Advertisement
Review| Volume 28, ISSUE 4, P119-137, June 2023

Download started.

Ok

In Vitro three-dimensional (3D) cell culture tools for spheroid and organoid models

Open AccessPublished:March 27, 2023DOI:https://doi.org/10.1016/j.slasd.2023.03.006

      Abstract

      Three-dimensional (3D) cell culture technology has been steadily studied since the 1990′s due to its superior biocompatibility compared to the conventional two-dimensional (2D) cell culture technology, and has recently developed into an organoid culture technology that further improved biocompatibility. Since the 3D culture of human cell lines in artificial scaffolds was demonstrated in the early 90′s, 3D cell culture technology has been actively developed owing to various needs in the areas of disease research, precision medicine, new drug development, and some of these technologies have been commercialized. In particular, 3D cell culture technology is actively being applied and utilized in drug development and cancer-related precision medicine research. Drug development is a long and expensive process that involves multiple steps—from target identification to lead discovery and optimization, preclinical studies, and clinical trials for approval for clinical use. Cancer ranks first among life-threatening diseases owing to intra-tumoral heterogeneity associated with metastasis, recurrence, and treatment resistance, ultimately contributing to treatment failure and adverse prognoses. Therefore, there is an urgent need for the development of efficient drugs using 3D cell culture techniques that can closely mimic in vivo cellular environments and customized tumor models that faithfully represent the tumor heterogeneity of individual patients. This review discusses 3D cell culture technology focusing on research trends, commercialization status, and expected effects developed until recently. We aim to summarize the great potential of 3D cell culture technology and contribute to expanding the base of this technology.

      Keywords

      1. Introduction

      The cultivation of animal cells began in the early 90′s through the cultivation of neuronal fibroblasts from the cochlea of the tadpole in the coagulated lymphatic fluid of the frog. An extracellular substrate (scaffold) was used to provide space for cells to grow and to construct a structure in vitro similar to the in vivo extracellular matrix (ECM) [
      • Harrison R.G.
      Observations on the living developing nerve fiber.
      ]. Thereafter, animal cells could be stably cultured in vitro with the development of cell lines with good reproducibility of proliferation, cell culture media, and apparatus and equipment. Animal cells were cultured by adhering and growing them on the surface of a plastic dish coated with a cell adhesion promoter. These animal cell cultures have been widely used in biotechnology and medicine for the discovery and production of physiologically active substances, the development of disease models, toxicity analysis, and drug development.
      However, animal cells adhered to a plastic dish grow into a single layer, which is morphologically different from the 3D growth of animal cells in the living body. This environment also affects gene expression in cells. Animal cells exhibit differential gene expression in 2D cell culture models and biocompatible 3D cell culture models [
      Genomic and morphological changes of neuroblastoma cells in response to three-dimensional matrices.
      ]. As a result, in vitro animal cell culture shows poor correspondence with animal cells in vivo. Therefore, cell-based experimental results should be cautiously applied to basic life science research and medical disease models. In particular, when animal cells are used for drug efficacy or toxicity analysis, drug reactivity in 3D cell culture models greatly differs from that in conventional 2D cell culture models [
      • Huang G.S.
      • Tseng T.C.
      • Dai N.T.
      • et al.
      Fast isolation and expansion of multipotent cells from adipose tissue based on chitosan-selected primary culture.
      ,
      • Gurski L.A.
      • Petrelli N.J.
      • Jia X.
      • et al.
      3D matrices for anti-cancer drug testing and development.
      ,
      • Wong H.L.
      • Wang M.X.
      • Cheung P.T.
      • et al.
      A 3D collagen microsphere culture system for GDNF-secreting HEK293 cells with enhanced protein productivity.
      ,
      • Wu M.-.H.
      • Chang Y.-.H.
      • Liu Y.-.T.
      • et al.
      Development of high throughput microfluidic cell culture chip for perfusion 3-dimensional cell culture-based chemosensitivity assay.
      ,
      • Gurski L.A.
      • Jha A.K.
      • Zhang C.
      • et al.
      Hyaluronic acid-based hydrogels as 3D matrices for in vitro evaluation of chemotherapeutic drugs using poorly adherent prostate cancer cells.
      ]. When cells derived from cancer patients are cultured in 3D cell culture, the cell-cell interaction and ECM changes the morphology of the cells, as well as the type and expression of major genes [
      • Huang G.S.
      • Tseng T.C.
      • Dai N.T.
      • et al.
      Fast isolation and expansion of multipotent cells from adipose tissue based on chitosan-selected primary culture.
      ,
      • Gurski L.A.
      • Petrelli N.J.
      • Jia X.
      • et al.
      3D matrices for anti-cancer drug testing and development.
      ,
      • Wong H.L.
      • Wang M.X.
      • Cheung P.T.
      • et al.
      A 3D collagen microsphere culture system for GDNF-secreting HEK293 cells with enhanced protein productivity.
      ]. Consequently, many reports suggest that cellular drug responses differ between 3D and 2D cell culture models [
      • Wu M.-.H.
      • Chang Y.-.H.
      • Liu Y.-.T.
      • et al.
      Development of high throughput microfluidic cell culture chip for perfusion 3-dimensional cell culture-based chemosensitivity assay.
      ,
      • Gurski L.A.
      • Jha A.K.
      • Zhang C.
      • et al.
      Hyaluronic acid-based hydrogels as 3D matrices for in vitro evaluation of chemotherapeutic drugs using poorly adherent prostate cancer cells.
      ]. In addition, there are many difficulties in deriving meaningful results at the clinical stage owing to the absence of disease-specific models in the human body during drug development. Human in vitro 3D cell cultures using stem cells from different organs has the potential to overcome these limitations. Recently, attempts have been made to use drug response data of organoids cultured in 3D in precision medicine for the personalized treatment of cancer patients as clinical data, as well as gene data [
      • Pauli C.
      • Hopkins B.D.
      • Prandi D.
      • et al.
      Personalized in vitro and in vivo cancer models to guide precision medicine.
      ,
      • Broutier L.
      • Mastrogiovanni G.
      • Verstegen M.M.
      • et al.
      Human primary liver cancer-derived organoid cultures for disease modeling and drug screening.
      ]. Such an organoid model is composed of a combination of cells, an ECM (scaffold), a cell culture container, a cell growth factor, etc., and the model varies depending on the combinations thereof. The organoid model has been used to biologically model human organs in vitro and has recently developed into an organoid culture technology that further improved biocompatibility. [
      • Kim J.
      • Koo B.K.
      • Knoblich J.A.
      Human organoids: model systems for human biology and medicine.
      ,
      • Fatehullah A.
      • Tan S.H.
      • Barker N.
      Organoids as an in vitro model of human development and disease.
      ] In particular, bioprinting technology using human-derived cells and organoid culture using a cell culture platform (organ-on-a-chip) has shown potential for drug screening and human disease research. Organoids are self-organizing 3D culture systems that closely resemble human organs [
      • Sato T.
      • Vries R.G.
      • Snippert H.J.
      • et al.
      Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche.
      ,
      • Sato T.
      • Stange D.E.
      • Ferrante M.
      • et al.
      Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett's epithelium.
      ,
      • Lancaster M.A.
      • Renner M.
      • Martin C.A.
      • et al.
      Cerebral organoids model human brain development and microcephaly.
      ,
      • Takasato M.
      • Er P.X.
      • Chiu H.S.
      • et al.
      Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis.
      ,
      • Fujii M.
      • Matano M.
      • Toshimitsu K.
      • et al.
      Human intestinal organoids maintain self-renewal capacity and cellular diversity in niche-inspired culture condition.
      ,
      • Hu H.
      • Gehart H.
      • Artegiani B.
      • et al.
      Long-Term Expansion of Functional Mouse and Human Hepatocytes as 3D Organoids.
      ]. Therefore, organoid formation and analysis studies using these complement the existing 2D cell culture model and are very useful in basic biological research, medical research, drug development, and drug research under conditions physiologically similar to the human environment. The potential of organoids is being increasingly recognized. However, compared to conventional cell lines and animal models, the development of organoid technology is still in its infancy, and there are still many challenges to overcome. During the last decades, organoid models have been variously developed, some of which have been commercialized but are still not standardized. In addition, attempts to increase the biocompatibility of existing models have been made. This review summarizes the latest technologies and commercialization trends of organoid models and discusses the future directions and applications of organoid models and technology.

