaction, leading to the production of reactive oxygen species, thus inducing tissue necrosis [145]. In 1993, as reported by Grant et al., 5-ALA is given orally for systemic use (previously used topically or by intravenous administration) for the first time in the treatment of advanced oral OSCC, inducing necrosis in three out of four patients [143]. This treatment is precise, selective, and noninvasive and can be used for potentially malignant, precancerous, and cancerous lesions even when other therapeutic strategies, such as surgery, are not applicable [140].
Today, the indications for photodynamic therapy have diversified, being effective against viral, bacterial or fungal infections, malignant and pre-malignant lesions of other regions of the body (prostate, bladder, stomach, breast, etc.). It is also used in the treatment of benign and malignant skin diseases, such as Actinic Keratosis, often with the use of 5-ALA, ensuring a good aesthetic outcome among the other benefits [146].

5. Limitations of the Study

An analysis of oral pathology focusing on three topics—malignant and premalignant lesions, and diagnostic aids—was conducted. However, due to the heterogeneity of the fields, a systematic search was not feasible; instead, an overview was performed. The substantial amount of data collected precluded meta-analysis, as it comprised a combination of in vivo and ex vivo studies, technological support for in vivo diagnostic imaging, and analyses involving microbiome or salivary assessments.

6. Conclusions

Oral Squamous Cell Carcinoma (OSCC) is a type of cancer that affects the cells lining the mouth and can be potentially life-threatening if not detected and treated early. Factors that increase the risk of oral cancer include tobacco use, excessive alcohol intake, UV light exposure, inadequate oral hygiene, and a compromised immune system. Regular dental exams and routine screenings are crucial for early diagnosis. Possible treatments include surgery, radiation, chemotherapy, and targeted therapy. Advances in technology and medical research offer hope for improving OSCC and the accuracy of its precursor entities’ diagnosis and early detection rates in the future. Diagnostic processes have always been crucial in the clinical course; an excellent diagnostic path should guarantee the lowest level of invasiveness, ease and quickness of execution, and the reproducibility of the procedure [25,53]. The potential role of innovative technological and molecular methods does not represent an alternative to histopathological examination [119]. Nonetheless, the current literature agrees with the suitability of these methods for oral tissue inspection, before or during a biopsy and in monitoring the evolution of oral lesions through follow-up appointments. Future investigations are highly desirable to alleviate the current delays in diagnosis and expedite the diagnostic process, ensuring early and non-invasive identification of Oral Malignant and Potentially Malignant Disorders.

Author Contributions

Conceptualization: D.M. and M.S.; methodology: C.E.B., G.M.D. and M.S.; investigation: D.M., C.E.B. and R.M.; data curation: C.E.B., R.M. and S.S.; writing—original draft preparation, D.M.; writing—review and editing, M.S. and D.G.; supervision: D.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

The original contributions presented in the study are included in the article, further inquiries can be directed to the corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Melsen, B. Tissue Reaction and Biomechanics. Front. Oral Biol. 2016, 18, 36–45. [Google Scholar] [CrossRef]
  2. Sbordone, C.; Toti, P.; Brevi, B.; Martuscelli, R.; Sbordone, L.; di Spirito, F. Computed Tomography-Aided Descriptive Analysis of Maxillary and Mandibular Atrophies. J. Stomatol. Oral Maxillofac. Surg. 2019, 120, 99–105. [Google Scholar] [CrossRef]
  3. Minervini, G.; Romano, A.; Petruzzi, M.; Maio, C.; Serpico, R.; Lucchese, A.; Candotto, V.; di Stasio, D. Telescopic Overdenture on Natural Teeth: Prosthetic Rehabilitation on (OFD) Syndromic Patient and a Review on Available Literature. J. Biol. Regul. Homeost. Agents 2018, 32 (Suppl. S1), 131–134. [Google Scholar]
  4. Contaldo, M.; Luzzi, V.; Ierardo, G.; Raimondo, E.; Boccellino, M.; Ferati, K.; Bexheti-Ferati, A.; Inchingolo, F.; di Domenico, M.; Serpico, R.; et al. Bisphosphonate-Related Osteonecrosis of the Jaws and Dental Surgery Procedures in Children and Young People with Osteogenesis Imperfecta: A Systematic Review. J. Stomatol. Oral Maxillofac. Surg. 2020, 121, 556–562. [Google Scholar] [CrossRef] [PubMed]
  5. Minervini, G.; Nucci, L.; Lanza, A.; Femiano, F.; Contaldo, M.; Grassia, V. Temporomandibular Disc Displacement with Reduction Treated with Anterior Repositioning Splint: A 2-Year Clinical and Magnetic Resonance Imaging (MRI) Follow-Up. J. Biol. Regul. Homeost. Agents 2020, 34 (Suppl. S1), 151–160. [Google Scholar] [PubMed]
  6. Contaldo, M.; Fusco, A.; Stiuso, P.; Lama, S.; Gravina, A.G.; Itro, A.; Federico, A.; Itro, A.; Dipalma, G.; Inchingolo, F.; et al. Oral Microbiota and Salivary Levels of Oral Pathogens in Gastro-Intestinal Diseases: Current Knowledge and Exploratory Study. Microorganisms 2021, 9, 1064. [Google Scholar] [CrossRef]
