Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 15399 2023-11-09 18:07:47 |
2 The text of the manuscript has been abridged and proofread according to the editors' recommendations -8564 word(s) 6835 2023-11-11 11:06:27 | |
3 Alignment in table cells has been edited Meta information modification 6835 2023-11-11 11:20:19 | |
4 format + 179 word(s) 7014 2023-11-20 07:15:59 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Arutyunyan, I.; Jumaniyazova, E.; Makarov, A.; Fatkhudinov, T. In Vitro Models of Head and Neck Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/51382 (accessed on 02 September 2024).
Arutyunyan I, Jumaniyazova E, Makarov A, Fatkhudinov T. In Vitro Models of Head and Neck Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/51382. Accessed September 02, 2024.
Arutyunyan, Irina, Enar Jumaniyazova, Andrey Makarov, Timur Fatkhudinov. "In Vitro Models of Head and Neck Cancer" Encyclopedia, https://encyclopedia.pub/entry/51382 (accessed September 02, 2024).
Arutyunyan, I., Jumaniyazova, E., Makarov, A., & Fatkhudinov, T. (2023, November 09). In Vitro Models of Head and Neck Cancer. In Encyclopedia. https://encyclopedia.pub/entry/51382
Arutyunyan, Irina, et al. "In Vitro Models of Head and Neck Cancer." Encyclopedia. Web. 09 November, 2023.
In Vitro Models of Head and Neck Cancer
Edit

For several decades now, researchers have been trying to answer the demand of clinical oncologists to create an ideal preclinical model of head and neck squamous cell carcinoma (HNSCC) that is accessible, reproducible, and relevant. Over the past years, the development of cellular technologies has naturally allowed people to move from primitive short-lived primary 2D cell cultures to complex patient-derived 3D models that reproduce the cellular composition, architecture, mutational, or viral load of native tumor tissue. Depending on the tasks and capabilities, a scientific laboratory can choose from several types of models: primary cell cultures, immortalized cell lines, spheroids or heterospheroids, tissue engineering models, bioprinted models, organoids, tumor explants, and histocultures. HNSCC in vitro models make it possible to screen agents with potential antitumor activity, study the contribution of the tumor microenvironment to its progression and metastasis, determine the prognostic significance of individual biomarkers (including using genetic engineering methods), study the effect of viral infection on the pathogenesis of the disease, and adjust treatment tactics for a specific patient or groups of patients. Promising experimental results have created a scientific basis for the registration of several clinical studies using HNSCC in vitro models.

head and neck squamous cell carcinoma in vitro models 3D models viral infection personalized medicine

1. Introduction

Head and neck squamous cell carcinoma (HNSCC), one of the ten most common cancers, mostly arises from mucosal epithelium in the oral cavity, pharynx, and larynx in close proximity to a number of vital structures, which complicates the radical surgical intervention. This tumor is characterized by late diagnosis, rapid progression, and resistance to antitumor treatment strategies [1][2]. HNSCCs are variably sensitive to radiation and drug treatment, while the locally advanced or metastastatic cases require a combination of surgery, radiation therapy, and chemotherapy [2]. However, even with the use of several types of antitumor treatment, the recurrence rate of HNSCC reaches 50% [3]. The scarcity of biomarkers of HNSCC response to anticancer therapy reflects the lack of relevant prognostic models for HNSCC [4][5][6].

2. History: transition from 2D to 3D HNSCC in vitro models

In the middle decades of the 20th century, scientists used cutting-edge cell technologies in order to provide convenient and usable systems for laboratory investigation of the biological properties of tumor cells. For the first time, primary cell cultures from clinical material of patients with HNSCC were successfully isolated almost half a century ago [7]. Many tumor cultures have been immortalized; to date, several hundred such HNSCC lines have been obtained [8][9][10][11]

Surprisingly, the list of 73 (60 main and 13 additional) human tumor cell lines used in The NCI-60 Human Tumor Cell Lines Screen, designed to identify and characterize novel compounds with growth inhibition or killing of tumor cells, does not include a single HNSCC line [12]. To study this tumor type, a global biological resource, the American Type Culture Collection (ATCC), offers a panel of six cell lines [13][14].: A-253, SCC-9, SCC-15, SCC-25, and FaDu.

In the work of Li et al. the genomic data of 39 HNSCC cell lines were compared with the genomic findings in 106 HNSCC tumors. TP53 was the most commonly mutated gene both in HNSCC tumors and cell lines. Amplification of eight genes (PIK3CA, EGFR, CCND2, KDM5A, ERBB2, PMS1, FGFR1, and WHSCIL1) and deletion of five genes (CDKN2A, SMAD4, NOTCH2, NRAS, and TRIM33) were found in both HNSCC cell lines and tumors; all of these genes had higher alteration frequencies in HNSCC cell lines compared with primary tumors. These findings suggest that mutations and gene copy-number alterations that exist in HNSCC cell lines may result from selection and/or propagation in cell culture that does not reflect critical biological properties of this cancer. Moreover, seventeen genes were only mutated in HNSCC cell lines, but not in human tumors, suggesting that these mutations may arise through immortalization in tissue culture [15].

It is obvious that 2D HNSCC cell lines have significant phenotypic, physiological, genetic, and epigenetic differences compared to the tumor tissue from which they were obtained, due to clonal selection induced by selective survival pressures intrinsic to the culture conditions [13][15][16][17][18]

The advantages of cell lines as the most simplified in vitro tumor model are clear: ease of use, precise control of exogenous factors, economic accessibility, and wide range of applications [19][20]. At the same time, the main problem is their inconsistency with the 3D architecture of tumor tissue, which provides a gradient of gases, nutrients, and biologically active substances that can be reproduced in in vivo models [21][22][23][24]. To overcome this discrepancy, immortalized cells can be transplanted subcutaneously or organotopically into immunodeficient animals, obtaining cell-line derived xenografts, which are voluminous neoplasms, often capable of metastasis [8].

The possibility of obtaining such xenografts has been shown for many HNSCC cell lines (for example, A-253 [25][26], FaDu [26][27], CAL27 [28], and Detroit 562 [29]) and is actively used to evaluate the effectiveness of antitumor agents, search for methods for imaging tumor cells in vivo, or study the mechanisms of carcinogenesis. However, this path of transition to a 3D model of HNSCC cannot be called optimal, since immunodeficient animals require enormous financial and labor costs, obtaining a tumor volume sufficient for research work (150–300 mm3) takes several weeks, and some of the planned studies may turn out to be completely unavailable, for example, due to high radiosensitivity of some strains of mice (NOD-SCID), which precludes experiments using radiation therapy [22][30]. Moreover, not all HNSCC cell lines are tumorigenic; among the ten lines first described in 1981, only three have been confirmed to grow as transplantable xenografts [7].
An attempt to find a balance between the availability and reproducibility of simple models based on 2D adherent tumor lines and the relevance of labor-intensive 3D in vivo animal-based models ultimately led researchers to the creation of 3D in vitro cell models, the variety of forms of which allows studying different aspects of tumor tissue biology [19][31][32]. It is important to note that the development of 3D cell models, and their improvement and implementation in preclinical practice, contributed to the rationalization of the use of animals in research, complying with the principles of “The Three R’s” (Reduction, Replacement, and Refinement) in animal research [33][34].

3. 3D in vitro cell models of HNSCC: main types and methods of production

Spheroids

Multicellular spheroids collected from tumor cells are considered the most simple and reproducible in vitro tumor model capable of reflecting the response of tumor tissue to therapy [35][36][37][38]. Changing cultivation conditions during the transition from 2D to 3D leads to the formation of volumetric multicellular conglomerates, in which cells interact with each other and the external environment without the participation of a culture substrate; as a result, pathophysiological gradients characteristic of tumor tissue are formed in the spheroids [39][40][41][42]. Spheroids are characterized by cellular zoning, which begins to clearly manifest itself as the size of the spheroid increases; upon reaching a diameter of 500 μm, a hypoxic zone and foci of necrosis are formed in the spheroid. Histological examination allows the identification of several layers in large spheroids: an outer layer represented by rapidly proliferating cells, a middle layer with senescent or quiescent cells, and an inner layer containing necrotic cells [35][42][43][44]. These heterogeneous layers are the result of limited diffusion of oxygen and nutrients into the multicellular structure. Cells in the outer layer multiply rapidly due to easier access to oxygen, nutrients, and growth factors; closer to the center of the spheroid, the supply of oxygen and nutrients decreases, and the amount of carbon dioxide and decay products increases [39][42][45].
Large spheroids with a diameter of 500 μm or more reproduce the oxygen gradient characteristic of tumor tissue, forming a focus of hypoxia in its center, which affects the processes of tumorigenesis, activating protumor transcription [46][47] and triggering mechanisms for the development of chemo- and radio-resistance, which lead to increased tumor growth and worsening patient outcomes [48][49][50]. Under conditions of insufficient tissue perfusion, protons and lactic acid accumulate in the extracellular space, leading to the formation of an acidic extracellular microenvironment and creating a pH gradient. This phenomenon is similar to the Warburg effect associated with lactate accumulation in solid tumors [51].
The formation of spheroids occurs spontaneously under conditions where cell–cell interactions predominate over interactions between cells and the culture substrate. In fact, all the main methods for forming spheroids are aimed at creating the following conditions: the hanging drop method, culturing cells in dishes or vials with an ultra-low adhesion surface, and culturing using rotational systems or magnetic fields [35][52][53][54]. In each of these methods, the main task is to minimize the contact of cells and the culture substrate; however, the methods for solving this problem differ:
(1)
completely remove the substrate, culturing cells in a drop supported by surface tension (hanging drop method) [35][55][56];
(2)
modify the substrate, eliminating the possibility of cells to attach to it (culturing in dishes or plates with an ultra-low adhesion surface) [57][58][59];
(3)
ensure continuous mixing of the cell suspension, preventing cells from settling and coming into contact with the substrate (agitation-based method, magnetic levitation) [42].
The first two methods are most commonly used to create spheroids from immortalized cell lines or primary cell cultures isolated from tumor tissue of patients with HNSCC [56][59]. Primary cultures are inherently heterogeneous and are obviously more relevant models for use in personalized medicine. However, the process of obtaining primary cultures from HNSCC biopsies does not always lead to the desired result:
(1)
part of the material may be initially contaminated with bacteria or fungi;
(2)
during the cultivation process, epithelial cells can be replaced by more rapidly proliferating stromal cells;
(3)
the rate of cell proliferation and the efficiency of spheroid formation decreases with an increase in the number of passages performed;
(4)
some methods for obtaining spheroids from such cultures are ineffective [60][61].
To reproduce the heterogeneous composition of tumor tissue, it was proposed to use heterospheroids (or heterotypic spheroids, co-culture spheroids) [62], consisting of a well-characterized immortalized line and an additional cellular component usually associated with tumor cells in vivo, for example, immune cells, fibroblasts, or endothelial cells. Currently, there is active research work using heterospheroids; however, in most cases they are used to model liver and pancreatic tumors, and only a few studies have been performed with heterospheroids based on HNSCC tumor lines.
Thus, reproducible and accessible protocols for obtaining spheroids from immortalized lines or primary cultures of HNSCC allow the creation of in vitro models, in which diffusion restrictions are reproduced not only for oxygen and nutrients, but also for the transport of biologically active substances, imitating the physiological barriers existing in vivo. In many respects, spheroids are closer to tumor tissue than 2D cell cultures, which allows them to be used as a relevant preclinical model for testing the efficacy and toxicity of many anticancer drugs [35][39][55][57][63][64].