      2. Review of 3D cell culture

      2.1 3D cell culture

      A 3D cell culture model refers to a cell culture model in which an environment similar to a living body is artificially formed outside the body, allowing cells to grow in all dimensions and interact with the surrounding environment. In particular, since cells are implemented by 3D culture in an environment similar to the human body, they show similar characteristics to cells and organs of the human body [
      • Sachs N.
      • Papaspyropoulos A.
      • Zomer-van Ommen D.D.
      • et al.
      Long-term expanding human airway organoids for disease modeling.
      ]. Fig. 1 shows a 3D cell culture model from Griffith University, Austria, published in the Biological International Journal in 2014 [
      • Lovitt C.J.
      • Shelper T.B.
      • Avery V.M.
      Advanced cell culture techniques for cancer drug discovery.
      ]. In a 3D space composed of an ECM (scaffold), nutrients, oxygen, and drugs are supplied to the cells through a diffusion gradient and permeation to provide a biologically similar environment. Cell-to-cell and/or cell-to-ECM interactions and paracrine signaling by diffusion of cell secretions, that do not exist in conventional 2D cell culture, are the distinguishing features of the 3D cell culture model. Therefore, with advances in biotechnology, the 3D cell culture model is expected to become an optimized and efficient biotechnology platform for studying human physiology and disease.
      Fig 1
      Fig. 1Characteristics of a 3D Cell Culture Model
      [
      • Lovitt C.J.
      • Shelper T.B.
      • Avery V.M.
      Advanced cell culture techniques for cancer drug discovery.
      ]
      .

      2.2 Definition and difference of spheroid and organoid models

      3D cell culture technology is a promising way to bridge the gap between conventional cell culture methods and animal models. 3D cell culture methods can be divided into spheroid and organoid models. Spheroids and organoids are 3D cell structures formed by gathering many cells. However, depending on the cell origin, type, and function of the cell structure implemented in 3D, the spheroids and organoids are classified as follows. Spheroids are simply the aggregation of cells into 3D structures, but organoids have self-differentiation capabilities, indicating structures and functions similar to those of human organs. To date, the terms—3D cell culture models, organoids, and spheroids—have been used interchangeably [
      • Simian M.
      • Bissell M.J
      Organoids: a historical perspective of thinking in three dimensions.
      ]. However, although these two models can both be applied to 3D cell-based research, they differ in their fabrication method and cell origin. Spheroids are 3D cell culture models having spherical cell units that are cultured as free-floating aggregates [
      • Gilazieva Z.
      • Ponomarev A.
      • Rutland C.
      • et al.
      Promising applications of tumor spheroids and organoids for personalized medicine.
      ]. Compared to the existing 2D models, this method has the advantage of effectively implementing and analyzing cell-cell interactions. However, because spheroids are sometimes cultured without the ECM as a scaffold, they show relatively low structural complexity. Furthermore, spheroids are simple clusters of a wide range of cells, such as tumor tissues, embryonic bodies, hepatocytes, neural tissues, and mammary glands [
      • Gunti S.
      • Hoke A.T.K.
      • Vu K.P.
      • et al.
      Organoid and spheroid tumor models: techniques and applications.
      ]. Therefore, since spheroids cannot self-assemble or regenerate, they are disadvantageous in that they are less biocompatible than organoids [
      • Zanoni M.
      • Cortesi M.
      • Zamagni A.
      • et al.
      Modeling neoplastic disease with spheroids and organoids.
      ]. However, in cancer-related research, tumor spheroids are popular because they enable the culture of cancer cells in 3D and implement cell-cell and cell-ECM interactions. In particular, tumor spheroids are characterized by mimicking vascularized or poorly vascularized tumors. In particular, when cultured larger than 500 µm, a multilayered structure can be formed to achieve the outer layer, the middle layer of cells, and the inner layer structure of necrotic cells that show hypoxia [
      • Nunes A.S.
      • Barros A.S.
      • Costa E.C.
      • et al.
      3D tumor spheroids as in vitro models to mimic in vivo human solid tumors resistance to therapeutic drugs.
      ]. In addition, tumor rotators generally exhibit anticancer drug resistance and radiation resistance + and are widely used in cancer cell migration and invasion and drug screening studies [
      • Nath S.
      • Devi G.R.
      Three-dimensional culture systems in cancer research: focus on tumor spheroid model.
      ].
      Organoids are complex clusters of organ-specific cells, such as those in the stomach, liver, or bladder. Organoids can be cultured from embryonic stem cells, induced pluripotent stem cells, and adult stem cells. They exhibit multi-stage organogenesis features that self-assemble and proliferate in a scaffolding extracellular environment such as Matrigel or collagen [
      • McCauley H.A.
      • Wells J.M.
      Pluripotent stem cell-derived organoids: using principles of developmental biology to grow human tissues in a dish.
      ,
      • Spence J.R.
      • Mayhew C.N.
      • Rankin S.A.
      • et al.
      Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro.
      ]. Organoids are structurally complex and recapitulate the development of organs in vitro, making them very useful in the study of organogenesis, heredity, and pathology, especially in organs with little or no regenerative capacity, such as the brain [
      • Bartfeld S.
      • Bayram T.
      • van de Wetering M.
      • et al.
      In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection.
      ,
      • Garcez P.P.
      • Loiola E.C.
      • Madeiro da Costa R.
      • et al.
      Zika virus impairs growth in human neurospheres and brain organoids.
      ,
      • Takebe T.
      • Sekine K.
      • Enomura M.
      • et al.
      Vascularized and functional human liver from an iPSC-derived organ bud transplant.
      ,
      • Dutta D.
      • Heo I.
      • Clevers H.
      Disease modeling in stem cell-derived 3D organoid systems.
      ,
      • Clevers H.
      Modeling development and disease with organoids.
      ]. The term cancer organoids or tumoroids may not be appropriate because cancerous tissue is not an organ that originally exists within the body. However, since cancer organoids are implemented using cancer stem cells, the term organoid is commonly used depending on the origin of the cell. Unlike normal cells, cancer stem cells can produce cancer cells and indefinitely reproduce cancer progenitor cells. Therefore, cancer organoids exhibit self-renewal properties through cancer progression and re-proliferation, and their differentiation ability enables mimicking the unique characteristics of cancer tissues, such as intra-tumoral heterogeneity [
      • Lin Y.C.
      • Murayama Y.
      • Hashimoto K.
      • et al.
      Role of tumor suppressor genes in the cancer-associated reprogramming of human induced pluripotent stem cells.
      ,
      • Al-Hajj M.
      • Wicha M.S.
      • Benito-Hernandez A.
      • et al.
      Prospective identification of tumorigenic breast cancer cells.
      ,
      • Singh S.K.
      • Clarke I.D.
      • Terasaki M.
      • et al.
      Identification of a cancer stem cell in human brain tumors.
      ,
      • Bonnet D.
      • Dick J.E.
      Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell.
      ]. Cancer organoids are more complex than spheroids and have features and differences that better recapitulate the histological and genetic features of intrinsic malignant tissues [
      • Sato T.
      • Vries R.G.
      • Snippert H.J.
      • et al.
      Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche.
      ,
      • Schutgens F.
      • Clevers H.
      Human organoids: tools for understanding biology and treating diseases.
      ].

