Previous Article in Journal
Assessing the Utility of Organoid Intelligence: Scientific and Ethical Perspectives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Three-Dimensional Culture Systems in Neuroblastoma Research

1
Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
2
Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
*
Authors to whom correspondence should be addressed.
Organoids 2025, 4(2), 10; https://doi.org/10.3390/organoids4020010
Submission received: 23 March 2025 / Revised: 25 April 2025 / Accepted: 29 April 2025 / Published: 8 May 2025

Abstract

:
Basic and translational cancer biology research requires model systems that recapitulate the features of human tumors. While two-dimensional (2D) cell cultures have been foundational and allowed critical advances, they lack the organizational complexity, cellular interactions, and extracellular matrix present in vivo. Mouse models have thus remained the gold standard for studying cancer. In addition to high cost and low throughput, mouse models can also suffer from reduced tumor heterogeneity and species-specific differences. Three-dimensional (3D) culture models have emerged as a key intermediary between 2D cell lines and mouse models, with lower cost and greater flexibility than mouse models and a more accurate representation of the tumor microenvironment than 2D cell lines. In neuroblastoma, an aggressive childhood cancer, 3D models have been applied to study drug responses, cell motility, and tumor–matrix interactions. Recent advances include the integration of immune cells for immunotherapy studies, mesenchymal stromal cells for tumor–stroma interactions, and bioprinted systems to manipulate matrix properties. This review examines the use of 3D culture systems in neuroblastoma, highlighting their advantages and limitations while emphasizing their potential to bridge gaps between in vitro, preclinical, and clinical applications. By improving our understanding of neuroblastoma biology, 3D models hold promise for advancing therapeutic strategies and outcomes in this childhood cancer.

1. Introduction

Research on the mechanisms governing cellular physiology and pathology has significantly benefited from the development and application of model systems. The foundation of cell culture dates back to 1907, when Harrison conducted pioneering experiments on the origin of nerve fibers, marking the first successful cultivation of cells outside the body [1,2]. Cancer cells were famously first cultured by George Gey, who established the Hela cell line from a biopsy of the cervical cancer of Henrietta Lacks [3,4]. Cell culture techniques, now referred to as two-dimensional (2D, monolayer) cell culture, have evolved but remain indispensable tools in biology [5,6].
Two-dimensional cell culture has been particularly influential in cancer research, providing a simple and accessible platform for studying cellular behavior [7]. Widely utilized cell lines offer distinct advantages: They are readily available, cost-effective, easy to handle, and capable of producing highly reproducible data. Despite its widespread use and utility, 2D cell culture presents significant limitations. These systems fail to capture the structural complexity and microenvironment of tissues in vivo, limiting their ability to model the behavior of cells within a living organism [8]. Two-dimensional cultures rapidly lose cell–cell and cell–matrix interactions, disrupting key signaling cascades essential for in vivo cell function [9]. These signaling pathways can be important in proliferation, migration, and apoptosis, all highly relevant to tumor biology and therapy [10,11,12]. While 2D cancer cells can grow robustly, this can also allow them to contaminate each other, and cross-contamination of cell lines has been reported to be as high as 25% [13,14]. Prolonged passaging can also lead to progressive genetic and phenotype evolution and further deviation from the original tumor [8].
Humans have used animals to model human biology going back to ancient Greece [15]. The first animal models of cancer were used in the early 20th century and included Peyton Rous’s study of viral transmission of sarcomas in chickens and the demonstration of chemical carcinogenesis in rabbits by Katsusaburo Yamagiwa and Koichi Ichikawa [16,17]. Mouse models have come to dominate modern cancer research due to their ease of use and the ability to genetically modify them. Mouse models of cancer are widely regarded as the gold standard for mimicking human biology in cancer research, providing valuable insights into tumor behavior and therapeutic responses within a living system. Mice share several anatomical, cellular, and molecular features with humans that are essential for cancer development, and approximately 80% of mouse genes have human orthologs [18]. Cancer models in mice include transgenic tumor-prone mice, carcinogen-induced models, syngeneic transplant models, and human tumors cells grown in immunodeficient or humanized mice directly as patient-derived xenografts or indirectly as cell line-derived xenografts. Each of these systems has advantages and disadvantages, but all suffer from high cost, low throughput, and the utilization of living animals.
In recent years, three-dimensional (3D) cancer cell culture has become a key intermediary between traditional 2D cell lines and mouse models [19,20]. These models provide some of the flexibility and throughput of cell lines while more closely mimicking the in vivo microenvironment, including cellular architecture, gene expression, metabolism, and signaling pathways [21,22,23]. Three-dimensional cancer models have been used extensively in a variety of cancers, particularly epithelial cancers like colon, breast, liver, pancreas, prostate, ovarian, lung, and gastric cancers [10,21,23,24,25,26,27,28,29,30,31]. Three-dimensional cultures can be grown from human or murine tumors and formed in suspension culture [10,28] or embedded in a scaffold formed by the addition of native or artificial matrix components [23,24]. To more closely mimic tumors, 3D cultured tumor cells have been grown together with other cell types, including fibroblasts [32,33] and immune cells [28,34,35].
Three-dimensional culture has been used to study basic mechanisms of tumorigenesis [26,36], test existing therapies [37,38,39,40], screen for new therapies [25], and develop personalized treatment regimens [41], among other applications. Tumorigenesis has been studied by introducing genetic alterations into non-cancerous human tissue organoids, then examining the impact on the proliferation of the organoids and their ability to form tumors in immunodeficient mice [36,42,43]. Such studies have functionally tested the ability of mutations found in human tumors to drive tumorigenesis in human cells in a physiological cell context, something not previously possible. While not as easy to work with as 2D cell lines, 3D culture allows the throughput necessary to test drug responses. This has been used to predict patient therapy response, particularly in identifying patients likely or unlikely to respond to standard of care treatments [44,45,46] and to identify biomarkers of responders [41]. Three-dimensional cultures have also been used in large-scale drug testing. This includes small molecules [25], but also antibody-based therapeutics [47]. Indeed, Herpers and colleagues screened colon cancer organoids and identified a bispecific antibody targeting EGFR and LGR5 using a high-content-based platform. They went on to demonstrate its efficacy in organoids and patient-derived xenografts. This antibody (Petosemtamab) was subsequently demonstrated to have efficacy in an early-phase clinical trial [48].
This review will describe the current state of 3D culture systems in neuroblastoma, a pediatric tumor arising from the developing peripheral nervous system. We begin with a discussion of 2D cell culture and in vivo models in neuroblastoma, including patient-derived xenografts and genetically engineered models. We then describe 3D culture systems and their application in neuroblastoma research. These include the most basic models that include only tumor cells as well more complex models that include other cell types or added matrix components. We conclude with a discussion of the limitations associated with current 3D culture methodologies and how 3D culture can continue to improve our understanding and treatment of this complex disease.

2. Neuroblastoma 2D and In Vivo Models

2.1. Neuroblastoma

Neuroblastoma is the most prevalent extracranial solid tumor in children. It accounts for between 6 and 10% of all pediatric cancers and about 11% of cancer-related deaths in children [49,50]. Neuroblastomas arise from migrating neural crest cells. Primary lesions are found in the adrenal medulla or the paraspinal sympathetic ganglia of the neck, chest, abdomen, or pelvis [51]. Neuroblastoma is typically sporadic, with familial cases accounting for 1–2% of diagnoses [51]. Metastases are present in about 50% of patients at diagnosis, with bone marrow metastases as the most common location. Metastases are also found in bones and regional lymph nodes, while the involvement of the central nervous system and lungs is less common [52,53].
Neuroblastoma exhibits remarkable heterogeneity in clinical behavior, with some tumors proving fatal despite extensive, multimodal, and toxic treatments and others cured with minimal or even no therapy, including some special cases of metastatic disease [54,55]. Patients are risk-stratified according to a number of characteristics, including age, stage, pathological grade, MYCN amplification status, and the presence of segmental chromosomal aberrations. Low and intermediate groups have 5-year survival of greater than 85–90% while outcomes are still poor for high-risk patient with approximately 50% surviving long term [52,56]. Thus, despite significant progress in therapeutic and research efforts, the prognosis for high-risk neuroblastoma remains poor, necessitating innovative approaches to study neuroblastoma biology.

2.2. Neuroblastoma Cell Lines

Short-term culture of neuroblastoma cell lines dates back to the 1940s [57], even prior to the establishment of Hela cells. Murray and Stout described the morphology of eight pairs of primary and metastatic tumors in 2D culture, features which they argued could be used for diagnosis [57]. Neuroblastoma cells generally grow very well in culture and large numbers of cell lines have since been established. This has allowed researchers to define and study the differential impact of specific biologic and genetic features [58] and perform large scale experiments, including RNAi [59], CRISPR-Cas9 [60], and small molecule screens [61,62].
The large number of available cell lines has also allowed the appreciation that neuroblastoma cells can adopt different cellular morphologies in culture, sometimes even between cells in the same dish [63,64,65]. These cell types have been divided into three categories: neuroblastic (N-type), substrate-adherent (S-type), and intermediate (I-type), which combines characteristics of both N- and S-types [66,67]. More recently, these morphologic differences in cells aided in the transcriptional and epigenomic definition of two lineage states, an adrenergic or noradrenergic state and a mesenchymal or neural crest stem cell-like state [68,69]. These states have different super enhancer landscapes and core regulatory circuits of transcription factors [70], distinct surface proteins that may serve as immunotherapeutic targets [71], and different susceptibility to immune-cell killing [72,73]. The role of these cell line-defined states in human tumors is still debated, but there is evidence that they are relevant in patient tumors [74,75].
Despite their continued value, neuroblastoma cell lines suffer from the same drawbacks as 2D cultures in other tumor types in their failure to model the complexity of tumors [76]. This is particularly true in modeling therapy response, exemplified by PIVOT (Pediatric Preclinical In Vivo Testing Consortium) removing the in vitro component that had been part of its predecessor, the PPTC (Pediatric Preclinical Testing Consortium) [77]. Thus, there is clearly a need for a more physiological culture model.

2.3. Human Neuroblastoma Grown in Mice

Human neuroblastoma cell lines can be injected into immunodeficient mice as a cell line-derived xenograft (CDX) and patient tumor fragments or tumor cells can be directly implanted without culture as a patient-derived xenograft (PDX). These can be subcutaneous or orthotopic. Orthotopic injection is challenging in neuroblastoma, as the orthotopic location is the adrenal gland or the sympathetic chain, both small and not amenable to injection. However, studies have shown that implantation of tumors cells in the adrenal fat pad or under the renal capsule can produce tumors with a higher rate of metastases than subcutaneous, suggesting some improved utility to such models [78,79,80,81]. In either location, PDXs can be repeatedly passed to recipients of the following generation, and these models maintain their molecular characteristics following serial passaging [79,82,83]. Large numbers of PDXs have been developed. Many of these have been extensively characterized through efforts such as the PPTC/PIVOT [84] and the Childhood Solid Tumor Network (CSTN) [85]. Neuroblastoma PDXs are available to the research community through the CSTN and the Alex’s Lemonade Stand Foundation-funded Childhood Cancer Repository [86].
PDXs are generally considered a more accurate representation of human disease than CDXs, as the cells in a PDX have not adapted to and changed in 2D culture [82]. However, drawbacks remain. PDXs are grown in immunodeficient mice, and functional T cells are absent from athymic nude mice, functional T and B cells are absent from NOD-scid mice, and functional T, B, and NK cells are absent from the NOD-scid-gamma (NSG) strain. This limits studies of immunotherapy. Furthermore, while the tumor microenvironment (TME) of PDXs may include cell types that resemble patient neuroblastomas, the majority of these cells are derived from mice [83]. It is unclear how human tumor cells and murine stroma can signal to each other, given the species incompatibility between many secreted signaling molecules [87]. Additionally, PDXs require mice and are thus expensive, and they are not amenable to genetic manipulation. To compensate for some of these limitations, alternative models have been developed. To allow tumor-immune interactions, immunocompromised mice have been transplanted with human hematopoietic stem cells, including in mice with humanization of cytokines to promote innate immune cell development [88]. To allow genetic manipulation, human neural crest stem cells have been isolated and manipulated to form neuroblastoma after injection into immunocompromised mice [89] or immunocompetent mouse embryos [90]. These newer techniques are promising but continue to be resource intensive.

2.4. Murine Neuroblastoma Grown in Mice

Fully murine tumors grown in mice allow for native interactions between tumor and host and a complete and functional immune system. Such models include Genetically Engineered Mouse Models (GEMMs), carcinogen-induced tumors, and murine cancer cell lines reimplanted to create syngeneic models [91]. GEMMs are particularly suited to the study of pediatric cancers that are a result of interrupted developmental programs and have less complex genomes than most adult cancers [92]. In neuroblastoma, both GEMMs and syngeneic tumors have been used extensively. The first and most widely used neuroblastoma GEMM is the TH-MYCN model, driven by MYCN overexpression under the tyrosine hydroxylase (TH) promoter. This produces tumors that arise along the sympathetic chain and share key histological and genetic features with human neuroblastomas [93], including expression of the immunotherapeutic target GD2 [94]. While the TH-MYCN model has become essential to neuroblastoma biological and preclinical investigations, additional modifications have also been made for specific purposes. These include p53 haploinsufficiency to investigate mechanisms of chemotherapy response [95,96], the addition of mutant ALK to model response and resistance to ALK inhibition [97], and the deletion of Caspase 8 to enhance bone marrow metastases [98]. Alternative models that produce neuroblastoma in mice include simian virus 40 T-antigen expression [99,100] and dopamine β-hydroxylase (DBH) promoter-driven Cre expression leading to overexpression of MYCN [101] or LIN28B [102]. GEMMs are generally excellent neuroblastoma models but are resource- and labor-intensive to maintain breeding colonies, challenging to genetically manipulate, and not amenable to substantial throughput.
Neuroblastoma cell lines that have been used to form syngeneic models include those from the TH-MYCN model, particularly the 9464D line that is in the C57Bl/6 background. The C1300 murine cell line was developed from a spontaneous tumor and two derivatives of this line, Neuro2a and NXS2, are frequently used and form tumors in A/J mice [103]. These lines can be implanted subcutaneously or orthotopically [104]. Cell lines from the TH-MYCN model have important differences from the autochthonous tumors [94]. While amenable to genetic manipulation, these models lose important features of spontaneous tumors and remain resource-intensive compared to in vitro approaches.