Tissue-engineered models

The 2D cultures, spheroids, and heterospheroids described above have a significant drawback as an in vitro model, namely the lack of interaction between tumor cells and the extracellular matrix (ECM), which plays an important role in the occurrence and progression mechanisms of HNSCC [65]. In order to increase the relevance of in vitro tumor models, tissue engineering methods were used, namely cell culture on 3D scaffolds. The first attempts to cultivate HNSCC cells in collagen gel were made at the end of the 20th century [66][67], and the first model of HNSCC using a synthetic scaffold was created in 2007, based on oral squamous cell carcinoma (OSCC-3) cells cultured within a highly porous poly(lactide-co-glycolide) scaffold [68]. Currently, type I collagen in the form of a gel [69] or a porous matrix [70] is most often used as a scaffold material for modeling HNSCC. Such scaffolds are populated with both immortalized tumor cells (SCC090, UM-SCC6 [70], FaDu [71]), and primary cultures obtained from tumor tissue of patients with HNSCC [69][70].
A more complex technology for creating a 3D model of tumor tissue using TRACER (the Tissue Roll for Analysis of Cellular Environment and Response) is also described. It is a novel model in which cells (HNSCC cell lines CAL33 or FaDu co-cultured with cancer-associated fibroblasts) are embedded in a collagen hydrogel infiltrate into a porous cellulose scaffold that is then rolled around an aluminum core to generate a multi-layered 3D tissue [72][73].
Such models are very convenient for studying the interaction of tumor cells and individual proteins of the extracellular matrix or stromal cells [73], and the variability of the scaffolds used makes it possible to control the conditions of this interaction, for example, providing normoxia [72] or hypoxia [70]. By analogy with already developed tissue engineering models of other types of solid tumors, it is, in principle, possible to model HNSCC using a wide variety of scaffolds based on biocompatible natural or synthetic polymeric materials and their composites [74][75][76][77].
Another option for this type of in vitro model is the cultivation of tumor cells on decellularized ECM, when cellular components are removed, and the biological composition is mostly preserved. A study by He et al. compared the proteome of normal and tumor decellularized ECM from oral cavity tissues. It was found that 26 proteins only showed in tumor ECM, 14 proteins only showed in late-stage tumor ECM, and most variant proteins were linked to metabolic regulation and tumor immunity. Tumor ECM influenced the proliferation, apoptosis, and migration of tumor cells, as well as polarized macrophages towards an anti-inflammatory phenotype, which once again confirms the hypothesis of a significant influence of the ECM on the progression of HNSCC [65].
Thus, tissue-engineered in vitro tumor models provide a native-like tissue context for studies of HNSCC biology, but their applications are still at an early stage.

Bioprinted models

In recent years, the method of 3D printing of tissues (including tumor tissues) has been actively developing. This method makes it possible to create complex, volumetrically defined structures, which are also anatomically accurate and relevant [78]. The method consists of constructing 3D structures by precise spatial superimposing layers of bioink that contain cells or cell conglomerates, cytokines, and extracellular matrix components [79][80]. Bioink is usually biocompatible hydrogel loaded with single cells or cell spheroids [81].
Three-dimensional printing was developed several decades ago, but only recently has this versatile technology been adapted to the biomedical field for the fabrication of complex structures using biocompatible materials [79][82]. Inkjet printing [83], extrusion printing [84], laser printing [85], and stereolithography [86] are used for 3D tissue bioprinting. These methods differ from each other in many respects: the cost of equipment and consumables, printing speed, resolution, restrictions on the maximum linear dimensions of the created object, the set of biogels suitable for work, etc. All these bioprinting methods enable a highly improved control of cell distribution within the 3D space compared to conventional approaches [79][87]. The created 3D models are structurally stable over a wide temperature range of 4 °C to 37 °C, which is a necessary condition for working with biological objects [78].
Currently, there are not many works describing the successful modeling of HNSCC using the bioprinting method. In a study by Kort-Mascort et al., emphasis was placed on the development of a new printable biogel containing alginate and gelatin as rheological modifiers, which impart mechanical integrity to the biologically active decellularized ECM (dECM), derived from porcine tongue after its decellularization and solubilization. The topographical characterization of the bioink showed a fibrous network with nanometer-sized pores. Immortalized cell lines UM-SCC-12 (larynx carcinoma) or UM-SCC-38 (oropharynx carcinoma) were selected as the cellular component. The model in the shape of discs with a 5 mm diameter and 500 µm height was printed using extrusion printing. The model was highly reproducible and allows proliferation and reorganization of HNSCC cells while maintaining cell viability above 90% for periods of nearly 3 weeks; cells produced spheroids having a cross-sectional area of at least 3000 μm2 by day 15 of culture and were positive for cytokeratin. The resulting 3D model was used to assess the response of tumor cells to cisplatin and 5-fluorouracil [88]. The authors continued their work in this direction and recently published data on an improved 3D model of HNSCC, in which the cellular component was represented by two types of cells—UM-SCC-38 and A8-HVFFs (immortalized human vocal fold fibroblasts) in a ratio of 1:2. In the process of culturing the model, spheroid development and growth over time with cancer cells in the core and fibroblasts in the periphery were observed [89].
It can be concluded that the 3D bioprinted model of HNSCC is essentially an improved tissue-engineered model, where a hydrogel is used as a scaffold, and the use of a printing method instead of the usual layering of the gel on a substrate allows for more accurate control of the compliance of the tissue architecture with the specified parameters. So far, bioprinted HNSCC models have been obtained only for immortalized lines, which is due to the need to use a large number of cells to obtain the desired result (in [89] cells were encapsulated in gel at a final concentration of 10 million cells per ml). Obviously, an important task for researchers is to obtain such 3D models with HNSCC patient-derived cells to closely match more biological features and potentially use this technique for personalized medicine.