      2.3 Pros and cons of 2D cell culture and 3D cell culture

      A 2D cell culture model in which cells are plated on a dish surface has been continuously developed over the past 100 years and is now standardized. This 2D cell culture model is easy to implement and commercially available products can be easily obtained, so users can conveniently perform experiments. However, a disadvantage of the 2D cell culture model is that it cannot accurately represent the response owing to its low biocompatibility. To solve this problem, 3D cell culture models have been developed. High Throughput Screening (HTS) assay was performed by uniformly culturing pancreatic organoids in 384 and 1536 well plates using a magnetic force. Cells were labeled with nanoparticles (NanoShuttle-PL) for 3D cell culture using the magnetic levitation method, and then the cells quickly formed 3D spheroids react with a magnetic plate. [
      • Baillargeon P.
      • Shumate J.
      • Hou S.
      • et al.
      Automating a magnetic 3D spheroid model technology for high-throughput screening.
      ,
      • Fernandez-Vega V.
      • Hou S.
      • Plenker D.
      • et al.
      Lead identification using 3D models of pancreatic cancer.
      ] Also, the advantages of the 3D-HTS platform were emphasized through comparison with the conventional 2D-HTS analysis method [
      • Broutier L.
      • Andersson-Rolf A.
      • Hindley C.J.
      • et al.
      Culture and establishment of self-renewing human and mouse adult liver and pancreas 3D organoids and their genetic manipulation.
      ,
      • Moreira L.
      • Bakir B.
      • Chatterji P.
      • et al.
      Pancreas 3D organoids: current and future aspects as a research platform for personalized medicine in pancreatic cancer.
      ,
      • Hou S.
      • Tiriac H.
      • Sridharan B.P.
      • et al.
      Advanced development of primary pancreatic organoid tumor models for high-throughput phenotypic drug screening.
      ,
      • Caleffi J.T.
      • Aal M.C.E.
      • Gallindo H.O.M.
      • et al.
      Magnetic 3D cell culture: state of the art and current advances.
      ]. However, there are many variables, such as the type and shape of the ECM, cell culture factors, and shapes of cell culture containers, for imitating the living environment. The culture system is complicated for the user to perform the experiment easily. Despite these disadvantages, the 3D cell culture model has the advantage of providing a more accurate response owing to its high biocompatibility (Fig. 2). As shown in Table 1, 3D cell culture models have been reported to have higher biocompatibility than conventional 2D cell culture in terms of cell shape, cell differentiation, and drug metabolism. In addition, studies using 3D cell culture models have many potential advantages over animal-model-based experiments. 3D cell culture models can provide large amounts of information more quickly and efficiently. The 3D-cultured cells that are implemented using human-derived cells, can more accurately mimic human tissues compared to animal models, and the time and cost required to build a research model are less. In particular, there was a case in which the cytotoxicity efficacy of ~3300 approved drugs was evaluated using four patient-derived pancreatic cancer KRAS mutation-associated primary cells, including cancer-related fibroblasts [
      • Hou S.
      • Tiriac H.
      • Sridharan B.P.
      • et al.
      Advanced development of primary pancreatic organoid tumor models for high-throughput phenotypic drug screening.
      ]. Consequently, researchers can simultaneously conduct studies using a large number of 3D cell culture models. To meet this demand, 3D cell culture models have been built to gradually overcome the problems of complicated systems and difficulties of use, and are being commercialized. Corning, Insphero, 3D Biomatrix, and Trevigen are representative examples of commercialization of these 3D cell culture products, while SPL and MBD sell 3D cell culture models such as artificial ECM, culture plates, and culture equipment.
      Fig 2
      Fig. 2Comparative analysis of 2D and 3D cell culture models.
      Table 1Characteristic Differences Between 2D Cell Culture and Organoid Models .
      Key Characteristics2D Cell Culture3D Cell Culture
      Cell Shape
      • Antoni D.
      • Burckel H.
      • Josset E.
      • et al.
      Three-dimensional cell culture: a breakthrough in vivo.
      It grows only on the bottom of the dish and is flat and stretched on the surface.It is possible to grow in various directions, thus forming a natural form (ellipsoid / polarized).
      Time to culture
      • Khetan S.
      • Burdick J.A.
      Patterning network structure to spatially control cellular remodeling and stem cell fate within 3-dimensional hydrogels.
      Within minutes to a few hours (short time)From a few hours to a few days (long time)
      Cell interface to medium [
      • Kim J.B.
      Three-dimensional tissue culture models in cancer biology.
      ,
      • Yip D.
      • Cho C.H.
      A multicellular 3D heterospheroid model of liver tumor and stromal cells in collagen gel for anti-cancer drug testing.
      ]
      Because it is attached flat on the bottom of the culture dish, all the cells are uniformly exposed to the culture medium and the drug at the same time.As the cells grow in many directions, the concentration gradient of the culture medium and drug permeates in the cell mass as in the in vivo environment, and the cells on the lump surface are more exposed than the cells on the inner surface.
      Cells interactions [
      • Lee J.
      • Cuddihy M.J.
      • Kotov N.A.
      Three-dimensional cell culture matrices: state of the art.
      ,
      • Bissell M.J.
      • Rizki A.
      • Mian I.S.
      Tissue architecture: the ultimate regulator of breast epithelial function.
      ]
      The cell-cell has no gap due to growth only on the side, and the cell-extracellular environment interaction is not similar to the living body.Cell-cells are multifaceted and capable of proper interactions of the cell-extracellular environment, similar to the in vivo environment.
      Cell Differentiation and mechanisms
      • Chitcholtan K.
      • Asselin E.
      • Parent S.
      • et al.
      Differences in growth properties of endometrial cancer in three dimensional (3D) culture and 2D cell monolayer.
      ,
      • Birgersdotter A.
      • Sandberg R.
      • Ernberg I.
      Gene expression perturbation in vitro–a growing case for three-dimensional (3D) culture systems.
      ,
      • Gómez-Lechón M.J.
      • Jover R.
      • Donato T.
      • et al.
      Long-term expression of differentiated functions in hepatocytes cultured in three-dimensional collagen matrix.
      ,
      • Semino C.E.
      • Merok J.R.
      • Crane G.G.
      • et al.
      Functional differentiation of hepatocyte-like spheroid structures from putative liver progenitor cells in three-dimensional peptide scaffolds.
      ,
      • Ghosh S.
      • Spagnoli G.C.
      • Martin I.
      • et al.
      Three-dimensional culture of melanoma cells profoundly affects gene expression profile: a high density oligonucleotide array study.
      Normal or low differentiation. There is a change in gene expression and mRNA biochemistry in cells, which is different from in vivo conditions.High eruption. Has gene expression and biochemistry of cells as in vivo.
      Drug metabolism [
      • Schyschka L.
      • Sánchez J.J.
      • Wang Z.
      • et al.
      Hepatic 3D cultures but not 2D cultures preserve specific transporter activity for acetaminophen-induced hepatotoxicity.
      ,
      • Elkayam T.
      • Amitay-Shaprut S.
      • Dvir-Ginzberg M.
      • et al.
      Enhancing the drug metabolism activities of C3A–a human hepatocyte cell line–by tissue engineering within alginate scaffolds.
      ]
      Metabolism observation is not easy.Increased expression of CYP enzyme enhances drug metabolism.
      Drug Sensitivity
      • Bokhari M.
      • Carnachan R.J.
      • Cameron N.R.
      • et al.
      Culture of HepG2 liver cells on three dimensional polystyrene scaffolds enhances cell structure and function during toxicological challenge.
      Because it is attached to the floor and is directly exposed to the drug, the cell responds sensitively to the drug.Cells that grow in a multifaceted form a spheroid, so cells often show resistance and drugs show low efficacy.
      Cell Proliferation [
      • Chopra V.
      • Dinh T.V.
      • Hannigan E.V.
      Three-dimensional endothelial-tumor epithelial cell interactions in human cervical cancers.
      ,
      • Torisawa Y.S.
      • Shiku H.
      • Yasukawa T.
      • et al.
      Multi-channel 3-D cell culture device integrated on a silicon chip for anticancer drug sensitivity test.
      ]
      It has a higher growth rate than 3D cell culture but is limited.The rate of cell proliferation can be high or low and varies depending on the cell type and the 3D cell culture model, and it is possible to grow in a spheroid form.
      Viability
      • Bokhari M.
      • Carnachan R.J.
      • Cameron N.R.
      • et al.
      Culture of HepG2 liver cells on three dimensional polystyrene scaffolds enhances cell structure and function during toxicological challenge.
      Sensitive to cytotoxins.Insensitive to external stimuli.
      Apoptosis
      • Li C.L.
      • Tian T.
      • Nan K.J.
      • et al.
      Survival advantages of multicellular spheroids vs. monolayers of HepG2 cells in vitro.
      Very sensitive to drug-induced cell death.Improved +A1:C11tolerance to drug induced cell death stimulation.