3. Neuroblastoma In Vitro 3D Models

To address the gap between the simplistic and high-throughput monolayer culture of tumor cells and the complex but resource-intensive in vivo models, researchers have increasingly turned to 3D culture systems as a bridge, aiming to develop easy-to-reproduce, fast, and translational models to study cancer [19,31]. While 3D culture methods have existed since at least the 1970s [105], they have become increasingly utilized over the last decade. This is also the case in neuroblastoma. In neuroblastoma, some cell lines grow as multicellular spheres in suspensions even in traditional 2D culture conditions [58]. More deliberate 3D culture models have also been adopted in neuroblastoma, though they remain less characterized and established compared to those in colon or breast cancers, for example [20,106,107,108]. Terminology has been a challenge, in part because the term “organoid” in cancer was originally defined for epithelial cancers, and it is not straightforward to apply the same framework to non-epithelial tumors [109]. In neuroblastoma, terms such as sphere-forming cells, spheroids, neurospheres, tumor spheroids, multicellular tumor spheroids, tumorspheres, and organoids have all been used without a clear distinction between methodologies. For the purpose of this review, we will distinguish between (1) 3D cultures of neuroblastoma cells without other cell types and without an added extracellular matrix component, (2) 3D cultures of neuroblastoma cells grown with other cell types, and (3) 3D cultures of neuroblastoma cells grown with an added matrix support. We will discuss differences between the techniques that have been applied in neuroblastoma and the advantages and drawbacks they have offered in comparison to 2D culture and in vivo systems, as summarized in Figure 1.

3.1. Human Neuroblastoma Cells Grown Alone in 3D

The simplest 3D models involve only neuroblastoma cells without other cell types or an added extracellular matrix. While some neuroblastoma cell lines grow as unattached clusters of cells regardless of tissue culture conditions, concerted efforts have been made to force neuroblastoma cells to assume 3D structures since at least the 1980s [110]. A variety of methods have been used to achieve this. Some dissociated tumor cells will grow as spheres when grown in neural basal media (a mixture of DMEM/F12 without serum and with B27 and other growth factor supplements) [111] and some of these continuously growing 3D cell lines have been used in a number of subsequent studies [112,113,114]. To prevent adherence to a dish, some studies used low adherence tissue culture dishes either with neural basal media [83,115,116,117] or with typical serum-containing media [118,119,120,121,122], while another used a rotary bioreactor to create a zero-gravity condition [123]. Most studies have allowed cells to either proliferate into spheres or spontaneously aggregate, but others have centrifuged cells together [124].
3D spheres have been grown from traditional 2D cultured cell lines [118,119,120,123,125,126,127,128,129] and also directly from dissociated human tumors or PDXs without 2D culture first [83,111,115,130]. While there is an obvious appeal to avoiding 2D culture entirely and the adaptations it requires, there have not been extensive studies that compare 3D cultures that originated from cell lines to those that originated from tumors. In both cases, 3D cultured cells form spherical aggregates of cells that grow to approximately 200–700 μm in diameter. Typically, spheres of this size will develop variation in oxygenation and a necrotic core similar to a tumor [131], although this has been shown in only a minority of neuroblastoma models. Studies have shown that 3D cultured neuroblastoma cells express more cancer stem cell markers and display increased tumorigenicity compared to 2D cultures [126,128,132]. They also have transcriptomic and epigenomic features that more closely resemble tumors [83,111,115].
Neuroblastoma cells cultured alone in 3D have been used extensively to study the response to therapy and identify new therapeutic vulnerabilities. Studies have found that 3D cultures are more resistant to chemotherapy than 2D culture [128] and that 3D cultures can be used to study synergistic interactions between chemotherapy agents [133]. A recent study from the Bexell group showed that 3D cultures formed from PDXs that relapsed after chemotherapy treatment retained chemoresistance in culture compared to 3D cultures formed from chemotherapy-naïve PDXs. Furthermore, cells that survived in vitro chemotherapy treatment had similar transcriptional changes as PDXs that relapsed after in vivo chemotherapy [134]. Three-dimensional culture has also been used to study other therapeutic modalities, including antibody–drug conjugates [118], individual and combinatorial targeted therapies [120,135], differentiation therapy [129,136], and oncolytic therapy [128]. Kaess et al. used 3D culture to test the RIST treatment protocol (Rapamycin Irinotecan Sunitinib/Dasatinib Temozolomide) and showed this markedly decreased 3D cultured neuroblastoma viability [126], which is important as this regimen showed increased survival in a recent clinical trial compared to the control arm [137]. Three-dimensional cultures have also been leveraged to identify new neuroblastoma susceptibilities. Sundaramoorthy et al. identified BMX as a new therapeutic targeting by identifying transcriptional and epigenetic features common between PDXs and 3D culture but not found in 2D cell lines [115]. Three-dimensional cultures have also been adapted to high-throughput small molecule screens, which identified KSP inhibitors [138] and tyrosine kinase inhibitors with anti-neuroblastoma activity [139].

3.2. Human Neuroblastoma Cells Grown with Other Cell Types in 3D

While 2D co-cultures are an important tool to study cell–cell interactions, 3D models allow more native-like interactions between cell types. Tumor-immune cell interactions are the cell–cell interactions that have been most extensively studied in neuroblastoma 3D models. These models typically involve neuroblastoma sphere formation prior to the addition of immune cells. Three recent studies demonstrated the utility of 3D culture to study the activity of anti-GD2 antibodies. Kholosy and colleagues reported an assay for quantifying the death induced by an anti-GD2 antibody and peripheral blood mononuclear cells (PBMCs) [140], Troschke-Meurer et al. tested how chemotherapy alters the impact of anti-GD2 therapy and PBMCs [124], and Nguyen et al. showed that IL-15 can enhance the anti-neuroblastoma activity of anti-GD2 therapy and NK cells [141]. Other groups have studied antibody-independent killing of tumor cells. This includes NK cell killing, particularly the ability of MDM2 inhibition to enhance NK cell-mediated killing [142]. It also includes T cell killing. One study found that HDAC inhibition with etinostat increased MHC-I expression and sensitized it to killing by PRAME-specific T cells [113]. Another engineered αβ-T cells to express the γδ-T cell receptor and founds these could kill 3D neuroblastoma cultures independent of MHC-I expression [112].
Fewer studies have grown neuroblastoma cells in 3D with non-immune cells from the tumor microenvironment. One such important population in neuroblastoma is cancer-associated fibroblasts (CAFs) [143]. The Larsson group has established and used a 3D model by combining human neuroblastoma cell lines with a human dermal fibroblast cell line in low attachment plates and allowing them to form mixed spheres [144,145]. They showed that the combined spheres had a tumor-like hypoxic core and were more compact than mono-neuroblastoma spheres, and that the fibroblasts started expressing CAF markers. They then showed that inhibition of prostaglandin synthesis in fibroblasts resulted in sensitization of spheres to chemotherapy [145]. In a subsequent study, they used this model to screen for synergistic combinations with prostaglandin synthesis inhibitors. Another group made co-culture spheres in a similar fashion but with centrifugation of the cells and investigated how adrenergic and mesenchymal neuroblastoma cells interacted differently with fibroblasts [146]. This model has been more recently used by another group to test the efficacy of a novel combination therapy [147].
Another important cell type in the microenvironment of tumors is endothelial cells. Several studies have combined 3D neuroblastoma cells with endothelial cells [148,149,150,151,152,153]. In all cases, these required the addition of matrix components and/or bioprinting of scaffolds (see more in subsequent section on these techniques). Ning and colleagues used a gelatin derivative to bioprint chambers with a central cavity and microchannels, lined them with human umbilical vein endothelial cells (HUVECs), and then added neuroblastoma spheres to the cavity. The HUVECs increased the invasiveness of the neuroblastoma cells [151]. Nothdurfter and colleagues also used bioprinting with HUVEC-lined channels using a gelatin and fibrin mixture and similarly found that some neuroblastoma cells could invade the endothelial cells [152].

3.3. Murine Neuroblastoma Cells Grown in 3D

While the described systems using human cells have some distinct advantages, it is challenging to translate tumor-immune interactions probed in 3D culture directly into the in vivo setting. This is because such experiments would either require either immunodeficient mice or transition to murine neuroblastoma cells. For these reasons, growing murine tumor cells in 3D is appealing. Murine neuroblastoma cell lines have been used for this purpose. Neuro2A cells grow in 3D spheres in NeuroCult media and are more resistant to chemotherapy and radiation therapy compared to adherent cells [63,154]. The Fruci lab has grown 3D cultures directly from 9464D and 975A2 cells as well as from tumors formed from these lines in standard serum-containing media. By combining them with chemotherapy, they were able to demonstrate the ability of chemotherapy to sensitize to killing by splenocytes, and validate this effect in vivo [155,156].
Using spontaneous murine tumors directly in 3D culture has been more challenging than with human tumors or with human or murine cell lines. Three different groups have successfully cultured dissociated TH-MYCN tumor cells in 3D [157,158,159,160,161]. Two of these reported failure of non-adherent cells to grow in the type of neural basal media that had been successful with human tumors [157,159]. One was subsequently successful using a neural crest media with 12% fetal bovine serum [157] and the other by adding 15% fetal bovine serum and β-mercaptoethanol [159]. These groups went on to use these systems to investigate metabolic reprogramming in neuroblastoma [157] and the roles of PRMT1 [158] and PES1 [160] in neuroblastoma cell survival. Embaie et al. recently took a different approach and leveraged analysis of scRNAseq data from TH-MYCN tumors to design a complex culture media without serum but with a number of different growth factors and inhibitors of p38 MAPK and the TGFβ receptor. They found this media formulation promoted the growth of dissociated tumor cells in 3D when grown in Matrigel and went on to perform scRNAseq on these cells. They found that 3D culture maintained the adrenergic cell states found in the TH-MYCN tumor [161]. This last point is important, as this state is lost in TH-MYCN cell lines grown in 2D culture [94].