Organoids

The technology of obtaining organoids, which represent an improved in vitro model that combines the advantages of spheroids (the ability of cells to 3D self-organization) and tissue engineering models (the presence of ECM) helped researchers to advance even further on the path to studying the interaction of tumor cells and the ECM [90]. Based on grammar, “-oid” is a suffix meaning “resembling”; organoid thus means “resembling an organ”, so organoids have to exhibit at least some organ functionalities of the modeled tissue [91]. Over the years of improving protocols for the production and use of organoids as an in vitro model of tumor tissue (in this case, the term “tumoroid” is also used), their main mandatory properties have been determined:
(1)
organoids contain tumor cells at different levels of differentiation, including cancer stem cells;
(2)
organoids consist of several cell types that self-organize in space, reproducing the architectonics of the original tumor tissue;
(3)
self-organization of the organoid occurs in the presence of the ECM;
(4)
the interaction of cells and the ECM allows reproduction of the functional properties of the simulated tumor tissue [91][92][93].
Thus, organoids have high genetic and phenotypic similarity to native tissue, maintaining the original intratumoral heterogeneity [94][95].
Most often, organoids for modeling HNSCC are obtained from resected tumor tissue of the patient. In the first stage, the biopsy sample is cleared of tissues of the peritumoral area (connective, muscle, fat), then washed very thoroughly, since the area of material collection (oral cavity, pharynx, nasal septum) is highly likely to be contaminated with bacteria or fungi. Next, the tissue is subjected to mechanical and enzymatic disaggregation using collagenases, dispase, hyaluronidase, trypsin, and EDTA (separately or as part of a cocktail of enzymes), after which sieved single cells or cell clusters are plated in a 3D extracellular matrix (ECM) hydrogel such as basement membrane extract (BME), where they form complex conglomerates [96]. This matrix is obtained from the basement membrane of sarcoma of Engelbreth-Holm-Swarm (EHS) mice, and is rich in such ECM proteins as laminin (a major component), collagen IV, heparan sulfate proteoglycans, entactin/nidogen, and a number of growth factors. It is easy to use and commercially available in standard, protein-enriched, or growth-factor reduced versions [97]. On the other hand, the EHS matrix is extremely complex; proteomic analysis shows that it contains more than 1800 unique proteins, the ratio of which varies from lot to lot, which can affect the reproducibility of the properties of organoids grown in it. In addition, the matrix obtained from mouse tumor tissue has an obvious drawback—it does not meet animal-derived-free standards, which will undoubtedly become mandatory for biomedical products in the near future, so researchers are actively looking for a replacement among synthetic or natural hydrogels [98][99].
Over the past few years, organoids have been successfully obtained from normal and tumor tissues of the brain, lungs, esophagus, stomach, intestines, liver, pancreas, kidneys, salivary glands, ovaries, mammary glands, prostate, and other organs [100]. For each source, a lengthy selection of optimal conditions for the formation of organoids was necessary. In protocols for obtaining organoids from head and neck tumor tissue, the steps for isolating individual cells or clusters do not differ fundamentally between different research groups, but the composition of the culture medium varies significantly
The number of studies that resulted in the successful production of HNSCC organoids is small due to difficult logistics (the time from resection of tumor tissue to delivery to the laboratory must be minimized), an often insufficient volume of biomaterial, as well as high requirements for the qualitative and quantitative composition of the culture medium, which demands the mandatory presence of growth factors, cytokines, and inhibitors of signaling pathways leading to the triggering of apoptosis or epithelial-mesenchymal transition. At the same time, the efficiency of obtaining organoids from tumor tissue of the head and neck does not exceed 60–70%, even in laboratories that have been actively working with this material for many years.
It is believed that the main factor influencing the efficiency of obtaining this 3D model of HNSCC is the quality of the initial biomaterial, since necrotic tumor tissue has poor organoid-forming efficacy. To minimize cell death and increase organoid outgrowth, Driehuis et al. recommend to transport tumor pieces in ice-cold basic medium containing 10 μM Rho kinase (ROCK) inhibitor compound Y-2763 or (less desired) ice-cold PBS [96]. It has also been shown that organoids can be established from cryopreserved tissues, although the efficacy of derivation is probably lower than that obtained when starting with fresh material [96]. According to Wang et al., the efficiency of obtaining HNSCC organoids is also affected by:
(1)
mass of biomaterial (for tumor samples weighing less than 50 mg, the efficiency of obtaining organoids from it is reduced to 3%);
(2)
time of transportation of tumor tissue to the laboratory (time exceeding 24 h leads to a decrease in efficiency from 60–70% to 22%);
(3)
composition of the culture medium (for example, concentration of Wnt3a, R-spondin-1, EGF, Y27632, Noggin, and FGF2);
(4)
ECM used (Matrigel is preferable to collagen I);
(5)
primary/recurrent tumor status (for nasopharyngeal carcinoma, the efficiency of obtaining organoids is 82% for recurrent tumor and only 47% for primary tumor) [101].
In the same study, Wang et al. showed that the effectiveness of obtaining organoids from nasopharyngeal carcinoma tissue does not depend on the gender and age of the patient, or on the clinical stage or lesion location (primary focus or metastasis) of the tumor [101].
Thus, obtaining organoids requires significant material and time, as well as highly qualified personnel and close coordination of clinicians and cell biologists. The result is a tumor model that can be maintained in vitro for a long time, while maintaining genetic stability [102][103] and fairly fully reproducing the morphological characteristics of the original tissue. For example, organoids from nasopharyngeal carcinoma do not express CK7, unlike organoids from normal mucosa obtained from the same patient, which corresponds to differential expression of this marker in the original tissues [101]. It has been shown that organoids, when cultivated, retain the expression of many other markers of head and neck tumors, including those used to identify cancer stem cells: CK13, CK18, ALDH1A1, BMI-1, CD44, and CD133 [101][103][104][105][106]. Interestingly, lactate, which is promoted by Wnt activity, is required to maintain the population of CD133+ cells in HNSCC organoids. Moreover, silencing monocarboxylate transporter 1 (MCT1), the prominent pathway for lactate uptake in human tumors, with siRNA significantly impaired organoid-forming capacity of oral squamous cell carcinoma cells, which allows MCT1 to be considered as a potential therapeutic target for the treatment of HNSCC [106].
Organoids are considered highly relevant in vitro tumor models because they are able to preserve a unique set of biomarkers from donor tissue [107][108]. Increasing evidence indicates that organoids can predict response to treatment of the tumor from which they are derived [109]. In a pilot study, organoids obtained from 21 patients with gastrointestinal tumors predicted response to various types of chemotherapy with 100% sensitivity and 93% specificity [110]. A number of studies have highlighted the prognostic value of organoids in predicting the response of solid tumors to radiotherapy and its combination with other treatments [111][112]. In addition, organoids can also be expanded in vitro and cryopreserved, facilitating the establishment of an organoid biobank of different cancer subtypes from a large number of patients, representing an extremely useful tool for preclinical research [7][43][113]..

Tumor explants and histocultures

Another type of 3D cell model is patient-derived tumor explant (PDE) cultures, in which a tissue fragment is kept alive under ex vivo culture conditions. Explants require minimal manipulation to obtain an in vitro tumor model: the tissue, after mechanical grinding (usually to fragments of 1–3 mm3 in size), is placed in a culture medium. This approach allows preserving the heterogeneous composition of the original tumors, including the extracellular matrix and tumor-associated cells [114]. Explants are often used to obtain adherent 2D primary cell cultures, but in the case of the PDE model this is not done, so researchers cultivate explants under special conditions that exclude the migration of plastic-adherent cells from the tissue fragment. To do this, HNSCC explants are placed on an additional supporting matrix, for example, a collagen sponge [10] or a dermal equivalent consisting of human fibroblasts cultured on a viscose fiber fabric [115].
Despite the ease of production and the possibility of preserving the architecture of tumor tissue, the scope of application of PDE as an in vitro model is significantly limited by its fragility; most often, PDE is used for rapid testing of the effectiveness and/or toxicity of drugs within 5–7 days after being obtained [114]. It has been shown that the viability of HNSCC explants decreases from 90% to 30% within a week after the start of cultivation [116]. The survival rate of HNSCC explants can be increased several times by modifying the culture protocol, for example, adding hydrocortisone, aprotinin, ascorbic acid, EGF, or folic acid to the medium [115][116], or placing the explant in more physiological conditions that imitate contact with normal tissue, for example, onto the surface of the dermal equivalent [115] or into cell sheet composing of epithelium and subepithelial stroma [117]. Under such conditions, explants can be maintained in culture for 21 [115] or even more than 30 days [117], while cancer cell heterogeneity and the microenvironment, including vital immune cells, as well as tissue foci of hypoxia, are maintained.
Another option for this type of 3D model is histoculture. When preparing it, tumor tissue, as when obtaining explants, is crushed mechanically, not with surgical instruments, but with the help of a vibratome, which makes it possible to obtain thick sections of unfixed tissue. In these sections, as in explants, tumor cells are retained in their original microenvironment, including the extracellular matrix and immune and stromal cells [118]. The main problem with culturing histocultures is rapid deterioration of tissue condition and loss of cell viability (from 2 [119] to 6 days [120]). However, this time is sufficient to assess the response of tumor tissue to the action of, for example, cisplatin, docetaxel, and cetuximab, while apoptotic fragmentation, activation of caspase 3, and cell loss were observed in treated tumor slices, which demonstrated a heterogeneous individual response [119][120].
Thus, explants and histocultures of HNSCC are easy to obtain and effective for quickly assessing the response of tumor tissue to drug therapy, which suggests their promise for use in personalized medicine.

Microfluidic Devices (Tumor-on-Chip)

At the very beginning, researchers will make an important note: the cultivation of tumor cells in microfluidic devices is not a separate type of model; rather, it is a special technology for culturing the models described above, providing continuous perfusion and removal of waste from cells and mimicking the function of the circulatory system. Microfluidic devices exploit the physical and chemical properties of liquids and gases at a microscale; in such systems, liquids circulate through channels with dimensions of tens to hundreds of micrometers [121]. Thanks to their small size, microfluidics allow the analysis and use of lower volumes of samples, chemicals, and reagents, reducing the global fees of applications. In addition, this technology makes it possible to very accurately control the impact on the object under study; in the future, it will also make it possible to automate routine research work. To assess the tumor response to exposure, it is possible to analyze both the system effluent (sampling can be carried out at any selected interval, for example, every 2 h) and the object itself after extraction [30][121].

When modeling HNSCC, microfluidic devices are most often used for culturing tumor tissue fragments (explants and histocultures).
Thus, tumor-on-chip technology is a valuable tool for personalized assessment of the effectiveness of various antitumor agents (alone or in combination), allowing dynamic monitoring of the response of tissue samples from patients with HNSCC to treatment. The development of microfluidic platforms can improve patient outcomes through the selection of an optimal personalized treatment strategy.

4. Comparison of in vitro HNSCC models

For several decades, preclinical studies of HNSCC have been carried out on in vitro models, which make it possible to evaluate the effectiveness of antitumor therapy, study resistance mechanisms, and improve early diagnostic and screening tests [7], [8], [18], [96]. To study the HNSCC biology, several types of in vitro models, differing in many parameters, are currently actively used (Table 1).

Table 1. In vitro models of HNSCC: main features

 

2D

3D

 

Immortalized

cell lines

Primary

cell cultures

Spheroids and

heterospheroids

Tissue-engineered

models

Bioprinted models

Organoids

Explants and

histocultures

Source

cell biobanks

patient-derived tissue

patient-derived tissue, primary cell cultures, immortalized cell lines

patient-derived tissue

Heterogeneity of tumor cellular composition

not preserved

partially preserved

depends on the source

preserved

ЕСМ

no

natural and synthetic polymers, decellularized tissue

bioink based on hydrogels

basement membrane matrix, collagen

native

Tissue architecture, pathophysiological gradients

absent

partially

reconstituted

reconstituted

preserved

In vitro culture duration

not limited

limited

Difficulty of obtaining

low

medium

high

medium

Major advantages

availability, stability of properties, many years of experience in use, ability to obtain a 3D model

availability, ability to obtain a 3D model

the most available 3D model

convenience of studying the ECM -cells interaction, possibility of getting a model with given linear dimensions

obtaining artificial tumor tissue with specified spatial characteristics

capability to support tumor cells at different levels of differentiation, mimicking the tumor microenvironment

minimally manipulated tumor tissue

Specific disadvantages

chromosomal instability, impossibility of use for personalized medicine

the initial ratio of tumor and tumor-associated cells and their properties may change during cultivation

prone to fusion to form conglomerates, difficulty in controlling size

a lot of cells are required for modelling

a lot of cells are required for modelling, sophisticated equipment is required

production efficiency about 60-70%

long-term in vitro cultivation requires supporting matrices or microfluidic devices

The choice of a specific model depends on the tasks and capabilities of the scientific laboratory. As follows from the bibliometric analysis of research papers on various topics in the PubMed database, the number of investigations in the area of 3D cancer models in vitro is growing every year. However, in 2001–2020, only 65 papers for all types of HNSCC 3D in vitro models were published, which is only 0.72% of all studies involving 3D cancer models. Such a small number of works (HNSCC claims the lives of 450 thousand people a year! [1]) is primarily due to the fact that obtaining a reproducible and relevant 3D model of HNSCC is in itself a rather complex scientific task.