  7. Contaldo, M.; Lucchese, A.; Romano, A.; della Vella, F.; di Stasio, D.; Serpico, R.; Petruzzi, M. Oral Microbiota Features in Subjects with Down Syndrome and Periodontal Diseases: A Systematic Review. Int. J. Mol. Sci. 2021, 22, 9251. [Google Scholar] [CrossRef]
  8. Contaldo, M.; Lucchese, A.; Lajolo, C.; Rupe, C.; di Stasio, D.; Romano, A.; Petruzzi, M.; Serpico, R. The Oral Microbiota Changes in Orthodontic Patients and Effects on Oral Health: An Overview. J. Clin. Med. 2021, 10, 780. [Google Scholar] [CrossRef]
  9. Contaldo, M.; Itro, A.; Lajolo, C.; Gioco, G.; Inchingolo, F.; Serpico, R. Overview on Osteoporosis, Periodontitis and Oral Dysbiosis: The Emerging Role of Oral Microbiota. Appl. Sci. 2020, 10, 6000. [Google Scholar] [CrossRef]
  10. Rebelo, M.B.; Oliveira, C.S.; Tavaria, F.K. Novel Strategies for Preventing Dysbiosis in the Oral Cavity. Front. Biosci. 2023, 15, 23. [Google Scholar] [CrossRef]
  11. di Spirito, F.; Toti, P.; Pilone, V.; Carinci, F.; Lauritano, D.; Sbordone, L. The Association between Periodontitis and Human Colorectal Cancer: Genetic and Pathogenic Linkage. Life 2020, 10, 211. [Google Scholar] [CrossRef]
  12. Contaldo, M.; della Vella, F.; Raimondo, E.; Minervini, G.; Buljubasic, M.; Ogodescu, A.; Sinescu, C.; Serpico, R. Early Childhood Oral Health Impact Scale (ECOHIS): Literature Review and Italian Validation. Int. J. Dent. Hyg. 2020, 18, 396–402. [Google Scholar] [CrossRef]
  13. Paoletti, I.; Fusco, A.; Grimaldi, E.; Perillo, L.; Coretti, L.; di Domenico, M.; Cozza, V.; Lucchese, A.; Contaldo, M.; Serpico, R.; et al. Assessment of Host Defence Mechanisms Induced by Candida Species. Int. J. Immunopathol. Pharmacol. 2013, 26, 663–672. [Google Scholar] [CrossRef]
  14. Milillo, L.; lo Muzio, L.; Carlino, P.; Serpico, R.; Coccia, E.; Scully, C. Candida-Related Denture Stomatitis: A Pilot Study of the Efficacy of an Amorolfine Antifungal Varnish. Int. J. Prosthodont. 2005, 18, 55–59. [Google Scholar] [PubMed]
  15. Contaldo, M.; Romano, A.; Mascitti, M.; Fiori, F.; della Vella, F.; Serpico, R.; Santarelli, A. Association between Denture Stomatitis, Candida Species and Diabetic Status. J. Biol. Regul. Homeost. Agents 2019, 33 (Suppl. S1), 35–41. [Google Scholar] [PubMed]
  16. di Stasio, D.; Lauritano, D.; Minervini, G.; Paparella, R.S.; Petruzzi, M.; Romano, A.; Candotto, V.; Lucchese, A. Management of Denture Stomatitis: A Narrative Review. J. Biol. Regul. Homeost. Agents 2018, 32 (Suppl. S1), 113–116. [Google Scholar]
  17. Lucchese, A.; di Stasio, D.; Romano, A.; Fiori, F.; de Felice, G.P.; Lajolo, C.; Serpico, R.; Cecchetti, F.; Petruzzi, M. Correlation between Oral Lichen Planus and Viral Infections Other Than HCV: A Systematic Review. J. Clin. Med. 2022, 11, 5487. [Google Scholar] [CrossRef] [PubMed]
  18. Donnarumma, G.; de Gregorio, V.; Fusco, A.; Farina, E.; Baroni, A.; Esposito, V.; Contaldo, M.; Petruzzi, M.; Pannone, G.; Serpico, R. Inhibition of HSV-1 Replication by Laser Diode-Irradiation: Possible Mechanism of Action. Int. J. Immunopathol. Pharmacol. 2010, 23, 1167–1176. [Google Scholar] [CrossRef]
  19. Lucchese, A.; Serpico, R.; Crincoli, V.; Shoenfeld, Y.; Kanduc, D. Sequence Uniqueness as a Molecular Signature of HIV-1-Derived B-Cell Epitopes. Int. J. Immunopathol. Pharmacol. 2009, 22, 639–646. [Google Scholar] [CrossRef]
  20. Kanduc, D.; Serpico, R.; Lucchese, A.; Shoenfeld, Y. Correlating Low-Similarity Peptide Sequences and HIV B-Cell Epitopes. Autoimmun. Rev. 2008, 7, 291–296. [Google Scholar] [CrossRef]
  21. Constantin, M.; Chifiriuc, M.C.; Mihaescu, G.; Vrancianu, C.O.; Dobre, E.G.; Cristian, R.E.; Bleotu, C.; Bertesteanu, S.V.; Grigore, R.; Serban, B.; et al. Implications of oral dysbiosis and HPV infection in head and neck cancer: From molecular and cellular mechanisms to early diagnosis and therapy. Front. Oncol. 2023, 13, 1273516. [Google Scholar] [CrossRef]
  22. Romano, A.; di Stasio, D.; Lauritano, D.; Lajolo, C.; Fiori, F.; Gentile, E.; Lucchese, A. Topical Photodynamic Therapy in the Treatment of Benign Oral Mucosal Lesions: A Systematic Review. J. Oral Pathol. Med. 2021, 50, 639–648. [Google Scholar] [CrossRef]
  23. Boccellino, M.; di Stasio, D.; Romano, A.; Petruzzi, M.; Lucchese, A.; Serpico, R.; Frati, L.; di Domenico, M. Lichen Planus: Molecular Pathway and Clinical Implications in Oral Disorders. J. Biol. Regul. Homeost. Agents 2018, 32 (Suppl. S1), 135–138. [Google Scholar]
  24. della Vella, F.; Lauritano, D.; Lajolo, C.; Lucchese, A.; di Stasio, D.; Contaldo, M.; Serpico, R.; Petruzzi, M. The Pseudolesions of the Oral Mucosa: Differential Diagnosis and Related Systemic Conditions. Appl. Sci. 2019, 9, 2412. [Google Scholar] [CrossRef]
  25. Romano, A.; di Stasio, D.; Petruzzi, M.; Fiori, F.; Lajolo, C.; Santarelli, A.; Lucchese, A.; Serpico, R.; Contaldo, M. Noninvasive Imaging Methods to Improve the Diagnosis of Oral Carcinoma and Its Precursors: State of the Art and Proposal of a Three-Step Diagnostic Process. Cancers 2021, 13, 2864. [Google Scholar] [CrossRef]
  26. di Stasio, D.; Romano, A.; Paparella, R.S.; Gentile, C.; Serpico, R.; Minervini, G.; Candotto, V.; Laino, L. How Social Media Meet Patients’ Questions: YouTube™ Review for Mouth Sores in Children. J. Biol. Regul. Homeost. Agents 2018, 32 (Suppl. S1), 117–121. [Google Scholar]
  27. Gioco, G.; Rupe, C.; Basco, A.; Contaldo, M.; Gallenzi, P.; Lajolo, C. Oral Juvenile Xanthogranuloma: An Unusual Presentation in an Adult Patient and a Systematic Analysis of Published Cases. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2022, 133, 42–49. [Google Scholar] [CrossRef]