3.4. Neuroblastoma Cells Grown with an Added Matrix

A major limitation of 2D culture is the absence of a surrounding extracellular matrix (ECM), a complex network of bioactive molecules that provides structural support and signaling cues essential for cellular function, tissue organization, and tumor progression, including initiation, growth, metastases, and treatment resistance [162]. Three-dimensional culture provides an opportunity to include extracellular matrix components to more closely mimic in vivo tumors, and this has been performed extensively in other tumor types [163]. Indeed, unlike in neuroblastoma, the most commonly used 3D culture conditions incorporate the use of ECM hydrogels such as Matrigel [164], though less biologically complex scaffolds are also common, including collagen and fibrinogen [165]. In general, approaches can be divided into three groups: native TME components (such as collagen, fibrin, and Matrigel), artificial components (non-native to the TME, including cellulose and synthetic polymers), and bioengineering approaches (which can combine both native and artificial scaffolds with methods like bioprinting and microfluidic devices) [162]. While not used as extensively as in other tumor types, all three approaches have been studied in neuroblastoma.
Several research groups have incorporated native TME components in 3D neuroblastoma culture. Collagen, a molecule abundantly present in the tumor ECM, has been used to study neuroblastoma invasion [166] and collagen enriched with glycosaminoglycans and nanohydroxyapatite has been used to accurately model neuroblastoma chemoresistance [167,168]. Gelatin methacrylate (GelMA), a biomaterial derived from gelatin, can be printed and used to support 3D culture. One group used this to incorporate endothelial cells as described above [151] and another added a synthetic polymer to increase its porosity and used this system to study metastatic behavior [169]. Matrigel, derived from the secretion of Engelbreth-Holm-Swarm mouse sarcoma cells [170], is widely used in adult 3D cancer models. It has been incorporated into 3D neuroblastoma systems to promote sphere formation in both human [171] and murine 3D culture and is helpful in culturing neuroblastoma cells with endothelial cells [148].
Artificial scaffolds include those made from cellulose, a naturally occurring but non-TME component. Two independent studies have explored its applications. One study enhanced a bacterial nanocellulose scaffold with trimethyl ammonium beta-hydroxypropyl and collagen [172]. The other study chemically oxidized cellulose and modified its blend, discovering that the resulting composites enhanced cytocompatibility and promoted neurite development [173]. Other artificial scaffolds include synthetic polymers, such as polyacrylonitrile (PAN) and polyethylene glycol (PEG). PAN has been shown to enhance the survival of neuroblastoma cells [174] and PEG, when enriched with vitronectin, improved neuroblastoma cell adhesion [175]. Other studies have used scaffolds composed of silk to create hypoxic gradients [176], to study the response to integrin inhibition after incorporation of vitronectin [177], and to model the interactions between neuroblastoma cells, macrophages, and NK cells [178]. Lastly, another group used a freeze-drying technique to combine several TME components with synthetic polymers to create a porous microstructure that could support neuroblastoma cell growth [179].
More complex systems can include combinations of natural and artificial scaffolds and bioengineering methods to provide variable structure, flow, and concentration gradients. In neuroblastoma, two studies used microfluidic devices [149,150]. Liu and colleagues incorporated a decellularized ECM and microvascular endothelial cells with a microfluidic device, producing a system that could produce drug concentration gradients [149]. Villasante et al. used a microfluidic device with a collagen/fibrin scaffold and also incorporated endothelial cells [150]. Several studies have used 3D bioprinting [151,152,180,181]. In addition to the previously described studies using gelatin to incorporate endothelial cells [151,152,153], another group used ink made from cellulose nanofibrils, alginate, and single-walled carbon nanotubes to bioprint scaffolds for neuroblastoma cell growth [180]. Quinn et al. printed using tumor cells mixed with gelatin and sodium alginate and showed histology similar to tumors and more resistance to hypoxia and chemotherapy than 2D cell lines. Another group has published two studies [182,183] using β-tricalcium phosphate to bioprint scaffolds to model bone marrow metastases, including the incorporation of mesenchymal stromal cells. While intriguing, these systems increase cost and complexity and reduce the accessibility of these approaches for the broader neuroblastoma community while also limiting experimental design. The utility of these systems to understand neuroblastoma biology remains to be seen.
Figure 1. Comparison of Examples of Preclinical Models for Neuroblastoma Research. An overview of the preclinical models used to study neuroblastoma, including monolayer cultures [58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73], 3D culture systems (cancer cells alone [83,110,111,115,116,117,118,119,120,121,122,123,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139], cancer cells co-cultured with other cell types such as immune cells and fibroblasts [112,113,114,124,140,141,142,143,144,145,146,147,148,149,150,151,152,153], and cancer cells grown in a 3D ECM scaffold [148,149,150,151,152,153,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183]), and in vivo models (GEMMs and PDXs) [78,79,80,81,82,83,84,85,86,92,93,94,95,96,97,98,99,100,101,102]. Each model is assessed based on key attributes, including complexity, translational relevance, and ability to recapitulate the tumor microenvironment. This image was created with BioRender (https://biorender.com/ accessed on: 31 March 2025).
Figure 1. Comparison of Examples of Preclinical Models for Neuroblastoma Research. An overview of the preclinical models used to study neuroblastoma, including monolayer cultures [58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73], 3D culture systems (cancer cells alone [83,110,111,115,116,117,118,119,120,121,122,123,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139], cancer cells co-cultured with other cell types such as immune cells and fibroblasts [112,113,114,124,140,141,142,143,144,145,146,147,148,149,150,151,152,153], and cancer cells grown in a 3D ECM scaffold [148,149,150,151,152,153,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183]), and in vivo models (GEMMs and PDXs) [78,79,80,81,82,83,84,85,86,92,93,94,95,96,97,98,99,100,101,102]. Each model is assessed based on key attributes, including complexity, translational relevance, and ability to recapitulate the tumor microenvironment. This image was created with BioRender (https://biorender.com/ accessed on: 31 March 2025).
Organoids 04 00010 g001

4. Limitations

Survival for patients with high-risk neuroblastoma has improved, but therapy is highly toxic and nearly half of patients do not survive [184]. Further advances have been hindered by in vitro models that do not reflect the tumor’s heterogeneity and its interactions with the microenvironment and in vivo models that are inflexible and difficult to use in substantial throughput. As a result, preclinical studies too often fail to predict clinical outcomes, hindering the development of effective therapies. Significant progress has been made in developing 3D models that mimic native tumors, enhancing the physiological relevance of cell-based biological and therapeutic investigations [185,186]. As we have discussed, these types of models are being adopted in neuroblastoma. It remains to be seen if these will be able to capture the heterogeneity of human tumors sufficiently to improve the success of drug development.
Creating 3D cultures for cancer, in general, presents several challenges. Serum-free media is often supplemented with more expensive alternatives [187]. Similarly, while extracellular scaffolds may improve model accuracy, they also increase the cost and complexity of the technique. Matrigel, the most commonly used matrix for 3D culture, includes growth factors in unpredictable and uncontrollable amounts, potentially influencing research results in both beneficial and detrimental ways [188]. Using 3D culture is generally more time-consuming than 2D models, often requiring increased monitoring and manipulation [189]. Due to the diverse range of 3D culture techniques, even within the same tissue or cancer type, achieving consistency remains a challenge. Analysis of cells is more difficult as well, as 3D cultures can require proteolytic degradation to separate the cells. In some cases, 3D methods have lower efficiency, shorter lifespans, and reduced replicability when compared to 2D systems [190]. Using cancer cell lines to create a 3D culture is convenient and can help in reproducibility, but this can reduce the fidelity of the model if culture in 3D does not restore the original cancer phenotype due to adaptations from prolonged 2D culture. Using freshly dissociated tumor tissue decreases accessibility and throughput, and extensive expansion may also result in further deviation from the features of the original tumor. Additionally, intra- and inter-technique variability in the size of 3D structures can impact the consistency and uniformity of the models [6,191,192]. Lastly, current models suitable for high-throughput screening are largely limited to cancer cells and a single additional cell type, such as fibroblasts [193], which may miss important cell–cell-drug interactions.
Despite these challenges, several groups have retrospectively demonstrated that drug sensitivity profiling in patient-derived 3D cultures correlates with patient response in adult cancers [41,44,46], prompting the exploration of the use of such an approach for the choice of therapy in patients [45,194,195]. These studies demonstrate the feasibility of using 3D culture to drive clinical decision making. However, additional challenges remain, including incomplete success in generating 3D cultures, prolonged time necessary to generate response profiles, and limitations in the number of drugs that can be tested [45,194,196]. Such challenges will need to be overcome to allow such testing to be adopted more universally.
While not as advanced as in some epithelial cancers, significant progress has been made in culturing neuroblastoma cells in 3D. These studies have performed drug screens and tested combination therapies, analyzed ECM tumor interactions, probed tumor-immune and tumor-endothelial cell interactions, and performed transcriptomic and proteomic analyses, as discussed in this review. Figure 2 summarizes some of these diverse applications. Despite these successes, many of the same challenges found with other tumors are also present for neuroblastoma. These include increased costs of media and ECM components, more time-intensive labor needs, and variability across models, conditions, and sphere sizes. Direct comparisons between the fidelity of cell line-derived and direct tumor-derived models are lacking and would be helpful to determine if less accessible tumor-derived models are necessary. There are also some more specific challenges in neuroblastoma, particularly the difficulty in growing primary murine tumors in 3D. Successful techniques have now been developed for murine tumors and should be more widely adapted. The development of direct clinical applications of patient-derived 3D culture is further behind the more common adult cancer types, in part due to the challenges inherent to the study of rare tumor types. Efforts have thus largely focused on using organoids to link genomic or transcriptomic features to drug response, rather than direct testing of patient-derived samples [130]. While direct drug-response testing may not be feasible unless histology-agnostic protocols are developed, ongoing research on 3D culture in neuroblastoma still holds great potential for advancing our understanding and treatment of neuroblastoma.

5. Conclusions and Future Directions

In this review, we characterize and organize the methods that have been used in 3D culture in neuroblastoma research. We identified and defined three major groups and methodologies for culturing, maintaining, and utilizing neuroblastoma 3D culture: (1) 3D culture of neuroblastoma cells without other cell types or an extracellular matrix, (2) 3D culture of neuroblastoma cells grown with other cell types, and (3) 3D culture of neuroblastoma cells grown with matrix support. We also distinguish whether these models were applied in murine or human models. We chose to use this classification to highlight the similarities and distinctions among these culture methods and avoided the words typically used (spheroids/tumorspheres/organoids), because in neuroblastoma they have been used to describe highly similar techniques without clear differences. While such terms are useful and convenient, we hope that this methodology-driven classification provides a more useful comparison of these models and their applications.
As in other cancer types, 3D culture systems in neuroblastoma have been used to make important advances in a tractable system that closely mimics in vivo conditions. These neuroblastoma models have been applied to investigate tumor heterogeneity, tumor–host cell interaction (immune cells, CAFs, and endothelial cells), metastasis, and tumor–ECM interactions. Despite these successes, there remains significant potential for further research and refinement. Future studies could focus more on integrating patient-derived immune cells to better understand immunotherapy responses. They should also aim to integrate a wider array of cell types and microenvironmental components, including diverse combinations of immune and vascular cells, to more faithfully replicate the in vivo tumor landscape. At the same time, efficiency, simplicity, and miniaturization are also important in developing better high-throughput platforms for drug screening and enabling personalized medicine approaches. Lastly, expanding the use of mouse-derived 3D models and advancing genetic engineering techniques will allow researchers to better understand anti-tumor immunity and investigate specific genetic and molecular mechanisms important in neuroblastoma progression and resistance.
As the field progresses, we expect that 3D culture technologies will become even more integral to neuroblastoma research. We expect that technological advances will enable discoveries that could lead to more effective therapies and improved outcomes for patients.

Author Contributions

Conceptualization, P.J. and A.J.W.; writing-original draft preparation, P.J.; writing-review and editing, P.J. and A.J.W.; funding acquisition, A.J.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded in part by grants from the NCI (K08 CA266914) and the Department of Defense (W81XWH2210474) and by a Hyundai Hope on Wheels Hope Scholar Award.

Data Availability Statement

No new data were created.

Acknowledgments

The artificial intelligence writing program Grammarly was used to assist in correcting English wording in the original draft preparation of the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
2DTwo-Dimensional
3DThree-Dimensional
CDXCell line-Derived Xenograft
CSTNChildhood Solid Tumor Network
DBHDopamine β-Hydroxylase
ECMExtracellular Matrix
gelMAGelatin Methacryloyl
GEMMGenetically Engineered Mouse Models
HDACHistone Deacetylase
HUVECHuman Umbilical Vein Endothelial Cells
KSPKinesin Spindle Protein
NKNatural Killer
NSGNOD-Scid-Gamma
PANPolyacrylonitrile
PBMC Peripheral Blood Mononuclear Cells
PDXPatient-Derived Xenografts
PEGPolyethylene Glycol
PIVOTPediatric Preclinical In Vivo Testing Consortium
PPTCPediatric Preclinical Testing Consortium
RISTRapamycin Irinotecan Sunitinib/Dasatinib Temozolomide
THTyrosine Hydroxylase