5. In vitro cell models of HNSCC and Human papillomavirus

More than 200 papillomavirus types infect humans. According to the World Health Organization International Agency for Research on Cancer (WHO IARC), HPV types 16 and 18 are classified as carcinogenic to humans (Group 1), HPV types 31 and 33 are probably carcinogenic (Group 2A), and HPV types 35, 39, 45, 51, 52, 56, 58, and 59 are possibly carcinogenic (Group 2B) [122]. Some researchers suggest considering HPV+ and HPV− HNSCC as separate groups of neoplasms. Compared to HPV-negative, HPV+ HNSCC more commonly affects the oropharynx (predominantly), hypopharynx, and larynx than the oral cavity. Moreover, HPV+ HNSCCs have different pathological patterns, immune signature, and mutation burden, i.e., greater infiltration of B cells into the tumor microenvironment, fewer genetic mutations, and intact apoptotic response, which may explain the improved prognosis and superior response to radio- and immunotherapy and significantly longer median survival than HPV-negative HNSCC (130 months vs. 20 months) [1].
Thus, it is not surprising that when modeling HNSCC in vitro, scientists always emphasize the HPV status of the cell line or tumor cells within the 3D model. HPV accounts for 72% of all head and neck squamous cell carcinoma cases in developed nations [1]; however, in fact, researchers rarely work with HPV+ HNSCC in vitro models. Just compare: the number of currently available HPV+ immortalized HNSCC cell lines is significantly limited, with eight lines known precisely (UMSCC-47, UMSCC-104, UPCI:SCC090, UPCI:SCC152, UPCI:SCC154, 93-VU-147T, HMS001, and LU-HNSCC-26) [10][105][123], while the number of HPV-negative lines is in the hundreds.
The development of primary cell lines from a naturally infected HPV+ cancer is rarely successful [11] due to the low mutational load; for example, the TP53 gene is almost always wild-type, both in tumor tissue and in primary cell cultures or immortalized cell lines derived from it [105][123]. The preservation of genes involved in cell cycle control and apoptosis induction leads to low proliferation rates of HPV+ tumor cells when transferred to a 2D monolayer; the cultures are highly radiation and drug-sensitive and readily enter senescence. It is important to note that immortalized HPV+ cell lines retain the radiation sensitivity both in 2D form and when cultured in the form of spheroids or cell line-derived xenografts [124][125]. Interestingly, some HPV+ immortalized lines are unable to form stable 3D spheroids (for example, UMSCC-47) or only form slowly growing spheroids (for example, UPCI:SCC090) [125].
Similar difficulties are observed with patient-derived 3D models. For a long period of time, the attempts to obtain organoids from HPV-associated HNSCC were unsuccessful. This was primarily for clinical reasons, i.e., the patients with HPV+ HNSCC less commonly receive surgery. One of the first organoids from HNSCC specimens was obtained by Tanaka et al., with the efficiencies similar for HPV+ (3/9) and for HPV− (10/34) patients [105]. In another study, the efficiency of obtaining organoids was higher: eight lines of HPV+ organoids were isolated from nine samples of initially infected tumor tissue [126]. Another approach for obtaining organoids from HPV+ tumor tissue involves the implantation of tumor fragments in NOD/SCID/IL-2Rγ−/− mice to a <25% stable engraftment rate; after the xenograft reaches a volume of 1 cm3, it was passaged at least twice, removed, dissociated into individual cells, and cultured on ECM to form organoids.

6. New trends in in vitro modeling of HNSCC

The development and implementation of a personalized approach in clinical oncology requires constant improvement of preclinical models aimed at maximizing the approximation of cell models to the structure of the native tumor and increasing the efficiency of obtaining such models. For in vitro models of HNSCC, several areas of research can be distinguished.

Models of vascularization

In the tumor in vivo microenvironment, there is an active interaction between tumor and endothelial cells, which is important for the recruitment of angiogenic cells, tumor cell survival, and migration [108][127]. In most cases, the method of co-cultivation of 2D cultures is used to analyze the interaction of tumor and endothelial cells [128]. It has been shown that exosomes produced by PCI-13 (HPV−) and UMSCC47 (HPV+) cell lines, as well as exosomes from plasma of HNSCC patients, stimulated proliferation, migration, and tube formation by human umbilical vein endothelial cells (HUVECs) in vitro and promoted formation of defined vascular structures in vivo [129]. To improve inter- and intratumoral vasculature, tumor cells actively produce proangiogenic factors. When 2D HNSCC cell lines are transferred to 3D culture conditions, their proangiogenic potential increases; transplantation of a suspension of primary cancer-associated fibroblasts and 3D spheroids from FaDu cells led to the growth of a well-vascularized tumor in a mouse model of xenografts, while, when transplanting cell suspensions from 2D cultures, gradual necrosis of the graft due to insufficient blood supply to the tissue was observed [130].
Angiogenesis in solid tumor tissue is a promising target for HNSCC-targeted therapy. Researchers are actively searching for signaling molecules and pathways of interaction between epithelial and tumor cells, and tumor-associated lymphocytes (including B cells and T cells) [131] or macrophages [32] can act as mediators in this interaction. To create a relevant model that is as close as possible to native tissue, it is necessary to reproduce in vitro one of the key processes of tumor progression and metastasis-angiogenesis [132][133]. The furthest along this path has been bioprinting technology, which can directly bioprint the vascular wall structures or create hollow channels inside a volumetric matrix that will later be populated by endothelial cells [127]. Microfluidic technologies also make it possible to study various aspects of the interaction between tumor and endothelial cells in vitro: metastasis (intra- and extravasation), chemotaxis, production of angiogenic factors, etc. [134][135].

New types of matrices for tumor cell culturing

Currently, there is an active process of abandoning xenogeneic reagents when modeling human tissues in vitro in favor of autogenous or allogeneic materials; it is assumed that such a replacement will make it possible to recreate a more favorable microenvironment for cells, simulating their original environment in vivo [136]. Thus, when modeling HNSCC in vitro, it was proposed to use allogeneic gels based on human tumor tissues—human uterine leiomyoma-derived Myogel or human pre-metastatic neck lymph node-derived Lymphogel, instead of Matrigel (solubilized basement membrane preparation extracted from the Engelbreth-Holm-Swarm mouse sarcoma) [137][138]. Such matrices differ in their protein composition, including the content of unique adhesion-related proteins [138]. An experiment involving 12 HNSCC cell lines revealed an increase in the resistance of tumor cells to the action of EGFR and MEK inhibitors compared to cells cultured on plastic or in Matrigel. The authors of the work suggest that the human tumor matrix improves the predictability of in vitro anticancer drug testing [137].

7. Applications of 3D in vitro cell models of HNSCC

Currently, the attention of researchers is aimed at the development of personalized medicine, where an important tool is 3D models obtained from tumor tissue of patients. The main goal is to obtain a model that can predict the patient’s response to therapy in order to optimize the treatment strategy [139].
Valuable information for clinicians can be obtained by using patient-derived explants (including those cultured using microfluidic devices) to test antitumor therapy [140]. Such studies already make it possible to identify inter- and intra-patient variability in response to irradiation and chemotherapy [141].
Tanaka et al. [105] managed to obtain HNSCC organoids that were as close as possible to the primary tumor; they had a similar morphology, and retained the expression of markers of mesenchymal (vimentin) and tumor (CD44 and ALDH1A1) cells, although CD68+ cells were found only in the tumor, but not in the organoids. When testing sensitivity to platinum and docetaxel therapy in the resulting organoids, consistency was found between the response in vitro and in vivo [105].
Driehuis et al., analyzing the expression of markers TP63, TP40, Ki67, CK13, and CK5, convincingly showed that organoids obtained from the mucous membrane of the oral cavity or tongue recapitulate the functional and morphological characteristics of the tumor [103][142]. The same study found that the response of organoids to radiation correlated with the response of patients; three organoid lines were among the most resistant when exposed to radiotherapy in vitro, and three corresponding patients relapsed after undergoing radiotherapy; moreover, a patient whose organoid line showed the highest sensitivity to irradiation had a lasting response to palliative radiotherapy [103].
Millen et al. describes the organoid biobank, which stores a collection of biomaterial from patients with HNSCC from various anatomical locations and histological cancer subtypes. During routine surgical resection or biopsy procedures in 2019–2022, samples were collected from 228 patients, but organoids were successfully obtained from only 97 patients (42.5% efficiency). To store organoids, the biobank uses cryopreservation technology; the proportion of successfully thawed organoids was 70.9%. The resulting organoids retained histopathological and molecular features of primary tumor tissue; tumor cells in their composition expressed CK13 and p63, and a genetic study revealed mutations in the tumor-associated genes TP53 (in 63% of organoids), NOTCH1, PIK3CA, FAT1, and APOB, gains of oncogenes including PIK3CA, FGF3, and FGF4, and loss of tumor suppressor CDKN2A. The collection included unique samples, for example, four salivary gland tumor organoid models that retained the ability to produce mucin and amylase, as well as organoids from tissues of a patient with Fanconi anemia, which had increased sensitivity to double-stranded DNA breaks induced by mitomycin-C. Despite the similarity of organoids and tumor tissue at the cellular and molecular levels, in combined treatment (radiation therapy + cisplatin) no clear correlation between organoid and patient response was observed. At the same time, the organoid response to radiotherapy (measured as organoid viability at 2 Gy) correlated with clinical relapse status in the adjuvant setting but not in the primary setting. In addition, the researchers successfully used CRISPR-Cas9 base-editing technology to introduce the E545K mutation (one of the most common PIK3CA mutations), which opens up new opportunities for scientists to use patient-derived 3D models to search for and validate potential biomarkers of HNSCC response to therapy [126].
Promising results of experimental work allowed scientists to move on to promoting clinical trials using in vitro models of HNSCC. In October 2023, only three clinical trials were registered, which is entirely attributable to the difficulty of obtaining patient-derived models of head and neck cancer.
The ORGAVADS (ORGAnoids + VADS (French “voies aérodigestives supérieures”) tumors) study is a multicenter observational trial (NCT04261192) conducted to investigate the feasibility of generating and testing patient-derived tumor organoids derived from HNSCC for the evaluation of sensitivity to treatments (chemotherapy, radiotherapy, PARP inhibitors, and immunotherapy). The planned number of study participants is determined taking into account that the expected efficiency of obtaining organoids from patient material will be about 60%. The sponsor (Centre Francois Baclesse) assumes that this screening could make it possible to refine the choice of treatments adapted to each patient and thus limit the undesirable effects [143][144].
Investigators hypothesize that high-throughput screening on patient-derived tumor organoids can be used as an adjunct tool to aid treatment selection in patients with cancer. The objective of the NCT04279509 study is to determine if a drug screen assay (panel of ten primary and five additional anticancer drugs) using personal organoid models can accurately select a chemotherapeutic agent that results in objective response in patients with refractory solid tumors (including HNSCC) [145].
The SOTO (Sensitivity of Organoids to Treatment Outcome) prospective observational study (NCT05400239) aims to determine the sensitivity of organoids obtained from patients with HNSCC to radiotherapy, platinum (cisplatin and/or carboplatin) chemotherapy, or cetuximab or their combination, with subsequent determination of the correlation of the treatment sensitivities of organoids with the treatment outcome of patients [146].
If successful, the clinical trials described above will mark a new stage in the development of personalized medicine in head and neck cancer.