      2.4 3D cell culture tools

      Methods for implementing 3D cell culture models have not yet been standardized. The 3D cell culture model may be developed using a scaffold-free method that allows cells to grow together without an ECM, and a scaffolding method that cultivates cells in the ECM space (Fig. 3) [
      • Lv D.
      • Hu Z.
      • Lu L.
      • et al.
      Three-dimensional cell culture: a powerful tool in tumor research and drug discovery.
      ]. The scaffold-free method is classified into four types depending on the method used to collect cells. Anti-adhesion methods prevent the adhesion of cells by coating the cell attachment material or flowing the culture media, including static suspension culture, spinner/rotational chamber culture, nano-pattern well culture, and magnetic levitation culture. The magnetic levitation method uses magnetic particles to aggregate cells. In this regard, there is the NanoShuttle™-PL product. Nanoshuttle™-PL is composed of gold, iron oxide, and poly-l-lysine, and magnetizes cells by electrostatically attaching to cell membranes during overnight incubation. NanoShuttle™-PL attached to the cell membrane in this way remains attached for up to 8 days and helps to form 3D spheroids by magnetic force. In addition, NanoShuttle™-PL is biocompatible, has no effect on metabolism, proliferation, and inflammatory stress, and does not interfere with experimental techniques such as fluorescence imaging and Western blotting []. In addition to adhesion prevention, hanging-drop cultures and U- or V-shaped well cultures are used where gravitational forces collect cells to form single spheroids; here, a 3D cell culture model is implemented by densely gathering cells in a narrow space (concave bottom of a water body, bottom surface of a U- or V-shaped well) to form a single cell mass (spheroid). The scaffold method can be divided into various types depending on the artificial polymers used, such as solid scaffolds or hydrogels, as an ECM. Structurally, they are divided into well, microchannel, and micropillar types, depending on the location of the ECM. As described, various models are being developed as 3D cell culture models to enable the application of various experimental parameters according to the purpose of the experiment. A description of each representative model is shown in Fig. 3, and comparisons of the 3D cell culture model of each model are shown in Table 2.
      Table 2Advantages and disadvantages of 3D cell culture models.
      Model (Type)BiocompatibilityConvenienceCommercialization
      Static Suspension★★★★★★
      Spinner/rotational Chamber★★★★★★
      Hang in drop★★
      U or V Shape well★★★★★★
      Nano Culture★★★★★★
      Magnetic Levitation
      Solid-Scaffold-in-well★★★★★★★
      Hydrogells-in-well★★★★★
      Hydrogells-on-micro pillar★★★★★★★
      Hydrogells-in-microchannel★★★
      Static suspension [
      • Lv D.
      • Hu Z.
      • Lu L.
      • et al.
      Three-dimensional cell culture: a powerful tool in tumor research and drug discovery.
      ] is the most common 3D cell culture model. Here, cells are aggregated above the surface of an agar or anti-adhesion coating to prevent cell adhesion to the bottom of the dish or well plate. Floating cell lines, such as leukemia and murine ascites tumor cells, can be cultured with this model without bottom coating.
      Spinner/rotation chamber [
      • Lv D.
      • Hu Z.
      • Lu L.
      • et al.
      Three-dimensional cell culture: a powerful tool in tumor research and drug discovery.
      ] is a 3D cell culture model that allows cells to float inside the media using a spinner or a rotational chamber to prevent the cells from adhering to the bottom. As the cells rotate in the culture media, cells with a high affinity adhere to each other through intercellular interactions. It is mainly used to induce cell metabolism or to mass-proliferate cells.
      Hang in drop [
      • Lv D.
      • Hu Z.
      • Lu L.
      • et al.
      Three-dimensional cell culture: a powerful tool in tumor research and drug discovery.
      ] is a 3D cell culture model in which a media droplet is hanging on the surface and cells in the media are aggregated in the lower part owing to gravity and cultured in a lump (spheroid).
      U- or V-shaped well [] is a 3D cell culture model in which cells are collected using gravity or centrifugal force on the bottom of a well with a U- or V-shape to cultivate cells in a lump (spheroid).
      Nanopatterned well [] is a 3D cell culture model in which nanopatterns are formed on the bottom of a well, and cells are cultured in a lump-like 3D cell without adhering to the bottom of the well.
      Magnetic levitation [
      • Giobbe G.G.
      • Crowley C.
      • Luni C.
      • et al.
      Extracellular matrix hydrogel derived from decellularized tissues enables endodermal organoid culture.
      ] is a 3D cell culture model in which cells are floated from the bottom of the well using magnet particles, and the cells are 3D cultured in a lump shape.
      Solid-scaffold-in-well [] is a 3D cell culture model in which a cell is placed on a microstructure and penetrates the microstructure and grows in a 3D structure.
      Hydrogels-in-well [
      • Lv D.
      • Hu Z.
      • Lu L.
      • et al.
      Three-dimensional cell culture: a powerful tool in tumor research and drug discovery.
      ] is a 3D cell culture model in which hydrogel, which changes its state depending on temperature or a specific ion, is mixed with cells and is distributed at the bottom of the well. The cells proliferate while forming a mass in the hydrogel.
      Hydrogel-on-micropillar [
      • Lee D.W.
      • Choi Y.S.
      • Seo Y.J.
      • et al.
      High-throughput screening (HTS) of anticancer drug efficacy on a micropillar/microwell chip platform.
      ] is a recently introduced technique and 3D cell culture model in which hydrogel-containing cells are immobilized on pillar surfaces and combined with well chips containing culture media.
      Hydrogels-in-microchannel [
      • Lv D.
      • Hu Z.
      • Lu L.
      • et al.
      Three-dimensional cell culture: a powerful tool in tumor research and drug discovery.
      ] is a 3D cell culture model in which hydrogels and cells are injected into a blood vessel-sized microchannel, and cells proliferate while forming a lump in the channel. In particular, this model shows excellent biocompatibility because it can flow a culture fluid or drug similar to blood vessels in vivo.

