Open Access

Exploring oncology treatment strategies with tyrosine kinase inhibitors through advanced 3D models (Review)

  • Authors:
    • Giorgia Isinelli
    • Sharon Failla
    • Roberto Plebani
    • Alessandro Prete
  • View Affiliations

  • Published online on: December 20, 2024     https://doi.org/10.3892/mi.2024.212
  • Article Number: 13
  • Copyright : © Isinelli et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The limitations of two‑dimensional (2D) models in cancer research have hindered progress in fully understanding the complexities of drug resistance and therapeutic failures. However, three‑dimensional (3D) models provide a more accurate representation of in vivo environments, capturing critical cellular interactions and dynamics that are essential in evaluating the efficacy and toxicity of tyrosine kinase inhibitors (TKIs). These advanced models enable researchers to explore drug resistance mechanisms with greater precision, optimizing treatment strategies and improving the predictive accuracy of clinical outcomes. By leveraging 3D models, it will be possible to deepen the current understanding of TKIs and drive forward innovations in cancer treatment. The present review discusses the limitations of 2D models and the transformative impact of 3D models on oncology research, highlighting their roles in addressing the challenges of 2D systems and advancing TKI studies.

1. Introduction

Two-dimensional (2D) cell cultures have long been used in oncology to study cancer behaviors and drug responses (1). However, these models fall short in replicating the complex interactions of the tumor microenvironment (TME), limiting their ability to accurately predict therapeutic outcomes and resistance mechanisms. As a result, the translational potential of preclinical findings based on 2D systems is constrained. To address these limitations, three-dimensional (3D) models have emerged, providing a more realistic simulation of in vivo tumor dynamics. By capturing key aspects of cellular architecture and TME interactions, 3D models enhance our understanding of cancer biology and improve the evaluation of chemotherapeutic agents, particularly tyrosine kinase inhibitors (TKIs). The present review discusses the limitations of 2D models and the transformative impact of 3D models on oncology research, highlighting their roles in overcoming the shortcomings of 2D systems and advancing TKI studies.

2. Limitations of 2D models in assessing chemotherapeutic drug responses

As presented in Table I, 2D models present multiple limitations. The evaluation of pharmacokinetics is limited in 2D models, as they often neglect crucial aspects, such as drug metabolism, distribution and excretion. For instance, penetration into tumor tissues and interactions with surrounding cells and extracellular matrix (ECM) components remain inadequately understood with 2D models (2). These elements, indeed, play a vital role in determining drug efficacy and toxicity but pose challenges for accurate assessment within 2D culture systems.

Table I

Limitations and implications of 2D models.

Table I

Limitations and implications of 2D models.

Limitation of 2D modelsImplications(Refs.)
Evaluation of pharmacokineticsOften neglect crucial aspects such as drug metabolism, distribution, and excretion(187)
Failure to replicate drug resistance mechanismsResistance to chemotherapy via various pathways, including genetic mutations, epigenetic alterations and interactions with the surrounding microenvironment may not be faithfully represented(3)
Lack of physiological gradientsPoor understanding of drug effects on complex biological systems(4-6)
Erroneous predictions of drug effectivenessDiminished predictive precision concerning drug reactions and adverse effects in humans(5,6)
Lack the three-dimensional arrangement and cellular interplayConsequently, there is no prove of the interactions with extracellular matrix, immune cells and blood vessels affecting the study of drug interactions in a physiologically relevant context(7-10)
Lack of dimensionality of the extracellular matrix surrounding cellsIt profoundly influences cell proliferation, differentiation, cell survival and cellular responses to external stimuli and challenges(11-15)
Tumorigenesis and metastasis are oversimplified in monoculturesThe study of metastasis process cannot be reduced to cell movement through a wound healing assay. It should also include evaluating whether these cells can invade tissues or/and enter vessels(27)
False positives or negatives may occur in drug screeningSimplistic 2D models fail to translate to success in more complex in vivo models or clinical trials(29)
Poorly understood interactions between malignant cells and stromal components within the tumor microenvironmentThese elements are crucial in influencing tumor initiation, progression, and response to treatment(35)

The failure to replicate drug resistance mechanisms is another significant drawback of 2D models. Tumor cells can develop resistance to chemotherapy via various pathways, including genetic mutations, epigenetic alterations and interactions with the surrounding microenvironment (3). Moreover, 2D models typically lack key physiological aspects, such as gradients of oxygen, nutrients and signaling molecules inherent in complex biological systems. These gradients play fundamental roles in influencing cellular responses to drugs, including developing resistance mechanisms (4). All these complexities may not be faithfully represented in 2D models and lead to erroneous predictions of drug effectiveness (5,6). The primary constraint of 2D models lies in their diminished predictive precision concerning drug reactions and adverse effects in humans.

The 2D models lack the 3D arrangement and cellular interplay found in vivo, such as ECM, immune cells and blood vessels, which can significantly affect the study of drug interactions in a physiologically relevant context (7-10). Although they provide valuable insight into fundamental cellular mechanisms, their capacity to faithfully recreate intricate in vivo circumstances remains constrained. Emerging evidence from recent studies suggests that augmenting the dimensionality of the ECM surrounding cells and transitioning from 2D to 3D models can profoundly influence cell proliferation, differentiation, cell survival, and, most importantly, cellular responses to external stimuli and challenges (11-15). The absence of mechanical forces, such as shear stress and compression, which are prevalent in the TME, further limits the relevance of 2D models in recapitulating in vivo conditions (16,17). With regards to this, various studies have used models able to respond to mechanical stimuli (18-20). In the study by Gill and West (21), it was demonstrated that their model was able to respond to mechanical stimuli and alter the bioactivity. Several models have been proposed to optimize the ECM composition and cell interactions, modeling tissues with higher fidelity and providing more suitable platforms to be used in drug testing and cancer treatment response (22-24) as shown by Mukubou et al (23) in a xenograft model with gemcitabine and radiotherapy treatment. For example, in anchorage-dependent cells, adhesive interactions with the extracellular matrix and neighboring cells are vital for determining the shape, spatial organization, gene expression, proliferation rate, response to stimuli and drug metabolism. These factors collectively regulate the close association between cell structure, signaling and function (25,26).

Numerous aspects of tumorigenesis and metastasis are frequently oversimplified in monocultures; for example, Riedl et al (27) proved that 3D models exhibited more potent antitumor responses to AKT/mTOR/S6K or MAPK pathway inhibitors compared to 2D models. In 2D cultures, blocking AKT/mTOR/S6K increased ERK phosphorylation; however, in 3D spheroids, ERK signaling was reduced under the same conditions (27). Consequently, 2D cultures often do not adequately recapitulate the complex TME, diffusion gradients, and cellular attributes characteristic of in vivo systems. This disparity contributes to deviations from the anticipated response observed in animal and computational modeling, as well as during clinical testing (13,28).

False positives or negatives may occur in drug screening when compounds, that appear effective in simplistic 2D models, fail to translate to success in more complex in vivo models or clinical trials. Horvath et al (29) noted that the recurrent inability to translate promising preclinical drug candidates into clinical success underscores the limited efficacy of current disease models employed in drug discovery. A clear reluctance to explore and embrace alternative cell- and tissue-based model systems, along with a disconnect from clinical practice during assay validation, contributes to ineffective translational research. As personalized medicine seeks to customize treatments according to individual patients' distinct genetic and molecular characteristics, 2D models may fall short in capturing the specific factors influencing drug responsiveness (30). This limitation curtails their effectiveness in personalized medicine strategies. For example, in ovarian cancer, the introduction of 3D models has allowed for the identification of various important features, as demonstrated by Kerslake et al (31). The top enriched gene sets for 2D vs. 3D included IFN-α and the IFN-γ response, TNF-α signaling, IL-6-JAK/STAT3 signaling, angiogenesis, hedgehog signaling, apoptosis, epithelial-mesenchymal transition (EMT), hypoxia and the inflammatory response (31).

Numerous investigations have underscored that the intricacies of tissue organization, differentiation and gene expression are more faithfully portrayed in 3D cell cultures (32,33). The configuration displayed in 3D models facilitates the growth of cells within an environment that maintains spatial complexities akin to in vivo conditions, enabling cells to differentiate and interact in a tissue-specific manner (34). In the field of cancer, interactions between malignant cells and stromal components within the TME, comprising both cancerous and non-cancerous cellular and non-cellular elements, are crucial in influencing tumor initiation, progression, and response to treatment. This reciprocal communication profoundly influences tumor behavior (35).

The present review discusses how 3D models have aided in the understanding of actual TKI treatment and their resistance mechanisms, owing to the ability to study the TME in a more detailed manner.

3. Most frequently used 3D models

The analysis of the TME plays a pivotal role in investigating novel anticancer therapies, particularly in elucidating mechanisms of resistance to immunotherapies. Utilizing 3D cell culture models enables the comprehensive analysis of tumor cell architecture and composition, facilitating the identification of potential responders to immunotherapy. 3D models, on the other hand, reproducing the TME more accurately, represent an exciting alternative in experimentation (36). Through 3D models, it is possible to perform more detailed experiments, often bridging the gap between 2D cell cultures and animal models (37). Ideally, a 3D model should simulate the specific microenvironment of a pathophysiological condition. However, the currently available models do not entirely fulfill these criteria, each presenting its strengths and limitations. Therefore, it is advisable to select the 3D model most appropriate to the specific need. 3D models can be divided into those with or without scaffolding (Fig. 1). In the first case, there is no need for an external support structure. In the second case, the seeding and cultivation of cells take place via structures that provide physical support. Models equipped with a scaffold can more easily imitate the interactions between the cell and the ECM, while models without a scaffold, such as spheroids, depending on their size, are more susceptible to physiological as well as cellular gradients (18,35).

Scaffold-free 3D models: Spheroids and organoids

Currently, several techniques do not use the support of a scaffold to generate a tissue of interest. The first method developed was the suspended drop technique introduced in 1944 for the culture of embryonic stem cells. This technique is now mainly used for the generation of spheroids. Scaffold-free culture methods are therefore based on the ability to form a specific tissue, exploiting cellular self-aggregation, thus producing a 3D matrix (38). The use of scaffold-free methods, through a high yield, provides the possibility of combining different cell types while exploiting simple methodologies (39). Among the techniques used for the production of scaffold-free 3D models, it is possible to include hanging drop, forced floating, micro molding, agitation-based techniques, magnetic levitation, microfluidics and pellet culture (40) (Fig. 1): i) In the hanging drop method, a single spheroid is formed following the accumulation of cells at the liquid-air interface due to the inversion of the tray in the plate. ii) The forced floating method results in high-performance data, using plates coated with hydrophilic material or completely uncoated (41); iii) in the micro molding techniques, micro molds used are generally made of agarose, in which the cells are seeded and left to self-aggregate (42). iv) Agitation-based techniques use bioreactors; the latter is equipped with a chamber that is filled with a cell suspension of appropriate density, which is constantly stirred to encourage aggregation (43-46). v) In the magnetic technique, cells with a 2D structure are incubated inside a specific apparatus equipped with magnetic nanoparticles. These have the function of binding in a non-specific electrostatic manner to the cell membrane, thus affecting its magnetization. Once magnetized, the cells are enzymatically dissociated, inserted into a 3D apparatus, and subjected to a further magnetic field generated through different techniques (such as levitation, ring formation, or bioprinting), thus inducing aggregation and generation of 3D structures (47). vi) In the microfluidic technique, a device consists of two states of polydimethylsiloxane (PDMS), of which the microfluidic channel constitutes the upper one, while the lower one presents the cell culture chambers. The size of the spheroid will thus depend on the geometry chosen for the culture chambers (48). vii) The pellet culture method allows for the control of the size of the spheroid as a function of the number of cells that are placed in suspension (49).

Spheroids. Spheroids are 3D cultures made of tumor cells that can assemble spontaneously into spheres during cell proliferation (50). Being cultured in ECM and the presence/absence of fibroblasts and immune cells allow the study of cell-cell and cell-ECM interactions (50). These structures generally have a diameter of 200 µm and have a spherical shape (50). Spheroids typically have a concentric cellular organization of three different layers: A nucleus comprised of necrotic cells, an intermediate cellular zone of quiescent cells, and finally, an external zone of proliferating migrating cells (50) (Fig. 2).

Spheroids represent one of the most commonly used 3D models in the study of tumor organization and functionality, being employed in the characterization of various tumor species such as breast, colon and different types of solid tumors. Depending on the cell type and preparation technique, tumor spheroids can be classified into four main classes, as follows: Tumorspheres obtained from solid tumors; tumorspheres derived from tissues remodeled following mechanical or enzymatic dissociation; multicellular tumor spheroids based on primary cell suspensions or cell lines; and finally, organotypic multicellular spheroids obtained from tumor fragment cultures (50-53).

