Reviews
Published: 2021-05-14
download
PDF

Oral cancer: changing the aim of the biopsy in the age of precision medicine. A review

Division of Otolaryngology and Head & Neck Surgery, European Institute of Oncology IRCCS, Milan, Italy
Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
Division of Otolaryngology and Head & Neck Surgery, European Institute of Oncology IRCCS, Milan, Italy
Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
Division of Otolaryngology and Head & Neck Surgery, European Institute of Oncology IRCCS, Milan, Italy
Division of Otolaryngology and Head & Neck Surgery, European Institute of Oncology IRCCS, Milan, Italy
Division of Otolaryngology and Head & Neck Surgery, European Institute of Oncology IRCCS, Milan, Italy
Division of Otolaryngology and Head & Neck Surgery, European Institute of Oncology IRCCS, Milan, Italy
Division of Otolaryngology and Head & Neck Surgery, European Institute of Oncology IRCCS, Milan, Italy
surgical biopsy liquid biopsy radiomics depth of invasion precision medicine

Abstract

Oral cancer is a heterogeneous disease that develops through a complex, multi-step process. Precision medicine should help to better understand its molecular basis, integrate traditional classifications and have a positive impact on cancer management. To apply this information in clinical practice, we need to define its histology and identify biomarkers expressed by the tumour that provide useful information for planning tailored treatment. The most reliable information currently derives from evaluation of biomarkers on post-operative samples. To plan personalised treatment, oncologists need to assess these markers on biopsy samples. We reviewed the recent literature and identified 6 of 184 publications that compared markers measured on biopsy and post-operative samples or assessed their predictivity for the development of lymph node metastases. Data from these studies suggest that markers measured on biopsy samples can provide useful indications for tailoring treatments. However, due to their heterogeneity and low level of evidence, these results need to be confirmed by clinical studies on a large population to standardise and validate biomarkers in biopsies and to assess their reliability in other diagnostic mini-invasive procedures such as radiomics and liquid biopsy.

Introduction

Cancer of the oral cavity is the eighth most common cancer worldwide: GLOBOCAN reported an estimated 354,900 new cases in 2018; about 90% of these tumours are oral squamous cell carcinomas (OSCCs) with a 5-year survival of < 60% 1. OSCCs can arise from the oral tongue, which is the most common sub-site and has the worse prognosis 2-4, followed by the floor of mouth, buccal mucosa, alveolar mucosa, and hard palate. Historically, OSCC affects older people, mainly men, smokers and alcohol drinkers. Recent clinical and epidemiological studies have reported an overall decline in the incidence of tobacco-related OSCCs, probably due to the anti-smoking campaigns 5-7, but an increasing trend of tongue and oropharyngeal cancers in younger patients, women and individuals with no exposure to traditional risk factors 8-10. While infection with human papillomavirus accounts for an increasing number of patients with oropharyngeal cancers, risk factors, especially viruses, in OSCC non-smokers and non-drinkers, are unclear 11,12. Head and neck cancer (HNC), including OSCC, is a heterogeneous disease that develops through a complex, multi-step process involving genetic alterations, growth regulation, apoptosis, angiogenesis, invasion and metastasis; moreover, it is influenced by the individual’s genetic predisposition and environmental exposure to carcinogens 13,14. This heterogeneity is confirmed in daily practice by the observation that patients with clinically similar tumours have different treatment responses and outcomes 15,16.

Precision medicine

To address this heterogeneity, the Cancer Genome Atlas project 17, classifications based on molecular pathology 18, and the Precision Medicine Initiative 19 have been proposed. These efforts should help to better understand the molecular basis of cancer, integrate traditional classifications and have a positive impact on cancer management 20. Given this complexity, both the diagnosis and therapy of OSCCs should be planned by a highly specialised multidisciplinary team that is able to globally assess the disease and patient 21,22. Such personalised diagnostic and treatment strategies which precisely target molecular alterations refer to Precision Medicine or Genomics-Driven Cancer Medicine 23-25. To apply precision medicine in clinical practice, we need to identify biomarkers expressed by tumours that reflect their individual characteristics 26-28.

Tissue biomarkers validated on post-operative surgical specimens

Many prognostic/predictive biomarkers have been taken into account and tested, such as c-erbB2, EGFR (Epidermal Growth Factor Receptor), tumour infiltrating lymphocytes, c-met, etc., and currently the most reliable information on high-risk patients derives from assessment of tissue biomarkers on post-operative samples. In 2004, two trials by the European Organization for Research and Treatment of Cancer (EORTC), and the United States Radiation Therapy Oncology Group (RTOG) compared adjuvant radiation alone vs chemoradiation in advanced head and neck cancers. The results showed that the addition of concurrent chemotherapy was associated with better oncologic outcomes compared to radiation alone in a subset of high risk patients. They evaluated pathological classification of the primary tumour, positive margins, tumour depth, positive nodes, extranodal extension (ENE), lympho-vascular invasion (LVI), perineural invasion (PNI) and histology grade 29,30. A subsequent comparative analysis of data pooled from these two trials showed that ENE and/or microscopically involved surgical margins were the only risk factors for which the impact of post-operative chemo-radiotherapy was significant in both trials 31. Cooper et al. in 2012 confirmed these data by evaluating the long-term results of patients enrolled in the RTOG study. They found that at 10 years, local recurrence and disease-free survival rates were significantly better only in patients included in the chemo-radiotherapy arm for ENE and/or microscopically involved margins 32. Subsequently, other studies confirmed these data and proposed risk classes for effective post-operative therapeutic planning 33-36. The 8th edition of the TNM staging system 20 suggests that HPV and depth of invasion (DOI) as well as specific biomarkers expressed by HNCs should be assessed. OSCCs are rarely related to HPV, while DOI is useful to evaluate biological behaviour, because prognosis worsens when the tumour is deeper 37,38. In the past, the DOI of early-stage tongue tumours (T1-T2) was considered a predictor of occult lymph node metastases, as in skin carcinomas, and was used to plan neck dissection 39,40. Before the publication of the TNM, 8th Edition, the terms “Tumour Thickness (TT)” and “Depth of Invasion (DOI)” were often considered synonymous. However, DOI, as defined in the AJCC (American Joint Committee on Cancer) Cancer Staging Manual 8th Edition, has a better predictive value than TT 38,41. Recent studies based on this definition suggest that DOI > 4 mm (range: 3.4 mm-6.6 mm) is a strong predictor of locoregional recurrence, and survival together with two or more markers such as PNI, LVI and Worst Pattern Of Invasion (WPOI-5) 42-44 which have been integrated into the 8th TNM edition. In addition, several studies have highlighted tumour budding (BD) and tumour-stroma ratio (TSR), considered in other non-TNM-based staging systems, as predictive factors for lymphatic diffusion and disease-free survival (DFS) 15,45-47. Table I shows the definition and purpose of these markers 2,15,37,38,42,45,46,48-57.