      2.5 Hydrogel scaffold

      The presence or absence of an ECM (scaffold) is a typical difference between 2D and 3D cell culture models. However, the effect of ECM composition on 3D cultured cells has not yet been standardized and defined. This uncertainty in ECM composition can affect the results of chemical and genetic screening studies using human 3D cultured cells [
      • Giobbe G.G.
      • Crowley C.
      • Luni C.
      • et al.
      Extracellular matrix hydrogel derived from decellularized tissues enables endodermal organoid culture.
      ,
      • Jee J.H.
      • Lee D.H.
      • Ko J.
      • et al.
      Development of collagen-based 3D matrix for gastrointestinal tract-derived organoid culture.
      ,
      • Cruz-Acuña R.
      • Quirós M.
      • Huang S.
      • et al.
      PEG-4MAL hydrogels for human organoid generation, culture, and in vivo delivery.
      ,
      • Ng S.
      • Tan W.J.
      • Pek M.M.X.
      • et al.
      Mechanically and chemically defined hydrogel matrices for patient-derived colorectal tumor organoid culture.
      ,
      • Gjorevski N.
      • Sachs N.
      • Manfrin A.
      • et al.
      Designer matrices for intestinal stem cell and organoid culture.
      ,
      • Broguiere N.
      • Isenmann L.
      • Hirt C.
      • et al.
      Growth of epithelial organoids in a defined hydrogel.
      ]. Therefore, it is important to study and understand the ECM. Among them, hydrogel scaffolds have numerous endogenous factors that promote the survival, proliferation, function, development of cells, and efficient chemical transfer. The structure and configuration of hydrogels can be tailored to achieve appropriate chemical, biological, and physical properties that promote specific structures outside the body, and provide an environment of endogenous signaling that promotes cellular interactions that underlie tissue or 3D cell formation. Hydrogel Scaffold has a variety of endogenous factors to promote cell survival, proliferation, and differentiation. In addition, migration can be observed by inducing dynamic 3D phenotypic changes of cells through cell-hydrogel interaction [
      • Cushing M.C.
      • Anseth K.S.
      Hydrogel cell cultures.
      ]. Each hydrogel scaffold differs in composition; hydrogels are inherently biocompatible because they are natural derivatives [
      • Dawson E.
      • Mapili G.
      • Erickson K.
      • et al.
      Biomaterials for stem cell differentiation.
      ]. Therefore, hydrogels are also used to 3D culture cells. The soft fibrous matrix provides appropriate physical parameters, and the Arg-Gly-Asp (RGD) adhesion domain is a key parameter for the formation of robust organisms [
      • Dawson E.
      • Mapili G.
      • Erickson K.
      • et al.
      Biomaterials for stem cell differentiation.
      ]. Along with the development of a 3D cell culture model, the hydrogel scaffold is also being studied for use in vitro experiments. Efforts are being made to overcome existing obstacles to implement a more robust human 3D cell culture model. Table 3 shows the types and characteristics of representative hydrogels currently available in the market.
      Table 3Types and properties of hydrogel scaffolds.
      Product Name (Type)Representation figureFeatures and Extraction
      Cultrex® 3D Culture Matrix® Collagen I , ,
      • Park D.W.
      • Choi D.S.
      • Ryu H.S.
      • et al.
      A well-defined in vitro three-dimensional culture of human endometrium and its applicability to endometrial cancer invasion.
      ,
      • Kokenyesi R.
      • Murray K.P.
      • Benshushan A.
      • et al.
      Invasion of interstitial matrix by a novel cell line from primary peritoneal carcinosarcoma, and by established ovarian carcinoma cell lines: role of cell-matrix adhesion molecules, proteinases, and E-cadherin expression.
      It is refined in the rat tail tendon and is provided at a high concentration that can polymerize to form hydrogel, and is a major structural component of extracellular matrix (ECM) found in connective tissue and internal organs.
      Corning® Matrigel® Basement Membrane Matrix [,]Soluble Hydrogel extracted from Engelbreth-Holm-Swarm (EHS) mice, laminin is the major component and has many growth factors.
      CultrexTM Basement Membrane Extracts (BME) ,
      • Chi H.C.
      • Tsai C.Y.
      • Wang C.S.
      • et al.
      DOCK6 promotes chemo- and radioresistance of gastric cancer by modulating WNT/β-catenin signaling and cancer stem cell traits.
      ,
      • Green Y.S.
      • Sargis T.
      • Reichert E.C.
      • et al.
      Hypoxia-Associated Factor (HAF) mediates neurofibromin ubiquitination and degradation leading to Ras-ERK pathway activation in hypoxia.
      Cultrex BMEs are a soluble form of basement membrane purified from Engelbreth-Holm-Swarm (EHS) tumor. Products include a full range of matrices and purified proteins including Basement Membrane Extracts, Laminin I, Collagen I, Collagen IV, Vitronectin, and Fibronectin.
      Glycosil® Hyaluronic Acid , ,
      • Maloney E.
      • Clark C.
      • Sivakumar H.
      • et al.
      Immersion bioprinting of tumor organoids in multi-well plates for increasing chemotherapy screening throughput.
      ,
      • Gaetani R.
      • Feyen D.A.
      • Verhage V.
      • et al.
      Epicardial application of cardiac progenitor cells in a 3D-printed gelatin/hyaluronic acid patch preserves cardiac function after myocardial infarction.
      Hyaluronic Acid is produced by a bacterial fermentation process through ISO 9001: 2000. It is a component of the natural extracellular matrix (ECM).