One of the fundamental requirements during the design and creation of these models is to generate biomimetic prototypes that can model the interactions within the TME, as well as the control of the pharmacological action. Therefore, it is possible to state that these 3D tumor models can mimic cellular interactions through adhesion molecules and soluble factors, reproducing ECM with characteristics typically observed in the tumor condition (51).

Due to the difficulties encountered in faithfully reproducing the polymorphic characteristics of a tumor, increasingly complex spheroid models have been introduced. Beginning from simple monoculture, these models have allowed for the integration of heterotypic cells, ultimately leading to ex vivo models derived directly from patients. Although the majority of studies in the literature have focused on spheroids derived from monoculture, due to tumor complexity, it is always preferable to use a heterotypic platform capable of integrating different cell types (52).

Organoids. Organoids are multicellular structures generated from 3D cultures of stem cells in vitro. These cells can be embryonic, pluripotent, or adult tissue cells. Presenting the ability to self-assemble, they can exactly mimic the functionality of the originating tissue. Owing to these exceptional characteristics, the organoid model successfully maintains genetic and phenotypic stability (53). They can overcome both 2D and in vivo model limitations (54).

In vivo models, including patient-derived tumor cells (PDCs) and patient-derived xenografts (PDX), are often used as tumor models (55). PDCs are primary cell cultures obtained directly from tumor cells extracted from fluids or tissues; although they can replace the traditional tumor cell lines used during studies, their main limitation is that they are 2D models, therefore failing to faithfully reproduce the tumor architecture (56). The PDX model is generated using immunodeficient mice that are administered a graft of tumor cells extracted directly from the patient (57).

Although these models provide rather reliable similarities in the human component, they are associated with a series of issues, such as a low success rate, limited screening possibilities and relatively long implementation times. However, owing to their versatility, organoids are commonly used in a number of fields, such as oncology and microbiology.

One of the most promising applications of organoids and cancer organoids, the so-called tumoroids, derived from patients is certainly that of creating a biobank that allows for the implementation of personalized therapy based on pharmacological screening implemented on these models (58). To date, multiple available models of tumor organoids are used in tumor modeling, pharmacological screening, immunotherapy and precision medicine. These models have been applied in tumor forms such as bladder cancer, ovarian cancer, prostate cancer, liver cancer, breast cancer, colorectal cancer, lung cancer and pancreatic cancer (55).

Despite their interesting structure, organoids lack vascularization. Therefore, to correctly mimic highly vascularized tissues with a dynamic microenvironment, such as the brain and heart, it is essential to implement this feature. For this purpose, it is possible to use different vascularization techniques based mainly on the in vivo approach in an immunodeficient host (59,60).

Moreover, in the case of models aimed at studying pathologies such as infections, autoimmune components and cancer, it becomes essential to enrich the organoid with immune cells through epithelial organoid-immune cell coculture systems (61). Furthermore, by inserting innate immune cells into this model, it is possible to improve the degree of tumor attachment as well as the vascular inflammation typical of metastases (62).

Scaffold-based 3D models: Organ-on-a-chip (OoC) and organ-on-a-plate

3D models equipped with scaffolds aim to reconstruct the dynamic interactions that are established between cells of the TME and the ECM (63). Compared with spheroids, 3D models equipped with scaffolds imitate the ECM and influence the mechanical and biochemical signals typical of cell-cell interaction. Therefore, these models represent excellent structures for cultivating primary tumor cultures, carrying out pharmacological safety and efficacy screening, and studying the role of stromal cells in cancer (64).

The biomaterials used in these models can be divided into the following main groups: i) Polymeric scaffolds are usually made in polymers, such as polyacrylamide, polylactin, poly (lactide-co-glycolic acid) or polyurethane (65). ii) Decellularized matrices are derived directly from diseased tissues or organs after the removal of cellular components, while preserving the ECM composition. This maintains the native structure, as well as the biomechanical characteristics, resulting in highly accurate models (66,67). iii) Hydrogels can be composed of natural gels, such as collagen and fibrinogen, synthetic polymers, or hybrids. Owing to their ability to mimic the ECM, hydrogels can allow the passage of soluble molecules, such as growth factors and cytokines, similar to tissues (68). iv) Hybrid scaffolds combine different materials, such as synthetic and natural polymers or hydrogels (69).

The devices used for these 3D cultures can include different models based on their fabrication. For example, they can be fabricated by solid freeform fabrication. This technique allows for the development of different types of scaffolds depending on the materials used, geometry and size of the pores. The 3D project is developed using software. Through computer-aided design technology, it is possible to better control manufacturing (70). This technique has aroused immense interest, particularly for its ability to create an architecture that imitates the native tissue microenvironment, as well as the interaction with the cells and components of the ECM (71).

Some of these are microfluidic devices comprise of circuits that allow continuous cell feeding via laminar flow. Through microfabrication, microchannels and micropillars <100 µm are generated, with an overall structure in the order of mm. This architecture, being equipped with a solid surface, mimics a physical environment such as the ECM. Moreover, the organization of channels, unlike other 3D culture technologies, allows for microfluidic devices to cultivate different cell types. The substrate used can be made of glass and/or silicon, polymeric material, or paper-based. Among the mainly used materials, PDMS is widely applied in biomedical technology. Due to its high biocompatibility and the possibility of modifying surface characteristics, such as chemical, physical and biological properties, exceptional applications can be obtained (72,73). Alternatively, devices for 3D cultures can be assembled manually to better exploit the properties of the chosen materials are exploited, in terms of biocompatibility to mimic intercellular interactions and the interaction with the matrix (71,72).

OoC. The first OoC was proposed in 2010 by Huh et al (74) as an artificial physiological system created on a chip. Depending on their functionalities and modalities, OoCs can be divided into single-OoCs, multi-OoCs (MOoCs), human-on-a-chip, and tumor-on-a-chip (ToC) (75).

The design of an OoC begins with a reductionist analysis of the target organ. It is essential to understand the structure of the target, simplifying its physiological functions. Subsequently, it is necessary to consider the selection of the cell culture, to control the dynamic flow perfusion, design the microstructure and account for the mechanical movement of biological organs (76).

OoCs are comprised of a physical compartment in which the cells are confined in different microfluidic channels (Fig. 3). The latter is used for the administration and transport of substances and the evaluation of measurable signals. Owing to their integrated compartment, it is possible to evaluate cellular behavior and the response of the microenvironment of the organ to the stimuli provided. Within the channels of an OoC, it is possible to incorporate polymeric membranes, thus modeling tissue interfaces and imitating barrier functions. Furthermore, in certain cases, it is possible to establish a vascular structure, increasing the complexity of the system and, therefore, the representativeness of the model (77).

Different OoC designs are dependent on the type of organ to be studied and the required characteristics. Typically, the structural classification is made based on the number of channels present and their organization. Therefore, there are single-channel chips, double-channel chips, respectively parallel or sandwich type and multi-channel chips. The dimensions are in the order of 1 cm, and the two channels are connected by a porous membrane so that interphase interactions between cells of the same tissue can be studied. To control the entry and exit of the fluid used and the introduction of biological material, the structure is equipped with entrances and exits. As regards the materials used, glass represents one of the oldest products, while PDMS is the most commonly used material; furthermore, the use of thermoplastics is currently spreading (78).

OoC systems can replicate a single unit or can be interconnected with each other. Thus, they can be classified into single- or MOoCs. These microfluidic cultures can also enable the connection of multiple units, leading to the concept of a body-on-a-chip or the replication of a TME, resulting in a ToC:

i) Single-OoCs. Contrary to what might be assumed, ‘single’ does not mean these models are simple or have only one channel/chamber, but rather that they can replicate a single unit. Single-OoCs can indeed host very complex co-cultures and differentiated tissues and have been developed to study the microphysiology and pathophysiology of numerous diseases, such as viral infections (79-81), lung diseases (82), or intestinal disorders (83), as well as for toxicological studies (84). They for allow the maturation and function of microtissues and the emulation of the physiological microenvironment. Examples of OoC can include lung-on-a-chip, which can reproduce the lung microenvironment, as well as the air-liquid interface and the barrier function performed for inhalation agents and can be used to carry out comparison studies between smoking and non-smoking conditions (85); brain-on-a-chip, which could be used to better mimic the chemical and electrical conditions of the human nervous system; liver-on-a-chip to mimic the structure of liver lobules; kidney-on-a-chip for glomerular study. Other examples are skin-on-a-chip, uterus-on-a-chip and vessels-on-a-chip models (86,87).

ii) Multiorgan- and body-on-a-chip. Developments in the field of OoCs have also led to the establishment of MOoCs. The connection of individual 3D organ models has allowed for the study of pharmacokinetics and pharmacodynamics, thus monitoring the intricate interactions between organs and studying their responses to drugs. MOoCs can also emulate blood circulation, allowing for the study of the dynamic response of the organs, in addition to the chronic cellular responses and the interaction between different organs in complex processes, such as absorption, distribution, metabolism and excretion (88). Through OoC, it is possible to generate different tissues capable of mimicking the complex functioning of the human body. These multi-tissue systems can be created with entirely human cells, becoming important metabolization models (89).

iii) ToC. The concept of a ToC involves creating a microfluidic 3D system that accurately mimics the behavior, biological activities, mechanical properties and various responses of tumor cells (90-92). One of the great advantages of the ToC is having both normal and cancer cells in the same model, thus better replicating the complex TME and allowing more accurate predictions of drug efficacy and toxicity, leading to better-informed decisions in both preclinical and clinical settings. This technology has been applied to a wide variety of tumors, such as pancreatic tumors (93,94), lung (95-97), brain (98,99) and blood (98,100-102). These advanced devices are particularly valuable in the context of breast cancer research; Chen et al (103) employed this technology for drug screening by evaluating the effects of different chemotherapeutic agents on the tumor model.

By understanding the factors that induce resistance in tumor cells, researchers can discover novel therapeutic strategies to combat disease. This knowledge may help to reduce the risks associated with current treatments. In addition, when primary cells from patients are used, this may pave the way for the development of personalized drugs, which represent the next frontier in cancer treatment (104).

Organ-on-a-plate. As with OoC systems, also known as organ plate models, organ-on-a-plate is an advanced approach within the field of tissue engineering and microfluidics that aims to replicate human organ functions in a laboratory setting. Previous studies have utilized this platform to perform high-throughput analysis for various physiological processes, such as: i) T-cell extravasation under flow: This process was analyzed using organ-on-a-plate technology, demonstrating its capability to model immune cell dynamics within the vascular system (105); ii) endothelial cell permeability: Researchers assessed the permeability of endothelial cells, which are crucial for understanding vascular barrier functions and pathological conditions, such as inflammation and cancer metastasis (106); iii) intestinal tract epithelium integrity: This technology has been employed to study the integrity of the intestinal epithelium, providing insight into gastrointestinal diseases and drug absorption (107).

This technology was also used for studying metastatic models, mimicking the early stages of breast cancer metastasis, including tumor cell invasion and intravasation (108). Utilizing this platform, a library of 86 targeted anticancer drugs with annotated molecular targets was screened. The results demonstrated the suitability of the platform for preclinical drug testing and drug discovery (108). Moreover, the metastasis-on-chip platform can be integrated with other established assays, such as tumor growth or motility assays, to enhance drug profiling outcomes (108). The development of automated imaging and analysis methods for the metastasis-on-chip platform has enabled high-throughput phenotypic drug screening. This automation not only improves the efficiency and reproducibility of experiments, but also allows for the detailed and comprehensive analysis of drug effects on cancer cell behavior.

All the aforementioned models, the pros and cons of which are highlighted in Table II, have contributed to research on drug resistance mechanisms, specifically in resistance mechanisms toward TKIs. TKIs represent a class of targeted cancer therapies that have revolutionized treatment since their discovery. Therefore, the present review discusses which models have provided insight into TKI resistance. It should be noted that while some studies highlight the general benefits of each 3D model type, scaffold-free and scaffold-based, they often do not address TKI-specific dynamics. This could indeed be an intriguing area for future research, possibly warranting a dedicated study to explore these unique applications in depth.

Table II

Pros and cons of 3D models per cancer type.

Table II

Pros and cons of 3D models per cancer type.