To plan personalised treatment, oncologists need to assess these markers, including DOI, on biopsy samples according to the AJCC TNM (8th edition) recommendations 38. Herein, we review the literature in order to evaluate the concordance between the values of tissue biomarkers measured in both pre-operative and post-operative samples from the same patient.

Methods

For this purpose, we reviewed the recent literature published in English (January 2015-December 2019) by consulting the PubMed, Medline, Web of Science, Embase and Cochrane databases. We used the following keywords (strings): oral biopsy and depth of infiltration/DOI; oral biopsy and tumour/tumor budding; oral cancer diagnosis and depth of infiltration/DOI; oral cancer diagnosis and tumour/tumor budding. Only peer-reviewed papers were considered. We thus identified 184 papers (Fig. 1); 90 of these were duplicates. The remaining 94 were evaluated based on the abstracts. Inclusion criteria were: A) histologically proven squamous cell carcinomas (SCC) of the oral cavity; B) both prospective and retrospective studies; C) papers that compared biopsy and post-operative samples from the same patient; D) papers that evaluated predictivity of biomarkers on biopsy specimens; E) articles based on a retrospective series whose biopsy and post-operative slides were reviewed by the authors. Exclusion criteria: a) reviews, case reports, abstracts; b) histology other than SCC; c) studies that did not review histological samples. As a result of this analysis, we discarded 72 papers unrelated to our aim; for 12 studies, criteria for patient selection or study design were not reported in detail. Fourteen papers (the 10 selected articles and 4 others identified through a manual search) were evaluated on the complete text. Following this analysis, 8 more papers were considered unsuitable for our purpose as they were reviews or the study design did not fall within the selection criteria.

Results

All six papers included were retrospective, and were divided into two groups based on the aim of the study (Tab. II) 48,49,58-61:

Comparison of tissue markers on biopsy and post-operative samples

Three papers 58-60 compared the values of biomarkers measured on patient’s biopsy and post-operative samples from the same patients in order to evaluate their concordance and reliability. Almangush et al. 58 reviewed the slides of 100 patients (81 = Stage I-II) with SCC of the mobile tongue treated with surgical excision. They measured TT and found that mean values were 4.1 mm (0.5-10 mm) in biopsy samples vs 6.3 mm (0.5-23 mm) in the corresponding post-operative specimens. They also evaluated TB (Tab. I) and their prognostic score (BD Model, Tab. III), and documented a sensitivity and specificity of pre-operative vs post-operative values of 59% and 100%, respectively, showing a 83% pre-operative/post-operative agreement. Seki et al. 59, among 91 OSCC patients treated with surgery alone or preoperative chemotherapy and resection, analysed 44 undergoing exclusive surgery. They evaluated TT, TB Score (TBS, Tab. III), infiltrative pattern (INF, Tab. III) and LVI, and found that the depth of infiltration measured on biopsies was lower than that on post-operative samples, while there was a good correlation of budding score in pre-treatment and post-treatment samples. Seki Soda et al. 60 recently analysed the relationship between TBS in biopsies and in resected specimens of 248 patients with OSCC and the effect of pre-operative chemotherapy on TBS. The authors did not report staging, but pointed out that 141 of 248 patients received preoperative chemotherapy. The mean TBS in biopsy and resected specimens showed no significant difference between patients with and without preoperative chemotherapy. Moreover, they found that pre-operative chemotherapy is effective in lowering the budding score and suppressing relapses.

Predictive value of pathological markers in biopsy samples

Four papers 48,49,59,61 evaluated whether markers assessed on biopsy samples were predictive of lymph node metastases (Tab. II). Seki et al. 59 examined 33 patients with tongue and floor SCC treated with tumour resection and neck dissection. They found that BD score ≥ 3 and TT > 3 mm was significantly predictive of nodal metastases, overall survival (OS) and relapse-free survival (RFS), while TT alone measured on biopsy samples was not. The same team 48 evaluated TBS on a larger series of 209 OSCC patients (76% cT1) treated with resection or with pre-operative chemotherapy and resection, without stratifying the patients according to treatment modalities. They found that ≥ 3 buds in the biopsy samples was predictive of lymph node metastases, and that TBS was significantly correlated with INF, Grading and LVI (p < 0.01). The authors pointed out that the TB cut-off point needs to be defined, and that the evaluation of TBS and TT needs to be standardised. Nayanar et al. 61 analysed 160 patients with oral cancer (Stage II = 48%) treated with wide excision and neck dissection and proposed a predictive score based on selected pathological features (rete pegs, pattern of invasion, TT, histologic differentiation) and site of tumour (buccal mucosa, tongue, other). The score ranged from 4 to 18; the authors found that a score of 4 is predictive of risk for lymph node metastasis of 5%; for scores of 10, 14 and 18, the risk was 31%, 67% and 91%, respectively. Sahoo et al. 49 studied 150 cT1-T2 patients treated with resection and neck dissection for OSCC. They compared three measurement modalities of the depth of invasion: TT1, TT2 (DOI) and TT3 (alternative DOI); according to the authors, TT2 corresponds to DOI, as defined in the TNM 8th Edition 37,38. They found that TT2 (DOI) and LVI correlate with nodal metastases.

Discussion

To our knowledge, this is the first comprehensive review correlating biological tumour characteristics between biopsy and corresponding pathologic specimen with an evidence-based method. Unfortunately, the studies that met the inclusion criteria had a level of evidence of 3 and 4 according to the AJCC on Cancer Levels of Evidence classification 20. It is worth stressing a criticality common to all: namely that they differ in the design of the study, selection of patients, oral subsite of cancer and in choice of pathological markers such as DOI, TT and budding score; for this reason, the results are not comparable. Furthermore, many authors have measured the depth of the invasion differently than what is foreseen in the TNM 8th Edition, even when published after its release. In this regard, the paper of Driven et al. that quantified the impact of the stage migration on patients’ prognosis using TT and DOI in a retrospective cohort of 456 OSCC patients is interesting. They found similar stage category and prognosis regardless of the measurement used 62. This review highlights several other critical issues.