  28. Minervini, G.; Romano, A.; Petruzzi, M.; Maio, C.; Serpico, R.; di Stasio, D.; Lucchese, A. Oral-Facial-Digital Syndrome (OFD): 31-Year Follow-up Management and Monitoring. J. Biol. Regul. Homeost. Agents 2018, 32 (Suppl. S1), 127–130. [Google Scholar] [PubMed]
  29. Romano, A.; Santarelli, A.; Lajolo, C.; della Vella, F.; Mascitti, M.; Serpico, R.; Contaldo, M. Analysis of Oral Mucosa Erosive-Ulcerative Lesions by Reflectance Confocal Microscopy. J. Biol. Regul. Homeost. Agents 2019, 33 (Suppl. S1), 11–17. [Google Scholar] [PubMed]
  30. de Benedittis, M.; Petruzzi, M.; Favia, G.; Serpico, R. Oro-Dental Manifestations in Hallopeau-Siemens-Type Recessive Dystrophic Epidermolysis Bullosa. Clin. Exp. Dermatol. 2004, 29, 128–132. [Google Scholar] [CrossRef] [PubMed]
  31. Angelini, G.; Bonamonte, D.; Lucchese, A.; Favia, G.; Serpico, R.; Mittelman, A.; Simone, S.; Sinha, A.A.; Kanduc, D. Preliminary Data on Pemphigus Vulgaris Treatment by a Proteomics-Defined Peptide: A Case Report. J. Transl. Med. 2006, 4, 43. [Google Scholar] [CrossRef] [PubMed]
  32. Lucchese, A.; Mittelman, A.; Tessitore, L.; Serpico, R.; Sinha, A.A.; Kanduc, D. Proteomic Definition of a Desmoglein Linear Determinant Common to Pemphigus Vulgaris and Pemphigus Foliaceous. J. Transl. Med. 2006, 4, 37. [Google Scholar] [CrossRef]
  33. di Stasio, D. An Overview of Burning Mouth Syndrome. Front. Biosci. 2016, 8, 762. [Google Scholar] [CrossRef] [PubMed]
  34. Mussap, M.; Beretta, P.; Esposito, E.; Fanos, V. Once upon a Time Oral Microbiota: A Cinderella or a Protagonist in Autism Spectrum Disorder? Metabolites 2023, 13, 1183. [Google Scholar] [CrossRef]
  35. Mascitti, M.; Togni, L.; Caponio, V.C.A.; Zhurakivska, K.; Bizzoca, M.E.; Contaldo, M.; Serpico, R.; lo Muzio, L.; Santarelli, A. Lymphovascular Invasion as a Prognostic Tool for Oral Squamous Cell Carcinoma: A Comprehensive Review. Int. J. Oral Maxillofac. Surg. 2022, 51, 1–9. [Google Scholar] [CrossRef] [PubMed]
  36. Romano, A.; di Stasio, D.; Gentile, E.; Petruzzi, M.; Serpico, R.; Lucchese, A. The Potential Role of Photodynamic Therapy in Oral Premalignant and Malignant Lesions: A Systematic Review. J. Oral Pathol. Med. 2021, 50, 333–344. [Google Scholar] [CrossRef]
  37. di Stasio, D.; Romano, A.; Russo, D.; Fiori, F.; Laino, L.; Caponio, V.C.A.; Troiano, G.; Lo Muzio, L.; Serpico, R.; Lucchese, A. Photodynamic Therapy Using Topical Toluidine Blue for the Treatment of Oral Leukoplakia: A Prospective Case Series. Photodiagnosis Photodyn. Ther. 2020, 31, 101888. [Google Scholar] [CrossRef]
  38. Mello, F.W.; Miguel, A.F.P.; Dutra, K.L.; Porporatti, A.L.; Warnakulasuriya, S.; Guerra, E.N.S.; Rivero, E.R.C. Prevalence of Oral Potentially Malignant Disorders: A Systematic Review and Meta-Analysis. J. Oral Pathol. Med. 2018, 47, 633–640. [Google Scholar] [CrossRef]
  39. Louredo, B.V.R.; de Lima-Souza, R.A.; Pérez-de-Oliveira, M.E.; Warnakulasuriya, S.; Kerr, A.R.; Kowalski, L.P.; Hunter, K.D.; Prado-Ribeiro, A.C.; Vargas, P.A.; Santos-Silva, A.R.D. Reported physical examination methods for screening of oral cancer and oral potentially malignant disorders: A systematic review. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2023. Epub ahead of print. [Google Scholar] [CrossRef]
  40. Contaldo, M.; Lauritano, D.; Carinci, F.; Romano, A.; di Stasio, D.; Lajolo, C.; della Vella, F.; Serpico, R.; Lucchese, A. Intraoral Confocal Microscopy of Suspicious Oral Lesions: A Prospective Case Series. Int. J. Dermatol. 2020, 59, 82–90. [Google Scholar] [CrossRef]
  41. Contaldo, M.; Boccellino, M.; Zannini, G.; Romano, A.; Sciarra, A.; Sacco, A.; Settembre, G.; Coppola, M.; di Carlo, A.; D’Angelo, L.; et al. Sex Hormones and Inflammation Role in Oral Cancer Progression: A Molecular and Biological Point of View. J. Oncol. 2020, 2020, 9587971. [Google Scholar] [CrossRef] [PubMed]
  42. Giannelli, G.; Milillo, L.; Marinosci, F.; lo Muzio, L.; Serpico, R.; Antonaci, S. Altered Expression of Integrins and Basement Membrane Proteins in Malignant and Pre-Malignant Lesions of Oral Mucosa. J. Biol. Regul. Homeost. Agents 2001, 15, 375–380. [Google Scholar]
  43. Dholariya, S.; Singh, R.D.; Sonagra, A.; Yadav, D.; Vajaria, B.N.; Parchwani, D. Integrating Cutting-Edge Methods to Oral Cancer Screening, Analysis, and Prognosis. Crit. Rev. Oncog. 2023, 28, 11–44. [Google Scholar] [CrossRef] [PubMed]
  44. Villa, A.; Sonis, S. Oral Leukoplakia Remains a Challenging Condition. Oral Dis. 2018, 24, 179–183. [Google Scholar] [CrossRef] [PubMed]
  45. Warnakulasuriya, S.; Johnson, N.W.; Van der Waal, I. Nomenclature and Classification of Potentially Malignant Disorders of the Oral Mucosa. J. Oral Pathol. Med. 2007, 36, 575–580. [Google Scholar] [CrossRef]
  46. van der Waal, I. Potentially Malignant Disorders of the Oral and Oropharyngeal Mucosa; Present Concepts of Management. Oral Oncol. 2010, 46, 423–425. [Google Scholar] [CrossRef]
  47. di Stasio, D.; Romano, A.; Gentile, C.; Maio, C.; Lucchese, A.; Serpico, R.; Paparella, R.; Minervini, G.; Candotto, V.; Laino, L. Systemic and Topical Photodynamic Therapy (PDT) on Oral Mucosa Lesions: An Overview. J. Biol. Regul. Homeost. Agents 2018, 32 (Suppl. S1), 123–126. [Google Scholar]