References

  1. Harrison, R.G. The outgrowth of the nerve fiber as a mode of protoplasmic movement. J. Exp. Zool. 1910, 9, 787–846. [Google Scholar] [CrossRef]
  2. Keshishian, H. Ross Harrison’s ‘The outgrowth of the nerve fiber as a mode of protoplasmic movement’. J. Exp. Zool. A Comp. Exp. Biol. 2004, 301A, 201–203. [Google Scholar] [CrossRef] [PubMed]
  3. Gey, G.O.; Coffman, W.D.; Kubicek, M.T. Tissue culture studies of the proliferative capacity of cervical carcinoma and normal epithelium. Cancer Res. 1952, 12, 264–265. [Google Scholar]
  4. Skloot, R. The Immortal Life of Henrietta Lacks; Crown Publishers: New York, NY, USA, 2010. [Google Scholar]
  5. Scudiero, D.A.; Shoemaker, R.H.; Paull, K.D.; Monks, A.; Tierney, S.; Nofziger, T.H.; Currens, M.J.; Seniff, D.; Boyd, M.R. Evaluation of a soluble tetrazolium/formazan assay for cell growth and drug sensitivity in culture using human and other tumor cell lines. Cancer Res. 1988, 48, 4827–4833. [Google Scholar]
  6. Kapałczyńska, M.; Kolenda, T.; Przybyła, W.; Zajączkowska, M.; Teresiak, A.; Filas, V.; Ibbs, M.; Bliźniak, R.; Łuczewski, Ł.; Lamperska, K. 2D and 3D cell cultures-a comparison of different types of cancer cell cultures. Arch. Med. Sci. Off. AMS 2018, 14, 910–919. [Google Scholar] [CrossRef]
  7. Voskoglou-Nomikos, T.; Pater, J.L.; Seymour, L. Clinical predictive value of the in vitro cell line, human xenograft, and mouse allograft preclinical cancer models. Clin. Cancer Res. 2003, 9, 4227–4239. [Google Scholar]
  8. Kaur, G.; Dufour, J.M. Cell lines: Valuable tools or useless artifacts. Spermatogenesis 2012, 2, 1–5. [Google Scholar] [CrossRef]
  9. Cukierman, E.; Pankov, R.; Yamada, K.M. Cell interactions with three-dimensional matrices. Curr. Opin. Cell Biol. 2002, 14, 633–640. [Google Scholar] [CrossRef]
  10. Godoy, P.; Hewitt, N.J.; Albrecht, U.; Andersen, M.E.; Ansari, N.; Bhattacharya, S.; Bode, J.G.; Bolleyn, J.; Borner, C.; Böttger, J.; et al. Recent advances in 2D and 3D in vitro systems using primary hepatocytes, alternative hepatocyte sources and non-parenchymal liver cells and their use in investigating mechanisms of hepatotoxicity, cell signaling and ADME. Arch. Toxicol. 2013, 87, 1315–1530. [Google Scholar] [CrossRef]
  11. Balamurugan, K.; Poria, D.K.; Sehareen, S.W.; Krishnamurthy, S.; Tang, W.; McKennett, L.; Padmanaban, V.; Czarra, K.; Ewald, A.J.; Ueno, N.T.; et al. Stabilization of E-cadherin adhesions by COX-2/GSK3β signaling is a targetable pathway in metastatic breast cancer. JCI Insight 2023, 8, e156057. [Google Scholar] [CrossRef]
  12. Marconi, G.D.; Porcheri, C.; Trubiani, O.; Mitsiadis, T.A. Three-Dimensional Culture Systems for Dissecting Notch Signalling in Health and Disease. Int. J. Mol. Sci. 2021, 22, 12473. [Google Scholar] [CrossRef] [PubMed]
  13. Ye, F.; Chen, C.; Qin, J.; Liu, J.; Zheng, C. Genetic profiling reveals an alarming rate of cross-contamination among human cell lines used in China. FASEB J. 2015, 29, 4268–4272. [Google Scholar] [CrossRef] [PubMed]
  14. Drexler, H.G.; Dirks, W.G.; Matsuo, Y.; MacLeod, R.A.F. False leukemia–lymphoma cell lines: An update on over 500 cell lines. Leukemia 2003, 17, 416–426. [Google Scholar] [CrossRef]
  15. Ericsson, A.C.; Crim, M.J.; Franklin, C.L. A brief history of animal modeling. Mo. Med. 2013, 110, 201–205. [Google Scholar] [PubMed]
  16. Rous, P. A sarcoma of the fowl transmissible by an agent separable from the tumor cells. J. Exp. Med. 1911, 13, 397–411. [Google Scholar] [CrossRef]
  17. Yamagiwa, K.; Ichikawa, K. Experimental Study of the Pathogenesis of Carcinoma. CA Cancer J. Clin. 1977, 27, 174–181. [Google Scholar] [CrossRef]
  18. Mouse Genome Sequencing Consortium Initial sequencing and comparative analysis of the mouse genome. Nature 2002, 420, 520–562. [CrossRef]
  19. Rodrigues, J.; Heinrich, M.A.; Teixeira, L.M.; Prakash, J. 3D In Vitro Model (R)evolution: Unveiling Tumor–Stroma Interactions. Trends Cancer 2021, 7, 249–264. [Google Scholar] [CrossRef]
  20. Corallo, D.; Frabetti, S.; Candini, O.; Gregianin, E.; Dominici, M.; Fischer, H.; Aveic, S. Emerging Neuroblastoma 3D In Vitro Models for Pre-Clinical Assessments. Front. Immunol. 2020, 11, 584214. [Google Scholar] [CrossRef]
  21. Nanki, Y.; Chiyoda, T.; Hirasawa, A.; Ookubo, A.; Itoh, M.; Ueno, M.; Akahane, T.; Kameyama, K.; Yamagami, W.; Kataoka, F.; et al. Patient-derived ovarian cancer organoids capture the genomic profiles of primary tumours applicable for drug sensitivity and resistance testing. Sci. Rep. 2020, 10, 12581. [Google Scholar] [CrossRef]
  22. Jacob, F.; Salinas, R.D.; Zhang, D.Y.; Nguyen, P.T.T.; Schnoll, J.G.; Wong, S.Z.H.; Thokala, R.; Sheikh, S.; Saxena, D.; Prokop, S.; et al. A Patient-Derived Glioblastoma Organoid Model and Biobank Recapitulates Inter- and Intra-tumoral Heterogeneity. Cell 2020, 180, 188–204.e22. [Google Scholar] [CrossRef] [PubMed]
  23. Sachs, N.; De Ligt, J.; Kopper, O.; Gogola, E.; Bounova, G.; Weeber, F.; Balgobind, A.V.; Wind, K.; Gracanin, A.; Begthel, H.; et al. A Living Biobank of Breast Cancer Organoids Captures Disease Heterogeneity. Cell 2018, 172, 373–386.e10. [Google Scholar] [CrossRef] [PubMed]
  24. Gao, D.; Vela, I.; Sboner, A.; Iaquinta, P.J.; Karthaus, W.R.; Gopalan, A.; Dowling, C.; Wanjala, J.N.; Undvall, E.A.; Arora, V.K.; et al. Organoid Cultures Derived from Patients with Advanced Prostate Cancer. Cell 2014, 159, 176–187. [Google Scholar] [CrossRef]
  25. Crespo, M.; Vilar, E.; Tsai, S.-Y.; Chang, K.; Amin, S.; Srinivasan, T.; Zhang, T.; Pipalia, N.H.; Chen, H.J.; Witherspoon, M.; et al. Colonic organoids derived from human induced pluripotent stem cells for modeling colorectal cancer and drug testing. Nat. Med. 2017, 23, 878–884. [Google Scholar] [CrossRef] [PubMed]
  26. Boj, S.F.; Hwang, C.-I.; Baker, L.A.; Chio, I.I.C.; Engle, D.D.; Corbo, V.; Jager, M.; Ponz-Sarvise, M.; Tiriac, H.; Spector, M.S.; et al. Organoid Models of Human and Mouse Ductal Pancreatic Cancer. Cell 2015, 160, 324–338. [Google Scholar] [CrossRef]
  27. Hwang, H.J.; Oh, M.-S.; Lee, D.W.; Kuh, H.-J. Multiplex quantitative analysis of stroma-mediated cancer cell invasion, matrix remodeling, and drug response in a 3D co-culture model of pancreatic tumor spheroids and stellate cells. J. Exp. Clin. Cancer Res. 2019, 38, 258. [Google Scholar] [CrossRef]
  28. Balážová, K.; Clevers, H.; Dost, A.F. The role of macrophages in non-small cell lung cancer and advancements in 3D co-cultures. eLife 2023, 12, e82998. [Google Scholar] [CrossRef]
  29. Ravi, M.; Ramesh, A.; Pattabhi, A. Contributions of 3D Cell Cultures for Cancer Research. J. Cell Physiol. 2017, 232, 2679–2697. [Google Scholar] [CrossRef]
  30. Abbas, Z.N.; Al-Saffar, A.Z.; Jasim, S.M.; Sulaiman, G.M. Comparative analysis between 2D and 3D colorectal cancer culture models for insights into cellular morphological and transcriptomic variations. Sci. Rep. 2023, 13, 18380. [Google Scholar] [CrossRef]
  31. Lv, D.; Hu, Z.; Lu, L.; Lu, H.; Xu, X. Three-dimensional cell culture: A powerful tool in tumor research and drug discovery. Oncol. Lett. 2017, 14, 6999–7010. [Google Scholar] [CrossRef]
  32. Zhao, H.; Jiang, E.; Shang, Z. 3D Co-culture of Cancer-Associated Fibroblast with Oral Cancer Organoids. J. Dent. Res. 2021, 100, 201–208. [Google Scholar] [CrossRef] [PubMed]
  33. Al Hrout, A.; Cervantes-Gracia, K.; Chahwan, R.; Amin, A. Modelling liver cancer microenvironment using a novel 3D culture system. Sci. Rep. 2022, 12, 8003. [Google Scholar] [CrossRef] [PubMed]
  34. Subtil, B.; Iyer, K.K.; Poel, D.; Bakkerus, L.; Gorris, M.A.J.; Escalona, J.C.; Dries, K.V.D.; Cambi, A.; Verheul, H.M.W.; De Vries, I.J.M.; et al. Dendritic cell phenotype and function in a 3D co-culture model of patient-derived metastatic colorectal cancer organoids. Front. Immunol. 2023, 14, 1105244. [Google Scholar] [CrossRef] [PubMed]
  35. Neal, J.T.; Li, X.; Zhu, J.; Giangarra, V.; Grzeskowiak, C.L.; Ju, J.; Liu, I.H.; Chiou, S.-H.; Salahudeen, A.A.; Smith, A.R.; et al. Organoid Modeling of the Tumor Immune Microenvironment. Cell 2018, 175, 1972–1988.e16. [Google Scholar] [CrossRef]
  36. Matano, M.; Date, S.; Shimokawa, M.; Takano, A.; Fujii, M.; Ohta, Y.; Watanabe, T.; Kanai, T.; Sato, T. Modeling colorectal cancer using CRISPR-Cas9–mediated engineering of human intestinal organoids. Nat. Med. 2015, 21, 256–262. [Google Scholar] [CrossRef]
  37. Lee, H.-K.; Noh, M.H.; Hong, S.-W.; Kim, S.-M.; Kim, S.H.; Kim, Y.S.; Broaddus, V.C.; Hur, D.Y. Erlotinib Activates Different Cell Death Pathways in EGFR-mutant Lung Cancer Cells Grown in 3D Versus 2D Culture Systems. Anticancer Res. 2021, 41, 1261–1269. [Google Scholar] [CrossRef]
  38. Berry, M.A.; Bland, A.R.; Major, G.S.; Ashton, J.C. Development of an ALK-positive Non-Small-Cell Lung Cancer in Vitro Tumor 3D Culture Model for Therapeutic Screening. J. Histochem. Cytochem. 2025, 73, 63–79. [Google Scholar] [CrossRef]
  39. Świerczewska, M.; Sterzyńska, K.; Ruciński, M.; Andrzejewska, M.; Nowicki, M.; Januchowski, R. The response and resistance to drugs in ovarian cancer cell lines in 2D monolayers and 3D spheroids. Biomed. Pharmacother. 2023, 165, 115152. [Google Scholar] [CrossRef]
  40. Tadić, V.; Zhang, W.; Brozovic, A. The high-grade serous ovarian cancer metastasis and chemoresistance in 3D models. Biochim. Biophys. Acta BBA-Rev. Cancer 2024, 1879, 189052. [Google Scholar] [CrossRef]
  41. Vlachogiannis, G.; Hedayat, S.; Vatsiou, A.; Jamin, Y.; Fernández-Mateos, J.; Khan, K.; Lampis, A.; Eason, K.; Huntingford, I.; Burke, R.; et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science 2018, 359, 920–926. [Google Scholar] [CrossRef]
  42. Dekkers, J.F.; Whittle, J.R.; Vaillant, F.; Chen, H.-R.; Dawson, C.; Liu, K.; Geurts, M.H.; Herold, M.J.; Clevers, H.; Lindeman, G.J.; et al. Modeling Breast Cancer Using CRISPR-Cas9-Mediated Engineering of Human Breast Organoids. J. Natl. Cancer Inst. 2020, 112, 540–544. [Google Scholar] [CrossRef] [PubMed]
  43. Vaishnavi, A.; Juan, J.; Jacob, M.; Stehn, C.; Gardner, E.E.; Scherzer, M.T.; Schuman, S.; Van Veen, J.E.; Murphy, B.; Hackett, C.S.; et al. Transposon Mutagenesis Reveals RBMS3 Silencing as a Promoter of Malignant Progression of BRAFV600E-Driven Lung Tumorigenesis. Cancer Res. 2022, 82, 4261–4273. [Google Scholar] [CrossRef]