References

  1. Barsouk, A.; Aluru, J.S.; Rawla, P.; Saginala, K.; Barsouk, A. Epidemiology, Risk Factors, and Prevention of Head and Neck Squamous Cell Carcinoma. Med. Sci. 2023, 11, 42.
  2. Johnson, D.E.; Burtness, B.; Leemans, C.R.; Lui, V.W.Y.; Bauman, J.E.; Grandis, J.R. Head and neck squamous cell carcinoma. Nat. Rev. Dis. Primers 2020, 6, 92.
  3. Nissi, L.; Suilamo, S.; Kytö, E.; Vaittinen, S.; Irjala, H.; Minn, H. Recurrence of head and neck squamous cell carcinoma in relation to high-risk treatment volume. Clin. Transl. Radiat. Oncol. 2021, 27, 139–146.
  4. Boguszewicz, Ł. Predictive Biomarkers for Response and Toxicity of Induction Chemotherapy in Head and Neck Cancers. Front. Oncol. 2022, 12, 900903.
  5. Schanne, D.H.; Koch, A.; Elicin, O.; Giger, R.; Medová, M.; Zimmer, Y.; Aebersold, D.M. Prognostic and Predictive Biomarkers in Head and Neck Squamous Cell Carcinoma Treated with Radiotherapy—A Systematic Review. Biomedicines 2022, 10, 3288.
  6. Budach, V.; Tinhofer, I. Novel prognostic clinical factors and biomarkers for outcome prediction in head and neck cancer: A systematic review. Lancet Oncol. 2019, 20, e313–e326.
  7. Easty, D.M.; Easty, G.C.; Carter, R.L.; Monaghan, P.; Butler, L.J. Ten human carcinoma cell lines derived from squamous carcinomas of the head and neck. Br. J. Cancer 1981, 43, 772–785.
  8. Chaves, P.; Garrido, M.; Oliver, J.; Pérez-Ruiz, E.; Barragan, I.; Rueda-Domínguez, A. Preclinical models in head and neck squamous cell carcinoma. Br. J. Cancer 2023, 128, 1819–1827.
  9. Lepikhova, T.; Karhemo, P.-R.; Louhimo, R.; Yadav, B.; Murumägi, A.; Kulesskiy, E.; Kivento, M.; Sihto, H.; Grénman, R.; Syrjänen, S.M.; et al. Drug-sensitivity screening and genomic characterization of 45 hpV-negative head and neck carcinoma cell lines for novel biomarkers of drug efficacy. Mol. Cancer Ther. 2018, 17, 2060–2071.
  10. Greaney-Davies, F.S.; Risk, J.M.; Robinson, M.; Liloglou, T.; Shaw, R.J.; Schache, A.G. Essential characterisation of human papillomavirus positive head and neck cancer cell lines. Oral Oncol. 2020, 103, 104613.
  11. 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.
  12. Close, D.A.; Wang, A.X.; Kochanek, S.J.; Shun, T.; Eiseman, J.L.; Johnston, P.A. Implementation of the NCI-60 Human Tumor Cell Line Panel to Screen 2260 Cancer Drug Combinations to Generate >3 Million Data Points Used to Populate a Large Matrix of Anti-Neoplastic Agent Combinations (ALMANAC) Database. SLAS Discov. Adv. Sci. Drug Discov. 2019, 24, 242–263.
  13. American Type Culture Collection. Head and Neck Cancer Panel TCP-1012TM. In Medical Radiology; Springer: Cham, Switzerland, 2022; pp. 137–157.
  14. Zhao, M.; Sano, D.; Pickering, C.R.; Jasser, S.A.; Henderson, Y.C.; Clayman, G.L.; Sturgis, E.M.; Ow, T.J.; Lotan, R.; Carey, T.; et al. Assembly and initial characterization of a panel of 85 genomically validated cell lines from diverse head and neck tumor sites. Clin. Cancer Res. 2011, 17, 7248–7264.
  15. Li, H.; Wawrose, J.S.; Gooding, W.E.; Garraway, L.A.; Lui, V.W.Y.; Peyser, N.D.; Grandis, J.R. Genomic analysis of head and neck squamous cell carcinoma cell lines and human tumors: A rational approach to preclinical model selection. Mol. Cancer Res. 2014, 12, 571–582.
  16. Kapałczyńska, M.; Kolenda, T.; Przybyła, W.; Zajączkowska, M.; Teresiak, A.; Filas, V.; Ibbs, M.; Bliźniak, R.; Łuczewski, L.; Lamperska, K. 2D and 3D cell cultures—A comparison of different types of cancer cell cultures. Arch. Med. Sci. 2018, 14, 910–919.
  17. Mseka, T.; Bamburg, J.R.; Cramer, L.P. ADF/cofilin family proteins control formation of oriented actin-filament bundles in the cell body to trigger fibroblast polarization. J. Cell Sci. 2007, 120, 4332–4344.
  18. Li, C.; Kato, M.; Shiue, L.; Shively, J.E.; Ares, M.; Lin, R.-J. Cell type and culture condition-dependent alternative splicing in human breast cancer cells revealed by splicing-sensitive microarrays. Cancer Res 2006, 66, 1990–1999.
  19. Seliger, B.; Al-Samadi, A.; Yang, B.; Salo, T.; Wickenhauser, C. In vitro models as tools for screening treatment options of head and neck cancer. Front. Med. 2022, 9, 971726.
  20. Burford-Mason, A.P.; Irish, J.C.; MacKay, A.J.; Gullane, P.J.; Bassett, R.; Dardick, I. Squamous cell carcinomas of the head and neck cultured in floating collagen gels: 1. The maintenance of stromal and epithelial elements in vitro without fibroblast overgrowth. Otolaryngol. Neck Surg. 1997, 116, 213–222.
  21. Jubelin, C.; Muñoz-Garcia, J.; Griscom, L.; Cochonneau, D.; Ollivier, E.; Heymann, M.-F.; Vallette, F.M.; Oliver, L.; Heymann, D. Three-dimensional in vitro culture models in oncology research. Cell Biosci. 2022, 12, 155.
  22. Cekanova, M.; Rathore, K. Animal models and therapeutic molecular targets of cancer: Utility and limitations. Drug Des. Dev. Ther. 2014, 8, 1911–1922.
  23. 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.
  24. Breslin, S.; O’driscoll, L. The relevance of using 3D cell cultures, in addition to 2D monolayer cultures, when evaluating breast cancer drug sensitivity and resistance. Oncotarget 2016, 7, 45745–45756.
  25. Park, S.-B.; Jung, W.K.; Kim, H.R.; Yu, H.-Y.; Kim, Y.H.; Kim, J. Esculetin has therapeutic potential via the proapoptotic signaling pathway in A253 human submandibular salivary gland tumor cells. Exp. Ther. Med. 2022, 24, 533.
  26. Colley, H.E.; Hearnden, V.; Jones, A.V.; Weinreb, P.H.; Violette, S.M.; MacNeil, S.; Thornhill, M.H.; Murdoch, C. Development of tissue-engineered models of oral dysplasia and early invasive oral squamous cell carcinoma. Br. J. Cancer 2011, 105, 1582–1592.
  27. Bernhard, W.; Barreto, K.; El-Sayed, A.; DeCoteau, J.; Geyer, C.R. Imaging Immune Cells Using Fc Domain Probes in Mouse Cancer Xenograft Models. Cancers 2022, 14, 300.
  28. Bernareggi, D.; Xie, Q.; Prager, B.C.; Yun, J.; Cruz, L.S.; Pham, T.V.; Kim, W.; Lee, X.; Coffey, M.; Zalfa, C.; et al. CHMP2A regulates tumor sensitivity to natural killer cell-mediated cytotoxicity. Nat. Commun. 2022, 13, 1899.
  29. Hsu, C.-M.; Yang, M.-Y.; Tsai, M.-S.; Chang, G.-H.; Yang, Y.-H.; Tsai, Y.-T.; Wu, C.-Y.; Chang, S.-F. Dihydroisotanshinone i as a treatment option for head and neck squamous cell carcinomas. Int. J. Mol. Sci. 2021, 22, 8881.
  30. Tevlek, A.; Kecili, S.; Ozcelik, O.S.; Kulah, H.; Tekin, H.C. Spheroid Engineering in Microfluidic Devices. ACS Omega 2023, 8, 3630–3649.
  31. Huo, K.-G.; D’arcangelo, E.; Tsao, M.-S. Patient-derived cell line, xenograft and organoid models in lung cancer therapy. Transl. Lung Cancer Res. 2020, 9, 2214–2232.
  32. Moya-Garcia, C.R.; Okuyama, H.; Sadeghi, N.; Li, J.; Tabrizian, M.; Li-Jessen, N.Y.K. In vitro models for head and neck cancer: Current status and future perspective. Front. Oncol. 2022, 12, 960340.
  33. Morrissey, B.; Blyth, K.; Carter, P.; Chelala, C.; Jones, L.; Holen, I.; Speirs, V. The Sharing Experimental Animal Resources, Coordinating Holdings (SEARCH) Framework: Encouraging Reduction, Replacement, and Refinement in Animal Research. PLoS Biol. 2017, 15, e2000719.
  34. Tenschert, E.; Kern, J.; Affolter, A.; Rotter, N.; Lammert, A. Optimisation of Conditions for the Formation of Spheroids of Head and Neck Squamous Cell Carcinoma Cell Lines for Use as Animal Alternatives. Altern. Lab. Anim. 2022, 50, 414–422.
  35. Mehta, G.; Hsiao, A.Y.; Ingram, M.; Luker, G.D.; Takayama, S. Opportunities and challenges for use of tumor spheroids as models to test drug delivery and efficacy. J. Control. Release 2012, 164, 192–204.