Biopsy technique

The patients evaluated by Almangush 58, Seki 48 and Seki-Soda 60 underwent incisional biopsy; those studied by Nayanar 61 had an incisional or wedge biopsy; those in the study of Sahoo 49 were treated with an excisional biopsy. The authors did not provide more details on the procedures applied to perform the incisional/excisional biopsy. This information is important since the reliability of the biopsy sample strictly depends on the quality and quantity of the tissue taken. Seki et al. 59 did not specify the biopsy modalities in their first report, but in a later study aimed to evaluate the relationship between TBS assessed in OSCC biopsy specimens and lymph node metastases (LNM), recommending that incisional biopsy be performed peripherally to the cancer so as to obtain a significant amount of non-necrotic tissue from the centre of the tumour 48.

Depth of biopsy

It should also be stressed that the biopsy sample should be deep enough to possibly include the entire tumour infiltration front and underlying healthy tissue 48. Reliability of biopsy (incisional, wedge, punch, excisional) is influenced by the depth of the sample because it is unlikely that specimens always include tumour stroma on the invasive front, which is adequate to evaluate the relationships between the various components of the tumour microenvironment (TME) 48,58,59. In fact, tissue biomarkers are the morphological expression of the TME, or the field in which the immune system and the tumour interplay. Inside TME cancer cells, non-tumour cells and stroma are in a continuous and dynamic relationship, and any change within the TME may reflect changes of the balance between the immune system and tumour 63. In this context, the role of biopsy is not only diagnostic, but also predictive, since biopsies should include the invasive front, the behaviour of tumour cells in the TME interface and biomarkers that provide useful information to guide the therapeutic choice 15,64,65-67. Leite et al. 66, in a recent preliminary study on biopsy samples from 56 OSCC cases, highlighted the possible importance of the correlation between grading, predominant mode of invasion and TB intensity. The authors classified the mode of invasion in 4 degrees, similar to the proposal of Shimuzu 54; they also graded TB as low-intensity (5 buds in one single x200 power field) and high- intensity (≥ 5 buds in one single x200 power field). The majority of cases studied (66.1%) were high-intensity TB; all cases with the worst mode of invasion showed high-intensity TB, while no association was found between TB and histopathological grading. They concluded that both TB and mode of invasion in OSCC diagnostic specimens could help to select patients who could benefit from more aggressive treatment. This paper has some critical issues, such as the biopsy performed in the central part of the tumour, which is rich in necrotic tissue that must be eliminated, and the lack of follow-up. Therefore, these promising data require confirmation on larger series with a longer follow-up. A large biopsy on the surface (at least 8 mm) and depth (at least 5 mm) could allow an assessment of the microenvironment in early cancers, but does not solve the problem 48,64. In this regard, Dhanda 64 compared TT measurement on biopsies and the corresponding post-operative samples reported in the medical records of 93 patients operated on for oral cancer; we have not included this paper in our review according to the exclusion criteria (point c) because those specimens were not revised by a pathologist. However, measurements reported in medical charts provide interesting information because in this series 20% of tumours had a tumour depth > 10 mm on post-operative specimens; on biopsies only 28% of samples had a thickness greater than the DOI calculated on the post-operative specimens. They therefore suggested performing a 10 mm deep punch biopsy, which should potentially include the invasive front in up to 80% of the cases analysed in this paper 64.

Additional biopsy related issues

It should also be pointed out that 17% of biopsy samples are not representative of the cancer due to technical errors such as fragmented, superficial, non-orientable samples and artifacts in the preparation of slides 58. For this reason, how the specimen is sectioned and oriented is important: slides are evaluable if sections are perpendicular and include all mucous, submucosal and muscle layers. On the contrary, tangential sectioning does not allow the evaluation of infiltration depth 68. In addition, biopsy and surgical procedures can sometimes make it difficult to measure the depth of invasion on post-operative specimens. This critical point is underlined by Berdugo who analysed the post-operative samples of 239 glossectomies 69: 5% had only a minimal residual of cancer and in 14% no tumour was found. He also emphasised that in 21/183 (11.5%) pT2 OSCCs the DOI was underestimated due to positivity of deep resection margins. Finally, it should be kept in mind that a large surgical biopsy may create an inflammatory reaction which alters the microenvironment and could promote the regional spread of cancer 70,71.

Changing the aim of biopsy

To plan the best treatment of a tumour, one must know its histopathological diagnosis, site, size and biological aggressiveness. Currently, the risk of developing lymph node metastases can be evaluated by measuring biomarkers such as Grading, PNI, LVI, positive resection margins, depth and mode of invasion and tumour budding on post-operative specimens. On the basis of this information, high-risk patients can be identified for treatment with post-operative radiotherapy or chemoradiotherapy. It is hoped that this predictive and prognostic information will be available for all patients, even those candidates for non-surgical therapies, at the end of the diagnostic process, so as to personalise the therapy for each. This review documents the possibility to reliably test these tissue biomarkers on biopsy samples. However, it is necessary that the biopsy technique and the methods of evaluation of biomarkers are standardised and validated in clinical trials on large case series before they can be used in clinical practice. For these reasons, the role of biopsy is changing and evaluation of a biopsy sample will allow not only to define the morphology of a tumour, but also to guide the therapeutic choice.

Clinical and imaging evaluation of depth of infiltration

The 8th edition of TNM suggests that the DOI of oral cancer is assessed pre-operatively with clinical and imaging investigations 38. It should be emphasised that clinical evaluation is subjective, depends on the experience and ability of each physician and is limited by anatomical conditions such as trismus and/or the patient’s intolerance to palpation for pain 72. Recently, prospective studies comparing pre-operative clinical and magnetic resonance imaging (MRI) evaluation 72, or MRI alone 50,73,74 with post-operative DOI have documented a good correlation of these investigations, especially in tumours with infiltration > 5 mm. On the contrary, other authors have reported an overestimation of T2 lesions and under-estimation of T4 cancers when clinical T staging is compared with histopathological data 75,76. These papers also highlight several critical issues related to imaging (MRI/computed tomography, CT) such as artifacts due to movement and dental fillings; the evaluation of peritumoural oedema and inflammation, and timing between imaging and biopsy (first imaging or biopsy? If imaging is performed after biopsy, how long does it take to get reliable information from MRI/CT?) 50,76,77. Tumour shrinkage of formalin-fixed post-operative specimens may interfere with comparing the DOI between MRI pre-operative imaging and post-operative specimens 72,78. A recent meta-analysis 79 documented a high correlation of TT measured by intraoral ultrasound and histopathology mainly in T1-2 tongue cancers (p < 0.001), although it is operator-dependent and often overestimates exophytic tumours 80,81.

Future directions

Other non-invasive diagnostic modalities are being considered to support evidence-based clinical decision-making, in particular radiomics and liquid biopsy.