3D modelsProsConsCancer types
SpheroidsImitate cellular interactions with adhesion molecules and soluble factors; Reproduce the tumor ECM (51).Difficulty in reproducing the polymorphic characteristics of the tumor (52).Breast, colon and different types of solid tumors (50-53).
OrganoidsOvercome 2D and in vivo models limitations (54); great versatility, as tumor modeling, drug screening, immunotherapy and precision medicine (55,58).Lack of vascularization (59,60).Bladder, ovarian, prostate, liver, breast, colorectal, lung cancer pancreatic cancer (55).
Organ-on-a-chipIncreasing the complexity of the system and the representativeness of the model (77); possibility of comparing tumor cells and healthy cells in the same model (90-92).Issue in identifying the biological target; difficulties to simplify its physiological functions by designing the microstructure (76).Pancreatic tumors (93,94), lung tumors (95-97), brain tumors (98,99), blood cancer (98,100-102), breast cancer (103).
Organ-on-a-platePossibility of studying multiple physiological processes and particularly metastatic models (105-108).Need to increase the reproducibility of the experiment (108).Breast cancer metastasis (108).

[i] The numbers in parentheses are the reference citations.

4. Cases of tyrosine kinase inhibitor resistance addressed using 3D models

TKIs specifically target enzymes known as tyrosine kinases, which play a crucial role in regulating cell growth, proliferation and survival pathways. The development of imatinib in the late 1990s marked a significant milestone in TKI discovery. Initially approved for the treatment of chronic myeloid leukemia (CML) (109), imatinib paved the way for subsequent TKIs targeting various tyrosine kinases implicated in various types of cancer (110). TKIs usually inhibit the enzymatic activity of tyrosine kinases by binding to their ATP-binding sites, thereby disrupting downstream signaling pathways involved in cancer progression. They are utilized in the treatment of various malignancies, including CML, gastrointestinal stromal tumors (GISTs) (111), non-small cell lung cancer (NSCLC) (112) and renal cell carcinoma (RCC) (113), among others. TKIs have demonstrated notable efficacy, significantly improving the outcomes and quality of life of patients.

Despite their therapeutic benefits, TKIs are not without limitations. One major challenge is the development of resistance (114,115), either through acquired mutations in the target kinase or the activation of alternative signaling pathways, leading to disease progression and treatment failure. Additionally, TKIs may exhibit off-target and adverse effects (114,116), impacting patient tolerability and treatment adherence. Addressing these limitations remains a key focus in oncology research, with ongoing efforts aimed at developing next-generation TKIs with improved efficacy, selectivity and safety profiles, as well as exploring combination therapies and personalized treatment approaches to overcome resistance and enhance clinical outcomes (117-120).

From a historical perspective, the development and introduction of imatinib and sorafenib were pivotal in the utilization of TKIs. These drugs marked significant advancements in targeted cancer therapies, laying the foundation for subsequent innovations in the field.

Imatinib, targeting BCR-ABL, KIT and PDGFR, has been FDA-approved for the treatment of CML and GISTs (121-124). Research has revealed that imatinib improves endothelial barrier stability and decreases invasive tumor cell activity (108). Ozer et al (108) explored their 3D model platform, metastasis-on-chip, for investigating cancer metastasis biology, drug discovery and drug reactions, providing potential for tailored anti-metastatic treatments for patients with triple-negative breast cancer. Imatinib notably decreased the intravasation of tumor cells, suggesting that its anti-metastatic effects may be linked to tumor-endothelial cell interactions. It enhanced the integrity and permeability of HUVEC monolayers when co-cultured with tumor cells/fibroblasts, mitigating endothelial barrier disruption and impeding tumor cell intravasation and extravasation (108).

Sorafenib is a multikinase inhibitor that targets several kinases, including Raf kinase (125-129), vascular endothelial growth factor receptor (VEGFR) (126,127) and platelet-derived growth factor receptor (PDGFR) (50,130,131). It is primarily utilized in the treatment of liver (129,132-135), kidney (126,136-140) and thyroid cancers (130,141-146). In this context, Ek et al (147) investigated the effects of sorafenib on a 3D cancer model of hepatocellular and colorectal carcinoma, identifying oxidative phosphorylation (OXPHOS) as a target vulnerability. They assessed global transcriptional responses in monolayer cell cultures, multicellular tumor spheroids, and tumoroids generated from a colorectal carcinoma patient. Cells in 3D models were sensitive to OXPHOS inhibitors, but resistant to other TKIs and chemotherapeutic drugs. Sorafenib and nitazoxanide exhibited an additive effect in reducing viability, regrowth potential, and inhibiting mitochondrial membrane potential at clinically relevant concentrations (147). An additional aspect of sorafenib resistance was investigated by Bielecka et al (148) in RCC cells treated with axitinib and sorafenib. Since hypoxia is known to reduce the efficacy of chemotherapy in solid tumors, Bielecka et al (148) developed a 3D model with suspended culture to mimic the molecular pathways characteristic of hypoxic conditions and the cell-cell dynamics found in the tumor environment. This model confirmed that resistance is linked to low oxygen levels, which induce conformational changes in the expression of key proteins, fulfilling TKI resistance (148).

Likewise, over the past years, sunitinib has been widely employed in RCC (kidney cancer) (149-152), GISTs (153-158) and pancreatic neuroendocrine tumors (159-162). Polena et al (163) investigated the effects of sunitinib, combined with bevacizumab, on VE-cadherin, a protein crucial for endothelial integrity. Using an endothelial monolayer model and both homotypic and heterotypic 3D cell models that mimic tumor growth, they assessed the effects of sunitinib and the antibody bevacizumab on VE-cadherin modifications and patient outcomes (163). The results revealed that sunitinib directly targeted VE-cadherin, inhibiting its phosphorylation and cleavage, and reducing endothelial cell migration in the 3D models. In patients with metastatic RCC, the baseline levels of soluble VE-cadherin (sVE) were higher than those in healthy donors. A decrease in sVE levels after 4 weeks of treatment was observed in responders to sunitinib, but not to bevacizumab (163). These findings obtained owing to the faithful 3D model employed suggest that sVE levels may be a valuable biomarker for monitoring the efficacy of targeted therapies, such as sunitinib in patients with cancer (163).

Sunitinib was also investigated in the study by Rausch et al (164), who conducted a study on clear cell RCC, revealing that resistance to sunitinib involves complex molecular changes, including dysregulated cell cycle progression and lysosomal sequestration of the drug. Using 3D heterotypic co-cultures, the researchers could mimic resistance mechanisms and test an optimized multidrug combination. This approach reduced metabolic activity in resistant cancer cells by >80%, highlighting the importance of 3D models in studying and overcoming TKI resistance, providing a more accurate platform for evaluating drug responses and developing effective treatments (164).

As regards more recent TKIs, the gefitinib inhibitor of the epidermal growth factor receptor (EGFR) tyrosine kinase was intensely studied in a 3D model. It is primarily used in the treatment of NSCLC with specific EGFR mutations. In the 3D lung carcinoma model, Stratmann et al (165) were able to prove the efficacy of TKIs, particularly gefitinib, in targeting EGFR. Employing 3D models on decellularized tissue matrices, the researchers created a complex TME that more accurately reflected clinical conditions. This approach revealed notable differences in drug responses when compared to conventional 2D cultures. In 3D models, gefitinib effectively inhibited EGFR activation in HCC827 cells, which harbor an activating EGFR mutation, leading to decreased proliferation and increased apoptosis (165). The 3D models enabled the quantitative measurements of proliferation, apoptosis and invasion, exhibiting strong associations with both in silico EGFR signaling models and clinical data. This thorough analysis established a reliable platform for assessing drug efficacy and tumor behavior. Additionally, applying TGFβ1 in the 3D model triggered EMT and tumor cell invasion, as evidenced by changes in cell morphology and EMT marker expression, effects not observed in 2D cultures, highlighting the enhanced predictive accuracy of 3D models (165).

Moreover, the efficacy and resistance of a number of gefitinib combinations were previously explored in 3D models. Liu et al (166) investigated the resistance mechanisms of lung cancer cells to EGFR-TKIs. The loss of wild-type EGFR reduced cell proliferation, migration and 3D-spheroid formation, while the loss of mutant EGFR or TKI resistance enhanced these processes. Wild-type EGFR disruption suppressed HER2/HER3 expression, but mutant EGFR loss or TKI resistance increased HER2/HER3 expression, promoting tumor cell survival and drug resistance via cyclin D1. Inhibiting cyclin D1/CDK4/6 re-sensitized erlotinib-resistant cells to erlotinib. Cyclin D1 expression in patients with NSCLC was associated with a worse survival (166). A 3D-spheroid tumor model was used to mimic the tumor microenvironment. In EGFR wild-type cell lines (A549 and H1299), EGFR ablation significantly inhibited cell proliferation, migration and tumor formation. Conversely, EGFR19del and erlotinib-resistant cells (H1650 and HCC827) exhibited increased proliferation, migration and 3D-spheroid formation upon EGFR19del ablation or TKI resistance (166). Another example of investigating the TKIs, gefitinib and erlotinib, is represented in the study conducted by Jacobi et al (167) that used 3D spheroids and organoids created with the hanging drop method embedded in Matrigel/collagen type I matrices. That study demonstrated the significant advantages of using 3D lung cancer cell cultures, specifically spheroid models, over traditional 2D cultures in proving the efficacy of TKIs targeting EGFR signaling. The 3D spheroid models revealed drug sensitivities that more closely match clinical outcomes. Notably, the addiction to the EGFR oncoprotein, crucial for the effectiveness of EGFR inhibitors, was observed only in 3D spheroid cultures. Furthermore, HCC827 cells with EGFR mutations exhibited significant sensitivity to EGFR inhibitors in 3D spheroids, but not in 2D cultures (167). Using the same cancer model, Zheng et al (168) investigated the novel STAT3 inhibitor, W2014-S, for its potential to enhance the efficacy of gefitinib and erlotinib in overcoming drug resistance in NSCLC. The study by Zheng et al (168), which included in vivo experiments using human NSCLC cell xenografts and PDX in mouse models, demonstrated that W2014-S enhanced the antitumor effects of gefitinib. By disrupting STAT3 signaling, a pathway often activated in NSCLC and associated with drug resistance, W2014-S effectively inhibited cancer cell proliferation, survival, migration, and invasion in 3D cultures. Additionally, it significantly sensitized TKI-resistant NSCLC cells to gefitinib and erlotinib, highlighting the importance of 3D cell cultures in evaluating the efficacy of new treatments (168).

In another cancer model, gefitinib and erlotinib were investigated by Hoque et al (169), who demonstrated that these TKIs more effectively inhibited cell viability, clonogenic growth and wound healing in human squamous epithelial A431-A6 cells compared to the controls in spheroids. Moreover, the study showed that increased AnxA6 levels enhance TKI efficacy by suppressing A431 cell motility and invasiveness. Likewise, in renal cancer cells, Fu et al (170) used 3D cultures to demonstrate the efficacy of TKIs, specifically gefitinib and erlotinib, targeting EGFR signaling. By employing spheroids and organoids, they found that these models reveal drug sensitivities that more closely align with clinical outcomes compared to traditional 2D cultures. The addiction to the EGFR oncoprotein, crucial for the effectiveness of EGFR inhibitors, was evident only in 3D cultures (170). Furthermore, 3D models were more predictive of clinical responses, as observed with HCC827 cells exhibiting significant sensitivity to EGFR inhibitors in 3D, but not in 2D cultures (170). The study Fu et al (170) underscores the enhanced predictability and clinical relevance of 3D models, advocating for their use in drug development and personalized cancer treatments to improve therapeutic outcomes (170).

As regards other combinations of gefitinib, Liu et al (171) explored the combination of gefitinib with rapamycin to address gefitinib resistance in NSCLC cells. The results of their study revealed that this combination significantly improved therapeutic outcomes, as rapamycin activated autophagy, synergizing with gefitinib to reduce cell viability and tumor formation both in vitro and in vivo. Additionally, an anti-EGFR aptamer-functionalized nanoparticle system was developed for targeted delivery, enhancing the cytotoxic effects of the drug combination (171).

Further combinations of other EGFR-TKIs were studied in 3D cell cultures of NSCLC. Kim et al (172) investigated the role of the receptor tyrosine kinase, AXL, in mediating resistance to EGFR-TKIs, including the third-generation TKI, osimertinib. Using both in vitro and in vivo models, including 3D cell cultures, xenograft tumors and PDX, their study found that AXL overexpression contributes to resistance by extending the protein degradation rate. Targeting AXL degradation with yuanhuadine (YD), an AXL degrader, restored sensitivity to EGFR-TKIs. The combination of YD and EGFR-TKIs effectively delayed or overcame resistance in EGFR-mutant NSCLC cells (172).

A more comprehensive approach to EFGR inhibition was offered by the study conducted by Hou et al (173), which used three-dimensional convolutional neural networks (CNN) with deep transfer learning to predict the treatment response in patients with stage IV lung adenocarcinoma with EGFR mutations. The 3D CNN model was able to stratify patients into subgroups with different progression risks and predict progression-free survival more accurately than models based solely on clinical features. This demonstrates the importance of 3D models in proving the efficacy of TKIs, providing more precise and reliable tools for clinical decision-making (173).