  36. Elliott, N.T.; Yuan, F. A review of three-dimensional in vitro tissue models for drug discovery and transport studies. J. Pharm. Sci. 2011, 100, 59–74.
  37. Kimlin, L.C.; Casagrande, G.; Virador, V.M. In vitro three-dimensional (3D) models in cancer research: An update. Mol. Carcinog. 2013, 52, 167–182.
  38. Jung, A.R.; Jung, C.-H.; Noh, J.K.; Lee, Y.C.; Eun, Y.-G. Epithelial-mesenchymal transition gene signature is associated with prognosis and tumor microenvironment in head and neck squamous cell carcinoma. Sci. Rep. 2020, 10, 3652.
  39. Han, S.J.; Kwon, S.; Kim, K.S. Challenges of applying multicellular tumor spheroids in preclinical phase. Cancer Cell Int. 2021, 21, 152.
  40. Costa, E.C.; Moreira, A.F.; De Melo-Diogo, D.; Gaspar, V.M.; Carvalho, M.P.; Correia, I.J. 3D tumor spheroids: An overview on the tools and techniques used for their analysis. Biotechnol. Adv. 2016, 34, 1427–1441.
  41. Thakuri, P.S.; Gupta, M.; Plaster, M.; Tavana, H. Quantitative Size-Based Analysis of Tumor Spheroids and Responses to Therapeutics. ASSAY Drug Dev. Technol. 2019, 17, 140–149.
  42. Cui, X.; Hartanto, Y.; Zhang, H. Advances in multicellular spheroids formation. J. R. Soc. Interface 2017, 14, 20160877.
  43. 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.
  44. Huang, B.-W.; Gao, J.-Q. Application of 3D cultured multicellular spheroid tumor models in tumor-targeted drug delivery system research. J. Control. Release 2018, 270, 246–259.
  45. Lu, H.; Stenzel, M.H. Multicellular Tumor Spheroids (MCTS) as a 3D In Vitro Evaluation Tool of Nanoparticles. Small 2018, 14, e1702858.
  46. Lin, R.; Chang, H. Recent advances in three-dimensional multicellular spheroid culture for biomedical research. Biotechnol. J. 2008, 3, 1172–1184.
  47. Raghavan, S.; Mehta, P.; Horst, E.N.; Ward, M.R.; Rowley, K.R.; Mehta, G. Comparative analysis of tumor spheroid generation techniques for differential in vitro drug toxicity. Oncotarget 2016, 7, 16948–16961.
  48. Luoto, K.R.; Kumareswaran, R.; Bristow, R.G. Tumor hypoxia as a driving force in genetic instability. Genome Integr. 2013, 4, 5.
  49. Das, V.; Bruzzese, F.; Konečný, P.; Iannelli, F.; Budillon, A.; Hajdúch, M. Pathophysiologically relevant in vitro tumor models for drug screening. Drug Discov. Today 2015, 20, 848–855.
  50. Yasui, H.; Kawai, T.; Matsumoto, S.; Saito, K.; Devasahayam, N.; Mitchell, J.B.; Camphausen, K.; Inanami, O.; Krishna, M.C. Quantitative imaging of pO2 in orthotopic murine gliomas: Hypoxia correlates with resistance to radiation. Free. Radic. Res. 2017, 51, 861–871.
  51. Gatenby, R.A.; Gillies, R.J. Why do cancers have high aerobic glycolysis? Nat. Rev. Cancer 2004, 4, 891–899.
  52. Kelm, J.M.; Fussenegger, M. Microscale tissue engineering using gravity-enforced cell assembly. Trends Biotechnol. 2004, 22, 195–202.
  53. Friedrich, J.; Ebner, R.; Kunz-Schughart, L.A. Experimental anti-tumor therapy in 3-D: Spheroids—Old hat or new challenge? Int. J. Radiat. Biol. 2007, 83, 849–871.
  54. Friedrich, J.; Seidel, C.; Ebner, R.; Kunz-Schughart, L.A. Spheroid-based drug screen: Considerations and practical approach. Nat. Protoc. 2009, 4, 309–324.
  55. Ham, S.L.; Joshi, R.; Thakuri, P.S.; Tavana, H. Liquid-based three-dimensional tumor models for cancer research and drug discovery. Exp. Biol. Med. 2016, 241, 939–954.
  56. Santi, M.; Mapanao, A.K.; Cappello, V.; Voliani, V. Production of 3D tumor models of head and neck squamous cell carcinomas for nanotheranostics assessment. ACS Biomater. Sci. Eng. 2020, 6, 4862–4869.
  57. Ivascu, A.; Kubbies, M. Rapid generation of single-tumor spheroids for high-throughput cell function and toxicity analysis. J Biomol. Screen. 2006, 11, 922–932.
  58. Tevis, K.M.; Colson, Y.L.; Grinstaff, M.W. Embedded Spheroids as Models of the Cancer Microenvironment. Adv. Biosyst. 2017, 1, 1700083.
  59. Hagemann, J.; Jacobi, C.; Gstoettner, S.; Welz, C.; Schwenk-Zieger, S.; Stauber, R.; Strieth, S.; Kuenzel, J.; Baumeister, P.; Becker, S. Therapy testing in a spheroid-based 3D cell culture model for head and neck squamous cell carcinoma. J. Vis. Exp. 2018, 2018, e57012.
  60. Shao, S.; Scholtz, L.U.; Gendreizig, S.; Martínez-Ruiz, L.; Florido, J.; Escames, G.; Schürmann, M.; Hain, C.; Hose, L.; Mentz, A.; et al. Primary head and neck cancer cell cultures are susceptible to proliferation of Epstein-Barr virus infected lymphocytes. BMC Cancer 2023, 23, 47.
  61. Hagemann, J.; Jacobi, C.; Hahn, M.; Schmid, V.; Welz, C.; Schwenk-Zieger, S.; Stauber, R.; Baumeister, P.; Becker, S. Spheroid-based 3D cell cultures enable personalized therapy testing and drug discovery in head and neck cancer. Anticancer. Res. 2017, 37, 2201–2210.
  62. Vakhshiteh, F.; Bagheri, Z.; Soleimani, M.; Ahvaraki, A.; Pournemat, P.; Alavi, S.E.; Madjd, Z. Heterotypic tumor spheroids: A platform for nanomedicine evaluation. J. Nanobiotechnol. 2023, 21, 249.
  63. Antoni, D.; Burckel, H.; Josset, E.; Noel, G. Three-dimensional cell culture: A breakthrough in vivo. Int. J. Mol. Sci. 2015, 16, 5517–5527.
  64. Kobayashi, H.; Man, S.; Graham, C.H.; Kapitain, S.J.; Teicher, B.A.; Kerbel, R.S. Acquired multicellular-mediated resistance to alkylating agents in cancer. Proc. Natl. Acad. Sci. USA 1993, 90, 3294–3298.
  65. He, Y.; Deng, P.; Yan, Y.; Zhu, L.; Chen, H.; Li, T.; Li, Y.; Li, J. Matrisome provides a supportive microenvironment for oral squamous cell carcinoma progression. J. Proteom. 2022, 253, 104454.
  66. Anbazhagan, R.; Sakakura, T.; Gusterson, B.A. The distribution of immuno-reactive tenascin in the epithelial-mesenchymal junctional areas of benign and malignant squamous epithelia. Virchows Arch. B Cell Pathol. Incl. Mol. Pathol. 1990, 59, 59–63.
  67. Berndt, A.; Hyckel, P.; Kosmehl, H.; Könneker, A. Dreidimensionales In-vitro-Invasionsmodell für orale Plattenepithelkarzinome. Mund-Kiefer-und Gesichtschirurgie 1998, 2, 256–260.
  68. Fischbach, C.; Chen, R.; Matsumoto, T.; Schmelzle, T.; Brugge, J.S.; Polverini, P.J.; Mooney, D.J. Engineering tumors with 3D scaffolds. Nat. Methods 2007, 4, 855–860.
  69. Rossi, L.; Corvò, R.; Videtic, G.M.; Paulus, R.; Singh, A.K.; Chang, J.Y.; Parker, W.; Olivier, K.R.; Timmerman, R.D.; Komaki, R.R.; et al. Retinoic acid modulates the radiosensitivity of head-and-neck squamous carcinoma cells grown in collagen gel. Int. J. Radiat. Oncol. 2002, 53, 1319–1327.
  70. 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.
  71. Gu, C.; Zhang, Y.; Chen, D.; Liu, H.; Mi, K. Tunicamycin-induced endoplasmic reticulum stress inhibits chemoresistance of FaDu hypopharyngeal carcinoma cells in 3D collagen I cultures and in vivo. Exp. Cell Res. 2021, 405, 112725.
  72. Young, M.; Rodenhizer, D.; Dean, T.; D’Arcangelo, E.; Xu, B.; Ailles, L.; McGuigan, A.P. A TRACER 3D Co-Culture tumour model for head and neck cancer. Biomaterials 2018, 164, 54–69.
  73. Dean, T.; Li, N.T.; Cadavid, J.L.; Ailles, L.; McGuigan, A.P. A TRACER culture invasion assay to probe the impact of cancer associated fibroblasts on head and neck squamous cell carcinoma cell invasiveness. Biomater. Sci. 2020, 8, 3078–3094.
  74. Ricci, C.; Moroni, L.; Danti, S. Cancer tissue engineering new perspectives in understanding the biology of solid tumours a critical review. OA Tissue Eng. 2013, 1, 1–7.
  75. Wang, J.-Z.; Zhu, Y.-X.; Ma, H.-C.; Chen, S.-N.; Chao, J.-Y.; Ruan, W.-D.; Wang, D.; Du, F.-G.; Meng, Y.-Z. Developing multi-cellular tumor spheroid model (MCTS) in the chitosan/collagen/alginate (CCA) fibrous scaffold for anticancer drug screening. Mater. Sci. Eng. C 2016, 62, 215–225.