Radiomics is an emerging translational field of research. Thanks to the extraction of mathematically defined parameters from routine medical images it is possible to generate large-scale sets of information that can be correlated with OS and treatment-related toxicity and can also be used to identify new biomarkers to be implemented in daily clinical practice 82. Currently, there are few but promising published studies on the application of radiomics in oral cancer with CT or MRI 83-85. Probably, if the role of radiomics will be confirmed with standardised methodology on a large number of patients, these results would help to promote cancer treatment towards personalised precision medicine 82,86.

Liquid biopsy is particularly promising as an alternative to surgical biopsy. It allows assessment of biomarkers in fluids (serum, saliva, urine, etc.) such as circulating tumour cells (CTC), DNA and RNA fragments (miRNA), exosomes that originate from cancerous cells, and their characteristics, particularly mutations and number of somatic alterations 87-90. Liquid biopsy theoretically provides the individual molecular profile of each cancer over time, and could allow physicians to diagnose a tumour, plan tailored therapy and monitor cancer evolution 91,92. Unfortunately, no circulating biomarker has yet been validated for routine use in clinical practice for OSCC, because alongside the promising potentials, there are also considerable critical issues 93,94. For example, the levels of circulating DNA fragments derived from the tumour (cfDNA) represent a small fraction (up to 10%) of the total cfDNA and variations between patients related to cancer stage, tumour vascularisation, tumour burden and apoptosis rate can compromise the accuracy of the tests 92,95-97.

Conclusions

The traditional role of biopsy is evolving; in the future, a single procedure will allow to define the histology of a tumour and to identify predictive and prognostic biomarkers. The data emerging from this review of the literature, even if heterogeneous and fragmented, suggest that the evaluation of markers on biopsy samples, mainly the DOI and growth characteristics of the invasive front (INF, TB) could provide useful indications for planning tailored treatments. Minimally invasive procedures such as liquid biopsy are also promising, although only preliminary results are currently available which require further confirmation. Further clinical studies are needed to standardise and validate clinical and imaging evaluation, surgical and liquid biopsy technique, sampling of specimens, and choice of biomarkers and their assessment before introducing these diagnostic and predictive modalities in daily clinical practice.

Figures and tables

Figure 1.Flow chart showing selection of papers.