As regards fibroblast growth factor receptor (FGFR) inhibition, a number of TKIs inhibiting FGFR1 have emerged as a promising strategy for cancer therapy. A novel in vitro 3D culture system was developed by Ko et al (174) to study FGFR signaling in prostate cancer stem cells within cell lines (PC3, DU145 and LNaP), and the induced pluripotent iPS87 cell line. This 3D model, which exhibited increased stemness markers, demonstrated that the desired inhibition, with BGJ398 or dovitinib, effectively reduced cell survival and proliferation. The use of the spheroids was crucial in demonstrating the efficacy of FGFR1-targeting TKIs, providing a more realistic representation of tumor behavior and drug response compared to 2D monolayer cells (174).

Finally, although resistance against TKIs is usually studied in cancer cell lines, they are not able to widely recapitulate patient-cancer model complexities. In the realm of lung cancer research, Kim et al (175) emphasized the critical importance of patient-derived organoids (PDOs) in demonstrating the effectiveness of TKIs for advanced-stage lung adenocarcinoma. PDOs were established from patients with advanced-stage lung adenocarcinoma and analyzed for genetic alterations and responses to targeted therapies. These organoids retained somatic alterations, including driver mutations, from the original patient tumors, and their responses to TKIs mirrored the clinical outcomes of the corresponding patients. PDOs with EGFR exon 19 deletions and BRAF G464A mutations responded to dabrafenib/trametinib, and those with EGFR L747P mutations were sensitive to afatinib, consistent with patient responses (175). They were used to identify effective therapies for novel molecular targets, demonstrating the efficacy of poziotinib against ERBB2 exon 20 insertions and pralsetinib against RET fusions. The use of 3D models such has PDOs directly from patients thus provides a more accurate and clinically relevant platform for evaluating the efficacy of TKIs (175).

Currently, despite all the advantages previously listed, it is important to also acknowledge the limitations of 3D models that hinder clinical translation. A number of these models require specialized equipment and are costly, limiting accessibility. They also exhibit high variability and low standardization, which affect reproducibility and complicate large-scale studies. Furthermore, advanced analytical techniques are often required, rendering 3D models less suited for high-throughput screening.

Nevertheless, the crucial role of clinical validation in establishing the relevance of findings from 3D models is commonly acknowledged, particularly for studying resistance to TKIs. While the present review synthesizes current preclinical data, additional clinical research is essential to evaluate how insight from 3D models can translate into real-world settings. Further studies using patient-derived samples or clinical trials could bridge this gap and underscore the translational potential of 3D models in oncology. Although robust clinical validations are limited, early advancements demonstrate the potential of 3D models to refine TKI efficacy predictions and support personalized cancer therapies. Few established clinical validations directly address TKI resistance findings from 3D models; however, recent studies have shown promise.

For example, recent research has combined computational and mechanistic modeling with in vitro 3D cultures to predict patient-specific responses. This approach has shown potential in lung adenocarcinoma treatments with EGFR-targeting TKIs (176), such as osimertinib, where data from clinical trials (e.g., NEJ002-NCT00322452 and FLAURA-NCT02296125) has been used to align model predictions with actual patient outcomes (177).

These advancements provide a foundation for future research to further validate and strengthen 3D model findings within clinical settings. As development progresses, 3D models can also assist in refining dosage strategies and minimizing off-target effects by simulating drug behavior in a controlled microenvironment, thus bridging the gap between preclinical insights and clinical outcomes.

5. Application of 3D models in other cancer drug classes

Having explored the valuable role of 3D models in evaluating the efficacy of TKIs and understanding their limitations in complex environments, the present review now provides a brief overview of how 3D models have brought innovation to other cancer drug classes. These include chemotherapeutic agents, hormone therapies and immunotherapies.

Hirschhaeuser et al (178), in a comprehensive review, discussed the benefits of using 3D spheroid models to simulate in vivo-like conditions for chemotherapy studies. These models allow for a better understanding of drug penetration, efficacy and resistance, as they replicate the architecture and gradients present in tumors. Wenzel et al (179) introduced a 3D platform to test a variety of compounds targeting dormant tumor cells, validating a screening system for substances that would not be adequately evaluated using traditional 2D models. Additionally, Imamura et al (180) evaluated the response of breast cancer cell lines cultured in 3D to assess drug sensitivity to paclitaxel, oxygen status, and significant markers of proliferation (181), such as Ki-67 and cell death, such as caspase. They concluded that the 3D model more accurately simulated in vivo tumor characteristics, revealing drug resistance mechanisms linked to hypoxia, dormancy and anti-apoptotic features (180).

Hormone therapies, such as tamoxifen, are widely used in the treatment of hormone receptor-positive breast cancer. Leeper et al (182) determined the sensitivity of breast cancer tissues to tamoxifen using a collagen-based 3D culture. The model shown by Leeper also demonstrated the histopathological changes caused by the drug, offering a more precise evaluation than traditional methods (182). Similarly, enzalutamide, a key drug in prostate cancer therapy, modulates androgen receptor activity as shown by Andolfi et al (183). The study demonstrate that using a 3D model allowed for clear evidence of reduced cell proliferation when MED12 and CDK8/19 were inhibited by enzalutamide, offering valuable insights into prostate cancer treatment (183).

Jenkins et al (184) highlighted how 3D models have been particularly useful in studying immune checkpoint inhibitors such as PD-1/PD-L1 and CTLA-4. These models enable researchers to recreate immune cell-tumor interactions in a more realistic tumor microenvironment, helping to elucidate mechanisms of immune resistance and response (184).

3D models have revolutionized cancer research by providing more physiologically relevant systems for evaluating drug efficacy, toxicity, and resistance mechanisms. While the present review has focused on the use of 3D models in studying TKIs, their application extends to other major drug classes, including chemotherapeutics, hormone therapies, immunotherapies and anti-angiogenic agents. These models enable researchers to capture complex tumor microenvironment dynamics that are often missed in 2D cultures, ultimately improving the translation of preclinical findings to clinical outcomes. Expanding the use of 3D models across different drug classes can lead to more effective cancer therapies and a better understanding of treatment responses.

6. Conclusion and future perspectives

The adoption of 3D models has revolutionized the strategies used to test chemotherapeutic agents, providing a more realistic representation of the TME and its complexities. These models have enhanced predictive power and accuracy in assessing drug efficacy and toxicity, streamlining the drug development process and reducing reliance on traditional 2D cultures. Researchers are now equipped with more robust platforms to inform clinical decision-making and treatment strategies.

Looking ahead, the continued refinement of 3D models holds immense potential for overcoming the limitations associated with TKIs. These models provide valuable tools for investigating interactions between cancer cells, stromal components, and immune cells within the TME. By unraveling the complex mechanisms behind TKI resistance, novel therapeutic strategies can be developed to enhance drug efficacy and overcome resistance barriers.

One exciting future direction is the integration of 3D models with multi-omics approaches, including genomics, transcriptomics, proteomics and metabolomics (185). By coupling these models with artificial intelligence (AI) and machine learning algorithms, researchers could gain deeper insight into tumor biology and drug responses (186). This integration would allow for the real-time monitoring of molecular changes in response to treatments, providing predictive data that could help preempt resistance and suggest alternative therapeutic pathways. These advancements have the potential to reshape the drug development pipeline by significantly improving the early detection of drug efficacy or failure, reducing the time and cost associated with clinical trials.

As 3D models continue to evolve, their integration with cutting-edge technologies, such as multi-omics, AI, immune-oncology models and microfluidics will push the boundaries of cancer research and therapy. These advancements will contribute to more personalized and precise treatment regimens, reshaping the landscape of oncology. Collaborative efforts across disciplines, driven by innovative technological developments, will not only enhance the understanding of cancer biology, but may also improve the translation of preclinical findings into clinical success. Ultimately, this progress promises to deliver more effective cancer therapies, ushering in a new era of precision oncology for patients worldwide.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

AP was involved in the conceptualization of the present review and in the final revisions. RP was involved in the final editing of the manuscript and in revisions. SF contributed to the study by creating the figures, collecting data from the literature and editing the text. GI collected data from the literature, edited the text and finalized the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Cao H, Duan L, Zhang Y, Cao J and Zhang K: Current hydrogel advances in physicochemical and biological response-driven biomedical application diversity. Signal Transduct Target Ther. 6(426)2021.PubMed/NCBI View Article : Google Scholar

2 

Munir MU: Nanomedicine penetration to tumor: Challenges, and advanced strategies to tackle this issue. Cancers (Basel). 14(2904)2022.PubMed/NCBI View Article : Google Scholar

3 

Mansoori B, Mohammadi A, Davudian S, Shirjang S and Baradaran B: The different mechanisms of cancer drug resistance: A brief review. Adv Pharm Bull. 7:339–348. 2017.PubMed/NCBI View Article : Google Scholar

4 

Gkretsi V, Stylianou A, Papageorgis P, Polydorou C and Stylianopoulos T: Remodeling components of the tumor microenvironment to enhance cancer therapy. Front Oncol. 5(214)2015.PubMed/NCBI View Article : Google Scholar

5 

Tosca EM, Ronchi D, Facciolo D and Magni P: Replacement, reduction, and refinement of animal experiments in anticancer drug development: The contribution of 3D in vitro cancer models in the drug efficacy assessment. Biomedicines. 11(1058)2023.PubMed/NCBI View Article : Google Scholar

6 

Khalil AS, Jaenisch R and Mooney DJ: Engineered tissues and strategies to overcome challenges in drug development. Adv Drug Deliv Rev. 158:116–139. 2020.PubMed/NCBI View Article : Google Scholar

7 

Booij TH, Price LS and Danen EHJ: 3D cell-based assays for drug screens: Challenges in imaging, image analysis, and high-content analysis. SLAS Discov. 24:615–627. 2019.PubMed/NCBI View Article : Google Scholar

8 

Candini O, Grisendi G, Foppiani EM, Brogli M, Aramini B, Masciale V, Spano C, Petrachi T, Veronesi E, Conte P, et al: A novel 3D in vitro platform for pre-clinical investigations in drug testing, gene therapy, and immuno-oncology. Sci Rep. 9(7154)2019.PubMed/NCBI View Article : Google Scholar

9 

Unger C, Kramer N, Walzl A, Scherzer M, Hengstschläger M and Dolznig H: Modeling human carcinomas: Physiologically relevant 3D models to improve anti-cancer drug development. Adv Drug Deliv Rev. 79-80:50–67. 2014.PubMed/NCBI View Article : Google Scholar

10 

Kelm JM, Lal-Nag M, Sittampalam GS and Ferrer M: Translational in vitro research: Integrating 3D drug discovery and development processes into the drug development pipeline. Drug Discov Today. 24:26–30. 2019.PubMed/NCBI View Article : Google Scholar

11 

Krisnawan VE, Stanley JA, Schwarz JK and DeNardo DG: Tumor microenvironment as a regulator of radiation therapy: New insights into stromal-mediated radioresistance. Cancers (Basel). 12(2916)2020.PubMed/NCBI View Article : Google Scholar

12 

Bonnier F, Keating ME, Wróbel TP, Majzner K, Baranska M, Garcia-Munoz A and Byrne HJ: Cell viability assessment using the Alamar blue assay: A comparison of 2D and 3D cell culture models. Toxicol In Vitro. 29:124–131. 2015.PubMed/NCBI View Article : Google Scholar

13 

Duval K, Grover H, Han LH, Mou Y, Pegoraro AF, Fredberg J and Chen Z: Modeling physiological events in 2D vs. 3D cell culture. Physiology (Bethesda). 32:266–277. 2017.PubMed/NCBI View Article : Google Scholar

14 

Yi HG, Jeong YH, Kim Y, Choi YJ, Moon HE, Park SH, Kang KS, Bae M, Jang J, Youn H, et al: A bioprinted human-glioblastoma-on-a-chip for the identification of patient-specific responses to chemoradiotherapy. Nat Biomed Eng. 3:509–519. 2019.PubMed/NCBI View Article : Google Scholar

15 

Gomez-Roman N, Chong MY, Chahal SK, Caragher SP, Jackson MR, Stevenson KH, Dongre SA and Chalmers AJ: Radiation responses of 2D and 3D glioblastoma cells: A novel, 3D-specific radioprotective role of VEGF/Akt signaling through functional activation of NHEJ. Mol Cancer Ther. 19:575–589. 2020.PubMed/NCBI View Article : Google Scholar

16 

Kalli M and Stylianopoulos T: Toward innovative approaches for exploring the mechanically regulated tumor-immune microenvironment. APL Bioeng. 8(011501)2024.PubMed/NCBI View Article : Google Scholar

17 

Zhou Z, Vessella T, Wang P, Cui F, Wen Q and Zhou HS: Mechanical cues in tumor microenvironment on chip. Biosens Bioelectron X. 14(100376)2023.