  44. Tiriac, H.; Belleau, P.; Engle, D.D.; Plenker, D.; Deschênes, A.; Somerville, T.D.D.; Froeling, F.E.M.; Burkhart, R.A.; Denroche, R.E.; Jang, G.-H.; et al. Organoid Profiling Identifies Common Responders to Chemotherapy in Pancreatic Cancer. Cancer Discov. 2018, 8, 1112–1129. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, H.-M.; Zhang, C.-Y.; Peng, K.-C.; Chen, Z.-X.; Su, J.-W.; Li, Y.-F.; Li, W.-F.; Gao, Q.-Y.; Zhang, S.-L.; Chen, Y.-Q.; et al. Using patient-derived organoids to predict locally advanced or metastatic lung cancer tumor response: A real-world study. Cell Rep. Med. 2023, 4, 100911. [Google Scholar] [CrossRef]
  46. Yao, Y.; Xu, X.; Yang, L.; Zhu, J.; Wan, J.; Shen, L.; Xia, F.; Fu, G.; Deng, Y.; Pan, M.; et al. Patient-Derived Organoids Predict Chemoradiation Responses of Locally Advanced Rectal Cancer. Cell Stem Cell 2020, 26, 17–26.e6. [Google Scholar] [CrossRef]
  47. Herpers, B.; Eppink, B.; James, M.I.; Cortina, C.; Cañellas-Socias, A.; Boj, S.F.; Hernando-Momblona, X.; Glodzik, D.; Roovers, R.C.; van de Wetering, M.; et al. Functional patient-derived organoid screenings identify MCLA-158 as a therapeutic EGFR × LGR5 bispecific antibody with efficacy in epithelial tumors. Nat. Cancer 2022, 3, 418–436. [Google Scholar] [CrossRef]
  48. Fayette, J.; Clatot, F.; Brana, I.; Saada, E.; Van Herpen, C.M.L.-; Mazard, T.; Perez, C.A.; Tabernero, J.; Le Tourneau, C.; Hollebecque, A.; et al. Petosemtamab (MCLA-158) with pembrolizumab as first-line (1L) treatment of recurrent/metastatic (r/m) head and neck squamous cell carcinoma (HNSCC): Phase 2 study. J. Clin. Oncol. 2024, 42 (Suppl. 16), 6014. [Google Scholar] [CrossRef]
  49. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef]
  50. Matthay, K.K.; Maris, J.M.; Schleiermacher, G.; Nakagawara, A.; Mackall, C.L.; Diller, L.; Weiss, W.A. Neuroblastoma. Nat. Rev. Dis. Primer 2016, 2, 16078. [Google Scholar] [CrossRef]
  51. Schleiermacher, G.; Janoueix-Lerosey, I.; Delattre, O. Recent insights into the biology of neuroblastoma. Int. J. Cancer 2014, 135, 2249–2261. [Google Scholar] [CrossRef]
  52. Irwin, M.S.; Naranjo, A.; Zhang, F.F.; Cohn, S.L.; London, W.B.; Gastier-Foster, J.M.; Ramirez, N.C.; Pfau, R.; Reshmi, S.; Wagner, E.; et al. Revised Neuroblastoma Risk Classification System: A Report From the Children’s Oncology Group. J. Clin. Oncol. 2021, 39, 3229–3241. [Google Scholar] [CrossRef] [PubMed]
  53. Morgenstern, D.A.; London, W.B.; Stephens, D.; Volchenboum, S.L.; Simon, T.; Nakagawara, A.; Shimada, H.; Schleiermacher, G.; Matthay, K.K.; Cohn, S.L.; et al. Prognostic significance of pattern and burden of metastatic disease in patients with stage 4 neuroblastoma: A study from the International Neuroblastoma Risk Group database. Eur. J. Cancer 2016, 65, 1–10. [Google Scholar] [CrossRef]
  54. D’Angio, G.; Evans, A.; Koop, C.E. Special pattern of widespread neuroblastoma with a favourable prognosis. Lancet 1971, 297, 1046–1049. [Google Scholar] [CrossRef]
  55. Brodeur, G.M. Spontaneous regression of neuroblastoma. Cell Tissue Res. 2018, 372, 277–286. [Google Scholar] [CrossRef] [PubMed]
  56. Gomez, R.L.; Ibragimova, S.; Ramachandran, R.; Philpott, A.; Ali, F.R. Tumoral heterogeneity in neuroblastoma. Biochim. Biophys. Acta BBA-Rev. Cancer 2022, 1877, 188805. [Google Scholar] [CrossRef]
  57. Murray, M.R.; Stout, A.P. Distinctive Characteristics of the Sympathicoblastoma Cultivated in Vitro: A Method for Prompt Diagnosis. Am. J. Pathol. 1947, 23, 429–441. [Google Scholar]
  58. Carol, J. Thiele Neuroblastoma Cell Lines. In Cell & Molecular Biology Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health; Springer: Berlin/Heidelberg, Germany, 1998; Volume 1, pp. 21–53. [Google Scholar]
  59. Cole, K.A.; Huggins, J.; Laquaglia, M.; Hulderman, C.E.; Russell, M.R.; Bosse, K.; Diskin, S.J.; Attiyeh, E.F.; Sennett, R.; Norris, G.; et al. RNAi screen of the protein kinome identifies checkpoint kinase 1 (CHK1) as a therapeutic target in neuroblastoma. Proc. Natl. Acad. Sci. USA 2011, 108, 3336–3341. [Google Scholar] [CrossRef]
  60. Chen, L.; Alexe, G.; Dharia, N.V.; Ross, L.; Iniguez, A.B.; Conway, A.S.; Wang, E.J.; Veschi, V.; Lam, N.; Qi, J.; et al. CRISPR-Cas9 screen reveals a MYCN-amplified neuroblastoma dependency on EZH2. J. Clin. Investig. 2017, 128, 446–462. [Google Scholar] [CrossRef]
  61. Hahn, C.K.; Ross, K.N.; Warrington, I.M.; Mazitschek, R.; Kanegai, C.M.; Wright, R.D.; Kung, A.L.; Golub, T.R.; Stegmaier, K. Expression-based screening identifies the combination of histone deacetylase inhibitors and retinoids for neuroblastoma differentiation. Proc. Natl. Acad. Sci. USA 2008, 105, 9751–9756. [Google Scholar] [CrossRef]
  62. Cheung, B.B.; Kleynhans, A.; Mittra, R.; Kim, P.Y.; Holien, J.K.; Nagy, Z.; Ciampa, O.C.; Seneviratne, J.A.; Mayoh, C.; Raipuria, M.; et al. A novel combination therapy targeting ubiquitin-specific protease 5 in MYCN-driven neuroblastoma. Oncogene 2021, 40, 2367–2381. [Google Scholar] [CrossRef]
  63. Chakrabarti, L. Reversible adaptive plasticity: A mechanism for neuroblastoma cell heterogeneity and chemo-resistance. Front. Oncol. 2012, 2, 82. [Google Scholar] [CrossRef] [PubMed]
  64. Dagogo-Jack, I.; Shaw, A.T. Tumour heterogeneity and resistance to cancer therapies. Nat. Rev. Clin. Oncol. 2018, 15, 81–94. [Google Scholar] [CrossRef] [PubMed]
  65. Ross, R.A.; Spengler, B.A.; Biedler, J.L. Coordinate morphological and biochemical interconversion of human neuroblastoma cells. J. Natl. Cancer Inst. 1983, 71, 741–747. [Google Scholar]
  66. Corey, J.M.; Gertz, C.C.; Sutton, T.J.; Chen, Q.; Mycek, K.B.; Wang, B.; Martin, A.A.; Johnson, S.L.; Feldman, E.L. Patterning N-type and S-type neuroblastoma cells with Pluronic F108 and ECM proteins. J. Biomed. Mater. Res. A 2010, 93A, 673–686. [Google Scholar] [CrossRef]
  67. Ross, R.A.; Biedler, J.L.; Spengler, B.A. A role for distinct cell types in determining malignancy in human neuroblastoma cell lines and tumors. Cancer Lett. 2003, 197, 35–39. [Google Scholar] [CrossRef]
  68. Boeva, V.; Louis-Brennetot, C.; Peltier, A.; Durand, S.; Pierre-Eugène, C.; Raynal, V.; Etchevers, H.C.; Thomas, S.; Lermine, A.; Daudigeos-Dubus, E.; et al. Heterogeneity of neuroblastoma cell identity defined by transcriptional circuitries. Nat. Genet. 2017, 49, 1408–1413. [Google Scholar] [CrossRef]
  69. Van Groningen, T.; Koster, J.; Valentijn, L.J.; Zwijnenburg, D.A.; Akogul, N.; Hasselt, N.E.; Broekmans, M.; Haneveld, F.; Nowakowska, N.E.; Bras, J.; et al. Neuroblastoma is composed of two super-enhancer-associated differentiation states. Nat. Genet. 2017, 49, 1261–1266. [Google Scholar] [CrossRef]
  70. Durbin, A.D.; Zimmerman, M.W.; Dharia, N.V.; Abraham, B.J.; Iniguez, A.B.; Weichert-Leahey, N.; He, S.; Krill-Burger, J.M.; Root, D.E.; Vazquez, F.; et al. Selective gene dependencies in MYCN-amplified neuroblastoma include the core transcriptional regulatory circuitry. Nat. Genet. 2018, 50, 1240–1246. [Google Scholar] [CrossRef]
  71. Kendsersky, N.M.; Odrobina, M.; Mabe, N.W.; Farrel, A.; Grossmann, L.; Tsang, M.; Groff, D.; Wolpaw, A.J.; Narch, A.; Zammarchi, F.; et al. Lineage dependence of the neuroblastoma surfaceome defines tumor cell state-dependent and -independent immunotherapeutic targets. Neuro-Oncol. 2025, noaf012. [Google Scholar] [CrossRef]
  72. Wolpaw, A.J.; Grossmann, L.D.; Dessau, J.L.; Dong, M.M.; Aaron, B.J.; Brafford, P.A.; Volgina, D.; Pascual-Pasto, G.; Rodriguez-Garcia, A.; Uzun, Y.; et al. Epigenetic state determines inflammatory sensing in neuroblastoma. Proc. Natl. Acad. Sci. USA 2022, 119, e2102358119. [Google Scholar] [CrossRef]
  73. Sengupta, S.; Das, S.; Crespo, A.C.; Cornel, A.M.; Patel, A.G.; Mahadevan, N.R.; Campisi, M.; Ali, A.K.; Sharma, B.; Rowe, J.H.; et al. Mesenchymal and adrenergic cell lineage states in neuroblastoma possess distinct immunogenic phenotypes. Nat. Cancer 2022, 3, 1228–1246. [Google Scholar] [CrossRef] [PubMed]
  74. Patel, A.G.; Ashenberg, O.; Collins, N.B.; Segerstolpe, Å.; Jiang, S.; Slyper, M.; Huang, X.; Caraccio, C.; Jin, H.; Sheppard, H.; et al. A spatial cell atlas of neuroblastoma reveals developmental, epigenetic and spatial axis of tumor heterogeneity. BioRxiv 2024. [Google Scholar] [CrossRef]
  75. Gartlgruber, M.; Sharma, A.K.; Quintero, A.; Dreidax, D.; Jansky, S.; Park, Y.-G.; Kreth, S.; Meder, J.; Doncevic, D.; Saary, P.; et al. Super enhancers define regulatory subtypes and cell identity in neuroblastoma. Nat. Cancer 2020, 2, 114–128. [Google Scholar] [CrossRef]
  76. Nolan, J.C.; Frawley, T.; Tighe, J.; Soh, H.; Curtin, C.; Piskareva, O. Preclinical models for neuroblastoma: Advances and challenges. Cancer Lett. 2020, 474, 53–62. [Google Scholar] [CrossRef] [PubMed]
  77. Smith, M.A.; Houghton, P.J.; Lock, R.B.; Maris, J.M.; Gorlick, R.; Kurmasheva, R.T.; Li, X.-N.; Teicher, B.A.; Chuang, J.H.; Dela Cruz, F.S.; et al. Lessons learned from 20 years of preclinical testing in pediatric cancers. Pharmacol. Ther. 2024, 264, 108742. [Google Scholar] [CrossRef]
  78. Teitz, T.; Stanke, J.J.; Federico, S.; Bradley, C.L.; Brennan, R.; Zhang, J.; Johnson, M.D.; Sedlacik, J.; Inoue, M.; Zhang, Z.M.; et al. Preclinical Models for Neuroblastoma: Establishing a Baseline for Treatment. PLoS ONE 2011, 6, e19133. [Google Scholar] [CrossRef]
  79. Stewart, E.; Shelat, A.; Bradley, C.; Chen, X.; Federico, S.; Thiagarajan, S.; Shirinifard, A.; Bahrami, A.; Pappo, A.; Qu, C.; et al. Development and characterization of a human orthotopic neuroblastoma xenograft. Dev. Biol. 2015, 407, 344–355. [Google Scholar] [CrossRef]
  80. Khanna, C.; Jaboin, J.J.; Drakos, E.; Tsokos, M.; Thiele, C.J. Biologically relevant orthotopic neuroblastoma xenograft models: Primary adrenal tumor growth and spontaneous distant metastasis. In Vivo Athens Greece 2002, 16, 77–85. [Google Scholar]
  81. Patterson, D.M.; Shohet, J.M.; Kim, E.S. Preclinical Models of Pediatric Solid Tumors (Neuroblastoma) and Their Use in Drug Discovery. Curr. Protoc. Pharmacol. 2011, 52, 14.17.1–14.17.18. [Google Scholar] [CrossRef]
  82. Braekeveldt, N.; Bexell, D. Patient-derived xenografts as preclinical neuroblastoma models. Cell Tissue Res. 2018, 372, 233–243. [Google Scholar] [CrossRef]
  83. Braekeveldt, N.; Wigerup, C.; Gisselsson, D.; Mohlin, S.; Merselius, M.; Beckman, S.; Jonson, T.; Börjesson, A.; Backman, T.; Tadeo, I.; et al. Neuroblastoma patient-derived orthotopic xenografts retain metastatic patterns and geno- and phenotypes of patient tumours. Int. J. Cancer 2015, 136, E252–E261. [Google Scholar] [CrossRef] [PubMed]
  84. Rokita, J.L.; Rathi, K.S.; Cardenas, M.F.; Upton, K.A.; Jayaseelan, J.; Cross, K.L.; Pfeil, J.; Egolf, L.E.; Way, G.P.; Farrel, A.; et al. Genomic Profiling of Childhood Tumor Patient-Derived Xenograft Models to Enable Rational Clinical Trial Design. Cell Rep. 2019, 29, 1675–1689.e9. [Google Scholar] [CrossRef] [PubMed]