  48. Hazarey, V.; Desai, K.M.; Warnakulasuriya, S. Proliferative verrucous leukoplakia/multifocal leukoplakia in patients with and without oral submucous fibrosis. Med. Oral Patol. Oral Cir. Bucal. 2024, 29, e119–e127. [Google Scholar] [CrossRef]
  49. Villa, A.; Woo, S.b. Leukoplakia—A Diagnostic and Management Algorithm. J. Oral Maxillofac. Surg. 2017, 75, 723–734. [Google Scholar] [CrossRef]
  50. Lan, Q.; Zhang, C.; Hua, H.; Hu, X. Compositional and functional changes in the salivary microbiota related to oral leukoplakia and oral squamous cell carcinoma: A case control study. BMC Oral Health 2023, 23, 1021. [Google Scholar] [CrossRef]
  51. Evren, I.; Najim, A.M.; Poell, J.B.; Brouns, E.R.; Wils, L.J.; Peferoen, L.A.N.; Brakenhoff, R.H.; Bloemena, E.; van der Meij, E.H.; de Visscher, J.G.A.M. The value of regular follow-up of oral leukoplakia for early detection of malignant transformation. Oral Dis. 2023. Epub ahead of print. [Google Scholar] [CrossRef]
  52. Holmstrup, P. Oral Erythroplakia-What Is It? Oral Dis. 2018, 24, 138–143. [Google Scholar] [CrossRef]
  53. Fiori, F.; Rullo, R.; Contaldo, M.; Inchingolo, F.; Romano, A. Noninvasive In-Vivo Imaging of Oral Mucosa: State-of-the-Art. Minerva Dent. Oral Sci. 2021, 70, 286–293. [Google Scholar] [CrossRef] [PubMed]
  54. Romano, A.; Contaldo, M.; della Vella, F.; Russo, D.; Lajolo, C.; Serpico, R.; di Stasio, D. Topical Toluidine Blue-Mediated Photodynamic Therapy for the Treatment of Oral Lichen Planus. J. Biol. Regul. Homeost. Agents 2019, 33 (Suppl. S1), 27–33. [Google Scholar] [PubMed]
  55. di Stasio, D.; Lucchese, A.; Romano, A.; Adinolfi, L.E.; Serpico, R.; Marrone, A. The Clinical Impact of Direct-Acting Antiviral Treatment on Patients Affected by Hepatitis C Virus-Related Oral Lichen Planus: A Cohort Study. Clin. Oral Investig. 2022, 26, 5409–5417. [Google Scholar] [CrossRef] [PubMed]
  56. Romano, A.; Grassi, R.; Fiori, F.; Nardi, G.M.; Borgia, R.; Contaldo, M.; Serpico, R.; Petruzzi, M. Oral Lichen Planus and Epstein-Barr Virus: A Narrative Review. J. Biol. Regul. Homeost. Agents 2022, 36, 87–92. [Google Scholar] [CrossRef]
  57. Romano, A.; Santoro, R.; Fiori, F.; Contaldo, M.; Serpico, R.; Lucchese, A. Image Postproduction Analysis as a Tool for Evaluating Topical Photodynamic Therapy in the Treatment of Oral Lichen Planus. Photodiagnosis Photodyn. Ther. 2022, 39, 102868. [Google Scholar] [CrossRef] [PubMed]
  58. Iocca, O.; Copelli, C.; Rubattino, S.; Sedran, L.; Di Maio, P.; Arduino, P.G.; Ramieri, G.; Garzino-Demo, P. Oral cavity carcinoma in patients with and without a history of lichen planus: A comparative analysis. Head Neck 2023, 45, 1367–1375. [Google Scholar] [CrossRef] [PubMed]
  59. Contaldo, M.; di Stasio, D.; Petruzzi, M.; Serpico, R.; Lucchese, A. In Vivo Reflectance Confocal Microscopy of Oral Lichen Planus. Int. J. Dermatol. 2019, 58, 940–945. [Google Scholar] [CrossRef] [PubMed]
  60. Agha-Hosseini, F.; Sheykhbahaei, N.; SadrZadeh-Afshar, M.S. Evaluation of Potential Risk Factors that contribute to Malignant Transformation of Oral Lichen Planus: A Literature Review. J. Contemp. Dent. Pract. 2016, 17, 692–701. [Google Scholar] [CrossRef]
  61. Arakeri, G.; Patil, S.G.; Aljabab, A.S.; Lin, K.-C.; Merkx, M.A.W.; Gao, S.; Brennan, P.A. Oral Submucous Fibrosis: An Update on Pathophysiology of Malignant Transformation. J. Oral Pathol. Med. 2017, 46, 413–417. [Google Scholar] [CrossRef]
  62. Phulari, R.G.S.; Dave, E.J. A Systematic Review on the Mechanisms of Malignant Transformation of Oral Submucous Fibrosis. Eur. J. Cancer Prev. 2020, 29, 470–473. [Google Scholar] [CrossRef]
  63. Hande, A.; Chaudhary, M.; Gawande, M.; Gadbail, A.; Zade, P.; Bajaj, S.; Patil, S.; Tekade, S. Oral Submucous Fibrosis: An Enigmatic Morpho-Insight. J. Cancer Res. Ther. 2019, 15, 463. [Google Scholar] [CrossRef] [PubMed]
  64. Chaturvedi, P.; Vaishampayan, S.S.; Nair, S.; Nair, D.; Agarwal, J.P.; Kane, S.V.; Pawar, P.; Datta, S. Oral Squamous Cell Carcinoma Arising in Background of Oral Submucous Fibrosis: A Clinicopathologically Distinct Disease. Head Neck 2012, 35, 1404–1409. [Google Scholar] [CrossRef] [PubMed]
  65. Chaturvedi, P.; Malik, A.; Nair, D.; Nair, S.; Mishra, A.; Garg, A.; Vaishampayan, S. Oral Squamous Cell Carcinoma Associated with Oral Submucous Fibrosis Have Better Oncologic Outcome than Those Without. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2017, 124, 225–230. [Google Scholar] [CrossRef] [PubMed]
  66. Balcere, A.; Konrāde-Jilmaza, L.; Pauliņa, L.A.; Čēma, I.; Krūmiņa, A. Clinical Characteristics of Actinic Keratosis Associated with the Risk of Progression to Invasive Squamous Cell Carcinoma: A Systematic Review. J. Clin. Med. 2022, 11, 5899. [Google Scholar] [CrossRef] [PubMed]
  67. EI-Naggar, A.K.; Chan, J.K.C.; Grandis, J.R.; Takata, T.; Slootweg, P.J. WHO Classification of Head and Neck Tumours; IARC: Lyon, France, 2017. [Google Scholar]
  68. Imanguli, M.; Alevizos, I.; Brown, R.; Pavletic, S.; Atkinson, J. Oral Graft-versus-Host Disease. Oral Dis. 2008, 14, 396–412. [Google Scholar] [CrossRef] [PubMed]
  69. Pannone, G.; Bufo, P.; Santoro, A.; Franco, R.; Aquino, G.; Longo, F.; Botti, G.; Serpico, R.; Cafarelli, B.; Abbruzzese, A.; et al. WNT Pathway in Oral Cancer: Epigenetic Inactivation of WNT-Inhibitors. Oncol. Rep. 2010, 24, 1035–1041. [Google Scholar] [CrossRef] [PubMed]