18 

Langhans SA: Three-dimensional in vitro cell culture models in drug discovery and drug repositioning. Front Pharmacol. 9(6)2018.PubMed/NCBI View Article : Google Scholar

19 

Rodrigues J, Heinrich MA, Teixeira LM and Prakash J: 3D in vitro model (R)evolution: Unveiling tumor-stroma interactions. Trends Cancer. 7:249–264. 2021.PubMed/NCBI View Article : Google Scholar

20 

Jensen C and Teng Y: Is it time to start transitioning from 2D to 3D cell culture? Front Mol Biosci. 7(33)2020.PubMed/NCBI View Article : Google Scholar

21 

Gill BJ and West JL: Modeling the tumor extracellular matrix: Tissue engineering tools repurposed towards new frontiers in cancer biology. J Biomech. 47:1969–1978. 2014.PubMed/NCBI View Article : Google Scholar

22 

Wishart G, Gupta P, Schettino G, Nisbet A and Velliou E: 3d tissue models as tools for radiotherapy screening for pancreatic cancer. Br J Radiol. 94(20201397)2021.PubMed/NCBI View Article : Google Scholar

23 

Mukubou H, Tsujimura T, Sasaki R and Ku Y: The role of autophagy in the treatment of pancreatic cancer with gemcitabine and ionizing radiation. Int J Oncol. 37:821–828. 2010.PubMed/NCBI View Article : Google Scholar

24 

Baselet B, Sonveaux P, Baatout S and Aerts A: Pathological effects of ionizing radiation: Endothelial activation and dysfunction. Cell Mol Life Sci. 76:699–728. 2019.PubMed/NCBI View Article : Google Scholar

25 

Baker BM and Chen CS: Deconstructing the third dimension: How 3D culture microenvironments alter cellular cues. J Cell Sci. 125(Pt 13):3015–3024. 2012.PubMed/NCBI View Article : Google Scholar

26 

Kapałczyńska M, Kolenda T, Przybyła W, Zajączkowska M, Teresiak A, Filas V, Ibbs M, Bliźniak R, Łuczewski Ł and Lamperska K: 2D and 3D cell cultures - a comparison of different types of cancer cell cultures. Arch Med Sci. 14:910–919. 2018.PubMed/NCBI View Article : Google Scholar

27 

Riedl A, Schlederer M, Pudelko K, Stadler M, Walter S, Unterleuthner D, Unger C, Kramer N, Hengstschläger M, Kenner L, et al: Comparison of cancer cells in 2D vs 3D culture reveals differences in AKT-mTOR-S6K signaling and drug responses. J Cell Sci. 130:203–218. 2017.PubMed/NCBI View Article : Google Scholar

28 

Cekanova M and Rathore K: Animal models and therapeutic molecular targets of cancer: Utility and limitations. Drug Des Devel Ther. 8:1911–1921. 2014.PubMed/NCBI View Article : Google Scholar

29 

Horvath P, Aulner N, Bickle M, Davies AM, Nery ED, Ebner D, Montoya MC, Östling P, Pietiäinen V, Price LS, et al: Screening out irrelevant cell-based models of disease. Nat Rev Drug Discov. 15:751–769. 2016.PubMed/NCBI View Article : Google Scholar

30 

Amoedo ND, Obre E and Rossignol R: Drug discovery strategies in the field of tumor energy metabolism: Limitations by metabolic flexibility and metabolic resistance to chemotherapy. Biochim Biophys Acta Bioenerg. 1858:674–685. 2017.PubMed/NCBI View Article : Google Scholar

31 

Kerslake R, Belay B, Panfilov S, Hall M, Kyrou I, Randeva HS, Hyttinen J, Karteris E and Sisu C: Transcriptional landscape of 3D vs. 2D ovarian cancer cell models. Cancers (Basel). 15(3350)2023.PubMed/NCBI View Article : Google Scholar

32 

Zhang B, Korolj A, Lai BFL and Radisic M: Advances in organ-on-a-chip engineering. Nat Rev Mater. 3:257–278. 2018.

33 

Luca AC, Mersch S, Deenen R, Schmidt S, Messner I, Schäfer KL, Baldus SE, Huckenbeck W, Piekorz RP, Knoefel WT, et al: Impact of the 3D microenvironment on phenotype, gene expression, and EGFR inhibition of colorectal cancer cell lines. PLoS One. 8(e59689)2013.PubMed/NCBI View Article : Google Scholar

34 

van Duinen V, Trietsch SJ, Joore J, Vulto P and Hankemeier T: Microfluidic 3D cell culture: From tools to tissue models. Curr Opin Biotechnol. 35:118–126. 2015.PubMed/NCBI View Article : Google Scholar

35 

Antonelli F: 3D cell models in radiobiology: Improving the predictive value of in vitro research. Int J Mol Sci. 24(10620)2023.PubMed/NCBI View Article : Google Scholar

36 

Boucherit N, Gorvel L and Olive D: 3D tumor models and their use for the testing of immunotherapies. Front Immunol. 11(603640)2020.PubMed/NCBI View Article : Google Scholar

37 

Barozzi D and Scielzo C: Emerging strategies in 3D culture models for hematological cancers. Hemasphere. 7(e932)2023.PubMed/NCBI View Article : Google Scholar

38 

Bédard P, Gauvin S, Ferland K, Caneparo C, Pellerin È, Chabaud S and Bolduc S: Innovative human three-dimensional tissue-engineered models as an alternative to animal testing. Bioengineering (Basel). 7(115)2020.PubMed/NCBI View Article : Google Scholar

39 

Li W, Zhou Z, Zhou X, Khoo BL, Gunawan R, Chin YR, Zhang L, Yi C, Guan X and Yang M: 3D biomimetic models to reconstitute tumor microenvironment in vitro: Spheroids, organoids, and Tumor-on-a-chip. Adv Healthc Mater. 12(e2202609)2023.PubMed/NCBI View Article : Google Scholar

40 

Barbosa MAG, Xavier CPR, Pereira RF, Petrikaitė V and Vasconcelos MH: 3D cell culture models as recapitulators of the tumor microenvironment for the screening of anti-cancer drugs. Cancers (Basel). 14(190)2021.PubMed/NCBI View Article : Google Scholar

41 

Amaral RLF, Miranda M, Marcato PD and Swiech K: Comparative analysis of 3D bladder tumor spheroids obtained by forced floating and hanging drop methods for drug screening. Front Physiol. 8(605)2017.PubMed/NCBI View Article : Google Scholar

42 

Strobel HA, Calamari EL, Alphonse B, Hookway TA and Rolle MW: Fabrication of custom agarose wells for cell seeding and tissue ring self-assembly using 3D-Printed Molds. J Vis Exp. (134)(56618)2018.PubMed/NCBI View Article : Google Scholar

43 

Rauh J, Milan F, Günther KP and Stiehler M: Bioreactor systems for bone tissue engineering. Tissue Eng Part B Rev. 17:263–280. 2011.PubMed/NCBI View Article : Google Scholar

44 

Antoni D, Burckel H, Josset E and Noel G: Three-dimensional cell culture: A breakthrough in vivo. Int J Mol Sci. 16:5517–5527. 2015.PubMed/NCBI View Article : Google Scholar

45 

Zhang S, Liu P, Chen L, Wang Y, Wang Z and Zhang B: The effects of spheroid formation of adipose-derived stem cells in a microgravity bioreactor on stemness properties and therapeutic potential. Biomaterials. 41:15–25. 2015.PubMed/NCBI View Article : Google Scholar

46 

Chaicharoenaudomrung N, Kunhorm P and Noisa P: Three-dimensional cell culture systems as an in vitro platform for cancer and stem cell modeling. World J Stem Cells. 11:1065–1083. 2019.PubMed/NCBI View Article : Google Scholar

47 

Marques IA, Fernandes C, Tavares NT, Pires AS, Abrantes AM and Botelho MF: Magnetic-based human tissue 3D cell culture: A systematic review. Int J Mol Sci. 23(12681)2022.PubMed/NCBI View Article : Google Scholar

48 

Patra B, Peng CC, Liao WH, Lee CH and Tung YC: Drug testing and flow cytometry analysis on a large number of uniform sized tumor spheroids using a microfluidic device. Sci Rep. 6(21061)2016.PubMed/NCBI View Article : Google Scholar

49 

Zanoni M, Piccinini F, Arienti C, Zamagni A, Santi S, Polico R, Bevilacqua A and Tesei A: 3D tumor spheroid models for in vitro therapeutic screening: A systematic approach to enhance the biological relevance of data obtained. Sci Rep. 6(19103)2016.PubMed/NCBI View Article : Google Scholar

50 

Pinto B, Henriques AC, Silva PMA and Bousbaa H: Three-dimensional spheroids as in vitro preclinical models for cancer research. Pharmaceutics. 12(1186)2020.PubMed/NCBI View Article : Google Scholar

51 

Rodrigues DB, Reis RL and Pirraco RP: Modelling the complex nature of the tumor microenvironment: 3D tumor spheroids as an evolving tool. J Biomed Sci. 31(13)2024.PubMed/NCBI View Article : Google Scholar

52 

Vakhshiteh F, Bagheri Z, Soleimani M, Ahvaraki A, Pournemat P, Alavi SE and Madjd Z: Heterotypic tumor spheroids: A platform for nanomedicine evaluation. J Nanobiotechnology. 21(249)2023.PubMed/NCBI View Article : Google Scholar

53 

Jiang X, Oyang L, Peng Q, Liu Q, Xu X, Wu N, Tan S, Yang W, Han Y, Lin J, et al: Organoids: opportunities and challenges of cancer therapy. Front Cell Dev Biol. 11(1232528)2023.PubMed/NCBI View Article : Google Scholar

54 

Hou X, Du C, Lu L, Yuan S, Zhan M, You P and Du H: Opportunities and challenges of patient-derived models in cancer research: Patient-derived xenografts, patient-derived organoid and patient-derived cells. World J Surg Oncol. 20(37)2022.PubMed/NCBI View Article : Google Scholar

55 

Qu J, Kalyani FS, Liu L, Cheng T and Chen L: Tumor organoids: Synergistic applications, current challenges, and future prospects in cancer therapy. Cancer Commun (Lond). 41:1331–1353. 2021.PubMed/NCBI View Article : Google Scholar

56 

Idrisova KF, Simon HU and Gomzikova MO: Role of patient-derived models of cancer in translational oncology. Cancers (Basel). 15(139)2023.PubMed/NCBI View Article : Google Scholar

57 

Sun H, Cao S, Mashl RJ, Mo CK, Zaccaria S, Wendl MC, Davies SR, Bailey MH, Primeau TM, Hoog J, et al: Comprehensive characterization of 536 patient-derived xenograft models prioritizes candidates for targeted treatment. Nat Commun. 12(5086)2021.PubMed/NCBI View Article : Google Scholar

58 

Tharehalli U, Svinarenko M and Lechel A: Remodelling and improvements in organoid technology to study liver carcinogenesis in a dish. Stem Cells Int. 2019(3831213)2019.PubMed/NCBI View Article : Google Scholar

59 

Yip S, Wang N and Sugimura R: Give them vasculature and immune cells: How to fill the gap of organoids. Cells Tissues Organs. 212:369–382. 2023.PubMed/NCBI View Article : Google Scholar

60 

Zhang S, Wan Z and Kamm RD: Vascularized organoids on a chip: Strategies for engineering organoids with functional vasculature. Lab Chip. 21:473–488. 2021.PubMed/NCBI View Article : Google Scholar

61 

Bar-Ephraim YE, Kretzschmar K, Asra P, de Jongh E, Boonekamp KE, Drost J, van Gorp J, Pronk A, Smakman N, Gan I, et al: Modelling cancer immunomodulation using epithelial organoid cultures. bioRxiv, p377655, 2018.