  85. Stewart, E.; Federico, S.M.; Chen, X.; Shelat, A.A.; Bradley, C.; Gordon, B.; Karlstrom, A.; Twarog, N.R.; Clay, M.R.; Bahrami, A.; et al. Orthotopic patient-derived xenografts of paediatric solid tumours. Nature 2017, 549, 96–100. [Google Scholar] [CrossRef]
  86. The Childhood Cancer Repository. Available online: https://www.cccells.org/xenografts.php (accessed on 30 April 2025).
  87. Scheerlinck, J.-P.Y. Cytokine Species-Specificity and Humanized Mice. In Humanized Mice for HIV Research; Poluektova, L.Y., Garcia, J.V., Koyanagi, Y., Manz, M.G., Tager, A.M., Eds.; Springer: New York, NY, USA, 2014; pp. 93–108. [Google Scholar] [CrossRef]
  88. Nguyen, R.; Patel, A.G.; Griffiths, L.M.; Dapper, J.; Stewart, E.A.; Houston, J.; Johnson, M.; Akers, W.J.; Furman, W.L.; Dyer, M.A. Next-generation humanized patient-derived xenograft mouse model for pre-clinical antibody studies in neuroblastoma. Cancer Immunol. Immunother. 2021, 70, 721–732. [Google Scholar] [CrossRef]
  89. Huang, M.; Fang, W.; Farrel, A.; Li, L.; Chronopoulos, A.; Nasholm, N.; Cheng, B.; Zheng, T.; Yoda, H.; Barata, M.J.; et al. ALK upregulates POSTN and WNT signaling to drive neuroblastoma. Cell Rep. 2024, 43, 113927. [Google Scholar] [CrossRef]
  90. Cohen, M.A.; Zhang, S.; Sengupta, S.; Ma, H.; Bell, G.W.; Horton, B.; Sharma, B.; George, R.E.; Spranger, S.; Jaenisch, R. Formation of Human Neuroblastoma in Mouse-Human Neural Crest Chimeras. Cell Stem Cell 2020, 26, 579–592.e6. [Google Scholar] [CrossRef]
  91. Guerin, M.V.; Finisguerra, V.; Van Den Eynde, B.J.; Bercovici, N.; Trautmann, A. Preclinical murine tumor models: A structural and functional perspective. eLife 2020, 9, e50740. [Google Scholar] [CrossRef]
  92. Chesler, L.; Weiss, W.A. Genetically engineered murine models--contribution to our understanding of the genetics, molecular pathology and therapeutic targeting of neuroblastoma. Semin. Cancer Biol. 2011, 21, 245–255. [Google Scholar] [CrossRef]
  93. Weiss, W.A. Targeted expression of MYCN causes neuroblastoma in transgenic mice. EMBO J. 1997, 16, 2985–2995. [Google Scholar] [CrossRef]
  94. McNerney, K.; Karageorgos, S.; Ferry, G.; Wolpaw, A.; Burudpakdee, C.; Khurana, P.; Toland, C.; Vemu, R.; Vu, A.; Hogarty, M.; et al. TH-MYCN tumors, but not tumor-derived cell lines, are adrenergic lineage, GD2+, and responsive to anti-GD2 antibody therapy. OncoImmunology 2022, 11, 2075204. [Google Scholar] [CrossRef]
  95. Yogev, O.; Barker, K.; Sikka, A.; Almeida, G.S.; Hallsworth, A.; Smith, L.M.; Jamin, Y.; Ruddle, R.; Koers, A.; Webber, H.T.; et al. p53 Loss in MYC-Driven Neuroblastoma Leads to Metabolic Adaptations Supporting Radioresistance. Cancer Res. 2016, 76, 3025–3035. [Google Scholar] [CrossRef] [PubMed]
  96. Chesler, L.; Goldenberg, D.D.; Collins, R.; Grimmer, M.; Kim, G.E.; Tihan, T.; Nguyen, K.; Yakovenko, S.; Matthay, K.K.; Weiss, W.A. Chemotherapy-Induced Apoptosis in a Transgenic Model of Neuroblastoma Proceeds Through p53 Induction. Neoplasia 2008, 10, 1268–1274, IN33–IN34. [Google Scholar] [CrossRef]
  97. Berry, T.; Luther, W.; Bhatnagar, N.; Jamin, Y.; Poon, E.; Sanda, T.; Pei, D.; Sharma, B.; Vetharoy, W.R.; Hallsworth, A.; et al. The ALKF1174L Mutation Potentiates the Oncogenic Activity of MYCN in Neuroblastoma. Cancer Cell 2012, 22, 117–130. [Google Scholar] [CrossRef] [PubMed]
  98. Teitz, T.; Inoue, M.; Valentine, M.B.; Zhu, K.; Rehg, J.E.; Zhao, W.; Finkelstein, D.; Wang, Y.-D.; Johnson, M.D.; Calabrese, C.; et al. Th-MYCN Mice with Caspase-8 Deficiency Develop Advanced Neuroblastoma with Bone Marrow Metastasis. Cancer Res. 2013, 73, 4086–4097. [Google Scholar] [CrossRef]
  99. Iwakura, H.; Ariyasu, H.; Kanamoto, N.; Hosoda, K.; Nakao, K.; Kangawa, K.; Akamizu, T. Establishment of a novel neuroblastoma mouse model. Int. J. Oncol. 2008, 33, 1195–1199. [Google Scholar] [CrossRef] [PubMed]
  100. Hadjidaniel, M.D.; Muthugounder, S.; Hung, L.T.; Sheard, M.A.; Shirinbak, S.; Chan, R.Y.; Nakata, R.; Borriello, L.; Malvar, J.; Kennedy, R.J.; et al. Tumor-associated macrophages promote neuroblastoma via STAT3 phosphorylation and up-regulation of c-MYC. Oncotarget 2017, 8, 91516–91529. [Google Scholar] [CrossRef]
  101. Althoff, K.; Beckers, A.; Bell, E.; Nortmeyer, M.; Thor, T.; Sprüssel, A.; Lindner, S.; De Preter, K.; Florin, A.; Heukamp, L.C.; et al. A Cre-conditional MYCN-driven neuroblastoma mouse model as an improved tool for preclinical studies. Oncogene 2015, 34, 3357–3368. [Google Scholar] [CrossRef]
  102. Molenaar, J.J.; Domingo-Fernández, R.; Ebus, M.E.; Lindner, S.; Koster, J.; Drabek, K.; Mestdagh, P.; Van Sluis, P.; Valentijn, L.J.; Van Nes, J.; et al. LIN28B induces neuroblastoma and enhances MYCN levels via let-7 suppression. Nat. Genet. 2012, 44, 1199–1206. [Google Scholar] [CrossRef]
  103. Ornell, K.J.; Coburn, J.M. Developing preclinical models of neuroblastoma: Driving therapeutic testing. BMC Biomed. Eng. 2019, 1, 33. [Google Scholar] [CrossRef]
  104. Kroesen, M.; Brok, I.C.; Reijnen, D.; Van Hout-Kuijer, M.A.; Zeelenberg, I.S.; Den Brok, M.H.; Hoogerbrugge, P.M.; Adema, G.J. Intra-adrenal murine TH-MYCN neuroblastoma tumors grow more aggressive and exhibit a distinct tumor microenvironment relative to their subcutaneous equivalents. Cancer Immunol. Immunother. 2015, 64, 563–572. [Google Scholar] [CrossRef]
  105. Yuhas, J.M.; Li, A.P.; Martinez, A.O.; Ladman, A.J. A simplified method for production and growth of multicellular tumor spheroids. Cancer Res. 1977, 37, 3639–3643. [Google Scholar] [PubMed]
  106. Hofmann, S.; Cohen-Harazi, R.; Maizels, Y.; Koman, I. Patient-derived tumor spheroid cultures as a promising tool to assist personalized therapeutic decisions in breast cancer. Transl. Cancer Res. 2022, 11, 134–147. [Google Scholar] [CrossRef]
  107. Tang, J.; Shi, J.; Liu, J. Editorial: Advances in 3D cell culture for drug screening and toxicology evaluation. Front. Bioeng. Biotechnol. 2023, 11, 1266506. [Google Scholar] [CrossRef] [PubMed]
  108. Yamada, K.M.; Cukierman, E. Modeling Tissue Morphogenesis and Cancer in 3D. Cell 2007, 130, 601–610. [Google Scholar] [CrossRef] [PubMed]
  109. Drost, J.; Clevers, H. Organoids in cancer research. Nat. Rev. Cancer 2018, 18, 407–418. [Google Scholar] [CrossRef]
  110. Weichselbaum, R.R.; Little, J.B.; Tomkinson, K.; Evans, S.; Yuhas, J. Repair of fractionated radiation in plateau phase cultures of human tumor cells and human multicellular tumor spheroids. Radiother. Oncol. 1984, 2, 41–47. [Google Scholar] [CrossRef]
  111. Bate-Eya, L.T.; Ebus, M.E.; Koster, J.; Den Hartog, I.J.M.; Zwijnenburg, D.A.; Schild, L.; Van Der Ploeg, I.; Dolman, M.E.M.; Caron, H.N.; Versteeg, R.; et al. Newly-derived neuroblastoma cell lines propagated in serum-free media recapitulate the genotype and phenotype of primary neuroblastoma tumours. Eur. J. Cancer 2014, 50, 628–637. [Google Scholar] [CrossRef]
  112. Strijker, J.G.M.; Pscheid, R.; Drent, E.; Van Der Hoek, J.J.F.; Koopmans, B.; Ober, K.; Van Hooff, S.R.; Kholosy, W.M.; Cornel, A.M.; Coomans, C.; et al. αβ-T Cells Engineered to Express γδ-T Cell Receptors Can Kill Neuroblastoma Organoids Independent of MHC-I Expression. J. Pers. Med. 2021, 11, 923. [Google Scholar] [CrossRef]
  113. Cornel, A.M.; Dunnebach, E.; Hofman, D.A.; Das, S.; Sengupta, S.; van den Ham, F.; Wienke, J.; Strijker, J.G.M.; van den Beemt, D.A.M.H.; Essing, A.H.W.; et al. Epigenetic modulation of neuroblastoma enhances T cell and NK cell immunogenicity by inducing a tumor-cell lineage switch. J. Immunother. Cancer 2022, 10, e005002. [Google Scholar] [CrossRef]
  114. Chan, C.; Stip, M.; Nederend, M.; Jansen, M.; Passchier, E.; Van Den Ham, F.; Wienke, J.; Van Tetering, G.; Leusen, J. Enhancing IgA-mediated neutrophil cytotoxicity against neuroblastoma by CD47 blockade. J. Immunother. Cancer 2024, 12, e008478. [Google Scholar] [CrossRef]
  115. Sundaramoorthy, S.; Colombo, D.F.; Sanalkumar, R.; Broye, L.; Balmas Bourloud, K.; Boulay, G.; Cironi, L.; Stamenkovic, I.; Renella, R.; Kuttler, F.; et al. Preclinical spheroid models identify BMX as a therapeutic target for metastatic MYCN nonamplified neuroblastoma. JCI Insight 2024, 9, e169647. [Google Scholar] [CrossRef] [PubMed]
  116. Zhang, H.; Kim, H.; Yuan, T.; Zhang, Z.; Kaul, S.C.; Wadhwa, R. Molecular Characterization of Cancer Preventive and Therapeutic Potential of Three Antistress Compounds, Triethylene Glycol, Withanone, and Withaferin A. Int. J. Mol. Sci. 2025, 26, 493. [Google Scholar] [CrossRef] [PubMed]
  117. Challagundla, K.B.; Pathania, A.S.; Chava, H.; Kantem, N.M.; Dronadula, V.M.; Coulter, D.W.; Clarke, M. FOXJ3, a novel tumor suppressor in neuroblastoma. Mol. Ther. Oncol. 2025, 33, 200914. [Google Scholar] [CrossRef] [PubMed]
  118. Brignole, C.; Calarco, E.; Bensa, V.; Giusto, E.; Perri, P.; Ciampi, E.; Corrias, M.V.; Astigiano, S.; Cilli, M.; Loo, D.; et al. Antitumor activity of the investigational B7-H3 antibody-drug conjugate, vobramitamab duocarmazine, in preclinical models of neuroblastoma. J. Immunother. Cancer 2023, 11, e007174. [Google Scholar] [CrossRef]
  119. Lukoseviciute, M.; Need, E.; Holzhauser, S.; Dalianis, T.; Kostopoulou, O.N. Combined targeted therapy with PI3K and CDK4/6, or FGFR inhibitors show synergistic effects in a neuroblastoma spheroid culture model. Biomed. Pharmacother. 2024, 177, 116993. [Google Scholar] [CrossRef]
  120. Chilamakuri, R.; Agarwal, S. Repurposing of c-MET Inhibitor Tivantinib Inhibits Pediatric Neuroblastoma Cellular Growth. Pharmaceuticals 2024, 17, 1350. [Google Scholar] [CrossRef]
  121. Chilamakuri, R.; Rouse, D.C.; Yu, Y.; Kabir, A.S.; Muth, A.; Yang, J.; Lipton, J.M.; Agarwal, S. BX-795 inhibits neuroblastoma growth and enhances sensitivity towards chemotherapy. Transl. Oncol. 2022, 15, 101272. [Google Scholar] [CrossRef]
  122. Sumer-Bayraktar, Z.; Fife, C.M.; Byrne, F.L.; Kavallaris, M.; Packer, N.H. Membrane glycome is impacted by the cell culturing mode of neuroblastoma cells with differing migration and invasion potential. Glycobiology 2022, 32, 588–599. [Google Scholar] [CrossRef]
  123. Redden, R.A.; Doolin, E.J. Microgravity assay of neuroblastoma: In vitro aggregation kinetics and organoid morphology correlate with MYCN expression. Vitr. Cell. Dev. Biol.-Anim. 2011, 47, 312–317. [Google Scholar] [CrossRef]
  124. Troschke-Meurer, S.; Zumpe, M.; Meißner, L.; Siebert, N.; Grabarczyk, P.; Forkel, H.; Maletzki, C.; Bekeschus, S.; Lode, H.N. Chemotherapeutics Used for High-Risk Neuroblastoma Therapy Improve the Efficacy of Anti-GD2 Antibody Dinutuximab Beta in Preclinical Spheroid Models. Cancers 2023, 15, 904. [Google Scholar] [CrossRef]