  70. Pannone, G.; Santoro, A.; Feola, A.; Bufo, P.; Papagerakis, P.; lo Muzio, L.; Staibano, S.; Ionna, F.; Longo, F.; Franco, R.; et al. The Role of E-Cadherin down-Regulation in Oral Cancer: CDH1 Gene Expression and Epigenetic Blockage. Curr. Cancer Drug Targets 2014, 14, 115–127. [Google Scholar] [CrossRef]
  71. Solomon, M.C.; Chandrashekar, C.; Kulkarni, S.; Shetty, N.; Pandey, A. Exosomes: Mediators of cellular communication in potentially malignant oral lesions and head and neck cancers. F1000Res 2023, 12, 58. [Google Scholar] [CrossRef]
  72. Wetzel, S.L.; Wollenberg, J. Oral Potentially Malignant Disorders. Dent. Clin. N. Am. 2020, 64, 25–37. [Google Scholar] [CrossRef]
  73. Caudell, J.J.; Gillison, M.L.; Maghami, E.; Spencer, S.; Pfister, D.G.; Adkins, D.; Birkeland, A.C.; Brizel, D.M.; Busse, P.M.; Cmelak, A.J.; et al. NCCN Guidelines® Insights: Head and Neck Cancers, Version 1.2022. J. Natl. Compr. Cancer Netw. 2022, 20, 224–234. [Google Scholar] [CrossRef]
  74. Lajolo, C.; Patini, R.; Limongelli, L.; Favia, G.; Tempesta, A.; Contaldo, M.; de Corso, E.; Giuliani, M. Brown Tumors of the Oral Cavity: Presentation of 4 New Cases and a Systematic Literature Review. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2020, 129, 575–584.e4. [Google Scholar] [CrossRef]
  75. Favia, G.; lo Muzio, L.; Serpico, R.; Maiorano, E. Angiosarcoma of the Head and Neck with Intra-Oral Presentation. A Clinico-Pathological Study of Four Cases. Oral Oncol. 2002, 38, 757–762. [Google Scholar] [CrossRef]
  76. Favia, G.; Lo Muzio, L.; Serpico, R.; Maiorano, E. Rhabdomyoma of the Head and Neck: Clinicopathologic Features of Two Cases. Head Neck 2003, 25, 700–704. [Google Scholar] [CrossRef] [PubMed]
  77. Lauritano, D.; Lucchese, A.; Contaldo, M.; Serpico, R.; lo Muzio, L.; Biolcati, F.; Carinci, F. Oral Squamous Cell Carcinoma: Diagnostic Markers and Prognostic Indicators. J. Biol. Regul. Homeost. Agents 2016, 30 (Suppl. S1), 169–176. [Google Scholar]
  78. Rivera, C. Essentials of Oral Cancer. Int. J. Clin. Exp. Pathol. 2015, 8, 11884–11894. [Google Scholar] [PubMed]
  79. lo Muzio, L.; Leonardi, R.; Mariggiò, M.A.; Mignogna, M.D.; Rubini, C.; Vinella, A.; Pannone, G.; Giannetti, L.; Serpico, R.; Testa, N.F.; et al. HSP 27 as Possible Prognostic Factor in Patients with Oral Squamous Cell Carcinoma. Histol. Histopathol. 2004, 19, 119–128. [Google Scholar] [CrossRef]
  80. lo Muzio, L.; Campisi, G.; Farina, A.; Rubini, C.; Pannone, G.; Serpico, R.; Laino, G.; de Lillo, A.; Carinci, F. P-Cadherin Expression and Survival Rate in Oral Squamous Cell Carcinoma: An Immunohistochemical Study. BMC Cancer 2005, 5, 63. [Google Scholar] [CrossRef] [PubMed]
  81. Lo Muzio, L.; Leonardi, R.; Mignogna, M.D.; Pannone, G.; Rubini, C.; Pieramici, T.; Trevisiol, L.; Ferrari, F.; Serpico, R.; Testa, N.; et al. Scatter Factor Receptor (c-Met) as Possible Prognostic Factor in Patients with Oral Squamous Cell Carcinoma. Anticancer Res. 2004, 24, 1063–1069. [Google Scholar] [PubMed]
  82. Pannone, G.; Bufo, P.; Caiaffa, M.F.; Serpico, R.; Lanza, A.; Lo Muzio, L.; Rubini, C.; Staibano, S.; Petruzzi, M.; de Benedictis, M.; et al. Cyclooxygenase-2 Expression in Oral Squamous Cell Carcinoma. Int. J. Immunopathol. Pharmacol. 2004, 17, 273–282. [Google Scholar] [CrossRef]
  83. Pannone, G.; Sanguedolce, F.; de Maria, S.; Farina, E.; Lo Muzio, L.; Serpico, R.; Emanuelli, M.; Rubini, C.; de Rosa, G.; Staibano, S.; et al. Cyclooxygenase Isozymes in Oral Squamous Cell Carcinoma: A Real-Time RT-PCR Study with Clinic Pathological Correlations. Int. J. Immunopathol. Pharmacol. 2007, 20, 317–324. [Google Scholar] [CrossRef] [PubMed]
  84. Fiorelli, A.; Ricciardi, C.; Pannone, G.; Santoro, A.; Bufo, P.; Santini, M.; Serpico, R.; Rullo, R.; Pierantoni, G.M.; di Domenico, M. Interplay between Steroid Receptors and Neoplastic Progression in Sarcoma Tumors. J. Cell Physiol. 2011, 226, 2997–3003. [Google Scholar] [CrossRef] [PubMed]
  85. Aquino, G.; Pannone, G.; Santoro, A.; Liguori, G.; Franco, R.; Serpico, R.; Florio, G.; de Rosa, A.; Mattoni, M.; Cozza, V.; et al. PEGFR-Tyr 845 Expression as Prognostic Factors in Oral Squamous Cell Carcinoma. Cancer Biol. Ther. 2012, 13, 967–977. [Google Scholar] [CrossRef] [PubMed]
  86. Saikia, P.J.; Pathak, L.; Mitra, S.; Das, B. The emerging role of oral microbiota in oral cancer initiation, progression and stemness. Front. Immunol. 2023, 14, 1198269. [Google Scholar] [CrossRef] [PubMed]
  87. di Spirito, F.; Amato, A.; Romano, A.; Dipalma, G.; Xhajanka, E.; Baroni, A.; Serpico, R.; Inchingolo, F.; Contaldo, M. Analysis of Risk Factors of Oral Cancer and Periodontitis from a Sex- and Gender-Related Perspective: Gender Dentistry. Appl. Sci. 2022, 12, 9135. [Google Scholar] [CrossRef]
  88. Pannone, G.; Santoro, A.; Carinci, F.; Bufo, P.; Papagerakis, S.M.; Rubini, C.; Campisi, G.; Giovannelli, L.; Contaldo, M.; Serpico, R.; et al. Double Demonstration of Oncogenic High Risk Human Papilloma Virus DNA and HPV-E7 Protein in Oral Cancers. Int. J. Immunopathol. Pharmacol. 2011, 24 (Suppl. S2), 95–101. [Google Scholar] [CrossRef]
  89. Favia, G.; Kanduc, D.; lo Muzio, L.; Lucchese, A.; Serpico, R. Possible Association between HPV16 E7 Protein Level and Cytokeratin 19. Int. J. Cancer 2004, 111, 795–797. [Google Scholar] [CrossRef]