  76. Curvello, R.; Kast, V.; Ordóñez-Morán, P.; Mata, A.; Loessner, D. Biomaterial-based platforms for tumour tissue engineering. Nat. Rev. Mater. 2023, 8, 314–330.
  77. Villasante, A.; Vunjak-Novakovic, G. Tissue-engineered models of human tumors for cancer research. Expert Opin. Drug Discov. 2015, 10, 257–268.
  78. Gu, Y.; Schwarz, B.; Forget, A.; Barbero, A.; Martin, I.; Shastri, V.P. Advanced bioink for 3D bioprinting of complex free-standing structures with high stiffness. Bioengineering 2020, 7, 141.
  79. Zhang, Y.S.; Duchamp, M.; Oklu, R.; Ellisen, L.W.; Langer, R.; Khademhosseini, A. Bioprinting the Cancer Microenvironment. ACS Biomater. Sci. Eng. 2016, 2, 1710–1721.
  80. Murphy, S.V.; Atala, A. 3D bioprinting of tissues and organs. Nat. Biotechnol. 2014, 32, 773–785.
  81. Hoarau-Véchot, J.; Rafii, A.; Touboul, C.; Pasquier, J. Halfway between 2D and animal models: Are 3D cultures the ideal tool to study cancer-microenvironment interactions? Int. J. Mol. Sci. 2018, 19, 181.
  82. Zhang, Y.S.; Yue, K.; Aleman, J.; Mollazadeh-Moghaddam, K.; Bakht, S.M.; Yang, J.; Jia, W.; Dell’erba, V.; Assawes, P.; Shin, S.R.; et al. 3D Bioprinting for Tissue and Organ Fabrication. Ann. Biomed. Eng. 2017, 45, 148–163.
  83. Boland, T.; Xu, T.; Damon, B.; Cui, X. Application of inkjet printing to tissue engineering. Biotechnol. J. 2006, 1, 910–917.
  84. Chang, C.C.; Boland, E.D.; Williams, S.K.; Hoying, J.B. Direct-write bioprinting three-dimensional biohybrid systems for future regenerative therapies. J. Biomed. Mater. Res. Part B Appl. Biomater. 2011, 98B, 160–170.
  85. Guillotin, B.; Souquet, A.; Catros, S.; Duocastella, M.; Pippenger, B.; Bellance, S.; Bareille, R.; Rémy, M.; Bordenave, L.; Amédée, J.; et al. Laser assisted bioprinting of engineered tissue with high cell density and microscale organization. Biomaterials 2010, 31, 7250–7256.
  86. Ma, X.; Qu, X.; Zhu, W.; Li, Y.; Yuan, S.; Zhang, H.; Liu, J.; Wang, P.; Lai, C.S.E.; Zanella, F.; et al. Deterministically patterned biomimetic human iPSC-derived hepatic model via rapid 3D bioprinting. Proc. Natl. Acad. Sci. USA 2016, 113, 2206–2211.
  87. Nie, J.; Gao, Q.; Fu, J.; He, Y. Grafting of 3D Bioprinting to In Vitro Drug Screening: A Review. Adv. Healthc. Mater. 2020, 9, e1901773.
  88. Kort-Mascort, J.; Bao, G.; Elkashty, O.; Flores-Torres, S.; Munguia-Lopez, J.G.; Jiang, T.; Ehrlicher, A.J.; Mongeau, L.; Tran, S.D.; Kinsella, J.M. Decellularized Extracellular Matrix Composite Hydrogel Bioinks for the Development of 3D Bioprinted Head and Neck in Vitro Tumor Models. ACS Biomater. Sci. Eng. 2021, 7, 5288–5300.
  89. Kort-Mascort, J.; Shen, M.L.; Martin, E.; Flores-Torres, S.; Pardo, L.A.; Siegel, P.M.; Tran, S.D.; Kinsella, J.M. Bioprinted cancer-stromal in-vitro models in a decellularized ECM-based bioink exhibit progressive remodeling and maturation. Biomed. Mater. 2023, 18, 045022.
  90. Saglam-Metiner, P.; Gulce-Iz, S.; Biray-Avci, C. Bioengineering-inspired three-dimensional culture systems: Organoids to create tumor microenvironment. Gene 2019, 686, 203–212.
  91. Barbet, V.; Broutier, L. Future Match Making: When Pediatric Oncology Meets Organoid Technology. Front. Cell Dev. Biol. 2021, 9, 674219.
  92. Kim, S.; Choung, S.; Sun, R.X.; Ung, N.; Hashemi, N.; Fong, E.J.; Lau, R.; Spiller, E.; Gasho, J.; Foo, J.; et al. Comparison of Cell and Organoid-Level Analysis of Patient-Derived 3D Organoids to Evaluate Tumor Cell Growth Dynamics and Drug Response. SLAS Discov. Adv. Sci. Drug Discov. 2020, 25, 744–754.
  93. Clevers, H. Modeling Development and Disease with Organoids. Cell 2016, 165, 1586–1597.
  94. Papaccio, F.; Cabeza-Segura, M.; Garcia-Micò, B.; Tarazona, N.; Roda, D.; Castillo, J.; Cervantes, A. Will Organoids Fill the Gap towards Functional Precision Medicine? J. Pers. Med. 2022, 12, 1939.
  95. Yan, H.H.N.; Siu, H.C.; Law, S.; Ho, S.L.; Yue, S.S.K.; Tsui, W.Y.; Chan, D.; Chan, A.S.; Ma, S.; Lam, K.O.; et al. A Comprehensive Human Gastric Cancer Organoid Biobank Captures Tumor Subtype Heterogeneity and Enables Therapeutic Screening. Cell Stem Cell 2018, 23, 882–897.e811.
  96. Driehuis, E.; Kretzschmar, K.; Clevers, H. Establishment of patient-derived cancer organoids for drug-screening applications. Nat. Protoc. 2020, 15, 3380–3409.
  97. Hughes, C.S.; Postovit, L.M.; Lajoie, G.A. Matrigel: A complex protein mixture required for optimal growth of cell culture. Proteomics 2010, 10, 1886–1890.
  98. Kaur, S.; Kaur, I.; Rawal, P.; Tripathi, D.M.; Vasudevan, A. Non-matrigel scaffolds for organoid cultures. Cancer Lett. 2021, 504, 58–66.
  99. Kozlowski, M.T.; Crook, C.J.; Ku, H.T. Towards organoid culture without Matrigel. Commun. Biol. 2021, 4, 1387.
  100. Gunti, S.; Hoke, A.T.K.; Vu, K.; London, N.R. Organoid and spheroid tumor models: Techniques and applications. Cancers 2021, 13, 874.
  101. Wang, X.-W.; Xia, T.-L.; Tang, H.-C.; Liu, X.; Han, R.; Zou, X.; Zhao, Y.-T.; Chen, M.-Y.; Li, G. Establishment of a patient-derived organoid model and living biobank for nasopharyngeal carcinoma. Ann. Transl. Med. 2022, 10, 526.
  102. Tuveson, D.A.; Clevers, H. Cancer modeling meets human organoid technology. Science 2019, 364, 952–955.
  103. 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.
  104. Kijima, T.; Nakagawa, H.; Shimonosono, M.; Chandramouleeswaran, P.M.; Hara, T.; Sahu, V.; Kasagi, Y.; Kikuchi, O.; Tanaka, K.; Giroux, V.; et al. Three-Dimensional Organoids Reveal Therapy Resistance of Esophageal and Oropharyngeal Squamous Cell Carcinoma Cells. Cell. Mol. Gastroenterol. Hepatol. 2019, 7, 73–91.
  105. 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.
  106. Zhao, H.; Hu, C.-Y.; Chen, W.-M.; Huang, P. Lactate Promotes Cancer Stem-like Property of Oral Sequamous Cell Carcinoma. Curr. Med. Sci. 2019, 39, 403–409.
  107. 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.
  108. Bonartsev, A.P.; Lei, B.; Kholina, M.S.; Menshikh, K.A.; Svyatoslavov, D.S.; Samoylova, S.I.; Sinelnikov, M.Y.; Voinova, V.V.; Shaitan, K.V.; Kirpichnikov, M.P.; et al. Models of head and neck squamous cell carcinoma using bioengineering approaches. Crit. Rev. Oncol. 2022, 175, 103724.
  109. Verduin, M.; Hoeben, A.; De Ruysscher, D.; Vooijs, M. Patient-Derived Cancer Organoids as Predictors of Treatment Response. Front. Oncol. 2021, 11, 820.
  110. Vlachogiannis, G.; Hedayat, S.; Vatsiou, A.; Jamin, Y.; Fernández-Mateos, J.; Khan, K.; Lampis, A.; Eason, K.; Huntingford, I.; Burke, R.; et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science 2018, 359, 920–926.
  111. Yao, Y.; Xu, X.; Yang, L.; Zhu, J.; Wan, J.; Shen, L.; Xia, F.; Fu, G.; Deng, Y.; Pan, M.; et al. Patient-Derived Organoids Predict Chemoradiation Responses of Locally Advanced Rectal Cancer. Cell Stem Cell 2020, 26, 17–26.E16.
  112. Park, M.; Kwon, J.; Kong, J.; Moon, S.M.; Cho, S.; Yang, K.Y.; Jang, W.I.; Kim, M.S.; Kim, Y.; Shin, U.S. A patient-derived organoid-based radiosensitivity model for the prediction of radiation responses in patients with rectal cancer. Cancers 2021, 13, 3760.
  113. Smith, R.C.; Tabar, V. Constructing and Deconstructing Cancers using Human Pluripotent Stem Cells and Organoids. Cell Stem Cell 2019, 24, 12–24.
  114. Wu, K.Z.; Adine, C.; Mitriashkin, A.; Aw, B.J.J.; Iyer, N.G.; Fong, E.L.S. Making In Vitro Tumor Models Whole Again. Adv. Healthc. Mater. 2023, 12, e2202279.