62 

Kim J, Koo BK and Knoblich JA: Human organoids: Model systems for human biology and medicine. Nat Rev Mol Cell Biol. 21:571–584. 2020.PubMed/NCBI View Article : Google Scholar

63 

Ferreira LP, Gaspar VM and Mano JF: Design of spherically structured 3D in vitro tumor models-Advances and prospects. Acta Biomater. 75:11–34. 2018.PubMed/NCBI View Article : Google Scholar

64 

Unnikrishnan K, Thomas LV and Ram Kumar RM: Advancement of scaffold-based 3D cellular models in cancer tissue engineering: An update. Front Oncol. 11(733652)2021.PubMed/NCBI View Article : Google Scholar

65 

Sun L, Wang X, He Y, Chen B, Shan B, Yang J, Wang R, Zeng X, Li J, Tan H and Liang R: Polyurethane scaffold-based 3D lung cancer model recapitulates in vivo tumor biological behavior for nanoparticulate drug screening. Regen Biomater. 10(rbad091)2023.PubMed/NCBI View Article : Google Scholar

66 

Li X, Shan J, Chen X, Cui H, Wen G and Yu Y: Decellularized diseased tissues: Current state-of-the-art and future directions. MedComm (2020). 4(e399)2023.PubMed/NCBI View Article : Google Scholar

67 

Zhang CY, Fu CP, Li XY, Lu XC, Hu LG, Kankala RK, Wang SB and Chen AZ: Three-dimensional bioprinting of decellularized extracellular matrix-based bioinks for tissue engineering. Molecules. 27(3442)2022.PubMed/NCBI View Article : Google Scholar

68 

Habanjar O, Diab-Assaf M, Caldefie-Chezet F and Delort L: 3D cell culture systems: Tumor application, advantages, and disadvantages. Int J Mol Sci. 22(12200)2021.PubMed/NCBI View Article : Google Scholar

69 

Abuwatfa WH, Pitt WG and Husseini GA: Scaffold-based 3D cell culture models in cancer research. J Biomed Sci. 31(7)2024.PubMed/NCBI View Article : Google Scholar

70 

Shivalkar S and Singh S: Solid freeform techniques application in bone tissue engineering for scaffold fabrication. Tissue Eng Regen Med. 14:187–200. 2017.PubMed/NCBI View Article : Google Scholar

71 

Suri S, Han LH, Zhang W, Singh A, Chen S and Schmidt CE: Solid freeform fabrication of designer scaffolds of hyaluronic acid for nerve tissue engineering. Biomed Microdevices. 13:983–993. 2011.PubMed/NCBI View Article : Google Scholar

72 

Tony A, Badea I, Yang C, Liu Y, Wells G, Wang K, Yin R, Zhang H and Zhang W: The additive manufacturing approach to polydimethylsiloxane (PDMS) microfluidic devices: Review and future directions. Polymers (Basel). 15(1926)2023.PubMed/NCBI View Article : Google Scholar

73 

Anthon SG and Valente KP: Vascularization strategies in 3D cell culture models: From scaffold-free models to 3D bioprinting. Int J Mol Sci. 23(14582)2022.PubMed/NCBI View Article : Google Scholar

74 

Huh D, Matthews BD, Mammoto A, Montoya-Zavala M, Hsin HY and Ingber DE: Reconstituting organ-level lung functions on a chip. Science. 328:1662–1668. 2010.PubMed/NCBI View Article : Google Scholar

75 

Wang Y, Gao Y, Pan Y, Zhou D, Liu Y, Yin Y, Yang J, Wang Y and Song Y: Emerging trends in organ-on-a-chip systems for drug screening. Acta Pharm Sin B. 13:2483–2509. 2023.PubMed/NCBI View Article : Google Scholar

76 

Li Z, Hui J, Yang P and Mao H: Microfluidic organ-on-a-chip system for disease modeling and drug development. Biosensors (Basel). 12(370)2022.PubMed/NCBI View Article : Google Scholar

77 

Vargas R, Egurbide-Sifre A and Medina L: Organ-on-a-chip systems for new drugs development. ADMET DMPK. 9:111–141. 2021.PubMed/NCBI View Article : Google Scholar

78 

Tajeddin A and Mustafaoglu N: Design and fabrication of organ-on-chips: Promises and challenges. Micromachines (Basel). 12(1443)2021.PubMed/NCBI View Article : Google Scholar

79 

Si L, Bai H, Rodas M, Cao W, Oh CY, Jiang A, Moller R, Hoagland D, Oishi K, Horiuchi S, et al: A humanairway-on-a-chip for the rapid identification of candidate antiviral therapeutics and prophylactics. Nat Biomed Eng. 5:815–829. 2021.PubMed/NCBI View Article : Google Scholar

80 

Bai H, Si L, Jiang A, Belgur C, Zhai Y, Plebani R, Oh CY, Rodas M, Patil A, Nurani A, et al: Mechanical control of innate immune responses against viral infection revealed in a human lung alveolus chip. Nat Commun. 13(1928)2022.PubMed/NCBI View Article : Google Scholar

81 

Plebani R, Bai H, Si L, Li J, Zhang C and Romano M: 3D lung tissue models for studies on SARS-CoV-2 pathophysiology and therapeutics. Int J Mol Sci. 23(10071)2022.PubMed/NCBI View Article : Google Scholar

82 

Plebani R, Potla R, Soong M, Bai H, Izadifar Z, Jiang A, Travis RN, Belgur C, Dinis A, Cartwright MJ, et al: Modeling pulmonary cystic fibrosis in a human lung airway-on-a-chip. J Cyst Fibros. 21:606–615. 2022.PubMed/NCBI View Article : Google Scholar

83 

Bein A, Fadel CW, Swenor B, Cao W, Powers RK, Camacho DM, Naziripour A, Parsons A, LoGrande N, Sharma S, et al: Nutritional deficiency in an intestine-on-a-chip recapitulates injury hallmarks associated with environmental enteric dysfunction. Nat Biomed Eng. 6:1236–1247. 2022.PubMed/NCBI View Article : Google Scholar

84 

Ewart L, Apostolou A, Briggs SA, Carman CV, Chaff JT, Heng AR, Jadalannagari S, Janardhanan J, Jang KJ, Joshipura SR, et al: Performance assessment and economic analysis of a human Liver-Chip for predictive toxicology. Commun Med (Lond). 2(154)2022.PubMed/NCBI View Article : Google Scholar

85 

Benam KH, Novak R, Nawroth J, Hirano-Kobayashi M, Ferrante TC, Choe Y, Prantil-Baun R, Weaver JC, Bahinski A, Parker KK and Ingber DE: Matched-comparative modeling of normal and diseased human airway responses using a microengineered breathing lung chip. Cell Syst. 3:456–466.e4. 2016.PubMed/NCBI View Article : Google Scholar

86 

Zommiti M, Connil N, Tahrioui A, Groboillot A, Barbey C, Konto-Ghiorghi Y, Lesouhaitier O, Chevalier S and Feuilloley MGJ: Organs-on-chips platforms are everywhere: A zoom on biomedical investigation. Bioengineering (Basel). 9(646)2022.PubMed/NCBI View Article : Google Scholar

87 

Monteduro AG, Rizzato S, Caragnano G, Trapani A, Giannelli G and Maruccio G: Organs-on-chips technologies-A guide from disease models to opportunities for drug development. Biosens Bioelectron. 231(115271)2023.PubMed/NCBI View Article : Google Scholar

88 

Zhao Y, Kankala RK, Wang SB and Chen AZ: Multi-organs-on-chips: Towards long-term biomedical investigations. Molecules. 24(675)2019.PubMed/NCBI View Article : Google Scholar

89 

R N, Aggarwal A, Sravani AB, Mallya P and Lewis S: Organ-on-a-chip: An emerging research platform. Organogenesis. 19(2278236)2023.PubMed/NCBI View Article : Google Scholar

90 

Mauriac H, Pannetier C and Casquillas GV: Organs on Chip Review. Elveflow. Available from: https://www.elveflow.com/microfluidic-reviews/organs-on-chip-3d-cell-culture/organs-chip-review/.

91 

Sun W, Luo Z, Lee J, Kim HJ, Lee K, Tebon P, Feng Y, Dokmeci MR, Sengupta S and Khademhosseini A: Organ-on-a-chip for cancer and immune organs modeling. Adv Healthc Mater. 8(1801363)2019.PubMed/NCBI View Article : Google Scholar

92 

van den Berg A, Mummery CL, Passier R and van der Meer AD: Personalised organs-on-chips: Functional testing for precision medicine. Lab Chip. 19:198–205. 2019.PubMed/NCBI View Article : Google Scholar

93 

Wu J, Dong M, Rigatto C, Liu Y and Lin F: Lab-on-chip technology for chronic disease diagnosis. NPJ Digit Med. 1(7)2018.PubMed/NCBI View Article : Google Scholar

94 

Glieberman AL, Pope BD, Zimmerman JF, Liu Q, Ferrier JP, Kenty JHR, Schrell AM, Mukhitov N, Shores KL, Tepole AB, et al: Synchronized stimulation and continuous insulin sensing in a microfluidic human Islet on a chip designed for scalable manufacturing. Lab Chip. 19:2993–3010. 2019.PubMed/NCBI View Article : Google Scholar

95 

Wang D, Cong Y, Deng Q, Han X, Zhang S, Zhao L, Luo Y and Zhang X: Physiological and disease models of respiratory system based on organ-on-a-chip technology. Micromachines (Basel). 12(1106)2021.PubMed/NCBI View Article : Google Scholar

96 

Kimura H, Sakai Y and Fujii T: Organ/body-on-a-chip based on microfluidic technology for drug discovery. Drug Metab Pharmacokinet. 33:43–48. 2018.PubMed/NCBI View Article : Google Scholar

97 

Lu RXZ and Radisic M: Organ-on-a-chip platforms for evaluation of environmental nanoparticle toxicity. Bioact Mater. 6:2801–2819. 2021.PubMed/NCBI View Article : Google Scholar

98 

Wikswo JP, Curtis EL, Eagleton ZE, Evans BC, Kole A, Hofmeister LH and Matloff WJ: Scaling and systems biology for integrating multiple organs-on-a-chip. Lab Chip. 13:3496–511. 2013.PubMed/NCBI View Article : Google Scholar

99 

Bang S, Jeong S, Choi N and Kim HN: Brain-on-a-chip: A history of development and future perspective. Biomicrofluidics. 13(051301)2019.PubMed/NCBI View Article : Google Scholar

100 

Mao M, Bei HP, Lam CH, Chen P, Wang S, Chen Y, He J and Zhao X: Human-on-leaf-chip: A biomimetic vascular system integrated with chamber-specific organs. Small. 16(e2000546)2020.PubMed/NCBI View Article : Google Scholar

101 

Ramadan Q and Gijs MA: In vitro micro-physiological models for translational immunology. Lab Chip. 15:614–636. 2015.PubMed/NCBI View Article : Google Scholar

102 

Mandrycky CJ, Howard CC, Rayner SG, Shin YJ and Zheng Y: Organ-on-a-chip systems for vascular biology. J Mol Cell Cardiol. 159:1–13. 2021.PubMed/NCBI View Article : Google Scholar

103 

Chen Y, Gao D, Wang Y, Lin S and Jiang Y: A novel 3D breast-cancer-on-chip platform for therapeutic evaluation of drug delivery systems. Anal Chim Acta. 1036:97–106. 2018.PubMed/NCBI View Article : Google Scholar

104 

Danku AE, Dulf EH, Braicu C, Jurj A and Berindan-Neagoe I: Organ-on-a-chip: A survey of technical results and problems. Front Bioeng Biotechnol. 10(840674)2022.PubMed/NCBI View Article : Google Scholar

105 

de Haan L, Suijker J, van Roey R, Berges N, Petrova E, Queiroz K, Strijker W, Olivier T, Poeschke O, Garg S and van den Broek LJ: A microfluidic 3D endothelium-on-a-chip model to study transendothelial migration of T cells in health and disease. Int J Mol Sci. 22(8234)2021.PubMed/NCBI View Article : Google Scholar

106 

van Duinen V, van den Heuvel A, Trietsch SJ, Lanz HL, van Gils JM, van Zonneveld AJ, Vulto P and Hankemeier T: 96 perfusable blood vessels to study vascular permeability in vitro. Sci Rep. 7(18071)2017.PubMed/NCBI View Article : Google Scholar

107 

Trietsch SJ, Naumovska E, Kurek D, Setyawati MC, Vormann MK, Wilschut KJ, Lanz HL, Nicolas A, Ng CP, Joore J, et al: Membrane-free culture and real-time barrier integrity assessment of perfused intestinal epithelium tubes. Nat Commun. 8(262)2017.PubMed/NCBI View Article : Google Scholar

108 

Ozer LY, Fayed HS, Ericsson J and Al Haj Zen A: Development of a cancer metastasis-on-chip assay for high throughput drug screening. Front Oncol. 13(1269376)2024.PubMed/NCBI View Article : Google Scholar

109 

Cohen MH, Williams G, Johnson JR, Duan J, Gobburu J, Rahman A, Benson K, Leighton J, Kim SK, Wood R, et al: Approval summary for imatinib mesylate capsules in the treatment of chronic myelogenous leukemia. Clin Cancer Res. 8:935–942. 2002.PubMed/NCBI