  90. Kanduc, D.; Lucchese, A.; Mittelman, A. Individuation of Monoclonal Anti-HPV16 E7 Antibody Linear Peptide Epitope by Computational Biology. Peptides 2001, 22, 1981–1985. [Google Scholar] [CrossRef]
  91. Katirachi, S.K.; Grønlund, M.P.; Jakobsen, K.K.; Grønhøj, C.; von Buchwald, C. The Prevalence of HPV in Oral Cavity Squamous Cell Carcinoma. Viruses 2023, 15, 451. [Google Scholar] [CrossRef]
  92. Mao, L. Oral Squamous Cell Carcinoma–Progresses from Risk Assessment to Treatment. Chin. J. Dent. Res. 2012, 15, 83–88. [Google Scholar] [PubMed]
  93. Contaldo, M.; Di Napoli, A.; Pannone, G.; Franco, R.; Ionna, F.; Feola, A.; De Rosa, A.; Santoro, A.; Sbordone, C.; Longo, F.; et al. Prognostic Implications of Node Metastatic Features in OSCC: A Retrospective Study on 121 Neck Dissections. Oncol. Rep. 2013, 30, 2697–2704. [Google Scholar] [CrossRef]
  94. Johnson, D.E.; Burtness, B.; Leemans, C.R.; Lui, V.W.Y.; Bauman, J.E.; Grandis, J.R. Head and neck squamous cell carcinoma. Nat. Rev. Dis. Primers. 2020, 6, 92, Erratum in Nat. Rev. Dis. Primers 2023, 9, 4. [Google Scholar] [CrossRef] [PubMed]
  95. de Maria, S.; Pannone, G.; Bufo, P.; Santoro, A.; Serpico, R.; Metafora, S.; Rubini, C.; Pasquali, D.; Papagerakis, S.M.; Staibano, S.; et al. Survivin Gene-Expression and Splicing Isoforms in Oral Squamous Cell Carcinoma. J. Cancer Res. Clin. Oncol. 2009, 135, 107–116. [Google Scholar] [CrossRef] [PubMed]
  96. Pannone, G.; Hindi, S.A.H.; Santoro, A.; Sanguedolce, F.; Rubini, C.; Cincione, R.I.; de Maria, S.; Tortorella, S.; Rocchetti, R.; Cagiano, S.; et al. Aurora B Expression as a Prognostic Indicator and Possible Therapeutic Target in Oral Squamous Cell Carcinoma. Int. J. Immunopathol. Pharmacol. 2011, 24, 79–88. [Google Scholar] [CrossRef]
  97. lo Muzio, L.; Mignogna, M.; Pannone, G.; Rubini, C.; Grassi, R.; Nocini, P.; Ferrari, F.; Serpico, R.; Favia, G.; de Rosa, G.; et al. Expression of Bcl-2 in Oral Squamous Cell Carcinoma: An Immunohistochemical Study of 90 Cases with Clinico-Pathological Correlations. Oncol. Rep. 2003, 10, 285–291. [Google Scholar] [CrossRef]
  98. Wehrhan, F.; Büttner-Herold, M.; Hyckel, P.; Moebius, P.; Preidl, R.; Distel, L.; Ries, J.; Amann, K.; Schmitt, C.; Neukam, F.W.; 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. [Google Scholar] [CrossRef]
  99. Zanoni, D.K.; Montero, P.H.; Migliacci, J.C.; Shah, J.P.; Wong, R.J.; Ganly, I.; Patel, S.G. Survival Outcomes after Treatment of Cancer of the Oral Cavity (1985–2015). Oral Oncol. 2019, 90, 115–121. [Google Scholar] [CrossRef]
  100. Somma, P.; lo Muzio, L.; Mansueto, G.; Delfino, M.; Fabbrocini, G.; Mascolo, M.; Mignogna, C.; di Benedetto, M.; Carinci, F.; de Lillo, A.; et al. Squamous Cell Carcinoma of the Lower LIP: Fas/Fasl Expression, Lymphocyte Subtypes and Outcome. Int. J. Immunopathol. Pharmacol. 2005, 18, 59–64. [Google Scholar] [CrossRef]
  101. Mittelman, A.; Lucchese, A.; Sinha, A.A.; Kanduc, D. Monoclonal and Polyclonal Humoral Immune Response to EC HER-2/NEU Peptides with Low Similarity to the Host’s Proteome. Int. J. Cancer 2002, 98, 741–747. [Google Scholar] [CrossRef]
  102. Staibano, S.; lo Muzio, L.; Pannone, G.; Somma, P.; Farronato, G.; Franco, R.; Bambini, F.; Serpico, R.; de Rosa, G. P53 and HMSH2 Expression in Basal Cell Carcinomas and Malignant Melanomas from Photoexposed Areas of Head and Neck Region. Int. J. Oncol. 2001, 19, 551–559. [Google Scholar] [CrossRef]
  103. López-Jornet, P.; Olmo-Monedero, A.; Peres-Rubio, C.; Pons-Fuster, E.; Tvarijonaviciute, A. Preliminary Evaluation Salivary Biomarkers in Patients with Oral Potentially Malignant Disorders (OPMD): A Case-Control Study. Cancers 2023, 15, 5256. [Google Scholar] [CrossRef]
  104. Lucchese, A.; Willers, J.; Mittelman, A.; Kanduc, D.; Dummer, R. Proteomic Scan for Tyrosinase Peptide Antigenic Pattern in Vitiligo and Melanoma: Role of Sequence Similarity and HLA-DR1 Affinity. J. Immunol. 2005, 175, 7009–7020. [Google Scholar] [CrossRef]
  105. Huang, S.H.; O’Sullivan, B. Overview of the 8th Edition TNM Classification for Head and Neck Cancer. Curr. Treat. Options Oncol. 2017, 18, 40. [Google Scholar] [CrossRef] [PubMed]
  106. Batool, S.; Sethi, R.K.V.; Wang, A.; Dabekaussen, K.; Egloff, A.M.; Del Vecchio Fitz, C.; Kuperwasser, C.; Uppaluri, R.; Shin, J.; Rettig, E.M. Circulating tumor-tissue modified HPV DNA testing in the clinical evaluation of patients at risk for HPV-positive oropharynx cancer: The IDEA-HPV study. Oral Oncol. 2023, 147, 106584. [Google Scholar] [CrossRef] [PubMed]
  107. Tiwari, R.; Geliebter, J.; Lucchese, A.; Mittelman, A.; Kanduc, D. Computational Peptide Dissection of Melan-a/MART-1 Oncoprotein Antigenicity. Peptides 2004, 25, 1865–1871. [Google Scholar] [CrossRef]
  108. Willers, J.; Lucchese, A.; Mittelman, A.; Dummer, R.; Kanduc, D. Definition of Anti-Tyrosinase MAb T311 Linear Determinant by Proteome-Based Similarity Analysis. Exp. Dermatol. 2005, 14, 543–550. [Google Scholar] [CrossRef] [PubMed]
  109. Kanduc, D.; Lucchese, A.; Mittelman, A. Non-Redundant Peptidomes from DAPs: Towards “The Vaccine”? Autoimmun. Rev. 2007, 6, 290–294. [Google Scholar] [CrossRef] [PubMed]
  110. di Stasio, D.; Romano, A.; Boschetti, C.E.; Montella, M.; Mosca, L.; Lucchese, A. Salivary MiRNAs Expression in Potentially Malignant Disorders of the Oral Mucosa and Oral Squamous Cell Carcinoma: A Pilot Study on MiR-21, MiR-27b, and MiR-181b. Cancers 2022, 15, 291. [Google Scholar] [CrossRef] [PubMed]