      PromoCell® 3D Cell Culture Matrix Kit (Alginate Hydrogel) , ,
      • Valenti F.
      • Sacconi A.
      • Ganci F.
      • et al.
      The miR-205-5p/BRCA1/RAD17 axis promotes genomic instability in head and neck squamous cell carcinomas.
      Alginate Hydrogel is based on Alginate, an anionic polysaccharide derived from algae cell walls, and provides cells with growth hormone and other ECM proteins as well as cells lacking cell attachment sites. In addition, three-dimensional cell cultivation is convenient by gelling calcium ions and liquefaction in the presence of calcium-chelating agents.
      Mebiol® Gel PNIPAAm-PEG 3D Thermoreversible Hydrogel , ,
      • Aeby E.A.
      • Misun P.M.
      • Hierlemann A.
      • et al.
      Microfluidic hydrogel hanging-drop network for long-term culturing of 3D microtissues and simultaneous high-resolution imaging.
      ,
      • Kanda K.
      • Sugama K.
      • Hayashida H.
      • et al.
      Eccentric exercise-induced delayed-onset muscle soreness and changes in markers of muscle damage and inflammation.
      PEG hydrogels are copolymers of poly (N-isopropylacrylamide) and poly (ethylene glycol) (PNIPAAm-PEG) for research purposes in the early 2000s. It is characterized by temperature reversible sol-gel transition and has efficient gaps for transmission, gas and material exchange, and protection of cellular tissue from the outside.
      Sigma F5386–1 G Fibrin from Human Plasma, Insoluble Powder ,
      • Dixon B.
      • Smith R.
      • Campbell D.J.
      • et al.
      The effect of etanercept on lung leukocyte margination and fibrin deposition after cardiac surgery.
      ,
      • Fischer C.P.
      • Bochicchio G.
      • Shen J.
      • et al.
      A prospective, randomized, controlled trial of the efficacy and safety of fibrin pad as an adjunct to control soft tissue bleeding during abdominal, retroperitoneal, pelvic, and thoracic surgery.
      Fibrin gel is gelled by Thrombin and can be used as Hydrogel Scaffold of Organoids.

      2.5.1 Spheroid model applications

      The spheroid culture system is a very useful three-dimensional cell culture method that can be applied variously in life science research. Unlike the cells in the conventional two-dimensional monolayer culture model, the spheroid model exhibits cell-cell or cell-ECM interactions, maintaining intrinsic phenotypic characteristics [
      • Murphy K.C.
      • Hoch A.I.
      • Harvestine J.N.
      • et al.
      Mesenchymal stem cell spheroids retain osteogenic phenotype through α2β1 signaling.
      ]. The spheroid model was first developed by Sutherland et al. in 1970 to study the functional phenotype of human tumor cells and the effects of radiation therapy [
      • Sutherland R.M.
      • Inch W.R.
      • McCredie J.A.
      • et al.
      A multi-component radiation survival curve using an in vitro tumour model.
      ,
      • Sutherland R.M.
      • McCredie J.A.
      • Inch W.R.
      Growth of multicell spheroids in tissue culture as a model of nodular carcinomas.
      ]. The spheroid model promotes the expression of stem cell markers, such as Oct-4 and Nanog, which influences cell proliferation, survival, and migration. It also secretes higher levels of cytokines and chemokines than those secreted by monolayer-cultured cells, leading to higher angiogenesis. The spheroid model can also better mimic the hypoxia model. In relation to hypoxia, it shows apoptotic resistance, improved viability, and secretion of angiogenic factors and chemokines [
      • Zhang Q.
      • Nguyen A.L.
      • Shi S.
      • et al.
      Three-dimensional spheroid culture of human gingiva-derived mesenchymal stem cells enhances mitigation of chemotherapy-induced oral mucositis.
      ,
      • Cheng N.C.
      • Chen S.Y.
      • Li J.R.
      • et al.
      Short-term spheroid formation enhances the regenerative capacity of adipose-derived stem cells by promoting stemness, angiogenesis, and chemotaxis.
      ]. The spheroid model can efficiently simulate the in vivo microenvironment of a tumor, and therefore, is a useful tool for researchers to predict drug efficacy in cancer research. Spheroids can also be used for stem cell research for developing embryonic bodies from induced pluripotent metaphase cells, as well as be used as high-purity neural stem cells for research on neurological diseases and related treatments. Recently, spheroid models have been used to study the cytotoxic effects of CAR-T cells, such as using the KILR® cytotoxicity assay developed by DiscoverX. Tumor spheroids transduced with CAR-T cells with KILR can be formed and cultured on the same microplate for spheroid culture, and antibody-mediated cytotoxicity (ADCC) effects can be analyzed with high uniformity and data reproducibility (Fig.4) [
      • Zhang B.L.
      • Qin D.Y.
      • Mo Z.M.
      • et al.
      Hurdles of CAR-T cell-based cancer immunotherapy directed against solid tumors.
      ,
      • Liu X.
      • Jiang S.
      • Fang C.
      • et al.
      Affinity-tuned ErbB2 or EGFR chimeric antigen receptor T cells exhibit an increased therapeutic index against tumors in mice.
      ].
      Fig 4
      Fig. 4ADCC assay with spheroid model KILR cell lines and KILR CD16 effector cells [
      • Zhang B.L.
      • Qin D.Y.
      • Mo Z.M.
      • et al.
      Hurdles of CAR-T cell-based cancer immunotherapy directed against solid tumors.
      ,
      • Liu X.
      • Jiang S.
      • Fang C.
      • et al.
      Affinity-tuned ErbB2 or EGFR chimeric antigen receptor T cells exhibit an increased therapeutic index against tumors in mice.
      ].