  115. Engelmann, L.; Thierauf, J.; Laureano, N.K.; Stark, H.-J.; Prigge, E.-S.; Horn, D.; Freier, K.; Grabe, N.; Rong, C.; Federspil, P.; et al. Organotypic co-cultures as a novel 3d model for head and neck squamous cell carcinoma. Cancers 2020, 12, 2330.
  116. Dohmen, A.J.; Sanders, J.; Canisius, S.; Jordanova, E.S.; Aalbersberg, E.A.; Brekel, M.W.v.D.; Neefjes, J.; Zuur, C.L. Sponge-supported cultures of primary head and neck tumors for an optimized preclinical model. Oncotarget 2018, 9, 25034–25047.
  117. Lee, J.; You, J.H.; Shin, D.; Roh, J.-L. Ex vivo culture of head and neck cancer explants in cell sheet for testing chemotherapeutic sensitivity. J. Cancer Res. Clin. Oncol. 2020, 146, 2497–2507.
  118. Demers, I.; Donkers, J.; Kremer, B.; Speel, E.J. Ex Vivo Culture Models to Indicate Therapy Response in Head and Neck Squamous Cell Carcinoma. Cells 2020, 9, 2527.
  119. Peria, M.; Donnadieu, J.; Racz, C.; Ikoli, J.; Galmiche, A.; Chauffert, B.; Page, C. Evaluation of individual sensitivity of head and neck squamous cell carcinoma to cetuximab by short-term culture of tumor slices. Head Neck 2016, 38, E911–E915.
  120. Gerlach, M.M.; Merz, F.; Wichmann, G.; Kubick, C.; Wittekind, C.; Lordick, F.; Dietz, A.; Bechmann, I. Slice cultures from head and neck squamous cell carcinoma: A novel test system for drug susceptibility and mechanisms of resistance. Br. J. Cancer 2014, 110, 479–488.
  121. Bower, R.; Green, V.L.; Kuvshinova, E.; Kuvshinov, D.; Karsai, L.; Crank, S.T.; Stafford, N.D.; Greenman, J. Maintenance of head and neck tumor on-chip: Gateway to personalized treatment? Futur. Sci. OA 2017, 3, FSO174.
  122. Mahmutović, L.; Bilajac, E.; Hromić-Jahjefendić, A. Meet the insidious players: Review of viral infections in head and neck cancer etiology with an update on clinical trials. Microorganisms 2021, 9, 1001.
  123. Forslund, O.; Sugiyama, N.; Wu, C.; Ravi, N.; Jin, Y.; Swoboda, S.; Andersson, F.; Bzhalava, D.; Hultin, E.; Paulsson, K.; et al. A novel human in vitro papillomavirus type 16 positive tonsil cancer cell line with high sensitivity to radiation and cisplatin. BMC Cancer 2019, 19, 265.
  124. Wegge, M.; Dok, R.; Dubois, L.J.; Nuyts, S. Use of 3D Spheroid Models for the Assessment of RT Response in Head and Neck Cancer. Int. J. Mol. Sci. 2023, 24, 3763.
  125. Vitti, E.T.; Kacperek, A.; Parsons, J.L. Targeting DNA double-strand break repair enhances radiosensitivity of HPV-positive and HPV-negative head and neck squamous cell carcinoma to photons and protons. Cancers 2020, 12, 1490.
  126. Millen, R.; De Kort, W.W.; Koomen, M.; van Son, G.J.; Gobits, R.; de Vries, B.P.; Begthel, H.; Zandvliet, M.; Doornaert, P.; Raaijmakers, C.P.; et al. Patient-derived head and neck cancer organoids allow treatment stratification and serve as a tool for biomarker validation and identification. Med 2023, 4, 290–310.e12.
  127. Almela, T.; Tayebi, L.; Moharamzadeh, K. 3D bioprinting for in vitro models of oral cancer: Toward development and validation. Bioprinting 2021, 22, e00132.
  128. Matuszczak, S.; Szczepanik, K.; Grządziel, A.; Drzyzga, A.; Cichoń, T.; Czapla, J.; Pilny, E.; Smolarczyk, R. The Effect of Radiotherapy on Cell Survival and Inflammatory Cytokine and Chemokine Secretion in a Co-Culture Model of Head and Neck Squamous Cell Carcinoma and Normal Cells. Biomedicines 2023, 11, 1773.
  129. Ludwig, N.; Yerneni, S.S.; Razzo, B.M.; Whiteside, T.L. Exosomes from HNSCC promote angiogenesis through reprogramming of endothelial cells. Mol. Cancer Res. 2018, 16, 1798–1808.
  130. Choi, S.-Y.; Kang, S.H.; Oh, S.Y.; Lee, K.Y.; Lee, H.-J.; Gum, S.; Kwon, T.-G.; Kim, J.-W.; Lee, S.-T.; Hong, Y.J.; et al. Differential angiogenic potential of 3-dimension spheroid of hnscc cells in mouse xenograft. Int. J. Mol. Sci. 2021, 22, 8245.
  131. Yang, J.; Wang, W.; Xia, H.; Yu, Z.; Li, H.; Ren, J.; Chen, G.; Wang, B.; Jia, J.; Zhang, W.; et al. Lymphotoxin-α promotes tumor angiogenesis in HNSCC by modulating glycolysis in a PFKFB3-dependent manner. Int. J. Cancer 2019, 145, 1358–1370.
  132. Gilazieva, Z.; Ponomarev, A.; Rutland, C.; Rizvanov, A.; Solovyeva, V. Promising applications of tumor spheroids and organoids for personalized medicine. Cancers 2020, 12, 2727.
  133. Ehsan, S.M.; Welch-Reardon, K.M.; Waterman, M.L.; Hughes, C.C.W.; George, S.C. A three-dimensional in vitro model of tumor cell intravasation. Integr. Biol. 2014, 6, 603–610.
  134. Buchanan, C.F.; Verbridge, S.S.; Vlachos, P.P.; Rylander, M.N. Flow shear stress regulates endothelial barrier function and expression of angiogenic factors in a 3D microfluidic tumor vascular model. Cell Adhes. Migr. 2014, 8, 517–524.
  135. Cauli, E.; Polidoro, M.A.; Marzorati, S.; Bernardi, C.; Rasponi, M.; Lleo, A. Cancer-on-chip: A 3D model for the study of the tumor microenvironment. J. Biol. Eng. 2023, 17, 53.
  136. Clarke, R. Introduction: Cancer Systems and Integrative Biology; Humana: New York, NY, USA, 2023; Volume 2660.
  137. Tuomainen, K.; Al-Samadi, A.; Potdar, S.; Turunen, L.; Turunen, M.; Karhemo, P.-R.; Bergman, P.; Risteli, M.; Åström, P.; Tiikkaja, R.; et al. Human tumor–derived matrix improves the predictability of head and neck cancer drug testing. Cancers 2020, 12, 92.
  138. Naakka, E.; Wahbi, W.; Tiikkaja, R.; Juurikka, K.; Sandvik, T.; Koivunen, P.; Autio, T.; Tikanto, J.; Väisänen, J.; Tuominen, H.; et al. Novel human lymph node-derived matrix supports the adhesion of metastatic oral carcinoma cells. BMC Cancer 2023, 23, 750.
  139. Drost, J.; Clevers, H. Organoids in cancer research. Nat. Rev. Cancer 2018, 18, 407–418.
  140. Pillai, S.; Kwan, J.C.; Yaziji, F.; Yu, H.; Tran, S.D. Mapping the Potential of Microfluidics in Early Diagnosis and Personalized Treatment of Head and Neck Cancers. Cancers 2023, 15, 3894.
  141. Cheah, R.; Srivastava, R.; Stafford, N.D.; Beavis, A.W.; Green, V.; Greenman, J. Measuring the response of human head and neck squamous cell carcinoma to irradiation in a microfluidic model allowing customized therapy. Int. J. Oncol. 2017, 51, 1227–1238.
  142. Sawant, S.; Dongre, H.; Singh, A.K.; Joshi, S.; Costea, D.E.; Mahadik, S.; Ahire, C.; Makani, V.; Dange, P.; Sharma, S.; et al. Establishment of 3D co-culture models from different stages of human tongue tumorigenesis: Utility in understanding neoplastic progression. PLoS ONE 2016, 11, e0160615.
  143. Establishment of Squamous Cell Organoids of the Head and Neck to Assess Their Response to Innovative Therapies (ORGAVADS); ClinicalTrials.gov Identifier: NCT04261192. Available online: https://clinicaltrials.gov/study/NCT04261192 (accessed on 1 October 2023).
  144. Perréard, M.; Florent, R.; Divoux, J.; Grellard, J.-M.; Lequesne, J.; Briand, M.; Clarisse, B.; Rousseau, N.; Lebreton, E.; Dubois, B.; et al. ORGAVADS: Establishment of tumor organoids from head and neck squamous cell carcinoma to assess their response to innovative therapies. BMC Cancer 2023, 23, 223.
  145. Selecting Chemotherapy with High-Throughput Drug Screen Assay Using Patient Derived Organoids in Patients with Refractory Solid Tumours (SCORE); ClinicalTrials.gov Identifier: NCT04279509. Available online: https://clinicaltrials.gov/study/NCT04279509 (accessed on 1 October 2023).
  146. SOTO: Treatment Sensitivity of Organoids to Predict Treatment Outcome; ClinicalTrials.gov Identifier: NCT05400239. Available online: https://classic.clinicaltrials.gov/ct2/history/NCT05400239?V_2=View (accessed on 1 October 2023).
More
Information
Subjects: Biology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 388
Revisions: 4 times (View History)
Update Date: 20 Nov 2023
1000/1000
ScholarVision Creations