  111. Ilhan, B.; Lin, K.; Guneri, P.; Wilder-Smith, P. Improving Oral Cancer Outcomes with Imaging and Artificial Intelligence. J. Dent. Res. 2020, 99, 241–248. [Google Scholar] [CrossRef] [PubMed]
  112. Stasio, D.d.; Mosca, L.; Lucchese, A.; Cave, D.D.; Kawasaki, H.; Lombardi, A.; Porcelli, M.; Caraglia, M. Salivary Mir-27b Expression in Oral Lichen Planus Patients: A Series of Cases and a Narrative Review of Literature. Curr. Top. Med. Chem. 2020, 19, 2816–2823. [Google Scholar] [CrossRef]
  113. Gupta, A.; Sharma, S.; Batra, M.; Abidullah, M.; Bhuvinder, S.; Katragadda, P. Role of E-Cadherin in Progression of Oral Squamous Cell Carcinoma: A Retrospective Immunohistochemical Study. J. Contemp. Dent. Pract. 2018, 19, 1105–1110. [Google Scholar] [PubMed]
  114. Carinci, F.; Stabellini, G.; Calvitti, M.; Pelucchi, S.; Targa, L.; Farina, A.; Pezzetti, F.; Pastore, A. CD44 as Prognostic Factor in Oral and Oropharyngeal Squamous Cell Carcinoma. J. Craniofacial Surg. 2002, 13, 85–89. [Google Scholar] [CrossRef] [PubMed]
  115. Contaldo, M.; Lajolo, C.; di Petrillo, M.; Ballini, A.; Inchingolo, F.; Serpico, R.; Romano, A. Analysis of Lip Pigmentations by Reflectance Confocal Microscopy: Report of Two Cases. J. Biol. Regul. Homeost. Agents 2019, 33 (Suppl. S1), 19–25. [Google Scholar] [PubMed]
  116. di Stasio, D.; Lauritano, D.; Loffredo, F.; Gentile, E.; della Vella, F.; Petruzzi, M.; Lucchese, A. Optical Coherence Tomography Imaging of Oral Mucosa Bullous Diseases: A Preliminary Study. Dentomaxillofac Radiol. 2020, 49, 20190071. [Google Scholar] [CrossRef] [PubMed]
  117. Contaldo, M.; Poh, C.F.; Guillaud, M.; Lucchese, A.; Rullo, R.; Lam, S.; Serpico, R.; MacAulay, C.E.; Lane, P.M. Oral Mucosa Optical Biopsy by a Novel Handheld Fluorescent Confocal Microscope Specifically Developed: Technologic Improvements and Future Prospects. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2013, 116, 752–758. [Google Scholar] [CrossRef] [PubMed]
  118. Contaldo, M.; Agozzino, M.; Moscarella, E.; Esposito, S.; Serpico, R.; Ardigò, M. In Vivo Characterization of Healthy Oral Mucosa by Reflectance Confocal Microscopy: A Translational Research for Optical Biopsy. Ultrastruct. Pathol. 2013, 37, 151–158. [Google Scholar] [CrossRef] [PubMed]
  119. Adam, M. Can blood and salivary biomarkers replace biopsy as a diagnostic tool? Evid. Based Dent. 2022, 23, 10–11. [Google Scholar] [CrossRef] [PubMed]
  120. Romano, A.; di Spirito, F.; Amato, A.; Ferraro, G.A.; Dipalma, G.; Xhajanka, E.; Serpico, R.; Inchingolo, F.; Contaldo, M. Dental Microstructural Imaging: From Conventional Radiology to In Vivo Confocal Microscopy. Appl. Sci. 2022, 12, 10654. [Google Scholar] [CrossRef]
  121. Lucchese, A.; Gentile, E.; Romano, A.; Maio, C.; Laino, L.; Serpico, R. The Potential Role of in Vivo Reflectance Confocal Microscopy for Evaluating Oral Cavity Lesions: A Systematic Review. J. Oral Pathol. Med. 2016, 45, 723–729. [Google Scholar] [CrossRef]
  122. Contaldo, M.; di Stasio, D.; della Vella, F.; Lauritano, D.; Serpico, R.; Santoro, R.; Lucchese, A. Real Time In Vivo Confocal Microscopic Analysis of the Enamel Remineralization by Casein Phosphopeptide-Amorphous Calcium Phosphate (CPP-ACP): A Clinical Proof-of-Concept Study. Appl. Sci. 2020, 10, 4155. [Google Scholar] [CrossRef]
  123. Grassia, V.; Gentile, E.; di Stasio, D.; Jamilian, A.; Matarese, G.; D’Apuzzo, F.; Santoro, R.; Perillo, L.; Serpico, R.; Lucchese, A. In Vivo Confocal Microscopy Analysis of Enamel Defects after Orthodontic Treatment: A Preliminary Study. Ultrastruct. Pathol. 2016, 40, 317–323. [Google Scholar] [CrossRef] [PubMed]
  124. Maitland, K.C.; Gillenwater, A.M.; Williams, M.D.; El-Naggar, A.K.; Descour, M.R.; Richards-Kortum, R.R. In Vivo Imaging of Oral Neoplasia Using a Miniaturized Fiber Optic Confocal Reflectance Microscope. Oral Oncol. 2008, 44, 1059–1066. [Google Scholar] [CrossRef]
  125. Agozzino, M.; Bhasne, P.; Franceschini, C.; Vincenza, G.; Catricalà, C.; Ardigò, M. Noninvasive, in Vivo Assessment of Oral Squamous Cell Carcinoma. Br. J. Dermatol. 2014, 170, 754–756. [Google Scholar] [CrossRef]
  126. di Stasio, D.; Lauritano, D.; Romano, A.; Salerno, C.; Minervini, G.; Minervini, G.; Gentile, E.; Serpico, R.; Lucchese, A. In Vivo Characterization of Oral Pemphigus Vulgaris by Optical Coherence Tomography. J. Biol. Regul. Homeost. Agents 2015, 29 (Suppl. S1), 39–41. [Google Scholar]
  127. Gentile, E.; Maio, C.; Romano, A.; Laino, L.; Lucchese, A. The Potential Role of in Vivo Optical Coherence Tomography for Evaluating Oral Soft Tissue: A Systematic Review. J. Oral Pathol. Med. 2017, 46, 864–876. [Google Scholar] [CrossRef]
  128. Sunny, S.P.; Agarwal, S.; James, B.L.; Heidari, E.; Muralidharan, A.; Yadav, V.; Pillai, V.; Shetty, V.; Chen, Z.; Hedne, N.; et al. Intra-Operative Point-of-Procedure Delineation of Oral Cancer Margins Using Optical Coherence Tomography. Oral Oncol. 2019, 92, 12–19. [Google Scholar] [CrossRef]
  129. Wilder-Smith, P.; Lee, K.; Guo, S.; Zhang, J.; Osann, K.; Chen, Z.; Messadi, D. In Vivo Diagnosis of Oral Dysplasia and Malignancy Using Optical Coherence Tomography: Preliminary Studies in 50 Patients. Lasers Surg. Med. 2009, 41, 353–357. [Google Scholar] [CrossRef]