      2.5.2 Organoid model applications

      Recently, organoid model studies have focused on the development of organotypic culture models. The organoid has structural and functional similarities to actual organs such as the stomach and liver. An organoid model with excellent biocompatibility was implemented in vitro for use in disease research and drug development (Fig. 5). When assessing whether the organoids have been successfully established, it is important to analyze whether the organoids consist of adequate cell types and whether the organoids accurately mimic the functions of the corresponding tissue in vivo [
      • Zhao Z.
      • Chen X.
      • Dowbaj A.M.
      • et al.
      Organoids.
      ]. Although it is not possible to make an organoid by simple lump formation of a cell, the organoid uses a hydrogel-in-well model, which is a conventional 3D cell culture method. However, cells are cultured in a suitable ECM for a long period with appropriate differentiation or growth factors to produce an artificial organ with functions similar to those of human organs. Because the ultimate goal of a 3D cell culture model is to create a highly biocompatible model in vitro, the final stage of development is considered to be an organoid. Organoid studies are led by the Hans Clevers Group [
      • Huch M.
      • Gehart H.
      • van Boxtel R.
      • et al.
      Long-term culture of genome-stable bipotent stem cells from adult human liver.
      ] and have been actively pursuing research into application in precision medicine by developing a biocompatible model. Because the organoid model for producing organoids uses a highly viscous ECM, it is difficult to dispense a precise amount of cells and ECM using the conventional pipet used in biological experiments. High-throughput screening studies are performed using 384- or 1536- well plates. Therefore, since three-dimensional organoid samples and drugs are dispensed into numerous individual well plates, 3D bioprinting equipment capable of dispensing samples at high speed is required. The use of such 3D bioprinting equipment provides the researcher convenience, and a very small amount of liquid sample (less than a few nL) can be handled with very high uniformity and reproducibility. Therefore, it is possible to ensure high reproducibility of the experimental results. To solve this problem, the production of organoids now employs 3D bioprinting capable of precise liquid supply, as shown in Fig. 6 [
      • Lee H.J.
      Trends and prospects of 3D bioprinting technology, information and communication technology.
      ,
      • Schneeberger K.
      • Spee B.
      • Costa P.
      • et al.
      Converging biofabrication and organoid technologies: the next frontier in hepatic and intestinal tissue engineering?.
      ,
      • Mironov V.
      • Visconti R.P.
      • Kasyanov V.
      • et al.
      Organ printing: tissue spheroids as building blocks.
      ]. Recent clinical trials and studies have analyzed the efficacy of anticancer drugs in organoids developed from cancer patient-derived cells for precision treatment. A study aimed to measure the efficacy of anticancer drugs against patient-derived cancer cells and compare them with clinical data to select optimal anticancer drugs or to analyze the efficacy of new drugs (Fig. 7) [
      • Kim S.T.
      • Kim J.
      • Shin S.
      • et al.
      3-Dimensional micropillar drug screening identifies FGFR2-IIIC overexpression as a potential target in metastatic giant cell tumor.
      ]. In particular, when a patient-derived cell-based organoid model is used for research, it is possible to effectively represent the different genomic mutations, cancer cell characteristics, and tumor microenvironments of individual patients. Through the comparative analysis of patient-derived cancer organoid models and clinical data, cancer organoid models can represent actual clinical patients [
      • Lee D.W.
      • Choi S.Y.
      • Kim S.Y.
      • et al.
      A novel 3D pillar/well array platform using patient-derived head and neck tumor to predict the individual radioresponse.
      ,
      • Lee D.W.
      • Kim J.E.
      • Lee G.H.
      • et al.
      High-throughput 3D tumor spheroid array platform for evaluating sensitivity of proton-drug combinations.
      ,
      • Ratliff M.
      • Kim H.
      • Qi H.
      • et al.
      Patient-derived tumor organoids for guidance of personalized drug therapies in recurrent glioblastoma.
      ,
      • Kim H.
      • El-Khoury V.
      • Schulte N.
      • et al.
      Personalized functional profiling using ex-vivo patient-derived spheroids points out the potential of an antiangiogenic treatment in a patient with a metastatic lung atypical carcinoid.
      ].
      Fig 5
      Fig. 5A schematic diagram used in the treatment of diseases after organogenesis using an organoid model developed by extracting patient-derived cells (PDCs) from patients
      [
      • Zhao Z.
      • Chen X.
      • Dowbaj A.M.
      • et al.
      Organoids.
      ]
      .
      Fig 6
      Fig. 6Development of an organoid model using bioprinting
      [
      • Schneeberger K.
      • Spee B.
      • Costa P.
      • et al.
      Converging biofabrication and organoid technologies: the next frontier in hepatic and intestinal tissue engineering?.
      ]
      .
      Fig 7
      Fig. 7Example of analysis of anticancer drug efficacy using organoid models
      [
      • Kim S.T.
      • Kim J.
      • Shin S.
      • et al.
      3-Dimensional micropillar drug screening identifies FGFR2-IIIC overexpression as a potential target in metastatic giant cell tumor.
      ]
      .

      2.6 Commercialization trend of 3D cell culture market

      Table 4 shows that the global 3D cell culture market is projected to grow from $1.3 billion in 2022 to $2.6 billion in 2027, with an average annual growth rate of 15.6% between 2022 and 2027 [, , ].
      Table 4Domestic and world market size of 3D cell culture technology.115–117.
      DivisionCurrent Market Size (2022)Expected Market Size (2027)
      World Market SizeUSD 1.3 billionUSD 2.6 billion
      Expectancy [%]Expected market size of the global market is estimated to be 15.6%
      In particular, 3D cell culture models can be actively used to solve problems such as the increasing interest in personalized medicine and the increasing incidence of chronic diseases. However, there is still a lack of infrastructure for 3D cell culture model-based research, and the high cost of tumor biology research and clinical staging is expected to restrain the growth of this market. Currently, various 3D cell culture technologies from Corning, Insphero, and 3D Biomatrices are available in the market. Table 5 shows companies that sell 3D cell culture products.
      Table 5Representative companies of 3D cell culture technology.
      CompanyProduct nameRepresentation figureScaffold Type
      Corning , , , , , , Corning Spheroid microplates [,]Scaffold-Free
      Matrigel Matrix-3D Plates [,]Scaffold (Plate)
      Elplasia Plates Scaffold-Free
      Elplasia 12 K Flask [,]Scaffold-Free
      InSphero [,]3D Insight™ MicrotissuesScaffold-Free
      EMD Millipore Corporation [,]CellASIC ONIXScaffold (Micro fluidics)
      MBD [,]Cellvitro™Scaffold (Micro pillar)
      SPL [

      Solutions for Life Science Research & Clinical Diagnostics | MBL. https://www.mblintl.com/assets/NCP_Handbook.pdf.