  125. Lundsten, S.; Berglund, H.; Jha, P.; Krona, C.; Hariri, M.; Nelander, S.; Lane, D.P.; Nestor, M. p53-Mediated Radiosensitization of 177Lu-DOTATATE in Neuroblastoma Tumor Spheroids. Biomolecules 2021, 11, 1695. [Google Scholar] [CrossRef]
  126. Kaess, C.; Matthes, M.; Gross, J.; Waetzig, R.; Heise, T.; Corbacioglu, S.; Sommer, G. Evaluating the RIST Molecular-Targeted Regimen in a Three-Dimensional Neuroblastoma Spheroid Cell Culture Model. Cancers 2023, 15, 1749. [Google Scholar] [CrossRef]
  127. Zheng, X.; Naiditch, J.; Czurylo, M.; Jie, C.; Lautz, T.; Clark, S.; Jafari, N.; Qiu, Y.; Chu, F.; Madonna, M.B. Differential effect of long-term drug selection with doxorubicin and vorinostat on neuroblastoma cells with cancer stem cell characteristics. Cell Death Dis. 2013, 4, e740. [Google Scholar] [CrossRef]
  128. Mahller, Y.Y.; Williams, J.P.; Baird, W.H.; Mitton, B.; Grossheim, J.; Saeki, Y.; Cancelas, J.A.; Ratner, N.; Cripe, T.P. Neuroblastoma Cell Lines Contain Pluripotent Tumor Initiating Cells That Are Susceptible to a Targeted Oncolytic Virus. PLoS ONE 2009, 4, e4235. [Google Scholar] [CrossRef] [PubMed]
  129. Marayati, R.; Williams, A.P.; Bownes, L.V.; Quinn, C.H.; Stewart, J.E.; Mroczek-Musulman, E.; Atigadda, V.R.; Beierle, E.A. Novel retinoic acid derivative induces differentiation and growth arrest in neuroblastoma. J. Pediatr. Surg. 2020, 55, 1072–1080. [Google Scholar] [CrossRef]
  130. Langenberg, K.P.S.; van Hooff, S.R.; Koopmans, B.; Strijker, J.G.M.; Kholosy, W.M.; Ober, K.; Zwijnenburg, D.A.; van der Hoek, J.J.F.; Keller, K.M.; Vernooij, L.; et al. Exploring high-throughput drug sensitivity testing in neuroblastoma cell lines and patient-derived tumor organoids in the era of precision medicine. Eur. J. Cancer. 2025, 218, 115275. [Google Scholar] [CrossRef]
  131. Rossi, M.; Blasi, P. Multicellular Tumor Spheroids in Nanomedicine Research: A Perspective. Front. Med. Technol. 2022, 4, 909943. [Google Scholar] [CrossRef]
  132. Coulon, A.; Flahaut, M.; Mühlethaler-Mottet, A.; Meier, R.; Liberman, J.; Balmas-Bourloud, K.; Nardou, K.; Yan, P.; Tercier, S.; Joseph, J.-M.; et al. Functional Sphere Profiling Reveals the Complexity of Neuroblastoma Tumor-Initiating Cell Model. Neoplasia 2011, 13, 991–1004, IN30. [Google Scholar] [CrossRef] [PubMed]
  133. Besançon, O.G.; Tytgat, G.A.M.; Meinsma, R.; Leen, R.; Hoebink, J.; Kalayda, G.V.; Jaehde, U.; Caron, H.N.; Van Kuilenburg, A.B.P. Synergistic interaction between cisplatin and gemcitabine in neuroblastoma cell lines and multicellular tumor spheroids. Cancer Lett. 2012, 319, 23–30. [Google Scholar] [CrossRef]
  134. Mañas, A.; Aaltonen, K.; Andersson, N.; Hansson, K.; Adamska, A.; Seger, A.; Yasui, H.; Van Den Bos, H.; Radke, K.; Esfandyari, J.; et al. Clinically relevant treatment of PDX models reveals patterns of neuroblastoma chemoresistance. Sci. Adv. 2022, 8, eabq4617. [Google Scholar] [CrossRef]
  135. Radke, K.; Hansson, K.; Sjölund, J.; Wolska, M.; Karlsson, J.; Esfandyari, J.; Pietras, K.; Aaltonen, K.; Gisselsson, D.; Bexell, D. Anti-tumor effects of rigosertib in high-risk neuroblastoma. Transl. Oncol. 2021, 14, 101149. [Google Scholar] [CrossRef]
  136. Cuperus, R.; Tytgat, G.A.M.; Leen, R.; Brites, P.; Bras, J.; Caron, H.N.; Van Kuilenburg, A.B.P. Pleiotropic effects of fenretinide in neuroblastoma cell lines and multicellular tumor spheroids. Int. J. Oncol. 2008, 32, 1011–1019. [Google Scholar] [CrossRef]
  137. Corbacioglu, S.; Lode, H.; Ellinger, S.; Zeman, F.; Suttorp, M.; Escherich, G.; Bochennek, K.; Gruhn, B.; Lang, P.; Rohde, M.; et al. Irinotecan and temozolomide in combination with dasatinib and rapamycin versus irinotecan and temozolomide for patients with relapsed or refractory neuroblastoma (RIST-rNB-2011): A multicentre, open-label, randomised, controlled, phase 2 trial. Lancet Oncol. 2024, 25, 922–932. [Google Scholar] [CrossRef] [PubMed]
  138. Hansson, K.; Radke, K.; Aaltonen, K.; Saarela, J.; Mañas, A.; Sjölund, J.; Smith, E.M.; Pietras, K.; Påhlman, S.; Wennerberg, K.; et al. Therapeutic targeting of KSP in preclinical models of high-risk neuroblastoma. Sci. Transl. Med. 2020, 12, eaba4434. [Google Scholar] [CrossRef]
  139. Sidarovich, V.; De Mariano, M.; Aveic, S.; Pancher, M.; Adami, V.; Gatto, P.; Pizzini, S.; Pasini, L.; Croce, M.; Parodi, F.; et al. A High-Content Screening of Anticancer Compounds Suggests the Multiple Tyrosine Kinase Inhibitor Ponatinib for Repurposing in Neuroblastoma Therapy. Mol. Cancer Ther. 2018, 17, 1405–1415. [Google Scholar] [CrossRef] [PubMed]
  140. Kholosy, W.M.; Derieppe, M.; Van Den Ham, F.; Ober, K.; Su, Y.; Custers, L.; Schild, L.; Van Zogchel, L.M.J.; Wellens, L.M.; Ariese, H.R.; et al. Neuroblastoma and DIPG Organoid Coculture System for Personalized Assessment of Novel Anticancer Immunotherapies. J. Pers. Med. 2021, 11, 869. [Google Scholar] [CrossRef]
  141. Nguyen, R.; Moustaki, A.; Norrie, J.L.; Brown, S.; Akers, W.J.; Shirinifard, A.; Dyer, M.A. Interleukin-15 Enhances Anti-GD2 Antibody-Mediated Cytotoxicity in an Orthotopic PDX Model of Neuroblastoma. Clin. Cancer Res. 2019, 25, 7554–7564. [Google Scholar] [CrossRef]
  142. Veneziani, I.; Infante, P.; Ferretti, E.; Melaiu, O.; Battistelli, C.; Lucarini, V.; Compagnone, M.; Nicoletti, C.; Castellano, A.; Petrini, S.; et al. Nutlin-3a Enhances Natural Killer Cell–Mediated Killing of Neuroblastoma by Restoring p53-Dependent Expression of Ligands for NKG2D and DNAM-1 Receptors. Cancer Immunol. Res. 2021, 9, 170–183. [Google Scholar] [CrossRef]
  143. Blavier, L.; Yang, R.-M.; DeClerck, Y.A. The Tumor Microenvironment in Neuroblastoma: New Players, New Mechanisms of Interaction and New Perspectives. Cancers 2020, 12, 2912. [Google Scholar] [CrossRef]
  144. Zaghmi, A.; Aybay, E.; Jiang, L.; Shang, M.; Steinmetz-Späh, J.; Wermeling, F.; Kogner, P.; Korotkova, M.; Östling, P.; Jakobsson, P.; et al. High-content screening of drug combinations of an mPGES -1 inhibitor in multicellular tumor spheroids leads to mechanistic insights into neuroblastoma chemoresistance. Mol. Oncol. 2024, 18, 317–335. [Google Scholar] [CrossRef]
  145. Kock, A.; Bergqvist, F.; Steinmetz, J.; Elfman, L.H.M.; Korotkova, M.; Johnsen, J.I.; Jakobsson, P.; Kogner, P.; Larsson, K. Establishment of an in vitro 3D model for neuroblastoma enables preclinical investigation of combined tumor-stroma drug targeting. FASEB J. 2020, 34, 11101–11114. [Google Scholar] [CrossRef] [PubMed]
  146. Corallo, D.; Nardelli, C.; Pantile, M.; Menegazzo, S.; Biffi, A.; Aveic, S. Exploring the Role of Fibroblasts in Promoting Neuroblastoma Cell Migration and Invasion. J. Nanotheranostics 2024, 5, 212–227. [Google Scholar] [CrossRef]
  147. Treis, D.; Lundberg, K.I.; Bell, N.; Polychronopoulos, P.A.; Tümmler, C.; Åkerlund, E.; Aliverti, S.; Lilienthal, I.; Pepich, A.; Seashore-Ludlow, B.; et al. Targeted inhibition of WIP1 and histone H3K27 demethylase activity synergistically suppresses neuroblastoma growth. Cell Death Dis. 2025, 16, 318. [Google Scholar] [CrossRef]
  148. Wan, X.; Wang, W.; Liang, Z. Epigallocatechin-3-gallate inhibits the growth of three-dimensional in vitro models of neuroblastoma cell SH-SY5Y. Mol. Cell Biochem. 2021, 476, 3141–3148. [Google Scholar] [CrossRef] [PubMed]
  149. Liu, X.; Cheng, J.; Zhao, Y. Tumor Microenvironment Based on Extracellular Matrix Hydrogels for On-Chip Drug Screening. Biosensors 2024, 14, 429. [Google Scholar] [CrossRef]
  150. Villasante, A.; Lopez-Martinez, M.J.; Quiñonero, G.; Garcia-Lizarribar, A.; Peng, X.; Samitier, J. Microfluidic model of the alternative vasculature in neuroblastoma. In Vitro Models 2024, 3, 49–63. [Google Scholar] [CrossRef]
  151. Ning, L.; Shim, J.; Tomov, M.L.; Liu, R.; Mehta, R.; Mingee, A.; Hwang, B.; Jin, L.; Mantalaris, A.; Xu, C.; et al. A 3D Bioprinted in vitro Model of Neuroblastoma Recapitulates Dynamic Tumor-Endothelial Cell Interactions Contributing to Solid Tumor Aggressive Behavior. Adv. Sci. 2022, 9, 2200244. [Google Scholar] [CrossRef]
  152. Nothdurfter, D.; Ploner, C.; Coraça-Huber, D.C.; Wilflingseder, D.; Müller, T.; Hermann, M.; Hagenbuchner, J.; Ausserlechner, M.J. 3D bioprinted, vascularized neuroblastoma tumor environment in fluidic chip devices for precision medicine drug testing. Biofabrication 2022, 14, 035002. [Google Scholar] [CrossRef]
  153. Amoli, M.S.; Rezapourdamanab, S.; Jin, L.; Cadena, M.A.; Kaw, K.; Sridhar, V.; Meselhe, M.; Krikor, S.; Mahmoudi, M.; Ning, L.; et al. Protocol for 3D bioprinting of a 3D in vitro model of neuroblastoma. STAR Protoc. 2025, 6, 103725. [Google Scholar] [CrossRef]
  154. Abou-Antoun, T.J.; Nazarian, J.; Ghanem, A.; Vukmanovic, S.; Sandler, A.D. Molecular and functional analysis of anchorage independent, treatment-evasive neuroblastoma tumorspheres with enhanced malignant properties: A possible explanation for radio-therapy resistance. PLoS ONE 2018, 13, e0189711. [Google Scholar] [CrossRef]
  155. Lucarini, V.; Melaiu, O.; D’Amico, S.; Pastorino, F.; Tempora, P.; Scarsella, M.; Pezzullo, M.; De Ninno, A.; D’Oria, V.; Cilli, M.; et al. Combined mitoxantrone and anti-TGFβ treatment with PD-1 blockade enhances antitumor immunity by remodelling the tumor immune landscape in neuroblastoma. J. Exp. Clin. Cancer Res. 2022, 41, 326. [Google Scholar] [CrossRef] [PubMed]
  156. Lucarini, V.; Melaiu, O.; Gragera, P.; Król, K.; Scaldaferri, V.; Damiani, V.; De Ninno, A.; Nardozi, D.; Businaro, L.; Masuelli, L.; et al. Immunogenic Cell Death Inducers in Cancer Immunotherapy to Turn Cold Tumors into Hot Tumors. Int. J. Mol. Sci. 2025, 26, 1613. [Google Scholar] [CrossRef]
  157. Liu, M.; Xia, Y.; Ding, J.; Ye, B.; Zhao, E.; Choi, J.-H.; Alptekin, A.; Yan, C.; Dong, Z.; Huang, S.; et al. Transcriptional Profiling Reveals a Common Metabolic Program in High-Risk Human Neuroblastoma and Mouse Neuroblastoma Sphere-Forming Cells. Cell Rep. 2016, 17, 609–623. [Google Scholar] [CrossRef] [PubMed]
  158. Hua, Z.-Y.; Hansen, J.N.; He, M.; Dai, S.-K.; Choi, Y.; Fulton, M.D.; Lloyd, S.M.; Szemes, M.; Sen, J.; Ding, H.-F.; et al. PRMT1 promotes neuroblastoma cell survival through ATF5. Oncogenesis 2020, 9, 50. [Google Scholar] [CrossRef] [PubMed]
  159. Cao, D.; Kishida, S.; Huang, P.; Mu, P.; Tsubota, S.; Mizuno, M.; Kadomatsu, K. A New Tumorsphere Culture Condition Restores Potentials of Self-Renewal and Metastasis of Primary Neuroblastoma in a Mouse Neuroblastoma Model. PLoS ONE 2014, 9, e86813. [Google Scholar] [CrossRef] [PubMed]