Marker Definition Notes
Depth of Invasion (DOI) DOI is measured from the level of the basement membrane of the closest adjacent normal mucosa. A “plumb line” is dropped from this plane to the deepest point of tumour invasion 38,42 Included in T1-3 Categories for Oral Cavity Cancer, TNM Staging Manual 8th Edition 37The T category increases with every interval of 5 mm
Tumour Thickness (TT) TT is measured from the surface of the tumour to the deepest point of invasion. In exofitic and ulcerated lesions TT is measured from the imaginary line reconstructing the intact mucosa to the deepest point of invasion 42,48TT1 is measured from the level of adjacent mucosa to the deepest point of tumour invasion 49TT2 is the distance from the bottom of most adjacent dysplastic abnormal rete pegs to the deepest point of invasion 49TT3 is measured as distance from the epithelial junction of the most adjacent dermal papillae to the deepest point of tumour invasion 49 As defined before publication of the 8th TNM Edition 42,48 or defined as an alternative to the proposed classification 49Rete pegs: epithelial extensions in the connective tissue underlying the mucosa 49
Radiological Depth of Invasion (rDOI) rDOI is measured by drawing a perpendicular line from the reference line to the deepest point of the tumour 50 Radiological definition of DOI is also reported in TNM 8th Edition 37. The reference line connects the junction of the tumour surfaces and of the normal mucosa on both sides 50
Tumour Budding (TB) TB is defined as the presence of either isolated single cells or small-cell clusters comprising fewer than five cells scattered in the stroma ahead of the invasive tumour front 15,45,48,51-54 TB is speculated to be the result of interactions between cancer cells and tumour microenvironments. It is expression of loss of cohesion and active invasive cellular movement 55. It is considered the first step in metastasis of a solid tumour
Pattern of Invasion (POI) POI at the tumour-host interface of oral cancer is graded 1 to 5. Tumour dispersion is assessed at the advancing tumour edge.The most common WPOI-5 phenotype is tumor dispersion through soft tissue. Dispersed extratumoral peri-neural invasion, or extratumoral lymphovascular invasion, also can qualify for classification as WPOI-5 37
Worst Pattern of Invasion (WPOI) WPOI-5 (POI Grade 5) consists of dispersed, discontinuous growth pattern, with a defined tumour dispersion cutoff of 1 mm 37,54
Tumour-Stroma Ratio (TSR) TSR defines the interactions between cancer cells and intra-tumoural stroma, which is the main component of the microenvironment 46,56,57 These interactions are important for both cancer initiation and progression: the proportion of this stroma acts as a key regulator in cancer biology and could provide strategies for biological cancer treatment
Peri Neural Invasion (PNI) PNI is defined as the tumour cell infiltration in any layer of the nerve sheath or tumour in close proximity involving more than one-third of the nerve circumference 49 PNI should be subclassified as either intratumoral or extratumoral, and as focal or multifocal. Extensive multifocal PNl is usually extratumoral and frequently associated with a “strand-like” tumour phenotype 37
Lympho Vascular Invasion (LVI) LIV is defined as the detection of tumour epithelial cells within or attached to the endothelial cell lining of the vascular space 49 LIV should be reported as either intratumoral or extratumoral, as well as focal or multifocal 37
Table I.Pathological markers predictive of loco-regional recurrence and prognosticators.
Author Study design Time period Site Stage or TNM teatment No. patients Biopsy technique Aim of the study Markers Results Notes
Almangush, 2018 58 Retrospective 1981-2016 Mobile oral tongueStage I-II = 81%Stage III-IV = 19%Surgery = 100% 100Incisional biopsy Analysis of the sensitivity and specificity of BD scores on biopsy and postoperative samples TBTTBD model score BD model scorepreo-op vs post-op- Sensitivity 59.1%- Specificity = 100%- Agreement pre/post op = 83%Depth values (mm)pre-op (mean) = 4.1 (0.5-10 mm)post-op (mean) = 6.3 (0.5-23 mm) 17% = non-representative biopsies (badly fragmented, too superficial, technical artifacts)TB and TT (Tab. I)Budding model score (Tab. II)
Seki, 2016 59 Retrospective2009-2013 Tongue, floor of the mouthT1 = 43%T2-T4 = 57%Preop CT = 52%Surgery = 48% 91Not described Detection of histopathologicparameters predictive forlympho nodal metastases in preoperative biopsy specimens Histologic GradingTTINFLIVBudding score Tumour depth: higher in resection specimens than in biopsy samples. It is predictive of lymph node metastases on resection specimens, not on biopsy samplesBudding score: good correlation between biopsy and postop specimensBD = > 3 and Tumour depth > 3 mm significantly predictive of lymph node metastases, OS and RFSBD = > 3 and INFa/INFb: predictive of early or late lymph node metastases or late in many cases TT (Tab. I)INF (Tab. I)Budding score (Tab. IIb)Comparison between TB, TT, and INF status on lymph node metastases was assessed on 33 patients who underwent resection and neck dissection
Seki Soda, 2019 60 Retrospective2009-2015 Oral cavityStage not reportedPreop CT = 57%Surgery = 43% 248Incisional biopsy Evaluation of relashionship between tumour budding score in biopsy and resected specimens and the effect of pre-op chemotherapy on tumour budding Budding score Mean budding scores: no significant differences in both groups; but tended to increase in resected patients without preop CTPreoperative CT-S1 treatment is effective for suppressing relapse and lowering the budding scores Budding score (Tab. II)
Seki, 2017 48 Retrospective2009-2014 Mobile tongue, buccal mucosa, palate, lipT1-T2 = 76%T3-T4 = 24%Preop CT and resection +/- ND = 53%Resection+/- ND = 47% 209Incisional biopsy Evaluation of the relashionship between tumour budding in biopsy specimen and lymphnode metastases TBINFTumour gradeTT TB => 3 predictive of Lymphnode metastases and poor prognosisP < 0.01 (mainly T1-T2 cN0)The budding score (high) together with INF and LIV were found to be independent risk factors for lymphnode metastases at multivariate analysesStrong correlation (p < 0.01) between the budding score and tumour grade, tumour depth, INF, and blood vessels invasionTB in early OSCC can be used as a reliable parameter for stratifying patients with different risks of lymph node metastases Authors describe biopsy technique;Tumour buddingcan be detected using routine haematoxylin andeosin-stained slides orcytokeratin-stained slides.Results may differ significantly depending on the staining used
Nayanar, 2019 61 Retrospective2014-2016 Oral cavity (lips, buccal mucosa, tongue, hard palate, retromolar trigone, floor of the mouth)Stage II = 48%Stage III = 20%Stage IV = 32%Wide excision with Neck dissection 160Incision or wedge biopsy Identification of clinical and histopathological predictors of lymphnode metastasesDevelopment of a predictive scoring system Shape of rete pegsPattern of invasionDepth of invasion (TT)Histologic differentiationSite of cancer Risk score (4-18)Score 4: predictive of N+ = 5%Score 18: predictive of N+ = 91% The risk score needs to be tested and validated in other patient populationsRete pegs (Tab. I)Risk score (Tab. II)
Sahoo, 2020 49 Retrospective2014-2016 Oral cavity (GB sulcus, tongue, floor of the mouth, retromolar and maxilla)cT1/T2Resection and elective neck dissection 150Excisional biopsy Comparison of prognostic performance of TT and of DOI (TT2 and TT3) in predicting lymphnode metastases.Predictive potential of PNI and LVI TT1, TT2, TT3, DOIPNI, LVI TT2 and LVI correlate with lymph node metastases TT1, TT2, TT3, DOI (Tab. I)
Table II.Evaluation of pathological markers on biopsy and postoperative samples in the 6 papers selected.
Author Definition Score
Almangush, 2018 58 BD Model Score (BDM):N° of buds and depth of invasive front (IF, mm) 0: < 5 buds at the IF and depth < 4 mm1: =/> 5 buds at the IF or depth > 4 mm2: =/> 5 buds at the IF and depth > 4mm
Seki, 2017 48 Tumour Budding Score (TBS):Number of buds Low: < 3 cellsIntermediate: 3-4 cellsHigh: => 5 cells
Seki, 2017 48 INF (Infiltrative Pattern):Cancer growth and mode of invasion INFa: Expanding growth with a distinct border from surrounding tissue;INFb: Intermediate pattern between INFa and INFc;INFc: Infiltrative growth with no distinct border from the surrounding tissue
Nayanar, 2019 61 Risk Score:Sum of single score of clinical and pathologic parameters Shape of rete pegs: Slender and fused = 1, Bulbous and uniform = 2; Irregular = 3Pattern of invasion: Pushing = 1; Minimally invasive = 2; Frankly invasive = 5Depth of invasion (TT, mm): 0-3 = 1; > 3 = 3Histogic differentiation: Well = 1; Moderate = 3; Poor = 5Site of cancer: Buccal mucosa = 0; Tongue = 1; Others = 2
Table III.Different scores for prognostic evaluation.

References

  1. Ferlay J, Colombet M, Soerjomaratam I. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 2019; 144:1941-1953. DOI
  2. Wu K, Wei J, Liu Z. Can pattern and depth of invasion predict lymph node relapse and prognosis in tongue squamous cell carcinoma. BMC Cancer. 2019; 9:714. DOI
  3. Fitzmaurice C, Abate D, Abbasi N. Global burden of disease cancer collaboration global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability and disability-adjusted life-years for 29 cancer groups. 1990 to 2017. Jama Oncol. 2019; 5:1749-1768. DOI
  4. Chen SW, Zhang Q, Guo ZM. Trends in clinical features and survival of oral cavity cancer: fifty years of experience with 3,362 consecutive cases from a single institution. Cancer Manag Res. 2018; 10:4523-4535. DOI
  5. Paderno A, Morello R, Piazza C. Tongue carcinoma in young adults: a review of the literature. Acta Otorhinolaryngol Ital. 2018; 38:175-180. DOI
  6. Kharwala SS, Hatsukami DK, Stepanov I. Patterns of tobacco cessation attempts and symptoms experienced among smokers with head and neck squamous cell carcinoma. JAMA Otolaryngol Head Neck Surg. 2018; 144:477-482. DOI
  7. Bala MM, Strzeszynski L, Topor-Madry R. Mass media interventions for smoking cessation in adults. Cochrane Database Syst Rev. 2013; 6:CD004704. DOI
  8. Challapalli SD, Simpson MC, Adjei Boakye E. Head and neck squamous cell carcinoma in adolescents and young adults: survivorship patterns and disparities. J Adolesc Young Adult Oncol. 2018; 7:472-479. DOI
  9. Ng HJ, Iyer NG, Tan MH. Changing epidemiology of oral squamous cell carcinoma of the tongue: a global study. Head Neck. 2017; 39:297-304. DOI
  10. Hussein AA, Helder MN, de Visscher JG. Global incidence of oral and oropharynx cancer in patients younger than 45 years versus older patients: a systematic review. Eur J Cancer. 2017; 82:115-127. DOI
  11. Yete S, D’Souza W, Saranath D. High-risk human papillomavirus in oral cancer: clinical implications. Oncology. 2018; 94:133-141. DOI
  12. Hübbers CU, Akgül B. HPV and cancer of the oral cavity. Virulence. 2015; 6:244-248. DOI
  13. Russo D, Merolla F, Varricchio S. Epigenetics of oral and oropharyngeal cancers (Review). Biomed Rep. 2018; 9:275-283. DOI
  14. Lam DK, Schmidt BL. Current therapy in oral and maxillofacial surgery. Elsevier Inc: Amsterdam; 2012. DOI
  15. Zhu Y, Liu H, Xie N. Impact of tumour budding in head and neck squamous cell carcinoma: a meta-analysis. Head Neck. 2019; 41:542-550. DOI
  16. Naruse T, Yanamoto S, Matsushita Y. Cetuximab for the treatment of locally advanced and recurrent/metastatic oral cancer: an investigation of distant metastasis. Mol Clin Oncol. 2016; 5:246-252. DOI
  17. Weinstein JN, Collisson EA, Mills GB. The Cancer Genome Atlas pan-cancer analysis project. Nat Genet. 2013; 45:113-120. DOI
  18. Leichsering J, Horak P, Kreutzfeldt S. Variant classification in precision oncology. Int J Cancer. 2019; 145:2996-3010. DOI
  19. Sankar PL, Parker LS. The Precision Medicine Initiative’s All of Us Research Program: an agenda for research on its ethical, legal, and social issues. Genet Med. 2017; 19:743-750. DOI
  20. Amin MB, Greene FL, Edge SB. The Eighth Edition AJCC Cancer Staging Manual: continuing to build a bridge from a population-based to a more “personalized” approach to cancer staging. Ca Cancer J Clin. 2017; 67:93-99. DOI
  21. NCCN Clinical Practice Guidelines in Oncology. Head and Neck Cancers v 1-2020 - February 12, 2020.Publisher Full Text
  22. Orlandi E, Alfieri S, Simon C. Treatment challenges in and outside a network setting: head and neck cancers. Eur J Surg Oncol. 2019; 45:40-45. DOI
  23. Orlandi E, Licitra L. Personalized medicine and the contradictions and limits of first-generation deescalation trials in patients with Human Papillomavirus-positive oropharyngeal cancer. JAMA Otolaryngol Head Neck Surg. 2018; 144:99-100. DOI
  24. Coyle KM, Boudreau JE, Marcato P. Genetic mutations and epigenetic modifications driving cancer and informing Precision Medicine. Biomed Res Int. 2017; 2017:9620870. DOI
  25. Garraway LA, Verweij J, Ballman KV. Precision oncology: an overview. J Clin Oncol. 2013; 31:1803-1805. DOI
  26. Zhong L, Liu Y, Wang K. Biomarkers: paving stones on the road towards the personalized precision medicine for oral squamous cell carcinoma. BMC Cancer. 2018; 18:911. DOI
  27. Nor JE, Gutkind JS. Head and neck cancer in the new era of precision medicine. J Dent Res. 2018; 97:601-602. DOI
  28. Giefing M, Wierzbicka M, Szyfter K. Moving towards personalized therapy in head and neck squamous cell carcinoma through analysis of next generation sequencing data. Eur J Cancer. 2016; 55:147-157. DOI
  29. Bernier J, Domenge C, Ozsahin M. Postoperative irradiation with or without concomitant chemotherapy for locally advanced head and neck cancer. N Engl J Med. 2004; 350:1945-1952. DOI
  30. Cooper JS, Pajak TF, Forastiere AA. Postoperative concurrent radiotherapy and chemotherapy for high-risk squamous-cell carcinoma of the head and neck. N Engl J Med. 2004; 350:1937-1944. DOI
  31. Bernier J, Cooper JS, Pajak TF. Defining risk levels in locally advanced head and neck cancers: a comparative analysis of concurrent postoperative radiation plus chemotherapy trials of the EORTC (#22931) and RTOG (#9501). Head Neck. 2005; 27:843-850. DOI
  32. Cooper JS, Zhang Q, Pajak TF. Long-term follow-up of the RTOG 9501/intergroup phase III trial: postoperative concurrent radiation therapy and chemotherapy in high-risk squamous cell carcinoma of the head and neck. Int J Radiat Oncol Biol Phys. 2012; 84:1198-1205. DOI
  33. Chen WC, Lai CH, Fang CC. Identification of high-risk subgroups of patients with oral cavity cancer in need of postoperative adjuvant radiotherapy or chemo-radiotherapy. Medicine (Baltimore). 2016; 95(22):e3770. DOI
  34. Fan KH, Chen YC, Lin CY. Postoperative radiotherapy with or without concurrent chemotherapy for oral squamous cell carcinoma in patients with three or more minor risk factors: a propensity score matching analysis. Radiat Oncol. 2017; 12:184. DOI
  35. Cheraghlou S, Schettino A, Zogg CK. Changing prognosis of oral cancer: an analysis of survival and treatment between 1973 and 2014. Laryngoscope. 2018; 128:2762-2769. DOI
  36. Ribeiro IP, Caramelo F, Esteves L. Genomic and epigenetic signatures associated with survival rate in oral squamous cell carcinoma patients. J Cancer. 2018; 9:1885-1895. DOI
  37. Amin MB, Edge S, Greene F. AJCC Cancer Staging Manual. Springer International Publishing: New York; 2017.
  38. Lydiatt WM, Patel SG, O’Sullivan B. Head and neck cancers. Major changes in the American Joint Committee on Cancer eighth edition Cancer Staging Manual. Ca Cancer J Clin. 2017; 67:122-137. DOI
  39. Fakih AR, Rao RS, Borges AM. Elective versus therapeutic neck dissection in squamous carcinoma of tongue a prospective randomized trial. Am J Surg. 1989; 158:309-313. DOI
  40. Fukano H, Matsuura H, Hasegawa Y. Depth of invasion as a predictive factor for cervical lymph node metastasis in tongue carcinoma. Head Neck. 1997; 19:205-10. DOI
  41. Kukreja P, Parekh D, Roy P. Practical challenges in measurement of invasion in oral squamous cell carcinoma: pictographical documentation to improve consistency of reporting per the AJCC 8th Edition recommendations. Head Neck Pathol. 2020; 14:419-427. DOI
  42. Almangush A, Bello IO, Coletta RD. For early-stage oral tongue cancer, depth of invasion and worst pattern of invasion are the strongest pathological predictors for locoregional recurrence and mortality. Virchows Arch. 2015; 467:39-46. DOI
  43. Larson AR, Kemmer J, Formeister E. Beyond depth of invasion: adverse pathologic tumour features in early oral tongue squamous cell carcinoma. Laryngoscope. 2020; 130:1715-1720. DOI
  44. den Toom IJ, Janssen LM, van Es RJJ. Depth of invasion in patients with early stage oral cancer staged by sentinel node biopsy. Head Neck. 2019; 41:2100-2106. DOI
  45. Almangush A, Pirinen M, Heikkinen I. Tumour budding in oral squamous cell carcinoma: a meta-analysis. Br J Cancer. 2018; 118:577-586. DOI
  46. Almangush A, Heikkinen I, Bakhti N. Prognostic impact of tumour –stroma ratio in early-stage oral tongue cancers. Histopathology. 2018; 72:1128-1135. DOI
  47. Hori Y, Kubota A, Yokose T. Association between pathological invasion patterns and late lymph node metastases in patients with surgically treated clinical N0 early tongue carcinoma. Head Neck. 2020; 42:238-243. DOI
  48. Seki M, Sano T, Yokoo S. Tumour budding evaluated in biopsy specimens is a useful predictor of prognosis in patients with cN0 early stage oral squamous cell carcinoma. Histopathology. 2017; 70:869-879. DOI
  49. Sahoo A, Panda S, Mohanty N. Perineural, lymphovascular and depths of invasion in extrapolating nodal metastasis in oral cancer. Clin Oral Investig. 2020; 24:747-755. DOI
  50. Vidiri A, Panfili M, Boellis A. The role of MRI-derived depth of invasion in staging oral tongue squamous cell carcinoma: inter-reader and radiological-pathological agreement. Ann Radiol. 2020; 61:344-352. DOI
  51. Prall F. Tumour budding in colorectal carcinoma. Histopathology. 2007; 50:151-162. DOI
  52. Jensen DH, Dabelsteen E, Specht L. Molecular profiling of tumour budding implicates TGFb-mediated epithelial-mesenchymal transition as a therapeutic target in oral squamous cell carcinoma. J Pathol. 2015; 236:505-516. DOI
  53. Attramadal CG, Kumar S, Boysen ME. Tumour budding, EMT and cancer stem cells in T1-2/N0 oral squamous cell carcinomas. Anticancer Res. 2015; 35:6111-6120.
  54. Shimizu S, Miyazaki A, Sonoda T. Tumour budding is an independent marker in early stage oral squamous cell carcinoma: with special reference to the mode of invasion and worst pattern of invasion. Plos One. 2018; 13:e0195451. DOI
  55. Almangush A, Youssef O, Pirinen H. Does evaluation of tumour budding in diagnostic biopsies have a clinical relevance? A systematic review. Histopathology. 2019; 74:536-544. DOI
  56. Wang K, Ma W, Wang J. Tumour-stroma ratio is an independent predictor for survival in esophageal squamous cell carcinoma. J Thorac Oncol. 2012; 7:1457-1461. DOI
  57. Wu J, Liang C, Chen M. Association between tumour-stroma ratio and prognosis in solid tumour patients: a systematic review and meta-analysis. Oncotarget. 2016; 7:68954-68965. DOI
  58. Almangush A, Leivo I, Siponen M. Evaluation of the budding and depth of invasion (BD) model in oral tongue cancer biopsies. Virchows Arch. 2018; 472:231-236. DOI
  59. Seki M, Sano T, Yokoo S. Histologic assessment of tumour budding in preoperative biopsies to predict nodal metastasis in squamous cell carcinoma of the tongue and floor of the mouth. Head Neck. 2016; 38:E1582-1590. DOI
  60. Seki-Soda M, Sano T, Koshi H. Histopathological changes in tumour budding between biopsy and resected specimens from patients treated with preoperative S-1 chemotherapy for oral cancer. J Oral Pathol Med. 2019; 48:880-887. DOI
  61. Nayanar SK, Tripathy JP, Duraisamy K. Prognostic efficiency of clinicopathologic score to predict cervical ymph node metastasis in oral squamous cell carcinoma. J Oral Maxillofac Pathol. 2019; 23:36-42. DOI
  62. Dirven R, Ebrahimi A, Moeckelmann N. Tumour thickness versus depth of invasion – Analysis of the 8th edition American Joint Committee on Cancer Staging for oral cancer. Oral Oncol. 2017; 74:30-33. DOI
  63. Merlano MC, Abbona A, Denaro N. Knowing the tumour microenvironment to optimise immunotherapy. Conference Paper. Acta Otorhinolaryngol Ital. 2019; 39:18-21. DOI
  64. Dhanda J, Uppal N, Chowlia H. Features and prognostic utility of biopsy in oral squamous cell carcinoma. Head Neck. 2016; 38:E1857-1862. DOI
  65. Kolenda T, Przybyla W, Kapalczynska M. Tumour microenvironment - Unknown niche with powerful therapeutic potential. Rep Pract Oncol Radiother. 2018; 23:143-153. DOI
  66. Leite CF, Silva KDD, Horta MCR. Can morphological features evaluated in oral cancer biopsies influence in decision-making? A preliminary study. Pathol Res Pract. 2020; 216:153138. DOI
  67. Elseragy A, Salo T, Coletta RD. A proposal to revise the histopathologic grading system of early oral tongue cancer incorporating tumour budding. Am J Surg Pathol. 2019; 43:703-709. DOI
  68. Heerema MGJ, Melchers LJ, Roodenburg JNL. Reproducibility and prognostic value of pattern of invasion scoring in low-stage oral squamous cell carcinoma. Histopathology. 2016; 68:338-397. DOI
  69. Berdugo J, Thompson LDR, Purgina B. Measuring depth of invasion in early squamous cell carcinoma of the oral tongue: positive deep margin, extratumoural perineural invasion, and other challenges. Head Neck Pathol. 2019; 13:154-161. DOI
  70. Seppala M, Tervo S, Pohjola K. The association and prognostic relevance of cancerous inhibitor of protein phosphatase 2A and inflammation in tongue squamous cell carcinoma. APMIS. 2015; 123:1007-1015. DOI
  71. Lundqvist L, Stenlund H, Laurell G. The importance of stroma inflammation in squamous cell carcinoma of the tongue. J Oral Pathol Med. 2012; 41:379-383. DOI
  72. Alsaffar HA, Goldstein DP, King EV. Correlation between clinical and MRI assessment of depth of invasion in oral tongue squamous cell carcinoma. J Otolaryngol Head Neck Surg. 2016; 45:61. DOI
  73. Jayasankaran SC, Chelakkot PG, Karippaliyil M. Magnetic resonance imaging: a predictor of pathological tumour dimensions in carcinoma of the anterior two-thirds of tongue. Prospective evaluation. Indian J Cancer. 2017; 54:508-513. DOI
  74. Morand GB, Ikenberg K, Vital DG. Preoperative assessment of CD44-mediated depth of invasion as predictor of occult metastases in early oral squamous cell carcinoma. Head Neck. 2019; 41:950-958. DOI
  75. Goel V, Parihar PS, Parihar A. Accuracy of MRI in prediction of tumour thickness and nodal stage in oral tongue and gingivobuccal cancer with clinical correlation and staging. J Clin Diagn Res. 2016; 10:TC01-5. DOI
  76. Mao MH, Wang S, Feng ZE. Accuracy of magnetic resonance imaging in evaluating the depth of invasion of tongue cancer. A prospective cohort study. Oral Oncol. 2019; 91:79-84. DOI
  77. Franceschi L, Santo JM, Abreu AM. Staging of oral cavity cancer in the 8th edition of the TNM classification: the role of computed tomography in the assessment of depth of invasion and extranodal extension. Arch Head Neck Surg. 2018; 47:e0869. DOI
  78. Chen CH, Hsu MY, Jiang RS. Shrinkage of head and neck cancer specimens after formalin fixation. J Chin Med Assoc. 2012; 75:109-113. DOI
  79. Klein Nulent TJW, Noorlag R, Van Cann EM. Intraoral ultrasonography to measure tumour thickness of oral cancer: a systematic review and meta-analysis. Oral Oncol. 2018; 77:29-36. DOI
  80. Tarabichi O, Bulbul MG, Kanamuri VV. Utility of intraoral ultrasound in managing oral tongue squamous cell carcinoma: systematic review. Laryngoscope. 2019; 129:662-670. DOI
  81. Iida Y, Kamijo T, Kusafuka K. Depth of invasion in superficial oral tongue carcinoma quantified using intraoral ultrasonography. Laryngoscope. 2018; 128:2778-2782. DOI
  82. Rizzo S, Botta F, Raimondi S. Radiomics: the facts and the challenges of image analysis. Eur Radiol Exp. 2018; 2:36. DOI
  83. Haider SP, Burtness B, Yarbrough WG. Applications of radiomics in precision diagnosis, prognostication and treatment planning of head and neck squamous cell carcinomas. Cancers Head Neck. 2020; 5:6. DOI
  84. Mes SW, van Velden FHP, Peltenburg B. Outcome prediction of head and neck squamous cell carcinoma by MRI radiomic signatures. Eur Radiol. 2020; 30:6311-6321. DOI
  85. Pan X, Zhang T, Yang Q. Survival prediction for oral tongue cancer patients via probabilistic genetic algorithm optimized neural network models. Br J Radiol. 2020; 93:20190825. DOI
  86. Romeo V, Cuocolo R, Ricciardi C. Prediction of tumor grade and nodal status in oropharyngeal and oral cavity squamous-cell carcinoma using a radiomic approach. Anticancer Res. 2020; 40:271-280. DOI
  87. Castagnola M, Scarano E, Passali GC. Salivary biomarkers and proteomics: future diagnostic and clinical utilities. Acta Otorhinolaryngol Ital. 2017; 37:94-101. DOI
  88. Xiao C, Song F, Zheng YL. Exosomes in head and neck squamous cell carcinoma. Front Oncol. 2019; 9:894. DOI
  89. Cree IA, Uttley L, Buckley Woods H. The evidence base for circulating tumour DNA blood-based biomarkers for the early detection of cancer: a systematic mapping review. BMC Cancer. 2017; 17:697. DOI
  90. Mazunder S, Datta S, Ray JG. Liquid biopsy: miRNA as a potential biomarker in oral cancer. Cancer Epidemiol. 2019; 58:137-145. DOI
  91. Lousada-Fernandez F, Rapado-Gonzalez O, Lopez-Cedrun JL. Liquid biopsy in oral cancer. Int J Mol Sci. 2018; 19:1704. DOI
  92. Arneth B. Update on the types and usage of liquid biopsies in the clinical setting: a systematic review. BMC Cancer. 2018; 18:527. DOI
  93. Ribeiro IP, Barbosa de Melo J, Carreira IM. Head and neck cancer: searching for genomic and epigenetic biomarkers in body fluids – the state of art. Mol Cytogenet. 2019; 12:33. DOI
  94. Hussein AA, Forouzanfar T, Bloemena E. A review of the most promising biomarkers for early diagnosis and prognosis prediction of tongue squamous cell carcinoma. Br J Cancer. 2018; 119:724-736. DOI
  95. Economopoulou P, Kotsanti I, Kyrodimos E. Liquid biopsy: an emerging prognostic and predictive tool in head and neck squamous cell carcinoma (HNSCC). Focus on circulating tumour cells (CTCs). Oral Oncol. 2017; 74:83-89. DOI
  96. Spector ME, Farlow JL, Haring CT. The potential for liquid biopsies in head and neck cancer. Discover Med. 2018; 25:251-257.
  97. Van Ginkel JH, Huibers MMH, Noorlag R. Liquid biopsy: a future tool for posttreatment surveillance in head and neck cancer?. Pathobiology. 2017; 84:115-120. DOI

Affiliations

Roberto Bruschini

Division of Otolaryngology and Head & Neck Surgery, European Institute of Oncology IRCCS, Milan, Italy

Fausto Maffini

Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy

Fausto Chiesa

Division of Otolaryngology and Head & Neck Surgery, European Institute of Oncology IRCCS, Milan, Italy

Daniela Lepanto

Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy

Rita De Berardinis

Division of Otolaryngology and Head & Neck Surgery, European Institute of Oncology IRCCS, Milan, Italy

Francesco Chu

Division of Otolaryngology and Head & Neck Surgery, European Institute of Oncology IRCCS, Milan, Italy

Marta Tagliabue

Division of Otolaryngology and Head & Neck Surgery, European Institute of Oncology IRCCS, Milan, Italy

Gioacchino Giugliano

Division of Otolaryngology and Head & Neck Surgery, European Institute of Oncology IRCCS, Milan, Italy

Mohssen Ansarin

Division of Otolaryngology and Head & Neck Surgery, European Institute of Oncology IRCCS, Milan, Italy

Copyright

© Società Italiana di Otorinolaringoiatria e chirurgia cervico facciale , 2021

  • Abstract viewed - 1575 times
  • PDF downloaded - 689 times