110 

Cohen P, Cross D and Jänne PA: Kinase drug discovery 20 years after imatinib: Progress and future directions. Nat Rev Drug Discov. 20:551–569. 2021.PubMed/NCBI View Article : Google Scholar

111 

Blay JY, Kang YK, Nishida T and von Mehren M: Gastrointestinal stromal tumours. Nat Rev Dis Primer. 7(22)2021.PubMed/NCBI View Article : Google Scholar

112 

Reita D, Pabst L, Pencreach E, Guérin E, Dano L, Rimelen V, Voegeli AC, Vallat L, Mascaux C and Beau-Faller M: Molecular mechanism of EGFR-TKI resistance in EGFR-mutated non-small cell lung cancer: Application to biological diagnostic and monitoring. Cancers (Basel). 13(4926)2021.PubMed/NCBI View Article : Google Scholar

113 

Makhov P, Joshi S, Ghatalia P, Kutikov A, Uzzo RG and Kolenko VM: Resistance to systemic therapies in clear cell renal cell carcinoma: Mechanisms and Management Strategies. Mol Cancer Ther. 17:1355–1364. 2018.PubMed/NCBI View Article : Google Scholar

114 

He J, Huang Z, Han L, Gong Y and Xie C: Mechanisms and management of 3rd-generation EGFR-TKI resistance in advanced non-small cell lung cancer (Review). Int J Oncol. 59(90)2021.PubMed/NCBI View Article : Google Scholar

115 

Araki T, Kanda S, Horinouchi H and Ohe Y: Current treatment strategies for EGFR-mutated non-small cell lung cancer: From first line to beyond osimertinib resistance. Jpn J Clin Oncol. 53:547–561. 2023.PubMed/NCBI View Article : Google Scholar

116 

Yoshifuji K and Sasaki K: Adverse events and dose modifications of tyrosine kinase inhibitors in chronic myelogenous leukemia. Front Oncol. 12(1021662)2022.PubMed/NCBI View Article : Google Scholar

117 

Li Y, Mao T, Wang J, Zheng H, Hu Z, Cao P, Yang S, Zhu L, Guo S, Zhao X, et al: Toward the next generation EGFR inhibitors: An overview of osimertinib resistance mediated by EGFR mutations in non-small cell lung cancer. Cell Commun Signal. 21(71)2023.PubMed/NCBI View Article : Google Scholar

118 

Kumar S and Agrawal R: Next generation tyrosine kinase inhibitor (TKI): Afatinib. Recent Pat Anticancer Drug Discov. 9:382–393. 2014.PubMed/NCBI View Article : Google Scholar

119 

Blaquier JB, Ortiz-Cuaran S, Ricciuti B, Mezquita L, Cardona AF and Recondo G: Tackling osimertinib resistance in EGFR-mutant non-small cell lung cancer. Clin Cancer Res. 29:3579–3591. 2023.PubMed/NCBI View Article : Google Scholar

120 

Du X, Yang B, An Q, Assaraf YG, Cao X and Xia J: Acquired resistance to third-generation EGFR-TKIs and emerging next-generation EGFR inhibitors. Innovation (Camb). 2(100103)2021.PubMed/NCBI View Article : Google Scholar

121 

Iqbal N and Iqbal N: Imatinib: A breakthrough of targeted therapy in cancer. Chemother Res Pract. 2014(357027)2014.PubMed/NCBI View Article : Google Scholar

122 

Sacha T: Imatinib in chronic myeloid leukemia: An overview. Mediterr J Hematol Infect Dis. 6(e2014007)2014.PubMed/NCBI View Article : Google Scholar

123 

Lopes LF and Bacchi CE: Imatinib treatment for gastrointestinal stromal tumour (GIST). J Cell Mol Med. 14:42–50. 2010.PubMed/NCBI View Article : Google Scholar

124 

Hochhaus A, Larson RA, Guilhot F, Radich JP, Branford S, Hughes TP, Baccarani M, Deininger MW, Cervantes F, Fujihara S, et al: Long-term outcomes of imatinib treatment for chronic myeloid leukemia. N Engl J Med. 376:917–927. 2017.PubMed/NCBI View Article : Google Scholar

125 

Gollob JA, Wilhelm S, Carter C and Kelley SL: Role of Raf kinase in cancer: Therapeutic potential of targeting the Raf/MEK/ERK signal transduction pathway. Semin Oncol. 33:392–406. 2006.PubMed/NCBI View Article : Google Scholar

126 

Escudier B, Szczylik C, Eisen T, Stadler WM, Schwartz B, Shan M and Bukowski RM: Randomized phase III trial of the Raf kinase and VEGFR inhibitor sorafenib (BAY 43-9006) in patients with advanced renal cell carcinoma (RCC). J Clin Oncol. 23 (Suppl 16)(LBA451)2005.

127 

Ramakrishnan V, Timm M, Haug JL, Kimlinger TK, Wellik LE, Witzig TE, Rajkumar SV, Adjei AA and Kumar S: Sorafenib, a dual Raf kinase/vascular endothelial growth factor receptor inhibitor has significant anti-myeloma activity and synergizes with common anti-myeloma drugs. Oncogene. 29:1190–1202. 2010.PubMed/NCBI View Article : Google Scholar

128 

Wilhelm SM, Adnane L, Newell P, Villanueva A, Llovet JM and Lynch M: Preclinical overview of sorafenib, a multikinase inhibitor that targets both Raf and VEGF and PDGF receptor tyrosine kinase signaling. Mol Cancer Ther. 7:3129–3140. 2008.PubMed/NCBI View Article : Google Scholar

129 

Liu L, Cao Y, Chen C, Zhang X, McNabola A, Wilkie D, Wilhelm S, Lynch M and Carter C: Sorafenib blocks the RAF/MEK/ERK pathway, inhibits tumor angiogenesis, and induces tumor cell apoptosis in hepatocellular carcinoma model PLC/PRF/5. Cancer Res. 66:11851–11858. 2006.PubMed/NCBI View Article : Google Scholar

130 

Pagano M, Sierra NM, Panebianco M, Rossi G, Gnoni R, Bisagni G and Boni C: Sorafenib efficacy in thymic carcinomas seems not to require c-KIT or PDGFR-alpha mutations. Anticancer Res. 34:5105–5110. 2014.PubMed/NCBI

131 

Adnane L, Trail PA, Taylor I and Wilhelm SM: Sorafenib (BAY 43-9006, Nexavar®), a dual-action inhibitor that targets RAF/MEK/ERK pathway in tumor cells and tyrosine kinases VEGFR/PDGFR in tumor vasculature. Methods Enzymol. 407:597–612. 2006.PubMed/NCBI View Article : Google Scholar

132 

Woo HY and Heo J: Sorafenib in liver cancer. Expert Opin Pharmacother. 13:1059–1067. 2012.PubMed/NCBI View Article : Google Scholar

133 

Zhu YJ, Zheng B, Wang HY and Chen L: New knowledge of the mechanisms of sorafenib resistance in liver cancer. Acta Pharmacol Sin. 38:614–622. 2017.PubMed/NCBI View Article : Google Scholar

134 

Cheng Z, Wei-Qi J and Jin D: New insights on sorafenib resistance in liver cancer with correlation of individualized therapy. Biochim Biophys Acta Rev Cancer. 1874(188382)2020.PubMed/NCBI View Article : Google Scholar

135 

Gauthier A and Ho M: Role of sorafenib in the treatment of advanced hepatocellular carcinoma: An update. Hepatol Res. 43:147–154. 2013.PubMed/NCBI View Article : Google Scholar

136 

Porta C, Paglino C, Imarisio I and Ferraris E: Sorafenib tosylate in advanced kidney cancer: Past, present and future. Anticancer Drugs. 20:409–415. 2009.PubMed/NCBI View Article : Google Scholar

137 

Larkin JM and Eisen T: Renal cell carcinoma and the use of sorafenib. Ther Clin Risk Manag. 2:87–98. 2006.PubMed/NCBI

138 

Bukowski R, Cella D, Gondek K and Escudier B: Sorafenib TARGETs Clinical Trial Group. Effects of sorafenib on symptoms and quality of life: Results from a large randomized placebo-controlled study in renal cancer. Am J Clin Oncol. 30:220–227. 2007.PubMed/NCBI View Article : Google Scholar

139 

Szczylik C, Cella D, Eisen T, Shah S, Laferriere N, Scheuring U and Escudier BJ: Comparison of kidney cancer symptoms and quality of life (QoL) in renal cell cancer (RCC) patients (pts) receiving sorafenib vs interferon-α (IFN). J Clin Oncol. 26 (Suppl 15)(S9603)2008.

140 

Strumberg D: Sorafenib for the treatment of renal cancer. Expert Opin Pharmacother. 13:407–419. 2012.PubMed/NCBI View Article : Google Scholar

141 

Capdevila J, Iglesias L, Halperin I, Segura Á, Martínez-Trufero J, Vaz MÁ, Corral J, Obiols G, Grande E, Grau JJ and Tabernero J: Sorafenib in metastatic thyroid cancer. Endocr Relat Cancer. 19:209–216. 2012.PubMed/NCBI View Article : Google Scholar

142 

Pitoia F and Jerkovich F: Selective use of sorafenib in the treatment of thyroid cancer. Drug Des Devel Ther. 10:1119–1131. 2016.PubMed/NCBI View Article : Google Scholar

143 

Thomas L, Lai SY, Dong W, Feng L, Dadu R, Regone RM and Cabanillas ME: Sorafenib in metastatic thyroid cancer: A systematic review. Oncologist. 19:251–258. 2014.PubMed/NCBI View Article : Google Scholar

144 

Chen P, Wang J, Yao Y, Qu Y, Ji M and Hou P: . Targeting DUSP5 suppresses malignant phenotypes of BRAF-mutant thyroid cancer cells and improves their response to sorafenib. Endocrine. 85:1268–1277. 2024.PubMed/NCBI View Article : Google Scholar

145 

Pereira A, Parra D, Alvarez M and Rincon O: Response to sorafenib in a locally advanced oncocytic cell carcinoma of the thyroid. BMJ Case Rep. 17(e257738)2024.PubMed/NCBI View Article : Google Scholar

146 

Yun KM and Cohen EEW: An era of advances in systemic therapies for advanced thyroid cancer. JCO Oncol Pract. 20:899–906. 2024.PubMed/NCBI View Article : Google Scholar

147 

Ek F, Blom K, Selvin T, Rudfeldt J, Andersson C, Senkowski W, Brechot C, Nygren P, Larsson R, Jarvius M and Fryknäs M: Sorafenib and nitazoxanide disrupt mitochondrial function and inhibit regrowth capacity in three-dimensional models of hepatocellular and colorectal carcinoma. Sci Rep. 12(8943)2022.PubMed/NCBI View Article : Google Scholar

148 

Bielecka ZF, Malinowska A, Brodaczewska KK, Klemba A, Kieda C, Krasowski P, Grzesiuk E, Piwowarski J, Czarnecka AM and Szczylik C: Hypoxic 3D in vitro culture models reveal distinct resistance processes to TKIs in renal cancer cells. Cell Biosci. 7(71)2017.PubMed/NCBI View Article : Google Scholar

149 

Yueh PF, Chiang CS, Tsai IJ, Tseng YL, Chen HR, Lan KL and Hsu FT: A multifunctional PEGylated liposomal-encapsulated sunitinib enhancing autophagy, immunomodulation, and safety in renal cell carcinoma. J Nanobiotechnology. 22(459)2024.PubMed/NCBI View Article : Google Scholar

150 

Yan XQ, Ye MJ, Zou Q, Chen P, He ZS, Wu B, He DL, He CH, Xue XY, Ji ZG, et al: Toripalimab plus axitinib versus sunitinib as first-line treatment for advanced renal cell carcinoma: RENOTORCH, a randomized, open-label, phase III study. Ann Oncol. 35:190–199. 2024.PubMed/NCBI View Article : Google Scholar

151 

Motzer RJ, Porta C, Eto M, Powles T, Grünwald V, Hutson TE, Alekseev B, Rha SY, Merchan J, Goh JC, et al: Lenvatinib plus pembrolizumab versus sunitinib in first-line treatment of advanced renal cell carcinoma: Final prespecified overall survival analysis of CLEAR, a Phase III Study. J Clin Oncol. 42:1222–1228. 2024.PubMed/NCBI View Article : Google Scholar

152 

Uhlig A, Bergmann L, Bögemann M, Fischer T, Goebell PJ, Leitsmann M, Reichert M, Rink M, Schlack K, Trojan L, et al: Sunitinib for metastatic renal cell carcinoma: Real-world data from the STAR-TOR registry and detailed literature review. Urol Int. 108:198–210. 2024.PubMed/NCBI View Article : Google Scholar