  130. Heidari, A.E.; Suresh, A.; Kuriakose, M.A.; Chen, Z.; Wilder-Smith, P.; Sunny, S.P.; James, B.L.; Lam, T.M.; Tran, A.V.; Yu, J.; et al. Optical Coherence Tomography as an Oral Cancer Screening Adjunct in a Low Resource Settings. IEEE J. Sel. Top. Quantum Electron. 2019, 25, 2869643. [Google Scholar] [CrossRef]
  131. Sengupta, S.; Balla, V.K. A Review on the Use of Magnetic Fields and Ultrasound for Non-Invasive Cancer Treatment. J. Adv. Res. 2018, 14, 97–111. [Google Scholar] [CrossRef]
  132. Romano, A.; Montella, M.; Ronchi, A.; Borgia, R.; Fiori, F.; Contaldo, M.; Serpico, R. An Overview of High-Definition Ultrasounds Applied in Oral Diseases. J. Biol. Regul. Homeost. Agents 2022, 36, 77–86. [Google Scholar] [CrossRef]
  133. di Stasio, D.; Montella, M.; Romano, A.; Colella, G.; Serpico, R.; Lucchese, A. High-Definition Ultrasound Characterization of Squamous Carcinoma of the Tongue: A Descriptive Observational Study. Cancers 2022, 14, 564. [Google Scholar] [CrossRef]
  134. di Stasio, D.; Lauritano, D.; Paparella, R.; Franco, R.; Montella, M.; Serpico, R.; Lucchese, A. Ultrasound Imaging of Oral Fibroma: A Case Report. J. Biol. Regul. Homeost. Agents 2017, 31 (Suppl. S1), 23–26. [Google Scholar]
  135. di Stasio, D.; Romano, A.; Montella, M.; Contaldo, M.; Petruzzi, M.; Hasan, I.; Serpico, R.; Lucchese, A. Quantitative Ultrasound Analysis of Oral Mucosa: An Observational Cross-Sectional Study. Appl. Sci. 2022, 12, 6829. [Google Scholar] [CrossRef]
  136. Iida, Y.; Kamijo, T.; Kusafuka, K.; Omae, K.; Nishiya, Y.; Hamaguchi, N.; Morita, K.; Onitsuka, T. Depth of Invasion in Superficial Oral Tongue Carcinoma Quantified Using Intraoral Ultrasonography. Laryngoscope 2018, 128, 2778–2782. [Google Scholar] [CrossRef]
  137. Contaldo, M.; Lucchese, A.; Gentile, E.; Zulli, C.; Petruzzi, M.; Lauritano, D.; Amato, M.R.; Esposito, P.; Riegler, G.; Serpico, R. Evaluation of the Intraepithelial Papillary Capillary Loops in Benign and Malignant Oral Lesions by in Vivo Virtual Chromoendoscopic Magnification: A Preliminary Study. J. Biol. Regul. Homeost. Agents 2017, 31 (Suppl. S1), 11–22. [Google Scholar]
  138. Huang, Z. A review of progress in clinical photodynamic therapy. Technol. Cancer Res. Treat. 2005, 4, 283–293. [Google Scholar] [CrossRef]
  139. Brancaleon, L.; Moseley, H. Laser and non-laser light sources for photodynamic therapy. Lasers Med. Sci. 2002, 17, 173–186. [Google Scholar] [CrossRef]
  140. Siddiqui, S.A.; Siddiqui, S.; Hussain, M.A.B.; Khan, S.; Liu, H.; Akhtar, K.; Hasan, S.A.; Ahmed, I.; Mallidi, S.; Khan, A.P.; et al. Clinical evaluation of a mobile, low-cost system for fluorescence guided photodynamic therapy of early oral cancer in India. Photodiagnosis Photodyn. Ther. 2022, 38, 102843. [Google Scholar] [CrossRef]
  141. Biel, M. Advances in photodynamic therapy for the treatment of head and neck cancers. Lasers Surg. Med. 2006, 38, 349–355. [Google Scholar] [CrossRef] [PubMed]
  142. Konopka, K.; Goslinski, T. Photodynamic therapy in dentistry. J. Dent. Res. 2007, 86, 694–707, Erratum in J. Dent. Res. 2007, 86, 1126. [Google Scholar] [CrossRef] [PubMed]
  143. Grant, W.E.; Hopper, C.; MacRobert, A.J.; Speight, P.M.; Bown, S.G. Photodynamic therapy of oral cancer: Photosensitisation with systemic aminolaevulinic acid. Lancet 1993, 342, 147–148. [Google Scholar] [CrossRef] [PubMed]
  144. Allison, R.R.; Downie, G.H.; Cuenca, R.; Hu, X.H.; Childs, C.J.; Sibata, C.H. Photosensitizers in clinical PDT. Photodiagnosis Photodyn. Ther. 2004, 1, 27–42. [Google Scholar] [CrossRef] [PubMed]
  145. Celli, J.P.; Spring, B.Q.; Rizvi, I.; Evans, C.L.; Samkoe, K.S.; Verma, S.; Pogue, B.W.; Hasan, T. Imaging and photodynamic therapy: Mechanisms, monitoring, and optimization. Chem. Rev. 2010, 110, 2795–2838. [Google Scholar] [CrossRef] [PubMed]
  146. Moy, L.S.; Frost, D.; Moy, S. Photodynamic Therapy for Photodamage, Actinic Keratosis, and Acne in the Cosmetic Practice. Facial Plast. Surg. Clin. N. Am. 2020, 28, 135–148. [Google Scholar] [CrossRef]
Figure 1. Oral Leukoplakia involving the tongue.
Figure 1. Oral Leukoplakia involving the tongue.
Bioengineering 11 00228 g001
Figure 2. Oral Squamous Cell Carcinoma involving the tongue.
Figure 2. Oral Squamous Cell Carcinoma involving the tongue.
Bioengineering 11 00228 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Menditti, D.; Santagata, M.; Guida, D.; Magliulo, R.; D’Antonio, G.M.; Staglianò, S.; Boschetti, C.E. State of the Art in the Diagnosis and Assessment of Oral Malignant and Potentially Malignant Disorders: Present Insights and Future Outlook—An Overview. Bioengineering 2024, 11, 228. https://doi.org/10.3390/bioengineering11030228

AMA Style

Menditti D, Santagata M, Guida D, Magliulo R, D’Antonio GM, Staglianò S, Boschetti CE. State of the Art in the Diagnosis and Assessment of Oral Malignant and Potentially Malignant Disorders: Present Insights and Future Outlook—An Overview. Bioengineering. 2024; 11(3):228. https://doi.org/10.3390/bioengineering11030228

Chicago/Turabian Style

Menditti, Dardo, Mario Santagata, David Guida, Roberta Magliulo, Giovanni Maria D’Antonio, Samuel Staglianò, and Ciro Emiliano Boschetti. 2024. "State of the Art in the Diagnosis and Assessment of Oral Malignant and Potentially Malignant Disorders: Present Insights and Future Outlook—An Overview" Bioengineering 11, no. 3: 228. https://doi.org/10.3390/bioengineering11030228

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop