Next Article in Journal
Clinical Utility of Personalized Serum IgG Subclass Ratios for the Differentiation of IgG4-Related Sclerosing Cholangitis (IgG4-SC) from Primary Sclerosing Cholangitis (PSC) and Cholangiocarcinoma (CCA)
Previous Article in Journal
Laser Doppler Flowmetry Combined with Spectroscopy to Determine Peripheral Tissue Perfusion and Oxygen Saturation: A Pilot Study in Healthy Volunteers and Patients with Peripheral Arterial Disease
Previous Article in Special Issue
IL4Rα and IL13Rα1 Are Involved in the Development of Human Gallbladder Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Precision Medicine in Head and Neck Cancers: Genomic and Preclinical Approaches

1
Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
2
Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
3
Department of Biology, University of Pisa, 55126 Pisa, Italy
4
Maxillofacial Surgery Unit, “Bufalini Hospital”, AUSL Romagna, 47521 Cesena, Italy
5
Pathology Unit, “Bufalini” Hospital, AUSL Romagna, 47521 Cesena, Italy
6
Otolaryngology and Head-Neck Surgery Unit, Department of Head-Neck Surgeries, Morgagni Pierantoni Hospital, AUSL Romagna, 47121 Forlì, Italy
7
Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
8
Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 8 April 2022 / Revised: 11 May 2022 / Accepted: 19 May 2022 / Published: 24 May 2022

Abstract

:
Head and neck cancers (HNCs) represent the sixth most widespread malignancy worldwide. Surgery, radiotherapy, chemotherapeutic and immunotherapeutic drugs represent the main clinical approaches for HNC patients. Moreover, HNCs are characterised by an elevated mutational load; however, specific genetic mutations or biomarkers have not yet been found. In this scenario, personalised medicine is showing its efficacy. To study the reliability and the effects of personalised treatments, preclinical research can take advantage of next-generation sequencing and innovative technologies that have been developed to obtain genomic and multi-omic profiles to drive personalised treatments. The crosstalk between malignant and healthy components, as well as interactions with extracellular matrices, are important features which are responsible for treatment failure. Preclinical research has constantly implemented in vitro and in vivo models to mimic the natural tumour microenvironment. Among them, 3D systems have been developed to reproduce the tumour mass architecture, such as biomimetic scaffolds and organoids. In addition, in vivo models have been changed over the last decades to overcome problems such as animal management complexity and time-consuming experiments. In this review, we will explore the new approaches aimed to improve preclinical tools to study and apply precision medicine as a therapeutic option for patients affected by HNCs.

Graphical Abstract

1. Introduction

Head and neck cancer (HNC) is the sixth most common non-skin cancer worldwide, resulting in around 900,000 new cases each year, with around 460,000 new deaths (50% mortality rate) [1,2]. Squamous cell carcinomas (SCCs) constitute around 90% of total HNCs, and represent a subset of malignancies which arise in the squamous epithelial cells located in the mucosa of the oropharynx, hypopharynx, nasopharyngeal, oral and nasal cavity, larynx, and salivary glands [2,3]. Alcohol, tobacco, genetic mutations, viral infections of human papilloma virus (HPV) and Epstein-Barr virus (EBV) represent the major risk factors of HNCs [2,3,4,5]. HPV-positive SCCs mainly arise in the oropharyngeal tissues, and show better clinical outcomes and different pathological characteristics compared to their HPV-negative counterparts [6,7]. In contrast, EBV-positive SCCs are associated with nasopharyngeal tumours, especially in east and southeast Asia, where this type of carcinoma is endemic [8,9].
HNSCC (head and neck squamous cell carcinoma) carcinogenesis includes a series of pathological steps involving different kinds of cell types, according to the origin site of the neoplasia. Usually, the disease arises with the development of epithelial cells hyperplasia, which results in mild, moderate, or severe dysplasia [8]. The progression to neoplasia can be driven by different cell types, given the heterogeneous nature of HNSCCs. Carcinogenesis is driven by gene mutations and chromosomal instability, which could play a different role in the case of a presence/absence of HPV or EBV infection [10]. Thus, mutations in TP53, PIK3CA, FAT1, and NOTCH, the amplification of CCND1, and a loss of CDKN2A represent the most common genetic alterations in HPV-negative HNSCCs [10,11]. On the other hand, HPV-positive HNSCCs show different primary genetic alterations, including the amplification of E2F1, a loss of TRAF3, and a higher rate of PIK3CA mutations [10,11]. Tobacco and alcohol abuse represent the two principal risk factors for the onset of HPV-negative HNSCCs. Indeed, most of the chemicals present in tobacco have been demonstrated to have carcinogenic properties. Metabolic activation and inflammation induced by tobacco-related compounds are two events involved in the dysregulation of DNA repair processes, and are responsible for oncogenic transformation leading to genetic mutations and abnormalities [12]. Instead, alcohol is metabolized to acetaldehyde, a substance which is able to develop DNA adducts, as well as to synergize with tobacco to promote carcinogenesis [13,14]. Conversely, HPV-related HNSCCs show different pathological features in terms of gene expression, immune profiles, and genetic alterations. The 70% of HPV-positive HNSCCs arise in oropharynx tissues, showing lower percentages in other sites [15]. HPV-16 represents the most widespread virus strain implicated in HNCs. Among the nine proteins produced by the viral genome, E6 and E7 can induce malignant transformation [7,16]. E6 forms complexes with the tumour suppressor p53, promoting its ubiquitylation and subsequent proteasomal degradation [8,16]. The alteration of p53 activity is observed also in HPV-negative HNCs, but it has different causes, since it depends on mutations or deletions of the TP53 gene [17,18]. Moreover, E7 promotes the proteasomal degradation of retinoblastoma-associated protein (Rb1), a cell cycle regulator [18]. The degradation of Rb1 involves the upregulation of p16, an inhibitor of cyclin-dependent kinases (CDK), which is a well-known marker of HPV+ tumours [18,19].
However, other elements are involved in tumour progression. In this scenario, carcinogenesis is mainly induced by the oncogenic transformation of either adult stem cells or progenitor cells into cancer stem cells (CSCs) [20]. CSCs are a minority subpopulation of all tumour subclones distinguished by self-renewal properties and the capability to generate differentiated progeny, characterised by only transient proliferative status [21]. The cell adhesion molecule CD44 and aldehyde dehydrogenase (ALDH), an enzyme responsible for the oxidisation of aldehydes to carbolic acid, represent two markers of CTCs phenotypes in numerous solid tumours [21,22,23]. Depending on the HPV status, HNSCCs show differences in the plasticity and frequency of CTCs [24,25]. Several studies report on CSCs frequency in HNSCC tissues, but the results are still contradictory, describing different levels of stem cells in both HPV statuses [25,26]. The self-renewal properties, typical of these kinds of cells, are one of the causes of their resistance to standard therapies [27]. The characterization of HNSCC CSCs has attempted to identify biomarkers of chemo- and radioresistance. Chromatin regulator Bmi-1 and CD133 are only two examples of drug-sensitivity markers of CSCs, but their expression does not discriminate between HPV-positive and negative patients [28,29,30]. The higher efficacy of CSCs to repair the DNA damage is the main cause of radioresistance. Biomarkers such as RAD51, involved in the homologous recombination and repair of DNA, are highly expressed in HNSCC CSCs [22,31]. A promising marker involved in the higher radiotherapy efficacy typical of HPV-positive HNSCCs is represented by the fluorescent fusion protein ZsGreen-cODC. This protein, indicative of tumorigenic cells, is associated with mechanisms of radiation-induced dedifferentiation in HNSCC cell lines. Indeed, the ZsGreen-cODC-negative cells of HPV-negative cell lines showed higher re-expression of ZsGreen-cODC and higher dedifferentiation after radiation, all of which are features related to radioresistance [26].
Cell surface signalling receptors are other important biomarkers frequently used as targets for tumour treatment [32]. Among them, epithelial growth factor receptor (EGFR), an ErbB tyrosine kinase receptor family member, represents an important target for HNC treatment [33]. SCCs cells show a hyperactivation of EGFR signalling, a genetic event involved in the deregulation of several processes, including proliferation, angiogenesis, and metastasis development [8,34]. The EGFR expression is significantly downregulated with increasing distance from tumour microvessels [35]. Indeed, hypoxia has been identified as one of the major causes of treatment resistance of different tumours, including HNCs [35,36]. The hypoxic microenvironment induces changes in the DNA and growth properties of cancer cells, which are features associated with chemo- and radioresistance [35,36,37,38]. Oxygen represents an ideal radiosensitizer, due to its ability to induce the fixation of radical-induced DNA damage [38,39]. One of the major modulators of the adaptation response of tumour cells to low oxygen levels is hypoxia-inducible factor (HIF-1) [40]. HIF-1 activation promotes the transcription of over 100 downstream genes involved in tumour progression and survival [41]. For all of these reasons, HIF-1 and its related genes may be promising biomarkers of HNCs, as well as therapeutic targets for innovative therapies.

2. HNCs Clinical Approaches

HNCs are a heterogeneous group of malignancies that make a personalised therapeutic approach necessary for each patient. As a matter of fact, clinicians have to evaluate several parameters before therapy selection, such as tumour site, stage, natural history of the disease, biological characteristics, and clinical status. The curative approach is highly effective in preserving the organ’s function. In locoregional or local disease, therapeutic options include surgery, radiotherapy, and systemic chemotherapy [6,9]. Oral cavity cancers are commonly resected, while sites such as the larynx and pharynx are treated with standard or hypo-fractionated radiation [42]. To better preserve the tissue integrity with a minimally invasive approach, robotic or laser resection and the involvement of reconstructive techniques represent advanced surgical treatments, especially in larynx tumours [43]. Malignancies at a high risk of recurrence are commonly treated with postoperative radiation or chemoradiotherapy (CRT) in order to prevent relapses and to increase patient survival [9,44]. The involvement of surgical margins, extra-nodal extension, and perineural invasion are pathological features that describe the group of patients with a higher risk of recurrence. In these cases, radiotherapy and high doses of cisplatin-based chemotherapy improve patient survival [45]. Conversely, tumours at an advanced stage are commonly treated with CRT. The chemotherapeutic approach is based on high dose cisplatin-based monotherapy (100 mg/m2) or on low dose treatment in patients affected by renal dysfunctions or hearing loss [46]. Cetuximab, an epithelial growth factor receptor (EGFR) monoclonal antibody (mAbs), has been demonstrated to improve the effectiveness of radiation treatment [46,47]. The restricted group of EBV-positive nasopharyngeal cancers display not only treatment sensitivity, but also a higher risk of metastasis development [9]. An improvement in survival was demonstrated in EBV-positive patients treated with induction therapy or with additional chemotherapeutic regimens after CRT [48]. The combination of different drugs represents one of the front-line options of treatment in recurrent or metastatic HNSCC settings [49,50]. In this scenario, clinical and preclinical research needs to identify new treatment approaches. Recently, immunotherapy (IT) has been studied as an alternative option for the treatment of HNSCCs. Immunotherapeutic agents act directly on the patients’ immune system-activated cells to fight the disease. Programmed death 1 (PD1) immune check-point inhibitors have shown significant effectiveness compared to standard treatments [50,51]. In 2016, the Food and Drug Administration (FDA) approved two anti-PD-1 antibodies, nivolumab and pembrolizumab, for the treatment of recurrent or metastatic HNCs based on the results of the phase I KEYNOTE-12, phase II KEYNOTE-055, phase III KEYNOTE-040, and phase III CheckMate 141 trials [2,50,51]. Recently, the phase III trial KEYNOTE-048 trial showed a higher activity with pembrolizumab plus chemotherapy versus the EXTREME regiment (based on the triplet of carbo- or cisplatin, 5-fluorouracil and cetuximab) in all patients, as well as with pembrolizumab alone in front-line of recurrent/metastatic tumours expressing PD-L1 [52].
We have previously described the importance of hypoxia in treatment response. Several studies have explored the effect of hypoxic radiosensitizer to overcome the radioresistence mechanisms induced by low oxygen levels inside the tumour mass [53,54,55,56,57]. Among them, the DAHANCA 5 trial tested the hypoxia modifier nimorazole in combination with radiotherapy on HNSCC patients [58,59]. The study results revealed that the multi-treatment was only beneficial in HPV-negative patients, with respect to HPV-positive tumours. These results suggest a possible interaction between hypoxia and HPV status in the response to radiation effects [60].
In conclusion, the different behaviour, pathological features, HNSCCs sub-groups prognosis, patients’ comorbidities, treatment side effects (especially in multidrug regimens), and the limited clinical options available make the identification of new treatment options challenging for future perspective.

3. Precision Medicine in HNSCCs

Precision medicine represents an emerging therapeutic approach focused on tailoring treatment for each individual patient. This discipline is based on the concept that the prediction of clinical outcomes is more accurate if driven by analysis on the features of single tumour heterogeneity, rather than by comparison between cohorts of patients [61]. This concept has been developed in two major approaches (Figure 1). The first one is based on big OMICS data analysis to predict tumour behaviours. Here, multiple omics analysis, including epigenetics, genomics, metabolomics, and proteomics, are performed on tumour samples, in order to characterise the molecular and genetic properties of every single tumour [61,62]. The second approach consists of the development of personalised preclinical platforms using in vitro and in vivo techniques to test behaviours and drug sensitivity of patient-derived samples [63,64]. In this review, we will summarize the main techniques and models available to study and improve the efficacy of precision medicine in the treatment of HNSCCs.

3.1. Genomic Approach

The rise of next genome sequencing (NGS) techniques has allowed for the collection of large sets of omics data in a relatively simple and easily accessible way. The storage of genomic and clinical results represents an important opportunity for translating genomic information in clinical application. In this scenario, institutions worldwide have been contributed to the sharing of big data through international projects, such as the American Association for Cancer Research (AACR)’s project, Genomics Evidence Neoplasia Information Exchange (GENIE) [65], and the American Society of Clinical Oncology (ASCO)’s CANCERLINQ [66].
The genotype-drug matching approach has been demonstrated a valid strategy in the treatment of several tumour types. An example is the treatment of non-small cell lung cancer (NSCLC) patients expressing EGFR mutations with EGFR tyrosine kinase inhibitors (TKIs) [67]. The HNSCCs mutation landscape is largely known, but a challenging future objective is the matching of the genomic profile features with specific treatments. For this reason, new biomarker-driven trials have been designed to explore the efficacy of genotype–drug matching. The EORTC 1559 study (NCT03088059), the first international biomarker-driven study including HNSCCs, will compare the activity of targeted agents (including afatinib, palbociclib, niraparib, and entrectinib) with immunotherapy [68]. NGS technology will be used to analyse copy number variations and somatic mutations of 13 tumour suppressor genes and oncogenes; PTEN and p16 expression will be evaluated through immunohistochemistry. Patients will be divided into the different treatment arms based on genomic results and a pre-defined algorithm [68]. However, biomarker-driven clinical trials still have some limitations, such as narrow gene panels and restrictive matching algorithms, which entail a low matching rate [69]. Moreover, personalised medicine trials do not consider the possibility of combined strategies [70]. Currently, clinical trials based on personalised combined treatments are needed in HNSCCs.
The evolution of sequencing technologies has improved assay sensitivity and the analysis of biological samples. These achievements raise the opportunity to analyse the presence and the features of circulating tumour DNA (ctDNA) through liquid biopsy technologies [71]. Compared to tissue samples, the collection of liquid biopsies is less invasive, and can be performed easily and more frequently. Therefore, this type of approach allows for the monitoring of the presence of ctDNA over time, making ctDNA a potential biomarker [71]. Liquid biopsy is applied in several clinical evaluations, such as the prediction of patients’ outcomes and the selection of patients for personalised therapies [72]. In this context, the INSPIRE trial (NCT02644369) explores the correlation between ctDNA and the clinical outcomes of patients treated with pembrolizumab, which also includes HNSCCs [73]. These studies allow for a better patient selection for personalised treatments, especially for HNCs, in which patients are most likely to benefit from immunotherapy.

3.2. Multi-Omics Approaches

HNCs are diseases closely related to the tissue microenvironment and characterised by complex and heterogeneous molecular profiles [74,75]. For this reason, a single omic analysis represents a limited approach for describing the entire genetic and molecular profile. Over the last several years, innovative combinations of high throughput omics technologies, including transcriptomic, epigenomics, metabolomics, and proteomics, have been developed [76,77].
Among them, transcriptomic analysis explores total RNA transcripts present in the sample, such as mRNA, microRNA, and long noncoding RNA transcripts (lncRNAs). The cancer expression profile is not static, but it is affected by microenvironment and treatment stimuli [76,77,78]. The utility of transcriptomics in genotype-drug matching clinical trials represents one of the hot topics of oncology research. In this field, the WINTHER trial (NCT01856296) is an open non-randomized study matching mutation and transcriptomic profiles with the selection of therapies in metastatic cancers [79]. The two processes used, genotype- and transcriptome-matched drugs, showed similar response rates, ranging between 20% and 30% [79].
A further omic approach is represented by the study of the microbiome. Humans host thousands of microorganism species in several organs, especially in the oral cavity tissues [80]. The microbiome interacts with malignancy components through the release of metabolites and molecules, producing systemic and local effects [80,81]. The microbiome-related alterations modify cancer pathogenesis, progression, and drug response, including the efficacy of immune-checkpoint inhibitors [81]. Several studies have reported different microbiome compositions in the saliva of healthy and HNSCC patients, demonstrating a reduced risk of cancerogenesis related to the presence of specific bacteria strains [82,83,84]. The role of microbiome as a tumour biomarker and in the selection of patients that will benefit from immunotherapy is currently being studied for the NCT03686202 and NCT03838601 clinical trials [85].
The heterogeneity of the tumour microenvironment makes it necessary for researchers to discriminate between the contributions of the different cell types in cancer development. An innovative option is represented by digital spatial profile (DSP) techniques [86,87]. DSP is a high-plex spatial profiling method to spatially discern regions populated by healthy cells from those populated by malignant cells starting from formalin-fixed paraffin-embedded (FFPE) samples [86,87]. The detection of different targets is performed using oligonucleotide tags conjugated to oligo-labelled primary Abs or a cocktail of RNA probes for the in situ detection of proteins or RNA transcripts [88]. Then, a mix of fluorescently labelled Abs, markers of different cell populations, is used to visualise the targets of interest directly on the slides [88]. These systems allow for a fast and extended analysis simultaneously of up to 96 proteins or 1400 mRNA in about 48 h [86,89]. In the field of precision medicine, DSP represents a promising tool to predict prognosis and treatment responses through the detection of discrepancies in mRNA and protein expression patterns. These features are particularly attractive for the study of immunotherapy efficacy, as well as to discriminate between responder and non-responder patients. The application of DSP to the evaluation of immunotherapy response has already been explored in many solid and haematological tumours, such as non-small cell lung cancer (NSCLC), melanoma, and lymphoma [90,91,92]. Kulasinghe A. et al. was the first group of researchers to apply DSP technology to HNSCCs [87]. The study aimed to correlate immune and tumour markers to the clinical outcomes of a cohort of patients treated with nivolumab and pembrolizumab. The authors used pan-cytokeratin and two immune markers, CD3 and CD8, to discriminate between malignant- and immune-infiltrate regions inside the tumour tissues. Interestingly, the immune cell markers involved in CD8 T-cells infiltration were not predictive of outcomes in patients treated with immunotherapy. Instead, a panel of immune cell protein markers, CD4, CD68, CD45, CD44, and CD66b, was related to progressive disease. Due to the low number of patients included, the project represents an explorative study that needs to be confirmed on a higher number of patients.
Imaging technologies, such as magnetic resonance imaging (MRI), positron emission tomography (PET), and computed tomography (CT), assist clinicians in making therapeutic decisions. Innovation in image processing through artificial intelligence tools have increased the information and details available which are impossible to understand with common diagnostic technologies [93]. This innovative omics approach, named radiomics, has also been employed in HNC studies on medical imaging for treatment monitoring. Indeed, radiomics is used to identify treatment-resistant sub-volumes where radiobiological events, such as hypoxia, CSC accumulation, or a high proliferative rate, can alter the treatment efficacy [94,95]. In HNSCs, radiomics has been employed to understand the radiological differences between HPV-positive and negative patients. For example, HPV-positive tumours showed well-defined margins compared to the poorly defined boundaries of their negative counterparts [93,96]. This feature is typical of malignancies that tend to invade adjacent tissues, as described for HPV-negative HNSCC tumours.
In the field of radiomics, the technology involved in the extraction of spatial information, which is used afterwards to construct prediction model, is named dosiomics. Through machine learning techniques, dosiomics extracts characteristics from 3D radiotherapy dose distribution by intensity, textural, and shape-based characteristics that allow a high complexity description of the dose distribution [97,98]. Dosiomics is increasingly used to improve the prediction of clinical outcomes and side effects, such as the locoregional recurrence in HNSCCs after intensity-modulated radiation therapy (IMRT) or local control after radiotherapy [99].
Another multi-omics approach, which combines genomics and pharmacology to study the role of the genome in drugs response, is represented by pharmacogenomics. The correlation between genetic variants and clinical outcomes helps clinicians to stratify patients in responder and non-responder groups, to prevent side effects to specific treatments [100]. Pharmacogenomics take advantage of machine learning tools to process large amount of omics data with the aim to personalize patients’ treatment based on a genotype-guided chemotherapy [93,101]. The implementation and sharing of patients’ pharmacogenomics data are encouraged by many large-scale initiatives with the aim of developing detailed drug-gene interaction databases and platforms. Two examples are represented by the USA project Clinical Pharmacogenetics Implementation Consortium (CPIC), created by the Pharmacogenomics Knowledge Base (PharmGKB) and the National Institutes of Health (NIH), and the European Ubiquitous Pharmacogenomics program (U-PGx) [102,103].
Despite the improvements in knowledge on the molecular and genetic profiles of HNSCCs tumour tissues and immune infiltrate populations, the genomic and multi-genomic-based therapeutic decisions currently have a limited role in clinical practice. Future clinical trials and additional research are needed to better understand how to use the tumour and immune cells genomic profiles in order to drive the selection of the best treatment option for each patient, as well as to develop panels of markers that help to identify patients with high or low risks of recurrence or metastasis.

3.3. In Vitro and In Vivo Platforms to Develop Precision Medicine in HNSCCs

The study of HNSCCs genomic profiles is just one of the possible approaches for increasing the impact of precision medicine in clinical practice. Indeed, a multitude of preclinical models have been developed to improve and validate personalised treatments. The available models include the use of both in vitro and ex vivo samples, such as immortalized cell lines and patient-derived primary cultures, and also in vivo approaches (Table 1). In this review, we will explore the combination of ex vivo and in vivo systems in the field of HNSCCs precision medicine.

3.3.1. Cell Lines and Primary Cultures in HNSCCs Preclinical Research

Currently, in vitro preclinical oncology has largely used immortalized cell lines cultured on a monolayer [104]. The loss of the mechanisms involved in proliferation control guarantees the availability of a large amount of material, allowing for the possibility of developing a high number of experimental replicates and sample storage. In the literature, around 300 human HNC cell lines are listed, the majority of them derived from HPV-negative oral cavity squamous cell carcinomas [105]. Samples typically undergo extensive adaptations to survive in the in vitro controlled microenvironment. Indeed, media compositions and plastic supports are just two of the parameters that contribute to genetic alterations and the phenotypic and morphological features of patients’ cancer cells [105,106,107,108]. The heterogeneity of tumour subclones is reduced by clone selection, with the promotion of a more homogeneous cell population. Moreover, the loss of stromal and immune cell components makes it impossible to replicate in vitro, using cancer cell lines, the microenvironment features, including molecules and tumour-stroma crosstalk, cell interactions, and the three-dimensional architecture of tumour niches [104,109,110].
Thanks to ex vivo samples isolated from patients’ tissue biopsies, it is possible to overcome some of these disadvantages. Indeed, primary cultures allow for the preservation of the malignant subclones and the genetic and phenotypic cell features [104,110,111]. All of these features actively contribute to mimicking the human tumour microenvironment, as well as its contribution to drug response [112]. Thus, primary cultures represent an important tool for translational research because the preservation of tumour microenvironment makes the data of drug efficacies more realistic than those obtained from in vitro experiments. The ex vivo samples are also widely used in HNC preclinical research. Primary HNC cultures are established through different protocols. Explant cultures, chemical, enzymatic, and mechanical dissociation represent some examples of the methods used to establish primary culture [105,108,113,114]. The main challenge in the management of HNSCC primary samples is represented by the achievement of appropriate culture conditions for cell expansion. The selection of medium (such as DMEM, DMEM-F12, or RPMI-1640), serum percentages (from 0% to 20%), and growth factors, (normally fibroblast growth factor (FGF) and epidermal growth factor (EGF)) are the main parameters to consider [104,115,116,117,118,119,120]. Usually, primary cells are cultured in serum-free medium. This condition is useful for supporting the growth of stem-like tumour cells and to interfere with cancer-associated fibroblasts (CAFs) expansion [121,122,123,124]. CAFs are considered contaminant cells due to their ability to proliferate and survive at the expense of HNSCC cells, often even under serum-free medium [125,126]. To overcome this problem, cell scraping or serial trypsinization passages, based on the CAFs’ characteristic of detaching earlier than epithelial malignant cells, represent two alternatives [105,113,127]. To date, a unique protocol for an efficient establishment of HNSCC primary cultures does not exist, and further investigations are needed to overcome this barrier.

3.3.2. 3D Culture Models in HNSCCs Precision Medicine

HNCs are solid tumours, a group of malignancies that take advantage of the crosstalk between cell and non-cell components [128,129]. These interactions regulate molecular processes involved in cancer progression, migration, drug sensitivity, and proliferation, taking part in the development of a protumorigenic microenvironment [104,130,131]. Primary cultures partially mimic tumour microenvironment features. Despite the preservation of the heterogeneity of original tumour cell populations, these models lack other important features of human cancer tissues, such as the three-dimensional cell distribution. Tissues architecture contribution must be considered and included by in vitro studies in precision medicine.
Several 3D scaffold free and scaffold based in vitro models have been developed to reproduce the tumour microenvironment. Regarding scaffold-based models, several strategies have been developed using synthetic or biologically derived matrix materials. In this scenario, non-polymeric materials or decellularized matrices are two options for faithfully reproducing the natural pathophysiological tumour features [132]. Collagen is the most represented component of connective tissues [133], and, consequently, collagen-based scaffolds are widely used as excellent ECM mimetic devices. Recent studies demonstrated the capability of collagen-based scaffolds to induce different drug resistance mechanisms and pathological features on the basis of cell types [134,135]. Zhang M. et al. studied the radiosensitivity of HPV-positive and negative oropharyngeal squamous cell carcinoma (OSCC) cell lines using a synthetic polyester-based scaffold [136]. Their results show a higher radiosensitivity of HPV-positive OSCC cells than the negative counterparts, supporting better clinical outcomes of this kind of patient, and thus demonstrating the translational value of the device [136]. Moreover, we have published the first study using collagen-based scaffolds harbouring both cell lines and primary cultures to study OSCCs [131]. Our findings show the ability of the 3D system to mimic some of the characteristics of OSCCs, such as aggressiveness, migration properties, and drug sensitivity, better than the common monolayer culture [131]. Taken together, these results suggest a high potential of 3D scaffold models as drug screening platforms for the selection of the most effective treatment on patients’ derived cells.
Tumours are described as dynamic masses in continuous evolution [134]. Indeed, malignant lesions are characterised by different areas, each displaying their own phenotypic and biological features [137]. Scaffold-free cultures are excellent options for reproducing the spatial distribution of cancer cell populations. These systems usually refer to a culture developed through a self-assembled or forced cell aggregation characterised by a spherical shape [128,138]. This physical parameter allows for the development of superficial and inner zones with different nutrients and oxygen gradients, a structural characteristic of the tumour mass [138,139,140]. Oxygen tension and permeability are features that affect the chemo- and radio-cytotoxicity on tumour cells [141,142,143]. For these reasons, scaffold-free cultures are excellent and representative systems for in vitro drug screening assays. Moreover, scaffold-free cultures can be obtained from patient-derived cells, such as organoids. These spherical cultures are defined as self-organized structures derived from stem cell subclones, able to develop a 3D architecture [144]. The possibilities of obtaining healthy and cancer tissue-derived cultures represent a powerful advantage for the development of drug screening platforms which are able to predict drug response, as well as to identify compounds targeting cancer cells rather than healthy components [144,145,146,147]. In 2011, Lim Y.C. et al. successfully generated, for the first time, organoids from HNSCC patients’ tissues (named squamospheres) [148]. The 3D cultures were obtained starting from a single cell suspension derived from the digestion of primary HNC samples and cultured in serum-free media. In this preliminary study, the authors tested four chemotherapeutic agents: cisplatin, 5-fluorouracil (FU), paclitaxel, and docetaxel [148]. Differently, Driehuis E et al. measured the effect of conventional therapies already used in clinical practice, together with others tested for future implementation, on a panel of oral mucosa- and 31 HNSCC patient-derived organoids [149]. Treatments included targeted therapies (everolimus, niraparib, alpelisib, AZD4547, and vemurafenib), chemotherapeutic agents, radiotherapy, and different combinations of the above-mentioned drugs. The organoid’s ability to predict drug response needs to be confirmed in prospective clinical trials. One such validation study is represented by ONCODE-P2018-0003, in which 80 HNSCC patients’ outcomes after standard first-line treatments will be compared to organoids drug sensibility [150].
Previously described models are generated through single cell suspensions obtained after tissue digestion. This process entails the loss of the 3D tissue architecture and the selection of some cell populations [151]. It represents an important disadvantage for many research topics, such as the study of immunotherapy effects, as the spatial interactions with TME represent a crucial point of these drug mechanisms. Patient-derived explants (PDE) are another option to perform tumour cultures established by tissue fragments. In this case, samples maintain the original ECM structure and the vascular, immune, and stromal cell populations retain the histological characteristics of the original tumour [151,152]. PDEs have been widely used in preclinical research since the early 1950s [153,154]. The success of PDE platforms to predict patient responses is supported by several studies [155,156]. For example, Vescio R.A. et al. tested the reliability of a histoculture drug response assay (HDRA) as a drug screening platform for stomach and colon cancers [155]. After the first preclinical promising results, the HDRA clinical trial showed a correlation rate of 92.1% in predicting treatment efficacy in patients affected by colorectal and gastric cancer [157]. In the field of HNSCCs, Majumder B. et al. studied the efficacy of an innovative ex vivo drug screening platform, named CANScript, in which fresh tissue samples were sliced and seeded in supports coated with matrix proteins or injected as a patient-derived xenograft (PDX) [158]. The cytotoxicity data from the two models showed a strong correlation. Moreover, the CANScript technology was also able to stratify the patients treated with the combination of docetaxel, cisplatin, and 5-fluorouracil (TPF) into partial or complete responders and non-responders [158]. Another recent study compares the drug effect of targeted therapy in tumour tissue explants of PDX and directly in mice models [159]. The results showed a significant correspondence between the drug efficacy of ex vivo analysis (TEVA) and in vivo in mice. Despite the excellent results of these platforms, PDEs are not yet applied as a support in clinical practice.

3.3.3. HNSCCs PDX in Murine Model

In vivo models of HNSCCs, named patient derived xenografts (PDX), are developed through the implantation of cancer specimens or the injection of cancer cell suspensions into host animals. Nowadays, immunocompromised mice represent the most common species used in preclinical research. Over 50 years ago, engineered mice did not yet exist, and to overcome the problem of human samples rejection, engraftments were performed using murine cell lines in syngeneic models [160,161]. Now, a number of immunodeficient mouse strains are available, characterised by different engineered methods used to produce an impaired immune system. For example, the first immunocompromised mice, BALB/c Nude (nu/nu), were obtained through a mutation in Foxn1 gene that induced a low number of T-cells [162]. In this new scenario, PDX has been gradually employed in almost all the research fields associated with pathologies, including HNSCCs.
Initially, HNSCC PDXs were performed using athymic nude mice as hosts, but these murine models showed low engraftment rates [163]. The development of strains characterised by a higher immunodeficiency, such as NOD SCID gamma (NSG) mice, have increased the engraftment efficacy to the 80% rate of success obtained by Li H. et al., with the development of 61 PDX out on a total of 76 samples tested [164,165,166]. The scientific efforts in this field provided important translational results. For example, sample engraftments within 8 weeks correlate with a worse patient outcome, and PDXs from tumours that have developed lymph node metastasis in patients displayed higher engraftment efficacy in vivo [165,166,167,168]. Moreover, the implantation of human tissue fragments allows for the conservation of the microenvironment structure, and recapitulates the cell population’s heterogeneity [150,169]. As in the primary culture model, PDXs tend to lose progressively the genetic and physiopathological original features [165,170]. In order to demonstrate the ability of PDX to preserve the human cancer characteristics and the sensitivity to treatments, many studies have compared the genetic landscape of tumour samples or public genome databases (such as The Cancer Genome Atlas) to PDX tumours [10,165]. Copy number profiles, mutation status, and gene expression levels of PDXs and HNSCC tumours resulted in high concordance across the whole genome [10,165,167,168,169,171,172]. Thus, the HNC cell lines did not provide the same results, showing different genetic status when compared to matched human cancer tissues [165]. The same concordances were showed by comparing proteomic analyses. Despite the substitution of human stromal components with murine counterparts, the majority of proteins are conserved in PDX tumours, except for the selection of proteins related to proliferative pathways [166,173].
All of these features make PDXs in immunodeficient mice a promising model to predict patient drugs response. In Table 2, the projects that have investigated the drug sensitivity of PDXs in mice are summarized (Table 2) [167,168,170,174,175]. Chemotherapeutic agents, radiation, and several tyrosine kinase inhibitors (TKI) were the principal tested therapies. We have already reported the importance of immunotherapy in the treatment of HNSCCs. The mechanism of action of these types of drugs is related to the interaction between drugs and the immune system [176,177]. The absence of a functional immunity in immune deficient mice makes this model inappropriate for evaluating the effect of immunotherapeutic drugs. To overcome this limit, several research groups developed humanised PDX models through the co-engraftment of cancer tissue fragments and haematopoietic stem cells [178,179]. Therefore, the addition of human immune cells increases the host human systemic reproduction, essential for mimicking the complexity of the tumour microenvironment. Morton J.J. et al. developed the XactMice model, in which the injection of human hematopoietic stem and progenitor cells reconstitutes the radiation-depleted bone marrow of a NOD/SCID/IL2rg (−/−) (NSG) mice [179]. The data showed how the addition of these immune cells increased the amount of immune infiltrate, induced lymphangiogenesis, and modified the tumour gene expression profile [179].
Table 2. Summary of the methods used for patient-derived xenografts of HNC cell cultures in mice.
Table 2. Summary of the methods used for patient-derived xenografts of HNC cell cultures in mice.
Mouse StrainCancer TypesImplantation SitePDX TestedTreatment TestedReference
NOD/Hsd: Athymic Nude-Foxn1nuTonsil, Base of Tongue, Floor of MouthSubcutaneously3Radiation, Cisplatin, Cetuximab[170]
NSG (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ)Tongue, Alveolar ridge, BuccalSubcutaneously3Flavopiridol, Belinostat, Docetaxel[174]
NSGTonsilSubcutaneously5Radiation[175]
NSGNSEnzymatic dissociation and single cell suspension subcutaneously injected10Abemaciclib[167]
Nu/Nu/(NOG, NOD/Shi-scid/IL-2Rγnull)Tongue, oropharynxSubcutaneously3Afatinib, BKM120[168]
NS: not specified.
However, PDX models retain some important limits. For example, the implant of a single tissue fragment does not represent the tumour cell heterogeneity [165,180]. Moreover, the implantation is performed subcutaneously in the mice’s flanks, and not orthotopically, with a consequent decrease of the translational powers of obtained results. On the other hand, orthotopic implantation into oral tissues entails many disadvantages, such as the development of a tumour mass in the oral cavity that prevents the animal from feeding [181].
In conclusion, the evidence of drug sensitivity correlation between patients and the relative derived-PDX mice shown by these studies and in larger patient cohorts demonstrates the reliability of this preclinical model as a drug screening platform for HNC precision medicine.

3.3.4. HNSCCs PDX in Zebrafish Models

Preclinical drug screening platforms represent promising tools to optimize the single-patient treatment. However, these approaches are characterised by some limits, as previously discussed, that harper the translation of results in clinical practice. Certainly, the complexity to culture ex vivo samples and the low engraftment rates obtained with PDX murine models are two important experimental limits [182]. Moreover, the low amount of biological material available from surgical specimens and the temporal gap between tissue manipulation and experiment readouts contribute to making preclinical platforms difficult to apply in clinics [104].
A valid and innovative scientific approach to overcome part of these limits is represented by the zebrafish model. The first time that the zebrafish was used as host for xenografts dates back to 2006, when human melanoma and multiple myeloma cells were injected in zebrafish embryos [183,184]. Different from murine models, the zebrafish is mostly used in preclinical research at embryonic and larval stages until reaching 5 day post-fertilization (dpf). Indeed, the zebrafish is a highly fecund animal, able to lay hundreds of eggs weekly. Moreover, embryos are transparent for at least 2 days, or more if treated with PFU, and this characteristic allows for the tracing of cell dynamics very easily during embryonic and larval development. The zebrafish is also an easy-to-handle animal model, and its easy genetic manipulation is useful to develop transgenic or mutant strains [185]. Among them, the casper zebrafish is a double mutated (roy−/− and nacre−/−) strain characterised by the absence of pigmentation for the entire life [186]. This feature allows for in vivo imaging of inner organs, and makes casper zebrafish an optimal recipient to trace cell dynamics. Zebrafish manipulation is simpler compared to mice, and gives the opportunity to inject hundreds of embryos per day by a single operator [182,187]. Another advantage is represented by the low number of cells necessary for each injection (normally between 100 and 500 cells per embryo), or the small tissue fragments necessary for each single transplantation [188]. For all of these reasons, the zebrafish model is particularly suitable for large scale and high throughput PDX studies.
The use of PDXs in zebrafish is a recent approach, and the first documented reports of drugs tested on embryos injected with human primary cells are not older than a decade [184,189]. Since then, the use of xenotransplantation of primary cultures in zebrafish larvae has been documented by many studies. The development of innate immunity starts in the early embryonic stages, and ends in around 2/3 weeks [190]. Therefore, if mice need to be immunocompromised to avoid human cells rejection, zebrafish embryos have an incomplete immune system that allows xenotransplantation at embryonic stages. Recently, several groups have tested PDX in zebrafish for many solid and haematological malignancies [191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207]. The first documented PDX of HNSCCs cells was performed by the Al-Samadi A. et al. group in a preliminary study based on the development of a PCR-based assay to measure drug efficacy [207]. The project aim was the achievement of a fast method to screen a high number of compounds based on the quantification of human DNA inside fish as an indirect count of viable cells after drug exposure. Despite the low number of samples tested (four HNC cell lines and a metastatic oral tongue cancer primary culture), it represents the first and promising attempt to develop a drug screening platform for HNC personalised medicine [207].
The more common samples xenotransplanted in zebrafish are represented by commercial cell lines and single-cell suspensions obtained by the digestion of patient tissues [152,182]. However, in contrast to the murine model, primary tissues can be successfully engrafted in zebrafish embryos. Marques I.J. et al. developed an easy and fast method to transplant small tissue fragments into the yolk sack of 2 dpf embryos [195]. Pancreas, colon, and stomach cancer tissues were monitored in the embryo’s body to test the metastatic potential of the primary cells [195]. Usai A. et al., for the first time, studied the feasibility of a drug screening platform using zebrafish embryos transplanted with malignant patient tissues, named Avatars, and they developed a method to identify the dose conversion criterion from human to zebrafish [208]. Avatars were created using tissues from 24 patients affected by pancreatic, colon, and gastric cancers, and different chemotherapeutic regimens were tested to measure the fragments area regression [208]. The samples belonged to patients enrolled in NCT03668418 (xenoZ) co-clinical trials, an observational prospective study based on the xenotransplantation of primary hepato–biliary–pancreatic and gastro–intestinal cancer samples in zebrafish embryos [209]. The XenoZ model demonstrated the prediction of the patient’s clinical outcomes, discriminating between responders and non-responders patients. Here, it can be considered a pioneering study focused on the reliability of co-clinical trials based on the use of zebrafish for the selection of personalised therapies. Transplantations of HNCs tissue fragments have not yet been reported in scientific literature, and they also represent an important limitation for the development of precision medicine platforms in these aggressive forms of cancers.

4. Conclusions

In this review, we have summarized the current approaches and the promising technologies in the field of personalised medicine for HNC patients. Genomic and drug screening platforms represent the most used options which are currently achievable, but new technologies and preclinical models are under development, which will be explored worldwide. It will be important to overcome the disadvantages of the current system, starting from the progress of innovative approaches which are able to study immunotherapeutic agents. However, we think that collaborations and data sharing represent the best scientific strategies for obtaining successful improvements of the current and future tools in precision medicine.

Author Contributions

Conceptualization, G.M. (Giacomo Miserocchi), M.S. and L.M.; writing—original draft preparation, G.M. (Giacomo Miserocchi), S.C., A.U., M.S. and L.M.; writing—review and editing, F.D.R., A.D.V., C.L., C.S., C.C. (Claudia Cocchi), S.V., C.C. (Chiara Calabrese), M.B., G.D.L. and L.M.; supervision, F.D.R. and A.D.V.; project administration, G.M. (Giuseppe Meccariello), T.I. and L.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to thank Marcella Tazzari for figure creation assistance using Biorender.com (accessed on 8 April 2022) This work was partly supported thanks to the contribution of Ricerca Corrente by the Italian Ministry of Health within the research line Precision, Gender, and ethnicity-Based Medicine and Geroscience: Genetic–Molecular Mechanisms in the Development, Characterisation, and Treatment of Tumours.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Chow, Q.M.L. Head and Neck Cancer. N. Engl. J. Med. 2020, 382, 60–72. [Google Scholar] [CrossRef] [PubMed]
  3. Sanderson, R.J.; Ironside, J.A.D. Squamous cell carcinomas of the head and neck. BMJ 2002, 325, 822–827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Hashibe, M.; Brennan, P.; Chuang, S.C.; Boccia, S.; Castellsague, X.; Chen, C.; Curado, M.P.; Dal Maso, L.; Daudt, A.W.; Fabianova, E.; et al. Interaction between tobacco and alcohol use and the risk of head and neck cancer: Pooled analysis in the International Head and Neck Cancer Epidemiology Consortium. Cancer Epidemiol. Biomark. Prev. 2009, 18, 541–550. [Google Scholar] [CrossRef] [Green Version]
  5. Sabatini, M.E.; Chiocca, S. Human papillomavirus as a driver of head and neck cancers. Br. J. Cancer 2020, 122, 306–314. [Google Scholar] [CrossRef] [PubMed]
  6. Kian Ang, K.; Harris, J.; Wheeler, R.; Weber, R.; Rosenthal, D.I.; Nguyen-Tân, P.F.; Westra, W.H.; Chung, C.H.; Jordan, R.C.; Lu, C.; et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N. Engl. J. Med. 2010, 363, 24–35. [Google Scholar] [CrossRef] [Green Version]
  7. Paver, E.C.; Currie, A.M.; Gupta, R.; Dahlstrom, J.E. Human papilloma virus related squamous cell carcinomas of the head and neck: Diagnosis, clinical implications and detection of HPV. Pathology 2020, 52, 179–191. [Google Scholar] [CrossRef] [Green Version]
  8. Johnson, D.E.; Burtness, B.; Leemans, C.R.; Wai Yan Lui, V.; Bauman, J.E.; Grandis, J.R. Head and neck squamous cell carcinoma. Nat. Rev. Dis. Primers 2020, 6, 92. [Google Scholar] [CrossRef]
  9. Chen, Y.P.; Chan, A.T.C.; Le, Q.T.; Blanchard, P.; Sun, Y.; Ma, J. Nasopharyngeal carcinoma. Lancet 2019, 394, 64–80. [Google Scholar] [CrossRef]
  10. The Cancer Genome Atlas Network. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 2015, 517, 576–582. [Google Scholar] [CrossRef] [Green Version]
  11. Cerami, E.; Gao, J.; Dogrusoz, U.; Gross, B.E.; Sumer, S.O.; Aksoy, B.A.; Jacobsen, A.; Byrne, C.J.; Heuer, M.L.; Larsson, E.; et al. The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012, 2, 401–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Sheth, S.H.; Johnson, D.E.; Kensler, T.W.; Bauman, J.E. Chemoprevention targets for tobacco-related head and neck cancer: Past lessons and future directions. Oral. Oncol. 2015, 51, 557–564. [Google Scholar] [CrossRef] [PubMed]
  13. Pai, S.I.; Westra, W.H. Molecular pathology of head and neck cancer: Implications for diagnosis, prognosis, and treatment. Annu. Rev. Pathol. 2009, 4, 49–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Brooks, P.J.; Theruvathu, J.A. DNA adducts from acetaldehyde: Implications for alcohol-related carcinogenesis. Alcohol 2005, 35, 187–193. [Google Scholar] [CrossRef] [PubMed]
  15. Stein, A.P.; Saha, S.; Kraninger, J.L.; Swick, A.D.; Yu, M.; Lambert, P.F.; Kimple, R.J. Prevalence of human papillomavirus in oropharyngeal cancer: A systematic review. Cancer J. 2015, 21, 138–146. [Google Scholar] [CrossRef] [Green Version]
  16. Alsahafi, E.; Begg, K.; Amelio, I.; Raulf, N.; Lucarelli, P.; Sauter, T.; Tavassoli, M. Clinical update on head and neck cancer: Molecular biology and ongoing challenges. Cell Death Dis. 2019, 10, 540. [Google Scholar] [CrossRef] [Green Version]
  17. Zhou, G.; Liu, Z.; Myers, J.N. TP53 Mutations in Head and Neck Squamous Cell Carcinoma and Their Impact on Disease Progression and Treatment Response. J. Cell. Biochem. 2016, 117, 2682–2692. [Google Scholar] [CrossRef] [Green Version]
  18. Wiest, T.; Schwarz, E.; Enders, C.; Flechtenmacher, C.; Bosch, F.X. Involvement of intact HPV16 E6/E7 gene expression in head and neck cancers with unaltered p53 status and perturbed pRb cell cycle control. Oncogene 2002, 21, 1510–1517. [Google Scholar] [CrossRef] [Green Version]
  19. Eckhardt, M.; Zhang, W.; Gross, A.M.; Von Dollen, J.; Johnson, J.R.; Franks-Skiba, K.E.; Swaney, D.L.; Johnson, T.L.; Jang, G.M.; Shah, P.S.; et al. Multiple routes to oncogenesis are promoted by the human papillomavirus-host protein network. Cancer Discov. 2018, 8, 1474–1489. [Google Scholar] [CrossRef] [Green Version]
  20. Krishnamurthy, S.; Dong, Z.; Vodopyanov, D.; Imai, A.; Helman, J.I.; Prince, M.E.; Wicha, M.S.; Nör, J.E. Endothelial cell-initiated signaling promotes the survival and self-renewal of cancer stem cells. Cancer Res. 2010, 70, 9969–9978. [Google Scholar] [CrossRef] [Green Version]
  21. Reid, P.; Marcu, L.G.; Olver, I.; Moghaddasi, L.; Staudacher, A.H.; Bezak, E. Diversity of cancer stem cells in head and neck carcinomas: The role of HPV in cancer stem cell heterogeneity, plasticity and treatment response. Radiother. Oncol. 2019, 135, 1–12. [Google Scholar] [CrossRef] [PubMed]
  22. Ritchie, K.E.; Nor, J.E. Perivascular stem cell niche in head and neck cancer. Cancer Lett. 2013, 338, 41–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Zanoni, M.; Bravaccini, S.; Fabbri, F.; Arienti, C. Emerging Roles of Aldehyde Dehydrogenase Isoforms in Anti-cancer Therapy Resistance. Front. Med. 2022, 9, 795762. [Google Scholar] [CrossRef] [PubMed]
  24. Gillison, M.L.; D’Souza, G.; Westra, W.; Sugar, E.; Xiao, W.; Begum, S.; Viscidi, R. Distinct risk factor profiles for human papillomavirus type 16-positive and human papillomavirus type 16-negative head and neck cancers. J. Natl. Cancer Inst. 2008, 100, 407–420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Vlashi, E.; Chen, A.M.; Boyrie, S.; Yu, G.; Nguyen, A.; Brower, P.A.; Hess, C.B.; Pajonk, F. Radiation-Induced Dedifferentiation of Head and Neck Cancer Cells Into Cancer Stem Cells Depends on Human Papillomavirus Status. Int. J. Radiat. Oncol. Biol. Phys. 2016, 94, 1198–1206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Zhang, M.; Kumar, B.; Piao, L.; Xie, X.; Schmitt, A.; Arradaza, N.; Cippola, M.; Old, M.; Agrawal, A.; Ozer, E.; et al. Elevated intrinsic cancer stem cell population in human papillomavirus-associated head and neck squamous cell carcinoma. Cancer 2014, 120, 992–1001. [Google Scholar] [CrossRef]
  27. Peitzsch, C.; Nathansen, J.; Schniewind, S.I.; Schwarz, F.; Dubrovska, A. Cancer Stem Cells in Head and Neck Squamous Cell Carcinoma: Identification, Characterization and Clinical Implications. Cancers 2019, 11, 616. [Google Scholar] [CrossRef] [Green Version]
  28. Nör, C.; Zhang, Z.; Warner, K.A.; Bernardi, L.; Visioli, F.; Helman, J.I.; Roesler, R.; Nör, J.E. Cisplatin induces Bmi-1 and enhances the stem cell fraction in head and neck cancer. Neoplasia 2014, 16, 137–146. [Google Scholar] [CrossRef] [Green Version]
  29. Chen, A.M.; Felix, C.; Wang, P.-C.; Hsu, S.; Basehart, V.; Garst, J.; Beron, P.; Wong, D.; Rosove, M.H.; Rao, S.; et al. Reduced-dose radiotherapy for human papillomavirus-associated squamous-cell carcinoma of the oropharynx: A single-arm, phase 2 study. Lancet Oncol. 2017, 18, 803–811. [Google Scholar] [CrossRef] [Green Version]
  30. Lee, J.; Park, M.; Ko, Y.; Kim, B.; Kim, O.; Hyun, H.; Kim, D.; Sohn, H.; Moon, Y.A.; Lim, W. Ectopic overexpression of CD133 in HNSCC makes it resistant to commonly used chemotherapeutics. Tumor. Biol. 2017, 39, 1010428317695534. [Google Scholar] [CrossRef] [Green Version]
  31. Cojoc, M.; Mäbert, K.; Muders, M.H.; Dubrovska, A. A role for cancer stem cells in therapy resistance: Cellular and molecular mechanisms. Semin. Cancer Biol. 2015, 31, 16–27. [Google Scholar] [CrossRef] [PubMed]
  32. Hunt, J.L.; Barnes, L.; Lewis Jr, J.S.; Mahfouz, M.E.; Slootweg, P.J.; Thompson, L.D.R.; Cardesa, A.; Devaney, K.O.; Gnepp, D.R.; Westra, W.H.; et al. Molecular diagnostic alterations in squamous cell carcinoma of the head and neck and potential diagnostic applications. Eur. Arch. Otorhinolaryngol. 2014, 271, 211–223. [Google Scholar] [CrossRef] [PubMed]
  33. Gong, W.; Xiao, Y.; Wei, Z.; Yuan, Y.; Qiu, M.; Sun, C.; Zeng, X.; Liang, X.; Feng, M.; Chen, Q. Toward the use of precision medicine for the treatment of head and neck squamous cell carcinoma. Oncotarget 2017, 8, 2141–2152. [Google Scholar] [CrossRef] [PubMed]
  34. Fasano, M.; Della Corte, C.M.; Viscardi, G.; Di Liello, R.; Paragliola, F.; Sparano, F.; Iacovino, M.L.; Castrichino, A.; Doria, F.; Sica, A.; et al. Head and neck cancer: The role of anti-EGFR agents in the era of immunotherapy. Ther. Adv. Med. Oncol. 2021, 13, 1758835920949418. [Google Scholar] [CrossRef]
  35. Mayer, A.; Zahnreich, S.; Brieger, J.; Vaupel, P.; Schmidberger, H. Downregulation of EGFR in hypoxic, diffusion-limited areas of squamous cell carcinomas of the head and neck. Br. J. Cancer 2016, 115, 1351–1358. [Google Scholar] [CrossRef] [Green Version]
  36. Wilson, W.R.; Hay, M.P. Targeting hypoxia in cancer therapy. Nat. Rev. Cancer 2011, 11, 393–410. [Google Scholar] [CrossRef]
  37. Gabalski, E.C.; Adam, M.; Pinto, H.; Brown, J.M.; Bloch, D.A.; Terris, D.J. Pretreatment and midtreatment measurement of oxygen tension levels in head and neck cancers. Laryngoscope 1988, 108, 1856–1860. [Google Scholar] [CrossRef]
  38. Wegge, M.; Dok, R.; Nuyts, S. Hypoxia and Its Influence on Radiotherapy Response of HPV-Positive and HPV-Negative Head and Neck Cancer. Cancers 2021, 13, 5959. [Google Scholar] [CrossRef]
  39. Begg, K.; Tavassoli, M. Inside the hypoxic tumour: Reprogramming of the DDR and radioresistance. Cell Death Discov. 2020, 6, 77. [Google Scholar] [CrossRef]
  40. Harris, A.L. Hypoxia—A key regulatory factor in tumour growth. Nat. Rev. Cancer 2002, 2, 38–47. [Google Scholar] [CrossRef]
  41. Masoud, G.N.; Li, W. HIF-1α pathway: Role, regulation and intervention for cancer therapy. Acta Pharm. Sin. B 2015, 5, 378–389. [Google Scholar] [CrossRef] [Green Version]
  42. Lee, N.C.J.; Kelly, J.R.; Park, H.S.; An, Y.; Judson, B.L.; Burtness, B.A.; Husain, Z.A. Patterns of failure in high-metastatic node number human papillomavirus-positive oropharyngeal carcinoma. Oral. Oncol. 2018, 85, 35–39. [Google Scholar] [CrossRef] [PubMed]
  43. Forastiere, A.A.; Ismaila, N.; Lewin, J.S.; Nathan, C.A.; Adelstein, D.J.; Eisbruch, A.; Fass, G.; Fisher, S.G.; Laurie, S.A.; Le, Q.T. Use of larynx-preservation strategies in the treatment of laryngeal cancer: American Society of Clinical Oncology clinical practice guideline update. J. Clin. Oncol. 2018, 36, 1143–1169. [Google Scholar] [CrossRef] [PubMed]
  44. Bernier, J.; Domenge, C.; Ozsahin, M.; Matuszewska, K.; Lefèbvre, J.L.; Greiner, R.H.; Giralt, J.; Maingon, P.; Rolland, F.; Bolla, M.; et al. Postoperative irradiation with or without concomitant chemotherapy for locally advanced head and neck cancer. N. Engl. J. Med. 2004, 350, 1945–1952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Furqan, M.; Snyders, T.P.; Saqlain, M.U.; Mott, S.L.; Laux, D.; Snow, A.; Anderson, C.M.; Watkins, J.M.; Clamon, J.H. Comparing high-dose cisplatin with cisplatin-based combination chemotherapy in definitive concurrent chemoradiation setting for locally advanced head and neck squamous cell carcinoma (LAHNSCC). Cancer Med. 2019, 8, 2730–2739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Bauml, J.M.; Vinnakota, R.; Park, Y.H.A.; Bates, S.E.; Fojo, T.; Aggarwal, C.; Limaye, S.; Damjanov, N.; Di Stefano, J.; Ciunci, C.; et al. Cisplatin every 3 weeks versus weekly with definitive concurrent radiotherapy for squamous cell carcinoma of the head and neck. J. Natl. Cancer Inst. 2019, 111, 490–497. [Google Scholar] [CrossRef] [Green Version]
  47. Bonner, J.A.; Harari, P.M.; Giralt, J.; Azarnia, N.; Shin, D.M.; Cohen, R.B.; Jones, C.U.; Sur, R.; Raben, D.; Jassem, J.; et al. Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N. Engl. J. Med. 2006, 354, 567–578. [Google Scholar] [CrossRef] [Green Version]
  48. Zhang, Y.; Chen, L.; Hu, G.Q.; Zhang, N.; Zhu, X.D.; Yang, K.Y.; Jin, F.; Shi, M.; Chen, Y.P.; Hu, W.H.; et al. Gemcitabine and cisplatin induction chemotherapy in nasopharyngeal carcinoma. N. Engl. J. Med. 2019, 381, 1124–1135. [Google Scholar] [CrossRef]
  49. Kitamura, N.; Sento, S.; Yoshizawa, Y.; Sasabe, E.; Kudo, Y.; Yamamoto, T. Current Trends and Future Prospects of Molecular Targeted Therapy in Head and Neck Squamous Cell Carcinoma. Int. J. Mol. Sci. 2020, 22, 240. [Google Scholar] [CrossRef]
  50. Cramer, J.D.; Burtness, B.; Ferris, R.L. Immunotherapy for head and neck cancer: Recent advances and future directions. Oral. Oncol. 2019, 99, 104460. [Google Scholar] [CrossRef]
  51. Moskovitz, J.; Moy, J.; Ferris, R.L. Immunotherapy for Head and Neck Squamous Cell Carcinoma. Curr. Oncol. Rep. 2018, 20, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Burtness, B.; Harrington, K.J.; Greil, R.; Soulières, D.; Tahara, M.; de Castro, G., Jr.; Psyrri, A.; Basté, N.; Neupane, P.; Bratland, A.; et al. Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): A randomised, open-label, phase 3 study. Lancet 2019, 394, 1915–1928. [Google Scholar] [CrossRef]
  53. Aiken, R.; Fine, H.; Butowski, N. ATIM-46. A multicenter, phase I, trial of radiation, temozolomide and rrx-001 followed by maintenance temozolomide with or without rrx-001 in newly diagnosed glioblastoma patients. Neuro-Oncology 2019, 21, vi11–vi12. [Google Scholar] [CrossRef]
  54. Vermunt, M.A.C.; van der Heijden, L.T.; Hendrikx, J.J.M.A.; Schinkel, A.H.; de Weger, V.A.; van der Putten, E.; van Triest, B.; Bergman, A.M.; Beijnen, J.H. Pharmacokinetics of docetaxel and ritonavir after oral administration of ModraDoc006/r in patients with prostate cancer versus patients with other advanced solid tumours. Cancer Chemother. Pharmacol. 2021, 87, 855–869. [Google Scholar] [CrossRef] [PubMed]
  55. Zoi, V.; Galani, V.; Lianos, G.D.; Voulgaris, S.; Kyritsis, A.P.; Alexiou, G.A. The Role of Curcumin in Cancer Treatment. Biomedicines 2021, 9, 1086. [Google Scholar] [CrossRef]
  56. Larue, R.T.H.M.; Van De Voorde, L.; Berbée, M.; van Elmpt, W.J.C.; Dubois, L.J.; Panth, K.M.; Peeters, S.G.J.A.; Claessens, A.; Schreurs, W.M.J.; Nap, M.; et al. A phase 1 ‘window-of-opportunity’ trial testing evofosfamide (TH-302), a tumour-selective hypoxia-activated cytotoxic prodrug, with preoperative chemoradiotherapy in oesophageal adenocarcinoma patients. BMC Cancer 2016, 16, 644. [Google Scholar] [CrossRef] [Green Version]
  57. Gong, L.; Zhang, Y.; Liu, C.; Zhang, M.; Han, S. Application of Radiosensitizers in Cancer Radiotherapy. Int. J. Nanomed. 2021, 16, 1083–1102. [Google Scholar] [CrossRef]
  58. Overgaard, J.; Hansen, H.S.; Overgaard, M.; Bastholt, L.; Berthelsen, A.; Specht, L.; Lindeløv, B.; Jørgensen, K. A randomized double-blind phase III study of nimorazole as a hypoxic radiosensitizer of primary radiotherapy in supraglottic larynx and pharynx carcinoma. Results of the Danish Head and Neck Cancer Study (DAHANCA) Protocol 5-85. Radiother. Oncol. 1998, 46, 135–146. [Google Scholar] [CrossRef]
  59. Toustrup, K.; Sørensen, B.S.; Lassen, P.; Wiuf, C.; Alsner, J.; Overgaard, J. Gene expression classifier predicts for hypoxic modification of radiotherapy with nimorazole in squamous cell carcinomas of the head and neck. Radiother. Oncol. 2012, 102, 122–129. [Google Scholar] [CrossRef]
  60. Lassen, P.; Eriksen, J.G.; Hamilton-Dutoit, S.; Tramm, T.; Alsner, J.; Overgaard, J. HPV-associated p16-expression and response to hypoxic modification of radiotherapy in head and neck cancer. Radiother. Oncol. 2010, 94, 30–35. [Google Scholar] [CrossRef]
  61. Mirnezami, R.; Nicholson, J.; Darzi, A. Preparing for precision medicine. N. Engl. J. Med. 2012, 366, 489–491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Hyman, D.M.; Taylor, B.S.; Baselga, J. Implementing genome-driven oncology. Cell 2017, 168, 584–599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Strauss, D.G.; Blinova, K. Clinical trials in a dish. Trends Pharmacol. Sci. 2017, 38, 4–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Tuveson, D.; Clevers, H. Cancer modeling meets human organoid technology. Science 2019, 364, 952–955. [Google Scholar] [CrossRef]
  65. AACR Project GENIE Consortium. AACR Project GENIE: Powering precision medicine through an international consortium. Cancer Discov. 2017, 7, 818–831. [Google Scholar] [CrossRef] [Green Version]
  66. Schilsky, R.L.; Michels, D.L.; Kearbey, A.H.; Yu, P.P.; Hudis, C.A. Building a rapid learning health care system for oncology: The regulatory framework of CancerLinQ. J. Clin. Oncol. 2014, 32, 2373–2379. [Google Scholar] [CrossRef]
  67. Rebuzzi, S.E.; Alfieri, R.; La Monica, S.; Minari, R.; Petronini, P.R.; Tiseo, M. Combination of EGFR-TKIs and chemotherapy in advanced EGFR mutated NSCLC: Review of the literature and future perspectives. Crit. Rev. Oncol. Hematol. 2020, 146, 102820. [Google Scholar] [CrossRef]
  68. Galot, R.; Le Tourneau, C.; Guigay, J.; Licitra, L.; Tinhofer, I.; Kong, A.; Caballero, C.; Fortpied, C.; Bogaerts, J.; Govaerts, A.S.; et al. Personalized biomarker-based treatment strategy for patients with squamous cell carcinoma of the head and neck: EORTC position and approach. Ann. Oncol. 2018, 29, 2313–2327. [Google Scholar] [CrossRef] [Green Version]
  69. Prasad, V. Perspective: The precision-oncology illusion. Nature 2016, 537, S63. [Google Scholar] [CrossRef]
  70. Chatterjee, N.; Bivona, T.G. Polytherapy and Targeted Cancer Drug Resistance. Trends Cancer 2019, 5, 170–182. [Google Scholar] [CrossRef]
  71. Cescon, D.W.; Bratman, S.V.; Chan, S.M.; Siu, L.L. Circulating tumor DNA and liquid biopsy in oncology. Nat. Cancer 2020, 1, 276–290. [Google Scholar] [CrossRef] [PubMed]
  72. Mattox, A.K.; Bettegowda, C.; Zhou, S.; Papadopoulos, N.; Kinzler, K.W.; Vogelstein, B. Applications of liquid biopsies for cancer. Sci. Transl. Med. 2019, 11, eaay1984. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Clouthier, D.L.; Lien, S.C.; Yang, S.Y.C.; Nguyen, L.T.; Manem, V.S.K.; Gray, D.; Ryczko, M.; Razak, A.R.A.; Lewin, J.; Lheureux, S. An interim report on the investigator-initiated phase 2 study of pembrolizumab immunological response evaluation (INSPIRE). J. Immunother. Cancer 2019, 7, 72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Wang, G.; Zhang, M.; Cheng, M.; Wang, X.; Li, K.; Chen, J.; Chen, Z.; Chen, S.; Chen, J.; Xiong, G.; et al. Tumor microenvironment in head and neck squamous cell carcinoma: Functions and regulatory mechanisms. Cancer Lett. 2021, 507, 55–69. [Google Scholar] [CrossRef]
  75. Qin, Y.; Zheng, X.; Gao, W.; Wang, B.; Wu, Y. Tumor microenvironment and immune-related therapies of head and neck squamous cell carcinoma. Mol. Ther. Oncolytics 2021, 20, 342–351. [Google Scholar] [CrossRef]
  76. Manzoni, C.; Kia, D.A.; Vandrovcova, J.; Hardy, J.; Wood, N.W.; Lewis, P.A.; Ferrari, R. Genome, transcriptome and proteome: The rise of omics data and their integration in biomedical sciences. Brief. Bioinform. 2018, 19, 286–302. [Google Scholar] [CrossRef]
  77. Serafini, M.S.; Lopez-Perez, L.; Fico, G.; Licitra, L.; De Cecco, L.; Resteghini, C. Transcriptomics and epigenomics in head and neck cancer: Available repositories and molecular signatures. Cancers Head Neck 2020, 5, 2. [Google Scholar] [CrossRef] [Green Version]
  78. Chakraborty, S.; Hosen, M.I.; Ahmed, M.; Shekhar, H.U. Onco-multi-OMICS approach: A new frontier in cancer research. BioMed Res. Int. 2018, 2018, 9836256. [Google Scholar] [CrossRef] [Green Version]
  79. Rodon, J.; Soria, J.C.; Berger, R.; Miller, W.H.; Rubin, E.; Kugel, A.; Tsimberidou, A.; Saintigny, P.; Ackerstein, A.; Braña, I.; et al. Genomic and transcriptomic profiling expands precision cancer medicine: The WINTHER trial. Nat. Med. 2019, 25, 751–758. [Google Scholar] [CrossRef]
  80. Sepich-Poore, G.D.; Zitvogel, L.; Straussman, R.; Hasty, J.; Wargo, J.A.; Knight, R. The microbiome and human cancer. Science 2021, 371, eabc4552. [Google Scholar] [CrossRef]
  81. Elinav, E.; Garrett, W.S.; Trinchieri, G.; Wargo, J. The cancer microbiome. Nat. Rev. Cancer 2019, 19, 371–376. [Google Scholar] [CrossRef] [PubMed]
  82. Pushalkar, S.; Ji, X.; Li, Y.; Estilo, C.; Yegnanarayana, R.; Singh, B.; Li, X.; Saxena, D. Comparison of oral microbiota in tumor and non-tumor tissues of patients with oral squamous cell carcinoma. BMC Microbiol. 2012, 12, 144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Hooper, S.J.; Wilson, M.J.; Crean, S.J. Exploring the link between microorganisms and oral cancer: A systematic review of the literature. Head Neck 2009, 31, 1228–1239. [Google Scholar] [CrossRef] [PubMed]
  84. Guerrero-Preston, R.; Godoy-Vitorino, F.; Jedlicka, A.; Rodríguez-Hilario, A.; González, H.; Bondy, J.; Lawson, F.; Folawiyo, O.; Michailidi, C.; Dziedzic, A.; et al. 16S rRNA amplicon sequencing identifies microbiota associated with oral cancer, human papilloma virus infection and surgical treatment. Oncotarget 2016, 7, 51320–51334. [Google Scholar] [CrossRef] [Green Version]
  85. Vermorken, J.B.; Budach, V.; Leemans, C.R.; Machiels, J.P.; Nicolai, P.; O’Sullivan, B. Critical Issues in Head and Neck Oncology; Key Concepts from the Fifth THNO Meeting; Springer: Cham, Switzerland, 2017. [Google Scholar]
  86. Nerurkar, S.N.; Goh, D.; Cheung, C.C.L.; Nga, P.Q.Y.; Lim, J.C.T.; Yeong, J.P.S. Transcriptional Spatial Profiling of Cancer Tissues in the Era of Immunotherapy: The Potential and Promise. Cancers 2020, 12, 2572. [Google Scholar] [CrossRef]
  87. Kulasinghe, A.; Taheri, T.; O’Byrne, K.; Hughes, B.G.M.; Kenny, L.; Punyadeera, C. Highly Multiplexed Digital Spatial Profiling of the Tumor Microenvironment of Head and Neck Squamous Cell Carcinoma Patients. Front. Oncol. 2020, 10, 607349. [Google Scholar] [CrossRef]
  88. Beechem, J.M. High-Plex Spatially Resolved RNA and Protein Detection Using Digital Spatial Profiling: A Technology Designed for Immuno-oncology Biomarker Discovery and Translational Research. Methods Mol. Biol. 2020, 2055, 563–583. [Google Scholar]
  89. Merritt, C.R.; Ong, G.T.; Church, S.E.; Barker, K.; Danaher, P.; Geiss, G.; Hoang, M.; Jung, J.; Liang, Y.; McKay-Fleisch, J.; et al. Multiplex digital spatial profiling of proteins and RNA in fixed tissue. Nat. Biotechnol. 2020, 38, 586–599. [Google Scholar] [CrossRef]
  90. Zugazagoitia, J.; Gupta, S.; Liu, Y.; Fuhrman, K.; Gettinger, S.; Herbst, R.S.; Schalper, K.A.; Rimm, D.L. Biomarkers associated with beneficial PD-1 checkpoint blockade in non-small-cell lung cancer (NSCLC) identified using high-plex digital spatial profiling. Clin. Cancer Res. 2020, 26, 4360–4368. [Google Scholar] [CrossRef] [Green Version]
  91. Toki, M.I.; Merritt, C.R.; Wong, P.F.; Smithy, J.W.; Kluger, H.M.; Syrigos, K.N.; Ong, G.T.; Warren, S.E.; Beechem, J.M.; Rimm, D.L. High-Plex Predictive Marker Discovery for Melanoma Immunotherapy–Treated Patients Using Digital Spatial Profiling. Clin. Cancer Res. 2019, 25, 5503–5512. [Google Scholar] [CrossRef]
  92. Jeyasekharan, A.D.; Hoppe, M.M.; Fan, S.; VanSchoiack, A.; Xin, L.; De Mel, S.; Poon, M.; Chan, E.; Lee, J.; Chee, Y.L.; et al. Digital Spatial Profiling of Immune Markers in R-chop Treated Di use Large B-cell Lymphoma Reveals a Dominant Prognostic Significance of M2 Macrophage Infiltration. In Proceedings of the 15th International Conference on Malignant Lymphoma Palazzo dei Congressi, Lugano, Switzerland, 18–22 June 2019. [Google Scholar]
  93. Marcu, L.G.; Marcu, D.C. Current Omics Trends in Personalised Head and Neck Cancer Chemoradiotherapy. J. Pers. Med. 2021, 11, 1094. [Google Scholar] [CrossRef] [PubMed]
  94. Bogowicz, M.; Pavic, M.; Riesterer, O.; Finazzi, T.; Schüler, H.G.; Holz-Sapra, E.; Rudofsky, L.; Basler, L.; Spaniol, M.; Ambrusch, A.; et al. Targeting Treatment Resistance in Head and Neck Squamous Cell Carcinoma—Proof of Concept for CT Radiomics-Based Identification of Resistant Sub-Volumes. Front. Oncol. 2021, 11, 664304. [Google Scholar] [CrossRef] [PubMed]
  95. Guiot, J.; Vaidyanathan, A.; Deprez, L.; Zerka, F.; Danthine, D.; Frix, A.; Lambin, P.; Bottari, F.; Tsoutzidis, N.; Miraglio, B.; et al. A review in radiomics: Making personalized medicine a reality via routine imaging. Med. Res. Rev. 2022, 42, 426–440. [Google Scholar] [CrossRef] [PubMed]
  96. Cantrell, S.C.; Peck, B.W.; Li, G.; Wei, Q.; Sturgis, E.M.; Ginsberg, L.E. Differences in Imaging Characteristics of HPV-Positive and HPV-Negative Oropharyngeal Cancers: A Blinded Matched-Pair Analysis. Am. J. Neuroradiol. 2013, 34, 2005–2009. [Google Scholar] [CrossRef] [PubMed]
  97. Drzymala, R.E.; Mohan, R.; Brewster, L.; Chu, J.; Goitein, M.; Harms, W.; Urie, M. Dose-Volume Histograms. Int. J. Radiat. Oncol. Biol. Phys. 1991, 21, 71–78. [Google Scholar] [CrossRef]
  98. Placidi, L.; Gioscio, E.; Garibaldi, C.; Rancati, T.; Fanizzi, A.; Maestri, D.; Massafra, R.; Menghi, E.; Mirandola, A.; Reggiori, G.; et al. A Multicentre Evaluation of Dosiomics Features Reproducibility, Stability and Sensitivity. Cancers 2021, 13, 3835. [Google Scholar] [CrossRef]
  99. Wu, A.; Li, Y.; Qi, M.; Lu, X.; Jia, Q.; Guo, F.; Dai, Z.; Liu, Y.; Chen, C.; Zhou, L.; et al. Dosiomics Improves Prediction of Locoregional Recurrence for Intensity Modulated Radiotherapy Treated Head and Neck Cancer Cases. Oral. Oncol. 2020, 104, 104625. [Google Scholar] [CrossRef]
  100. van der Lee, M.; Kriek, M.; Guchelaar, H.; Swen, J.J. Technologies for Pharmacogenomics: A Review. Genes 2020, 11, 1456. [Google Scholar] [CrossRef]
  101. Matsusaka, S.; Lenz, H.-J. Pharmacogenomics of fluorouracil-based chemotherapy toxicity. Expert Opin. Drug Metab. Toxicol. 2015, 11, 811–821. [Google Scholar] [CrossRef]
  102. Relling, M.V.; Klein, T.E.; Gammal, R.S.; Whirl-Carrillo, M.; Hoffman, J.M.; Caudle, K.E. The Clinical Pharmacogenetics Implementation Consortium: 10 Years Later. Clin. Pharmacol. Ther. 2020, 107, 171–175. [Google Scholar] [CrossRef]
  103. Cecchin, E.; Roncato, R.; Guchelaar, H.J.; Toffoli, G.; Ubiquitous Pharmacogenomics Consortium. Ubiquitous Pharmacogenomics (U-PGx): The Time for Implementation is Now. An Horizon2020 Program to Drive Pharmacogenomics into Clinical Practice. Curr. Pharm. Biotechnol. 2017, 18, 204–209. [Google Scholar] [CrossRef] [PubMed]
  104. Miserocchi, G.; Mercatali, L.; Liverani, C.; De Vita, A.; Spadazzi, C.; Pieri, F.; Bongiovanni, A.; Recine, F.; Amadori, D.; Ibrahim, T. Management and potentialities of primary cancer cultures in preclinical and translational studies. J. Transl. Med. 2017, 15, 229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Lin, C.J.; Grandis, J.R.; Carey, T.E.; Gollin, S.M.; Whiteside, T.L.; Koch, W.M.; Ferris, R.L.; Lai, S.Y. Head and neck squamous cell carcinoma cell lines: Established models and rationale for selection. Head Neck 2007, 29, 163–188. [Google Scholar] [CrossRef] [Green Version]
  106. Choi, S.Y.C.; Lin, D.; Gout, P.W.; Collins, C.C.; Xu, Y.; Wang, Y. Lessons from patient-derived xenografts for better in vitro modeling of human cancer. Adv. Drug Deliv. Rev. 2014, 79, 222–237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Daniel, V.C.; Marchionni, L.; Hierman, J.S.; Rhodes, J.T.; Devereux, W.L.; Rudin, C.M.; Yung, R.; Parmigiani, G.; Dorsch, M.; Peacock, C.D.; et al. A primary xenograft model of small-cell lung cancer reveals irreversible changes in gene expression imposed by culture in vitro. Cancer Res. 2009, 69, 3364–3373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Miserocchi, G.; De Vita, A.; Mercatali, L.; Recine, F.; Liverani, C.; Spadazzi, C.; Pieri, F.; Riva, N.; Bongiovanni, A.; Casadei, R.; et al. Characterization and Drug Sensitivity of a New High-Grade Myxofibrosarcoma Cell Line. Cells 2018, 7, 186. [Google Scholar] [CrossRef] [Green Version]
  109. Cheng, H.; Yang, X.; Si, H.; Saleh, A.D.; Xiao, W.; Coupar, J.; Gollin, S.M.; Ferris, R.L.; Issaeva, N.; Yarbrough, W.G.; et al. Genomic and Transcriptomic Characterization Links Cell Lines with Aggressive Head and Neck Cancers. Cell Rep. 2018, 25, 1332–1345.e5. [Google Scholar] [CrossRef] [Green Version]
  110. De Vita, A.; Recine, F.; Mercatali, L.; Miserocchi, G.; Liverani, C.; Spadazzi, C.; Casadei, R.; Bongiovanni, A.; Pieri, F.; Riva, N.; et al. Myxofibrosarcoma primary cultures: Molecular and pharmacological profile. Ther. Adv. Med. Oncol. 2017, 9, 755–767. [Google Scholar] [CrossRef] [Green Version]
  111. De Vita, A.; Recine, F.; Mercatali, L.; Miserocchi, G.; Spadazzi, C.; Liverani, C.; Bongiovanni, A.; Pieri, F.; Casadei, R.; Riva, N.; et al. Primary Culture of Undifferentiated Pleomorphic Sarcoma: Molecular Characterization and Response to Anticancer Agents. Int. J. Mol. Sci. 2017, 18, 2662. [Google Scholar] [CrossRef] [Green Version]
  112. Zanoni, M.; Sarti, A.C.; Zamagni, A.; Cortesi, M.; Pignatta, S.; Arienti, C.; Tebaldi, M.; Sarnelli, A.; Romeo, A.; Bartolini, D.; et al. Irradiation causes senescence, ATP release, and P2X7 receptor isoform switch in glioblastoma. Cell Death Dis. 2022, 24, 80. [Google Scholar] [CrossRef]
  113. Owen, J.H.; Graham, M.P.; Chinn, S.B.; Darr, O.F.; Chepeha, D.B.; Wolf, G.T.; Bradford, C.R.; Carey, T.E.; Prince, M.E.P. Novel method of cell line establishment utilizing fluorescence-activated cell sorting resulting in 6 new head and neck squamous cell carcinoma lines. Head Neck 2016, 38 (Suppl. 1), E459–E467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Garcia-Inclan, C.; López-Hernández, A.; Alonso-Guervós, M.; Allonca, E.; Potes, S.; Melón, S.; López, F.; Llorente, J.L.; Hermsen, M. Establishment and genetic characterization of six unique tumor cell lines as preclinical models for sinonasal squamous cell carcinoma. Sci. Rep. 2014, 4, 4925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Lee, D.H.; Lee, J.K. Establishment of a Cell Line (CNUH-HNSCC-1) Derived from an Advanced Laryngeal Squamous Cell Carcinoma. Chonnam. Med. J. 2011, 47, 85–89. [Google Scholar] [CrossRef] [PubMed]
  116. De Vita, A.; Mercatali, L.; Miserocchi, G.; Liverani, C.; Spadazzi, C.; Recine, F.; Bongiovanni, A.; Pieri, F.; Cavaliere, D.; Fausti, V.; et al. Establishment of a Primary Culture of Patient-derived Soft Tissue Sarcoma. J. Vis. Exp. 2018, 56767. [Google Scholar] [CrossRef]
  117. Svobodova, M.; Raudenska, M.; Gumulec, J.; Balvan, J.; Fojtu, M.; Kratochvilova, M.; Polanska, H.; Horakova, Z.; Kostrica, R.; Babula, P.; et al. Establishment of oral squamous cell carcinoma cell line and magnetic bead-based isolation and characterization of its CD90/CD44 subpopulations. Oncotarget 2017, 8, 66254–66269. [Google Scholar] [CrossRef] [Green Version]
  118. Pozzi, V.; Sartini, D.; Rocchetti, R.; Santarelli, A.; Rubini, C.; Morganti, S.; Giuliante, R.; Calabrese, S.; Di Ruscio, G.; Orlando, F.; et al. Identification and characterization of cancer stem cells from head and neck squamous cell carcinoma cell lines. Cell. Physiol. Biochem. 2015, 36, 784–798. [Google Scholar] [CrossRef]
  119. De Vita, A.; Miserocchi, G.; Recine, F.; Mercatali, L.; Pieri, F.; Medri, L.; Bongiovanni, A.; Cavaliere, D.; Liverani, C.; Spadazzi, C.; et al. Activity of Eribulin in a Primary Culture of Well-Differentiated/Dedifferentiated Adipocytic Sarcoma. Molecules 2016, 21, 1662. [Google Scholar] [CrossRef] [Green Version]
  120. Liebertz, D.J.; Lechner, M.G.; Masood, R.; Sinha, U.K.; Han, J.; Puri, R.K.; Correa, A.J.; Epstein, A.L. Establishment and characterization of a novel head and neck squamous cell carcinoma cell line USC-HN1. Head Neck Oncol. 2010, 2, 5. [Google Scholar] [CrossRef] [Green Version]
  121. Ishiguro, T.; Ohata, H.; Sato, A.; Yamawaki, K.; Enomoto, T.; Okamoto, K. Tumor-derived spheroids: Relevance to cancer stem cells and clinical applications. Cancer Sci. 2017, 108, 283–289. [Google Scholar] [CrossRef] [Green Version]
  122. Oppel, F.; Shao, S.; Schürmann, M.; Goon, P.; Albers, A.E.; Sudhoff, H. An Effective Primary Head and Neck Squamous Cell Carcinoma In Vitro Model. Cells 2019, 8, 555. [Google Scholar] [CrossRef] [Green Version]
  123. Dong, Y.; Ochsenreither, S.; Cai, C.; Kaufmann, A.M.; Albers, A.E.; Qian, X. Aldehyde dehydrogenase 1 isoenzyme expression as a marker of cancer stem cells correlates to histopathological features in head and neck cancer: A meta-analysis. PLoS ONE 2017, 12, e0187615. [Google Scholar] [CrossRef] [Green Version]
  124. Di Donato, M.; Zamagni, A.; Galasso, G.; Di Zazzo, E.; Giovannelli, P.; Barone, M.V.; Zanoni, M.; Gunelli, R.; Costantini, M.; Auricchio, F.; et al. The androgen receptor/filamin A complex as a target in prostate cancer microenvironment. Cell Death Dis. 2021, 12, 127. [Google Scholar] [CrossRef]
  125. Tang, A.L.; Hauff, S.J.; Owen, J.H.; Graham, M.P.; Czerwinski, M.J.; Park, J.J.; Walline, H.; Papagerakis, S.; Stoerker, J.; McHugh, J.B.; et al. UM-SCC-104: A new human papillomavirus-16-positive cancer stem cell-containing head and neck squamous cell carcinoma cell line. Head Neck 2012, 34, 1480–1491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Ehrenberg, K.R.; Gao, J.; Oppel, F.; Frank, S.; Kang, N.; Dieter, S.M.; Herbst, F.; Möhrmann, L.; Dubash, T.D.; Schulz, E.R.; et al. Systematic Generation of Patient-Derived Tumor Models in Pancreatic Cancer. Cells 2019, 8, 142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Chang, C.W.; Chen, Y.S.; Chou, S.H.; Han, C.L.; Chen, Y.J.; Yang, C.C.; Huang, C.Y.; Lo, J.F. Distinct subpopulations of head and neck cancer cells with different levels of intracellular reactive oxygen species exhibit diverse stemness, proliferation, and chemosensitivity. Cancer Res. 2014, 74, 6291–6305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Egeblad, M.; Nakasone, E.S.; Werb, Z. Tumors as organs: Complex tissues that interface with the entire organism. Dev. Cell 2010, 18, 884–901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Zanoni, M.; Cortesi, M.; Zamagni, A.; Arienti, C.; Pignatta, S.; Tesei, A. Modeling neoplastic disease with spheroids and organoids. J. Hematol. Oncol. 2020, 13, 97. [Google Scholar] [CrossRef] [PubMed]
  130. Cortesi, M.; Zanoni, M.; Pirini, F.; Tumedei, M.M.; Ravaioli, S.; Rapposelli, I.G.; Frassineti, G.L.; Bravaccini, S. Pancreatic Cancer and Cellular Senescence: Tumor Microenvironment under the Spotlight. Int. J. Mol. Sci. 2021, 23, 254. [Google Scholar] [CrossRef]
  131. Miserocchi, G.; Cocchi, C.; De Vita, A.; Liverani, C.; Spadazzi, C.; Calpona, S.; Di Menna, G.; Bassi, M.; Meccariello, G.; De Luca, G.; et al. Three-dimensional collagen-based scaffold model to study the microenvironment and drug-resistance mechanisms of oropharyngeal squamous cell carcinomas. Cancer Biol. Med. 2021, 18, 502–516. [Google Scholar] [CrossRef]
  132. Rijal, G.; Li, W. 3D scaffolds in breast cancer research. Biomaterials 2016, 81, 135–156. [Google Scholar] [CrossRef]
  133. Egeblad, M.; Rasch, M.G.; Weaver, V.M. Dynamic interplay between the collagen scaffold and tumor evolution. Curr. Opin. Cell Biol. 2010, 22, 697–706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Liverani, C.; De Vita, A.; Minardi, S.; Kang, Y.; Mercatali, L.; Amadori, D.; Bongiovanni, A.; La Manna, F.; Ibrahim, T.; Tasciotti, E. A biomimetic 3D model of hypoxia-driven cancer progression. Sci. Rep. 2019, 9, 12263. [Google Scholar] [CrossRef] [Green Version]
  135. Liverani, C.; De Vita, A.; Spadazzi, C.; Miserocchi, G.; Cocchi, C.; Bongiovanni, A.; De Lucia, A.; La Manna, F.; Fabbri, F.; Tebaldi, M.; et al. Lineage-specific mechanisms and drivers of breast cancer chemoresistance revealed by 3D biomimetic culture. Mol. Oncol. 2022, 16, 921–939. [Google Scholar] [PubMed]
  136. Zhang, M.; Rose, B.; Soon Lee, C.; Hong, A.M. In vitro 3-dimensional tumor model for radiosensitivity of HPV positive OSCC cell lines. Cancer Biol. Ther. 2015, 16, 1231–1240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Thoma, C.R.; Zimmermann, M.; Agarkova, I.; Kelm, J.M.; Krek, W. 3D cell culture systems modeling tumor growth determinants in cancer target discovery. Adv. Drug Deliv. Rev. 2014, 69–70, 29–41. [Google Scholar] [CrossRef] [PubMed]
  138. Piccinini, F.; Tesei, A.; Zanoni, M.; Bevilacqua, A. ReViMS: Software tool for estimating the volumes of 3-D multicellular spheroids imaged using a light sheet fluorescence microscope. Biotechniques 2017, 63, 227–229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Han, S.J.; Kwon, S.; Kim, K.S. Challenges of applying multicellular tumor spheroids in preclinical phase. Cancer Cell Int. 2021, 21, 152. [Google Scholar] [CrossRef]
  140. Colliez, F.; Gallez, B.; Jordan, B.F. Assessing Tumor Oxygenation for Predicting Outcome in Radiation Oncology: A Review of Studies Correlating Tumor Hypoxic Status and Outcome in the Preclinical and Clinical Settings. Front. Oncol. 2017, 7, 10. [Google Scholar] [CrossRef] [Green Version]
  141. Peirsman, A.; Blondeel, E.; Ahmed, T.; Anckaert, J.; Audenaert, D.; Boterberg, T.; Buzas, K.; Carragher, N.; Castellani, G.; Castro, F.; et al. MISpheroID: A knowledgebase and transparency tool for minimum information in spheroid identity. Sci. Rep. 2016, 6, 19103, Erratum in Nat. Methods 2021, 18, 1294–01303. [Google Scholar] [CrossRef]
  142. Frankel, A.; Buckman, R.; Kerbel, R.S. Abrogation of taxol-induced G2-M arrest and apoptosis in human ovarian cancer cells grown as multicellular tumor spheroids. Cancer Res. 1997, 57, 2388–2393. [Google Scholar]
  143. Dubessy, C.; Merlin, J.M.; Marchal, C.; Guillemin, F. Spheroids in radiobiology and photodynamic therapy. Crit. Rev. Oncol. Hematol. 2000, 36, 179–192. [Google Scholar] [PubMed]
  144. Drost, J.; Clevers, H. Organoids in cancer research. Nat. Rev. Cancer 2018, 18, 407–418. [Google Scholar] [CrossRef] [PubMed]
  145. Zanoni, M.; Pignatta, S.; Arienti, C.; Bonafè, M.; Tesei, A. Anticancer drug discovery using multicellular tumor spheroid models. Expert. Opin. Drug Discov. 2019, 14, 289–301. [Google Scholar] [CrossRef] [PubMed]
  146. Driehuis, E.; Spelier, S.; Hernández, I.B.; de Bree, R.; Willems, S.M.; Clevers, H.; Oliveira, S. Patient-Derived Head and Neck Cancer Organoids Recapitulate EGFR Expression Levels of Respective Tissues and Are Responsive to EGFR-Targeted Photodynamic Therapy. J. Clin. Med. 2019, 8, 1880. [Google Scholar] [CrossRef] [Green Version]
  147. Tanaka, N.; Osman, A.A.; Takahashi, Y.; Lindemann, A.; Patel, A.A.; Zhao, M.; Takahashi, H.; Myers, J.N. Head and neck cancer organoids established by modification of the CTOS method can be used to predict in vivo drug sensitivity. Oral. Oncol. 2018, 87, 49–57. [Google Scholar] [CrossRef]
  148. Lim, Y.C.; Oh, S.Y.; Cha, Y.Y.; Kim, S.H.; Jin, X.; Kim, H. Cancer stem cell traits in squamospheres derived from primary head and neck squamous cell carcinomas. Oral. Oncol. 2011, 47, 83–91. [Google Scholar] [CrossRef]
  149. Driehuis, E.; Kolders, S.; Spelier, S.; Lõhmussaar, K.; Willems, S.M.; Devriese, L.A.; de Bree, R.; de Ruiter, E.J.; Korving, J.; Begthel, H.; et al. Oral Mucosal Organoids as a Potential Platform for Personalized Cancer Therapy. Cancer Discov. 2019, 9, 852–871. [Google Scholar]
  150. Lee, T.W.; Lai, A.; Harms, J.K.; Singleton, D.C.; Dickson, B.D.; Macann, A.M.J.; Hay, M.P.; Jamieson, S.M.F. Patient-Derived Xenograft and Organoid Models for Precision Medicine Targeting of the Tumour Microenvironment in Head and Neck Cancer. Cancers 2020, 12, 3743. [Google Scholar]
  151. Affolter, A.; Lammert, A.; Kern, J.; Scherl, C.; Rotter, N. Precision Medicine Gains Momentum: Novel 3D Models and Stem Cell-Based Approaches in Head and Neck Cancer. Front. Cell Dev. Biol. 2021, 8, 666515. [Google Scholar] [CrossRef]
  152. Powley, I.R.; Powley, I.R.; Patel, M.; Miles, G.; Pringle, H.; Howells, L.; Thomas, A.; Kettleborough, C.; Bryans, J.; Hammonds, T.; et al. Patient-derived explants (PDEs) as a powerful preclinical platform for anti-cancer drug and biomarker discovery. Br. J. Cancer 2020, 122, 735–744. [Google Scholar] [CrossRef] [Green Version]
  153. Leighton, J. A sponge matrix method for tissue culture; formation of organized aggregates of cells in vitro. J. Natl. Cancer Inst. 1951, 12, 545–561. [Google Scholar] [PubMed]
  154. Leighton, J. The growth patterns of some transplantable animal tumors in sponge matrix tissue culture. J. Natl. Cancer Inst. 1954, 15, 275–293. [Google Scholar] [PubMed]
  155. Vescio, R.A.; Connors, K.M.; Kubota, T.; Hoffman, R.M. Correlation of histology and drug response of human tumors grown in native-state three-dimensional histoculture and in nude mice. Proc. Natl Acad. Sci. USA 1991, 88, 5163–5166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Yoshimasu, T.; Oura, S.; Hirai, I.; Tamaki, T.; Kokawa, Y.; Hata, K.; Ohta, F.; Nakamura, R.; Kawago, M.; Tanino, H.; et al. Data acquisition for the histoculture drug response assay in lung cancer. J. Thorac. Cardiovasc. Surg. 2007, 133, 303–308. [Google Scholar] [CrossRef] [Green Version]
  157. Furukawa, T.; Kubota, T.; Hoffman, R.M. Clinical applications of the histoculture drug response assay. Clin. Cancer Res. 1995, 1, 305–311. [Google Scholar]
  158. Majumder, B.; Baraneedharan, U.; Thiyagarajan, S.; Radhakrishnan, P.; Narasimhan, H.; Dhandapani, M.; Brijwani, N.; Pinto, D.D.; Prasath, A.; Shanthappa, B.U.; et al. Predicting clinical response to anticancer drugs using an ex vivo platform that captures tumour heterogeneity. Nat. Commun. 2015, 6, 6169. [Google Scholar] [CrossRef] [Green Version]
  159. Ghosh, S.; Prasad, M.; Kundu, K.; Cohen, L.; Yegodayev, K.M.; Zorea, J.; Joshua, B.Z.; Lasry, B.; Dimitstein, O.; Bahat-Dinur, A.; et al. Tumor Tissue Explant Culture of Patient-Derived Xenograft as Potential Prioritization Tool for Targeted Therapy. Front. Oncol. 2019, 9, 17. [Google Scholar] [CrossRef] [Green Version]
  160. Griswold, D.P.; Corbett, T.H. A colon tumor model for anticancer agent evaluation. Cancer 1975, 36, 2441–2444. [Google Scholar] [CrossRef]
  161. Wang, Y.; Tseng, J.C.; Sun, Y.; Beck, A.H.; Kung, A.L. Noninvasive Imaging of Tumor Burden and Molecular Pathways in Mouse Models of Cancer. Cold Spring Harb. Protoc. 2015, 2015, 135–144. [Google Scholar] [CrossRef] [Green Version]
  162. Flanagan, S.P. ‘Nude’, a new hairless gene with pleiotropic effects in the mouse. Genet. Res. 1966, 8, 295–309. [Google Scholar] [CrossRef] [Green Version]
  163. Braakhuis, B.J.M.; Sneeuwloper, G.; Snow, G.B. The potential of the nude mouse xenograft model for the study of head and neck cancer. Arch. Otorhinolaryngol. 1984, 239, 69–79. [Google Scholar] [CrossRef] [PubMed]
  164. Walcher, L.; Müller, C.; Hilger, N.; Kretschmer, A.; Stahl, L.; Wigge, S.; Rengelshausen, J.; Müller, A.M.; Fricke, S. Effect of combined sublethal X-ray irradiation and cyclosporine A treatment in NOD scid gamma (NSG) mice. Exp. Anim. 2019, 68, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Peng, S.; Creighton, C.J.; Zhang, Y.; Sen, B.; Mazumdar, T.; Myers, J.N.; Lai, S.Y.; Woolfson, A.; Lorenzi, M.V.; Bell, D.; et al. Tumor grafts derived from patients with head and neck squamous carcinoma authentically maintain the molecular and histologic characteristics of human cancers. J. Transl. Med. 2013, 11, 198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Li, H.; Wheeler, S.; Park, Y.; Ju, Z.; Thomas, S.M.; Fichera, M.; Egloff, A.M.; Lui, V.W.; Duvvuri, U.; Bauman, J.E.; et al. Proteomic characterization of head and neck cancer patient-derived xenografts. Mol. Cancer Res. 2016, 14, 278–286. [Google Scholar] [CrossRef] [Green Version]
  167. Karamboulas, C.; Bruce, J.P.; Hope, A.J.; Meens, J.; Huang, S.H.; Erdmann, N.; Hyatt, E.; Pereira, K.; Goldstein, D.P.; Weinreb, I.; et al. Patient-Derived Xenografts for Prognostication and Personalized Treatment for Head and Neck Squamous Cell Carcinoma. Cell Rep. 2018, 25, 1318–1331.e4. [Google Scholar] [CrossRef] [Green Version]
  168. Kang, H.N.; Kim, J.H.; Park, A.Y.; Choi, J.W.; Lim, S.M.; Kim, J.; Shin, E.J.; Hong, M.H.; Pyo, K.H.; Yun, M.R.; et al. Establishment and characterization of patient-derived xenografts as paraclinical models for head and neck cancer. BMC Cancer 2020, 20, 316. [Google Scholar] [CrossRef] [Green Version]
  169. Campbell, K.M.; Lin, T.; Zolkind, P.; Barnell, E.K.; Skidmore, Z.L.; Winkler, A.E.; Law, J.H.; Mardis, E.R.; Wartman, L.D.; Adkins, D.R.; et al. Oral Cavity Squamous Cell Carcinoma Xenografts Retain Complex Genotypes and Intertumor Molecular Heterogeneity. Cell Rep. 2018, 24, 2167–2178. [Google Scholar] [CrossRef] [Green Version]
  170. Kimple, R.J.; Harari, P.M.; Torres, A.D.; Yang, R.Z.; Soriano, B.J.; Yu, M.; Armstrong, E.A.; Blitzer, G.C.; Smith, M.A.; Lorenz, L.D.; et al. Development and characterization of HPV-positive and HPV-negative head and neck squamous cell carcinoma tumorgrafts. Clin. Cancer Res. 2013, 19, 855–864. [Google Scholar] [CrossRef] [Green Version]
  171. Keysar, S.B.; Astling, D.P.; Anderson, R.T.; Vogler, B.W.; Bowles, D.W.; Morton, J.J.; Paylor, J.J.; Glogowska, M.J.; Le, P.N.; Eagles-Soukup, J.R.; et al. A patient tumor transplant model of squamous cell cancer identifies PI3K inhibitors as candidate therapeutics in defined molecular bins. Mol. Oncol. 2013, 7, 776–790. [Google Scholar] [CrossRef]
  172. Swick, A.D.; Stein, A.P.; McCulloch, T.M.; Hartig, G.K.; Ong, I.M.; Sampene, E.; Prabakaran, P.J.; Liu, C.Z.; Kimple, R.J. Defining the boundaries and expanding the utility of head and neck cancer patient derived xenografts. Oral. Oncol. 2017, 64, 65–72. [Google Scholar] [CrossRef] [Green Version]
  173. Izumchenko, E.; Paz, K.; Ciznadija, D.; Sloma, I.; Katz, A.; Vasquez-Dunddel, D.; Ben-Zvi, I.; Stebbing, K.; McGuire, W.; Harris, W.; et al. Patient-derived xenografts effectively capture responses to oncology therapy in a heterogeneous cohort of patients with solid tumors. Ann. Oncol. 2017, 28, 2595–2605. [Google Scholar] [CrossRef] [PubMed]
  174. Chia, S.; Low, J.L.; Zhang, X.; Kwang, X.L.; Chong, F.T.; Sharma, A.; Bertrand, D.; Toh, S.Y.; Leong, H.S.; Thangavelu, M.T.; et al. Phenotype-driven precision oncology as a guide for clinical decisions one patient at a time. Nat. Commun. 2017, 8, 435. [Google Scholar] [CrossRef] [PubMed]
  175. Lilja-Fischer, J.K.; Ulhøi, B.P.; Alsner, J.; Stougaard, M.; Thomsen, M.S.; Busk, M.; Lassen, P.; Steiniche, T.; Nielsen, V.E.; Overgaard, J. Characterization and radiosensitivity of HPV-related oropharyngeal squamous cell carcinoma patient-derived xenografts. Acta Oncol. 2019, 58, 1489–1494. [Google Scholar] [CrossRef] [PubMed]
  176. Huang, C.; Chen, L.; Savage, S.R.; Eguez, R.V.; Dou, Y.; Li, Y.; da Veiga Leprevost, F.; Jaehnig, E.J.; Lei, J.T.; Wenet, B.; et al. Proteogenomic insights into the biology and treatment of HPV-negative head and neck squamous cell carcinoma. Cancer Cell 2021, 39, 361–379.e16. [Google Scholar] [CrossRef]
  177. Hinshaw, D.C.; Shevde, L.A. The tumor microenvironment innately modulates cancer progression. Cancer Res. 2019, 79, 4557–4567. [Google Scholar] [CrossRef] [Green Version]
  178. Byrne, A.T.; Alférez, D.G.; Amant, F.; Annibali, D.; Arribas, J.; Biankin, A.V.; Bruna, A.; Budinská, E.; Caldas, C.; Chang, D.K.; et al. Interrogating open issues in cancer precision medicine with patient-derived xenografts. Nat. Rev. Cancer 2017, 17, 254–268. [Google Scholar] [CrossRef]
  179. Morton, J.J.; Bird, G.; Keysar, S.B.; Astling, D.P.; Lyons, T.R.; Anderson, R.T.; Glogowska, M.J.; Estes, P.; Eagles, J.R.; Le, P.N.; et al. XactMice: Humanizing mouse bone marrow enables microenvironment reconstitution in a patient-derived xenograft model of head and neck cancer. Oncogene 2016, 35, 290–300. [Google Scholar] [CrossRef] [Green Version]
  180. Eirew, P.; Steif, A.; Khattra, J.; Ha, G.; Yap, D.; Farahani, H.; Gelmon, K.; Chia, S.; Mar, C.; Wan, A.; et al. Dynamics of genomic clones in breast cancer patient xenografts at single-cell resolution. Nature 2015, 518, 422–426. [Google Scholar] [CrossRef]
  181. Cosper, P.F.; Abel, L.; Lee, Y.S.; Paz, C.; Kaushik, S.; Nickel, K.P.; Alexandridis, R.; Scott, J.G.; Bruce, J.Y.; Kimple, R.J. Patient derived models to study head and neck cancer radiation response. Cancers 2020, 12, 419. [Google Scholar] [CrossRef] [Green Version]
  182. Fazio, M.; Ablain, J.; Chuan, Y.; Langenau, D.M.; Zon, L.I. Zebrafish patient avatars in cancer biology and precision cancer therapy. Nat. Rev. Cancer 2020, 20, 263–273. [Google Scholar] [CrossRef]
  183. Haldi, M.; Ton, C.; Seng, W.L.; McGrath, P. Human melanoma cells transplanted into zebrafish proliferate, migrate, produce melanin, form masses and stimulate angiogenesis in zebrafish. Angiogenesis 2006, 9, 139–151. [Google Scholar] [CrossRef] [PubMed]
  184. Lin, J.; Zhang, W.; Zhao, J.J.; Kwart, A.H.; Yang, C.; Ma, D.; Ren, X.; Tai, Y.T.; Anderson, K.C.; Handin, R.I.; et al. A clinically relevant in vivo zebrafish model of human multiple myeloma to study preclinical therapeutic efficacy. Blood 2016, 128, 249–252. [Google Scholar] [CrossRef] [PubMed]
  185. Xiao, J.; Glasgow, E.; Agarwal, S. Zebrafish Xenografts for Drug Discovery and Personalized Medicine. Trends Cancer 2020, 6, 569–579. [Google Scholar] [CrossRef] [PubMed]
  186. White, R.M.; Sessa, A.; Burke, C.; Bowman, T.; LeBlanc, J.; Ceol, C.; Bourque, C.; Dovey, M.; Goessling, W.; Burns, C.E.; et al. Transparent adult zebrafish as a tool for in vivo transplantation analysis. Cell Stem Cell 2008, 2, 183–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Smith, A.C.H.; Raimondi, A.R.; Salthouse, C.D.; Ignatius, M.S.; Blackburn, J.S.; Mizgirev, I.V.; Storer, N.Y.; de Jong, J.L.O.; Chen, A.T.; Zhou, Y.; et al. High- throughput cell transplantation establishes that tumor- initiating cells are abundant in zebrafish T- cell acute lymphoblastic leukemia. Blood 2010, 115, 3296–3303. [Google Scholar] [CrossRef] [Green Version]
  188. Costa, B.; Estrada, M.F.; Mendes, R.V.; Fior, R. Zebrafish Avatars towards Personalized Medicine—A Comparative Review between Avatar Models. Cells 2020, 9, 293. [Google Scholar] [CrossRef] [Green Version]
  189. Bentley, V.L.; Veinotte, C.J.; Corkery, D.P.; Pinder, J.B.; LeBlanc, M.A.; Bedard, K.; Weng, A.P.; Berman, J.N.; Dellaire, G. Focused chemical genomics using zebrafish xenotransplantation as a pre-clinical therapeutic platform for T-cell acute lymphoblastic leukemia. Haematologica 2015, 100, 70–76. [Google Scholar] [CrossRef]
  190. Trede, N.S.; Langenau, D.M.; Traver, D.; Look, A.T.; Zon, L.I. The use of zebrafish to understand immunity. Immunity 2004, 20, 367–379. [Google Scholar] [CrossRef] [Green Version]
  191. Mercatali, L.; La Manna, F.; Groenewoud, A.; Casadei, R.; Recine, F.; Miserocchi, G.; Pieri, F.; Liverani, C.; Bongiovanni, A.; Spadazzi, C.; et al. Development of a patient-derived xenograft (PDX) of breast cancer bone metastasis in a zebrafish model. Int. J. Mol. Sci. 2016, 17, 1375. [Google Scholar] [CrossRef]
  192. Mizgireuv, I.V.; Revskoy, S.Y. Transplantable tumor lines generated in clonal zebrafish. Cancer Res. 2006, 66, 3120–3125. [Google Scholar] [CrossRef] [Green Version]
  193. Sabaawy, H.E.; Azuma, M.; Embree, L.J.; Tsai, H.J.; Starost, M.F.; Hickstein, D.D. TEL-AML1 transgenic zebrafish model of precursor B cell acute lymphoblastic leukemia. Proc. Natl. Acad. Sci. USA 2006, 103, 15166–15171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Beghini, A.; Corlazzoli, F.; Del Giacco, L.; Re, M.; Lazzaroni, F.; Brioschi, M.; Valentini, G.; Ferrazzi, F.; Ghilardi, A.; Righi, M.; et al. Regeneration-associated WNT signaling is activated in long-term reconstituting AC133bright acute myeloid leukemia cells 1,2. Neoplasia 2012, 14, 1236–2124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Marques, I.J.; Weiss, F.U.; Vlecken, D.H.; Nitsche, C.; Bakkers, J.; Lagendijk, A.K.; Partecke, L.I.; Heidecke, C.D.; Lerch, M.M.; Bagowski, C.P. Metastatic behaviour of primary human tumours in a zebrafish xenotransplantation model. BMC Cancer 2009, 9, 128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Weiss, F.U.; Marques, I.J.; Woltering, J.M.; Vlecken, D.H.; Aghdassi, A.; Partecke, L.I.; Heidecke, C.D.; Lerch, M.M.; Bagowski, C.P. Retinoic acid receptor antagonists inhibit miR-10a expression and block metastatic behavior of pancreatic cancer. Gastroenterology 2009, 137, 2136–2145.e7. [Google Scholar] [CrossRef] [PubMed]
  197. Eden, C.J.; Ju, B.; Murugesan, M.; Phoenix, T.N.; Nimmervoll, B.; Tong, Y.; Ellison, D.W.; Finkelstein, D.; Wright, K.; Boulos, N.; et al. Orthotopic models of pediatric brain tumors in zebrafish. Oncogene 2015, 34, 1736–1742. [Google Scholar] [CrossRef] [Green Version]
  198. Wenger, A.; Larsson, S.; Danielsson, A.; Elbæk, K.J.; Kettunen, P.; Tisell, M.; Sabel, M.; Lannering, B.; Nordborg, C.; Schepke, E.; et al. Stem cell cultures derived from pediatric brain tumors accurately model the originating tumors. Oncotarget 2017, 8, 18626–18639. [Google Scholar] [CrossRef] [Green Version]
  199. Gaudenzi, G.; Albertelli, M.; Dicitore, A.; Würth, R.; Gatto, F.; Barbieri, F.; Cotelli, F.; Florio, T.; Ferone, D.; Persani, L.; et al. Patient-derived xenograft in zebrafish embryos: A new platform for translational research in neuroendocrine tumors. Endocrine 2016, 57, 214–219. [Google Scholar] [CrossRef]
  200. Pruvot, B.; Jacquel, A.; Droin, N.; Auberger, P.; Bouscary, D.; Tamburini, J.; Muller, M.; Fontenay, M.; Chluba, J.; Solary, E. Leukemic cell xenograft in zebrafish embryo for investigating drug efficacy. Haematologica 2011, 96, 612–616. [Google Scholar] [CrossRef] [Green Version]
  201. Cirello, V.; Gaudenzi, G.; Grassi, E.S.; Colombo, C.; Vicentini, L.; Ferrero, S.; Persani, L.; Vitale, G.; Fugazzola, L. Tumor and normal thyroid spheroids: From tissues to zebrafish. Minerva Endocrinol. 2017, 43, 1–10. [Google Scholar] [CrossRef]
  202. Peverelli, E.; Giardino, E.; Treppiedi, D.; Meregalli, M.; Belicchi, M.; Vaira, V.; Corbetta, S.; Verdelli, C.; Verrua, E.; Serbanet, A.L.; et al. Dopamine receptor type 2 (DRD2) and somatostatin receptor type 2 (SSTR2) agonists are effective in inhibiting proliferation of progenitor/stem-like cells isolated from nonfunctioning pituitary tumors. Int. J. Cancer 2017, 140, 1870–1880. [Google Scholar] [CrossRef]
  203. Würth, R.; Barbieri, F.; Pattarozzi, A.; Gaudenzi, G.; Gatto, F.; Fiaschi, P.; Ravetti, J.L.; Zona, G.; Daga, A.; Persani, L.; et al. Phenotypical and pharmacological characterization of stem-like cells in human pituitary adenoma. Mol. Neurobiol. 2016, 54, 4879–4895. [Google Scholar] [CrossRef] [PubMed]
  204. Bansal, N.; Davis, S.; Tereshchenko, I.; Budak-Alpdogan, T.; Zhong, H.; Stein, M.N.; Kim, I.Y.; Dipaola, R.S.; Bertino, J.R.; Sabaawy, H.E. Enrichment of human prostate cancer cells with tumor initiating properties in mouse and zebrafish xenografts by differential adhesion. Prostate 2014, 74, 187–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Langenau, D.M.; Keefe, M.D.; Storer, N.Y.; Guyon, J.R.; Kutok, J.L.; Le, X.; Goessling, W.; Neuberg, D.S.; Kunkel, L.M.; Zon, L.I. Effects of RAS on the genesis of embryonal rhabdomyosarcoma. Genes Dev. 2007, 21, 1382–1395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Gill, J.A.; Lowe, L.; Nguyen, J.; Liu, P.P.; Blake, T.; Venkatesh, B.; Aplan, P.D. Enforced expression of Simian virus 40 large T-antigen leads to testicular germ cell tumors in zebrafish. Zebrafish 2010, 7, 333–341. [Google Scholar] [CrossRef]
  207. Al-Samadi, A.; Tuomainen, K.; Kivimäki, A.; Salem, A.; Al-Kubati, S.; Hyytiäinen, A.; Parikka, M.; Mesimäki, K.; Wilkman, T.; Mäkitie, A.; et al. PCR-based zebrafsh model for personalised medicine in head and neck cancer. J. Transl. Med. 2019, 17, 235. [Google Scholar] [CrossRef]
  208. Usai, A.; Di Franco, G.; Colucci, P.; Pollina, L.E.; Vasile, E.; Funel, N.; Palmeri, M.; Dente, L.; Falcone, A.; Morelli, L.; et al. A Model of a Zebrafish Avatar for Co-Clinical Trials. Cancers 2020, 12, 677. [Google Scholar] [CrossRef] [Green Version]
  209. Usai, A.; Di Franco, G.; Piccardi, M.; Cateni, P.; Pollina, L.; Vivaldi, C.; Vasile, E.; Funel, N.; Palmeri, M.; Dente, L.; et al. Zebrafish Patient-Derived Xenografts Identify Chemo-Response in Pancreatic Ductal Adenocarcinoma Patients. Cancers 2021, 13, 4131. [Google Scholar] [CrossRef]
Figure 1. The two main precision medicine options in HNC oncology: genomic and drug screening approaches. Image created with BioRender.com.
Figure 1. The two main precision medicine options in HNC oncology: genomic and drug screening approaches. Image created with BioRender.com.
Jpm 12 00854 g001
Table 1. Main characteristics of in vitro and in vivo preclinical cancer models.
Table 1. Main characteristics of in vitro and in vivo preclinical cancer models.
2D Cell Line Cultures2D Primary CulturesBiomimetic Scaffold CulturesOrganoidsMouse PDXsZebrafish PDXs
Easy manipulation++++++++++
CostVery lowVery lowLowMediumHighLow
Experiment durationDaysDaysDaysDays/MonthsMonthsDays
Number of cells needed for drug screening assaysLowLowHighMediumHighLow
Drug screening throughtput+++++++++++++++
Cancer subclones conservation/+++++++
TME concervation/+++++++++
Immune components conservation/+++++
(+) sufficient, (++) good, (+++) optimal, (/) not suitable.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Miserocchi, G.; Spadazzi, C.; Calpona, S.; De Rosa, F.; Usai, A.; De Vita, A.; Liverani, C.; Cocchi, C.; Vanni, S.; Calabrese, C.; et al. Precision Medicine in Head and Neck Cancers: Genomic and Preclinical Approaches. J. Pers. Med. 2022, 12, 854. https://0-doi-org.brum.beds.ac.uk/10.3390/jpm12060854

AMA Style

Miserocchi G, Spadazzi C, Calpona S, De Rosa F, Usai A, De Vita A, Liverani C, Cocchi C, Vanni S, Calabrese C, et al. Precision Medicine in Head and Neck Cancers: Genomic and Preclinical Approaches. Journal of Personalized Medicine. 2022; 12(6):854. https://0-doi-org.brum.beds.ac.uk/10.3390/jpm12060854

Chicago/Turabian Style

Miserocchi, Giacomo, Chiara Spadazzi, Sebastiano Calpona, Francesco De Rosa, Alice Usai, Alessandro De Vita, Chiara Liverani, Claudia Cocchi, Silvia Vanni, Chiara Calabrese, and et al. 2022. "Precision Medicine in Head and Neck Cancers: Genomic and Preclinical Approaches" Journal of Personalized Medicine 12, no. 6: 854. https://0-doi-org.brum.beds.ac.uk/10.3390/jpm12060854

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