153 

Heinrich MC, Jones RL, George S, Gelderblom H, Schöffski P, von Mehren M, Zalcberg JR, Kang YK, Razak AA, Trent J, et al: Ripretinib versus sunitinib in gastrointestinal stromal tumor: ctDNA biomarker analysis of the phase 3 INTRIGUE trial. Nat Med. 30:498–506. 2024.PubMed/NCBI View Article : Google Scholar

154 

Li J, Zhang J, Zhang Y, Qiu H, Zhou Y, Zhou Y, Zhang X, Zhou Y, Zhu Y, Li Y, et al: Efficacy and safety of ripretinib vs. sunitinib in patients with advanced gastrointestinal stromal tumor previously treated with imatinib: A phase 2, multicenter, randomized, open-label study in China. Eur J Cancer. 196(113439)2024.PubMed/NCBI View Article : Google Scholar

155 

Giraud EL, Westerdijk K, van der Kleij MBA, Guchelaar NAD, Meertens M, Bleckman RF, Rieborn A, Mohammadi M, Roets E, Mathijssen RHJ, et al: Sunitinib for the treatment of metastatic gastrointestinal stromal tumors: The effect of TDM-guided dose optimization on clinical outcomes. ESMO Open. 9(103477)2024.PubMed/NCBI View Article : Google Scholar

156 

Hopkins TG, Marples M and Stark D: Sunitinib in the management of gastrointestinal stromal tumours (GISTs). Eur J Surg Oncol. 34:844–850. 2008.PubMed/NCBI View Article : Google Scholar

157 

Mulet-Margalef N and Garcia-del-Muro X: Sunitinib in the treatment of gastrointestinal stromal tumor: Patient selection and perspectives. OncoTargets Ther. 9:7573–7582. 2016.PubMed/NCBI View Article : Google Scholar

158 

Le Tourneau C, Raymond E and Faivre S: Sunitinib: A novel tyrosine kinase inhibitor. A brief review of its therapeutic potential in the treatment of renal carcinoma and gastrointestinal stromal tumors (GIST). Ther Clin Risk Manag. 3:341–348. 2007.PubMed/NCBI View Article : Google Scholar

159 

Raymond E, Hammel P, Dreyer C, Maatescu C, Hentic O, Ruszniewski P and Faivre S: Sunitinib in pancreatic neuroendocrine tumors. Target Oncol. 7:117–125. 2012.PubMed/NCBI View Article : Google Scholar

160 

Kulke MH, Lenz HJ, Meropol NJ, Posey J, Ryan DP, Picus J, Bergsland E, Stuart K, Tye L, Huang X, et al: Activity of sunitinib in patients with advanced neuroendocrine tumors. J Clin Oncol. 26:3403–3410. 2008.PubMed/NCBI View Article : Google Scholar

161 

Raymond E, Dahan L, Raoul JL, Bang YJ, Borbath I, Lombard-Bohas C, Valle J, Metrakos P, Smith D, Vinik A, et al: Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 364:501–513. 2011.PubMed/NCBI View Article : Google Scholar

162 

Valle JW, Borbath I, Rosbrook B, Fernandez K and Raymond E: Sunitinib in patients with pancreatic neuroendocrine tumors: Update of safety data. Future Oncol. 15:1219–1230. 2019.PubMed/NCBI View Article : Google Scholar

163 

Polena H, Creuzet J, Dufies M, Sidibé A, Khalil-Mgharbel A, Salomon A, Deroux A, Quesada JL, Roelants C, Filhol O, et al: The tyrosine-kinase inhibitor sunitinib targets vascular endothelial (VE)-cadherin: A marker of response to antitumoural treatment in metastatic renal cell carcinoma. Br J Cancer. 118:1179–1188. 2018.PubMed/NCBI View Article : Google Scholar

164 

Rausch M, Rutz A, Allard PM, Delucinge-Vivier C, Docquier M, Dormond O, Wolfender JL and Nowak-Sliwinska P: Molecular and functional analysis of sunitinib-resistance induction in human renal cell carcinoma cells. Int J Mol Sci. 22(6467)2021.PubMed/NCBI View Article : Google Scholar

165 

Stratmann AT, Fecher D, Wangorsch G, Göttlich C, Walles T, Walles H, Dandekar T, Dandekar G and Nietzer SL: Establishment of a human 3D lung cancer model based on a biological tissue matrix combined with a Boolean in silico model. Mol Oncol. 8:351–365. 2014.PubMed/NCBI View Article : Google Scholar

166 

Liu B, Chen D, Chen S, Saber A and Haisma H: Transcriptional activation of cyclin D1 via HER2/HER3 contributes to EGFR-TKI resistance in lung cancer. Biochem Pharmacol. 178(114095)2020.PubMed/NCBI View Article : Google Scholar

167 

Jacobi N, Seeboeck R, Hofmann E, Schweiger H, Smolinska V, Mohr T, Boyer A, Sommergruber W, Lechner P, Pichler-Huebschmann C, et al: Organotypic three-dimensional cancer cell cultures mirror drug responses in vivo: Lessons learned from the inhibition of EGFR signaling. Oncotarget. 8:107423–107440. 2017.PubMed/NCBI View Article : Google Scholar

168 

Zheng Q, Dong H, Mo J, Zhang Y, Huang J, Ouyang S, Shi S, Zhu K, Qu X, Hu W, et al: A novel STAT3 inhibitor W2014-S regresses human non-small cell lung cancer xenografts and sensitizes EGFR-TKI acquired resistance. Theranostics. 11:824–840. 2021.PubMed/NCBI View Article : Google Scholar

169 

Hoque M, Elmaghrabi YA, Köse M, Beevi SS, Jose J, Meneses-Salas E, Blanco-Muñoz P, Conway JRW, Swarbrick A, Timpson P, et al: Annexin A6 improves anti-migratory and anti-invasive properties of tyrosine kinase inhibitors in EGFR overexpressing human squamous epithelial cells. FEBS J. 287:2961–2978. 2020.PubMed/NCBI View Article : Google Scholar

170 

Fu C, Dong J, Zhang J, Li X, Zuo S, Zhang H, Gao S and Chen L: Using three-dimensional model-based tumour volume change to predict the symptom improvement in patients with renal cell cancer. 3 Biotech. 14(148)2024.PubMed/NCBI View Article : Google Scholar

171 

Liu Y, Dai X, Jiang S, Qahar M, Fang C, Guo D, Wang L, Ma S and Huang L: Targeted Co-delivery of gefitinib and rapamycin by aptamer-modified nanoparticles overcomes EGFR-TKI Resistance in NSCLC via Promoting Autophagy. Int J Mol Sci. 23(8025)2022.PubMed/NCBI View Article : Google Scholar

172 

Kim D, Bach DH, Fan YH, Luu TT, Hong JY, Park HJ and Lee SK: AXL degradation in combination with EGFR-TKI can delay and overcome acquired resistance in human non-small cell lung cancer cells. Cell Death Dis. 10(361)2019.PubMed/NCBI View Article : Google Scholar

173 

Hou R, Li X, Xiong J, Shen T, Yu W, Schwartz LH, Zhao B, Zhao J and Fu X: Predicting tyrosine kinase inhibitor treatment response in stage IV lung adenocarcinoma patients with EGFR mutation using model-based deep transfer learning. Front Oncol. 11(679764)2021.PubMed/NCBI View Article : Google Scholar

174 

Ko J, Meyer AN, Haas M and Donoghue DJ: Characterization of FGFR signaling in prostate cancer stem cells and inhibition via TKI treatment. Oncotarget. 12:22–36. 2021.PubMed/NCBI View Article : Google Scholar

175 

Kim SY, Kim SM, Lim S, Lee JY, Choi SJ, Yang SD, Yun MR, Kim CG, Gu SR, Park C, et al: Modeling clinical responses to targeted therapies by patient-derived organoids of advanced lung adenocarcinoma. Clin Cancer Res. 27:4397–4409. 2021.PubMed/NCBI View Article : Google Scholar

176 

Darré H, Masson P, Nativel A, Villain L, Lefaudeux D, Couty C, Martin B, Jacob E, Duruisseaux M, Palgen JL, et al: Comparing the efficacy of two generations of EGFR-TKIs: An integrated drug-disease mechanistic model approach in EGFR-mutated lung adenocarcinoma. Biomedicines. 12(704)2024.PubMed/NCBI View Article : Google Scholar

177 

Zhao X, Zhang X, Chen H, Bao H, Wu X, Wang H, Bao H, Pang J, Wang S and Wang J: Mechanisms of resistance to tyrosine kinase inhibitors in ROS1 fusion-positive nonsmall cell lung cancer. Clin Chem. 70:629–641. 2024.PubMed/NCBI View Article : Google Scholar

178 

Hirschhaeuser F, Menne H, Dittfeld C, West J, Mueller-Klieser W and Kunz-Schughart LA: Multicellular tumor spheroids: An underestimated tool is catching up again. J Biotechnol. 148:3–15. 2010.PubMed/NCBI View Article : Google Scholar

179 

Wenzel C, Riefke B, Gründemann S, Krebs A, Christian S, Prinz F, Osterland M, Golfier S, Räse S, Ansari N, et al: 3D high-content screening for the identification of compounds that target cells in dormant tumor spheroid regions. Exp Cell Res. 323:131–143. 2014.PubMed/NCBI View Article : Google Scholar

180 

Imamura Y, Mukohara T, Shimono Y, Funakoshi Y, Chayahara N, Toyoda M, Kiyota N, Takao S, Kono S, Nakatsura T and Minami H: Comparison of 2D- and 3D-culture models as drug-testing platforms in breast cancer. Oncol Rep. 33:1837–1843. 2015.PubMed/NCBI View Article : Google Scholar

181 

Hashimoto K, Nishimura S, Ito T, Oka N, Kakinoki R and Akagi M: Clinicopathological assessment of cancer/testis antigens NY-ESO-1 and MAGE-A4 in osteosarcoma. Eur J Histochem. 66(3377)2022.PubMed/NCBI View Article : Google Scholar

182 

Leeper AD, Farrell J, Williams LJ, Thomas JS, Dixon JM, Wedden SE, Harrison DJ and Katz E: Determining tamoxifen sensitivity using primary breast cancer tissue in collagen-based three-dimensional culture. Biomaterials. 33:907–915. 2012.PubMed/NCBI View Article : Google Scholar

183 

Andolfi C, Bartolini C, Morales E, Gündoğdu B, Puhr M, Guzman J, Wach S, Taubert H, Aigner A, Eder IE, et al: MED12 and CDK8/19 modulate androgen receptor activity and enzalutamide response in prostate cancer. Endocrinology. 165(bqae114)2024.PubMed/NCBI View Article : Google Scholar

184 

Jenkins RW, Barbie DA and Flaherty KT: Mechanisms of resistance to immune checkpoint inhibitors. Br J Cancer. 118:9–16. 2018.PubMed/NCBI View Article : Google Scholar

185 

Hasin Y, Seldin M and Lusis A: Multi-omics approaches to disease. Genome Biol. 18(83)2017.PubMed/NCBI View Article : Google Scholar

186 

Libbrecht MW and Noble WS: Machine learning applications in genetics and genomics. Nat Rev Genet. 16:321–332. 2015.PubMed/NCBI View Article : Google Scholar

187 

Wang H, Brown PC, Chow ECY, Ewart L, Ferguson SS, Fitzpatrick S, Freedman BS, Guo GL, Hedrich W, Heyward S, et al: 3D cell culture models: Drug pharmacokinetics, safety assessment, and regulatory consideration. Clin Transl Sci. 14:1659–1680. 2021.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

March-April 2025
Volume 5 Issue 2

Print ISSN: 2754-3242
Online ISSN:2754-1304

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Isinelli G, Failla S, Plebani R and Prete A: Exploring oncology treatment strategies with tyrosine kinase inhibitors through advanced 3D models (Review). Med Int 5: 13, 2025.
APA
Isinelli, G., Failla, S., Plebani, R., & Prete, A. (2025). Exploring oncology treatment strategies with tyrosine kinase inhibitors through advanced 3D models (Review). Medicine International, 5, 13. https://doi.org/10.3892/mi.2024.212
MLA
Isinelli, G., Failla, S., Plebani, R., Prete, A."Exploring oncology treatment strategies with tyrosine kinase inhibitors through advanced 3D models (Review)". Medicine International 5.2 (2025): 13.
Chicago
Isinelli, G., Failla, S., Plebani, R., Prete, A."Exploring oncology treatment strategies with tyrosine kinase inhibitors through advanced 3D models (Review)". Medicine International 5, no. 2 (2025): 13. https://doi.org/10.3892/mi.2024.212