  160. Nakaguro, M.; Kiyonari, S.; Kishida, S.; Cao, D.; Murakami-Tonami, Y.; Ichikawa, H.; Takeuchi, I.; Nakamura, S.; Kadomatsu, K. Nucleolar protein PES 1 is a marker of neuroblastoma outcome and is associated with neuroblastoma differentiation. Cancer Sci. 2015, 106, 237–243. [Google Scholar] [CrossRef]
  161. Embaie, B.T.; Sarkar, H.; Alchahin, A.M.; Otte, J.; Olsen, T.K.; Tümmler, C.; Kameneva, P.; Artemov, A.V.; Akkuratova, N.; Adameyko, I.; et al. Comparative Single-Cell Transcriptomics of Human Neuroblastoma and Preclinical Models Reveals Conservation of an Adrenergic Cell State. Cancer Res. 2025, 85, 1015–1034. [Google Scholar] [CrossRef]
  162. Abuwatfa, W.H.; Pitt, W.G.; Husseini, G.A. Scaffold-based 3D cell culture models in cancer research. J. Biomed. Sci. 2024, 31, 7. [Google Scholar] [CrossRef] [PubMed]
  163. Jubelin, C.; Muñoz-Garcia, J.; Griscom, L.; Cochonneau, D.; Ollivier, E.; Heymann, M.-F.; Vallette, F.M.; Oliver, L.; Heymann, D. Three-dimensional in vitro culture models in oncology research. Cell Biosci. 2022, 12, 155. [Google Scholar] [CrossRef]
  164. Driehuis, E.; Kretzschmar, K.; Clevers, H. Establishment of patient-derived cancer organoids for drug-screening applications. Nat. Protoc. 2020, 15, 3380–3409. [Google Scholar] [CrossRef]
  165. Li, C.; Qiu, S.; Liu, X.; Guo, F.; Zhai, J.; Li, Z.; Deng, L.; Ge, L.; Qian, H.; Yang, L.; et al. Extracellular matrix-derived mechanical force governs breast cancer cell stemness and quiescence transition through integrin-DDR signaling. Signal Transduct. Target. Ther. 2023, 8, 247. [Google Scholar] [CrossRef]
  166. Mitchell, C.B.; O’Neill, G.M. Rac GTPase regulation of 3D invasion in neuroblastomas lacking MYCN amplification. Cell Adhes. Migr. 2017, 11, 68–79. [Google Scholar] [CrossRef]
  167. Curtin, C.; Nolan, J.C.; Conlon, R.; Deneweth, L.; Gallagher, C.; Tan, Y.J.; Cavanagh, B.L.; Asraf, A.Z.; Harvey, H.; Miller-Delaney, S.; et al. A physiologically relevant 3D collagen-based scaffold–neuroblastoma cell system exhibits chemosensitivity similar to orthotopic xenograft models. Acta Biomater. 2018, 70, 84–97. [Google Scholar] [CrossRef] [PubMed]
  168. Gallagher, C.; Murphy, C.; Kelly, G.; O’Brien, F.J.; Piskareva, O. Three-dimensional In Vitro Biomimetic Model of Neuroblastoma using Collagen-based Scaffolds. J. Vis. Exp. 2021, 62627. [Google Scholar] [CrossRef]
  169. Bova, L.; Maggiotto, F.; Micheli, S.; Giomo, M.; Sgarbossa, P.; Gagliano, O.; Falcone, D.; Cimetta, E. A Porous Gelatin Methacrylate-Based Material for 3D Cell-Laden Constructs. Macromol. Biosci. 2023, 23, 2200357. [Google Scholar] [CrossRef]
  170. Kozlowski, M.T.; Crook, C.J.; Ku, H.T. Towards organoid culture without Matrigel. Commun. Biol. 2021, 4, 1387. [Google Scholar] [CrossRef] [PubMed]
  171. Fusco, P.; Parisatto, B.; Rampazzo, E.; Persano, L.; Frasson, C.; Di Meglio, A.; Leslz, A.; Santoro, L.; Cafferata, B.; Zin, A.; et al. Patient-derived organoids (PDOs) as a novel in vitro model for neuroblastoma tumours. BMC Cancer 2019, 19, 970. [Google Scholar] [CrossRef]
  172. Innala, M.; Riebe, I.; Kuzmenko, V.; Sundberg, J.; Gatenholm, P.; Hanse, E.; Johannesson, S. 3D Culturing and differentiation of SH-SY5Y neuroblastoma cells on bacterial nanocellulose scaffolds. Artif. Cells Nanomed. Biotechnol. 2014, 42, 302–308. [Google Scholar] [CrossRef]
  173. Torresan, V.; Dedroog, L.M.; Deschaume, O.; Koos, E.; Lettinga, M.P.; Gandin, A.; Pelosin, M.; Zanconato, F.; Brusatin, G.; Bartic, C. Nanocellulose-collagen composites as advanced biomaterials for 3D in-vitro neuronal model systems. Carbohydr. Polym. 2025, 348, 122901. [Google Scholar] [CrossRef]
  174. Chemmarappally, J.M.; Pegram, H.C.N.; Abeywickrama, N.; Fornari, E.; Hargreaves, A.J.; De Girolamo, L.A.; Stevens, B. A co-culture nanofibre scaffold model of neural cell degeneration in relevance to Parkinson’s disease. Sci. Rep. 2020, 10, 2767. [Google Scholar] [CrossRef]
  175. Monferrer, E.; Dobre, O.; Trujillo, S.; González Oliva, M.A.; Trubert-Paneli, A.; Acevedo-León, D.; Noguera, R.; Salmeron-Sanchez, M. Vitronectin-based hydrogels recapitulate neuroblastoma growth conditions. Front. Cell Dev. Biol. 2022, 10, 988699. [Google Scholar] [CrossRef]
  176. Ornell, K.J.; Mistretta, K.S.; Ralston, C.Q.; Coburn, J.M. Development of a stacked, porous silk scaffold neuroblastoma model for investigating spatial differences in cell and drug responsiveness. Biomater. Sci. 2021, 9, 1272–1290. [Google Scholar] [CrossRef] [PubMed]
  177. Granados-Aparici, S.; Vieco-Martí, I.; López-Carrasco, A.; Navarro, S.; Noguera, R. Real-time morphometric analysis of targeted therapy for neuroblastoma cells in monolayer and 3D hydrogels using digital holographic microscopy. iScience 2024, 27, 111231. [Google Scholar] [CrossRef]
  178. Mistretta, K.S.; Coburn, J.M. Three-dimensional silk fibroin scaffolded co-culture of human neuroblastoma and innate immune cells. Exp. Cell Res. 2024, 443, 114289. [Google Scholar] [CrossRef] [PubMed]
  179. Barberio, C.; Saez, J.; Withers, A.; Nair, M.; Tamagnini, F.; Owens, R.M. Conducting Polymer-ECM Scaffolds for Human Neuronal Cell Differentiation. Adv. Healthc. Mater. 2022, 11, 2200941. [Google Scholar] [CrossRef] [PubMed]
  180. Quinn, C.H.; Beierle, A.M.; Julson, J.R.; Erwin, M.E.; Alrefai, H.; Markert, H.R.; Stewart, J.E.; Hutchins, S.C.; Bownes, L.V.; Aye, J.M.; et al. Using 3D-bioprinted models to study pediatric neural crest-derived tumors. Int. J. Bioprinting 2023, 9, 723. [Google Scholar] [CrossRef]
  181. Bordoni, M.; Karabulut, E.; Kuzmenko, V.; Fantini, V.; Pansarasa, O.; Cereda, C.; Gatenholm, P. 3D Printed Conductive Nanocellulose Scaffolds for the Differentiation of Human Neuroblastoma Cells. Cells 2020, 9, 682. [Google Scholar] [CrossRef]
  182. Aveic, S.; Seidelmann, M.; Davtalab, R.; Corallo, D.; Vogt, M.; Rütten, S.; Fischer, H. Three-dimensional in vitro model of bone metastases of neuroblastoma as a tool for pharmacological evaluations. Nanotheranostics 2024, 8, 1–11. [Google Scholar] [CrossRef]
  183. Aveic, S.; Janßen, S.; Nasehi, R.; Seidelmann, M.; Vogt, M.; Pantile, M.; Rütten, S.; Fischer, H. A 3D printed in vitro bone model for the assessment of molecular and cellular cues in metastatic neuroblastoma. Biomater. Sci. 2021, 9, 1716–1727. [Google Scholar] [CrossRef]
  184. Maris, J.M.; Hogarty, M.D.; Bagatell, R.; Cohn, S.L. Neuroblastoma. Lancet 2007, 369, 2106–2120. [Google Scholar] [CrossRef]
  185. Breslin, S.; O’Driscoll, L. Three-dimensional cell culture: The missing link in drug discovery. Drug Discov. Today 2013, 18, 240–249. [Google Scholar] [CrossRef] [PubMed]
  186. Asthana, A.; Kisaalita, W.S. Microtissue size and hypoxia in HTS with 3D cultures. Drug Discov. Today 2012, 17, 810–817. [Google Scholar] [CrossRef] [PubMed]
  187. Vega, V.F.; Yang, D.; Jordán, L.O.; Ye, F.; Conway, L.; Chen, L.Y.; Shumate, J.; Baillargeon, P.; Scampavia, L.; Parker, C.; et al. Protocol for 3D screening of lung cancer spheroids using natural products. SLAS Discov. 2023, 28, 20–28. [Google Scholar] [CrossRef] [PubMed]
  188. Habanjar, O.; Diab-Assaf, M.; Caldefie-Chezet, F.; Delort, L. 3D Cell Culture Systems: Tumor Application, Advantages, and Disadvantages. Int. J. Mol. Sci. 2021, 22, 12200. [Google Scholar] [CrossRef]
  189. Lee, S.-Y.; Koo, I.-S.; Hwang, H.J.; Lee, D.W. In Vitro three-dimensional (3D) cell culture tools for spheroid and organoid models. SLAS Discov. 2023, 28, 119–137. [Google Scholar] [CrossRef]
  190. McKee, C.; Chaudhry, G.R. Advances and challenges in stem cell culture. Colloids Surf. B Biointerfaces 2017, 159, 62–77. [Google Scholar] [CrossRef]
  191. Salinas-Vera, Y.M.; Valdés, J.; Pérez-Navarro, Y.; Mandujano-Lazaro, G.; Marchat, L.A.; Ramos-Payán, R.; Nuñez-Olvera, S.I.; Pérez-Plascencia, C.; López-Camarillo, C. Three-Dimensional 3D Culture Models in Gynecological and Breast Cancer Research. Front. Oncol. 2022, 12, 826113. [Google Scholar] [CrossRef]
  192. Law, A.M.K.; Rodriguez De La Fuente, L.; Grundy, T.J.; Fang, G.; Valdes-Mora, F.; Gallego-Ortega, D. Advancements in 3D Cell Culture Systems for Personalizing Anti-Cancer Therapies. Front. Oncol. 2021, 11, 782766. [Google Scholar] [CrossRef]
  193. Lovitt, C.; Shelper, T.; Avery, V. Advanced Cell Culture Techniques for Cancer Drug Discovery. Biology 2014, 3, 345–367. [Google Scholar] [CrossRef]
  194. Kim, S.-Y.; van de Wetering, M.; Clevers, H.; Sanders, K. The future of tumor organoids in precision therapy. Trends Cancer, 2025; online ahead of print. [Google Scholar] [CrossRef]
  195. Narasimhan, V.; Wright, J.A.; Churchill, M.; Wang, T.; Rosati, R.; Lannagan, T.R.M.; Vrbanac, L.; Richardson, A.B.; Kobayashi, H.; Price, T.; et al. Medium-throughput Drug Screening of Patient-derived Organoids from Colorectal Peritoneal Metastases to Direct Personalized Therapy. Clin. Cancer Res. 2020, 26, 3662–3670. [Google Scholar] [CrossRef] [PubMed]
  196. Xu, S.; Yin, S.-Y.; Bie, Z.-X.; Li, Y.-M.; Qi, J.; Ma, Y.-D.; Wang, Z.; Xi, J.J.; Li, X.-G. Personalized drug screening of patient-derived tumor-like cell clusters based on specimens obtained from percutaneous transthoracic needle biopsy in patients with lung malignancy: A real-world study. BMC Cancer 2025, 25, 649. [Google Scholar] [CrossRef] [PubMed]
Figure 2. Applications of 3D Models in Neuroblastoma Research. Key applications of various 3D culture models in neuroblastoma research, including their use in drug screening, studying chemotherapy resistance, or transcriptomic and epigenomic profiling. This image was created with BioRender (https://biorender.com/ accessed on 31 March 2025).
Figure 2. Applications of 3D Models in Neuroblastoma Research. Key applications of various 3D culture models in neuroblastoma research, including their use in drug screening, studying chemotherapy resistance, or transcriptomic and epigenomic profiling. This image was created with BioRender (https://biorender.com/ accessed on 31 March 2025).
Organoids 04 00010 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jung, P.; Wolpaw, A.J. Three-Dimensional Culture Systems in Neuroblastoma Research. Organoids 2025, 4, 10. https://doi.org/10.3390/organoids4020010

AMA Style

Jung P, Wolpaw AJ. Three-Dimensional Culture Systems in Neuroblastoma Research. Organoids. 2025; 4(2):10. https://doi.org/10.3390/organoids4020010

Chicago/Turabian Style

Jung, Piotr, and Adam J. Wolpaw. 2025. "Three-Dimensional Culture Systems in Neuroblastoma Research" Organoids 4, no. 2: 10. https://doi.org/10.3390/organoids4020010

APA Style

Jung, P., & Wolpaw, A. J. (2025). Three-Dimensional Culture Systems in Neuroblastoma Research. Organoids, 4(2), 10. https://doi.org/10.3390/organoids4020010

Article Metrics

Back to TopTop