      ,]
      SPL3D™Scaffold-free
      Organogenix
      • Ogino T.
      • Matsunaga N.
      • Tanaka T.
      • et al.
      Post-transcriptional repression of circadian component CLOCK regulates cancer-stemness in murine breast cancer cells.
      Nano Culture plateScaffold (Plate)
      Trevigen [,
      • Ramos R.I.
      • Bustos M.A.
      • Wu J.
      • et al.
      Upregulation of cell surface GD3 ganglioside phenotype is associated with human melanoma brain metastasis.
      , , ]
      Cultrex® 3D Spheroid Cell Invasion AssayScaffold (Plate)
      3D Biomatrix [
      • Barretina J.
      • Caponigro G.
      • Stransky N.
      • et al.
      The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity.
      ,
      • Smith S.C.
      • Baras A.S.
      • Lee J.K.
      • et al.
      The COXEN principle: translating signatures of in vitro chemosensitivity into tools for clinical outcome prediction and drug discovery in cancer.
      ]
      Perfecta3D® hanging drop plateScaffold-Free

      2.6.1 Corning [, , , , , , ]

      Corning sells spheroid microplates in either 96-, 384- or 1536- plate form (Fig. 8). Scaffold-free U-shaped wells use gravity to aggregate cells in one place for 3D cell culture. DU145 cell line, a human prostate cancer cell, was cultured in a corning spheroid 384-microplate, and then treated with doxorubicin for 24, 48, and 72 h to measure cell viability. In addition, drug efficacy can be quantitatively analyzed based on the dose-response curve (DRC) by treating the drug according to the concentration gradient. Although there is an advantage of enabling high-throughput screening (HTS) analysis using such 3D cultured cells, they are limited in that they cannot be cultured by mixing with the ECM (Fig. 9). In addition, corning sells various types of plate products such as Matrigel Matrix-3D Plates, Elplasia Plates, and Elplasia 12 K Flask for 3D cell culture experiments. By using Matrigel Matrix-3D Plates, polarized epithelial cells such as kidney, colon, lung, and mammary gland can create barriers separating the inside. Pre-coated Matrigel matrix-3D plates can be used to generate MDCK cysts for screening modulators of forskolininduced cyst swelling, which can be used to identify potential therapies for polycystic kidney disease (Fig. 10). [,] Corning Elplasia plates generate a high density of spheroids in a scaffold-free model using microcavity technology. The Elplasia plate is composed of individual U-bottom wells in the array well plate, so cells are cultured as 3D-spheroid without a scaffold (Fig. 11) []. Elplasia 12 K Flask consists of individual round wells with ultra-low attachment (ULA) surface on the bottom of the T-75 flask, so when cells are cultured, they form 3D-spheroids (Fig. 12) [,].
      Fig 8
      Fig. 8Corning spheroid microplates from Corning Inc. and 3D cell culture experiments .

      2.6.2 InSphero [,]

      InSphero has commercialized a 3D cell culture container with a product called Akura™ PLUS Hanging Drop System. It features a 96- or 384-plate culture vessel designed for HTS analysis based on the hanging drop method. First, perform 3D spheroid culture in hanging drop plates to aggregate and grow single cells into spheroid shapes. The generated 3D cultured cells are again moved to the “V shape” well so that various tests can be performed. Fig. 13 shows the protocol for transferring the sphere to the V-shaped well after cell culture and the results of the uniform formation of the sphere in the 384-well plates. The product is limited in that it is not useful for culturing by mixing various ECM and cells.

      2.6.3 EMD millipore corporation [,]

      EMD has commercialized a plate platform called CellASIC ONIX that can be used to 3D culture cells with ECMs using microfluidic technology. Fig. 14 illustrates the fluid control module that allows the culture and drug to flow through the plate. It is characterized by the use of microfluidic technology to provide biocompatibility by supplying continuous fluid to the cells. However, to continuously supply fluids, such as culture media and drugs, separate equipment, such as a fluid pump, is required. This has disadvantages in terms of cost and convenience during maintaining the culture.

      2.6.4 MBD (medical & bio decision) [
      • Lee D.W.
      • Choi Y.S.
      • Seo Y.J.
      • et al.
      High-throughput screening (HTS) of anticancer drug efficacy on a micropillar/microwell chip platform.
      ,]

      MBD has commercialized a 3D cell culture plate and chip called Cellvitro. Fig. 15 shows the development of a 3D cell culture chip with immobilized organoids and ECM on the micropillar structure of the MBD product and immersion of the micropillar in microwells for 3D cell culture. This is advantageous in that the culture liquid and drug can be easily replaced by moving the micropillar. This structure overcomes the difficulty of replacing the culture media, drug, and staining reagent in 3D cell culture models using ECM. The corresponding 3D cell culture chip was miniaturized to the size of a glass slide. This has the advantage of minimizing the consumption of organoid samples, expensive staining reagents, and drug samples required for experiments. In the case of organoid samples derived from patients, the amount that can be obtained is limited, and intrinsic genetic characteristics are lost when cultured in vitro for a long time to obtain excess samples. Therefore, the 3D cell culture chip is advantageous as it uses a very small amount of patient-derived organoid samples. Fig. 16 shows the drug efficacy analysis and genomic data obtained using the developed pillar/well chips.
      Fig 15
      Fig. 15The concept of pillar/well and the organoid-based high-throughput screening product from the product of CellVitroTM .
      Fig 16
      Fig. 16Analysis of drug efficacy and comparison with dielectric data of organoids Derived from Brain Tumor Patients Using Micro pillar/micro well chip, a product of CellVitroTM
      [
      • Lee D.W.
      • Choi Y.S.
      • Seo Y.J.
      • et al.
      High-throughput screening (HTS) of anticancer drug efficacy on a micropillar/microwell chip platform.
      ]
      .

      2.6.5 SPL [,]

      SPL has commercialized a 3D cell culture method using a scaffold that can fit into a well, called SPL3DTM. The achievement of a 3D cell culture environment similar to the in vivo environment makes it possible to maintain the physiological characteristics of cells in vitro and maximize the cell culture surface area through scaffold construction. In addition, a spheroid forming unit (SFU) based on suspension cell culture was designed to facilitate the generation of spheroids and size up the generated spheroids. The SFU lid is an air-permeable cap with a hydrophobic filter membrane that is designed to optimize cell uptake by allowing the necessary gas supply during cell culture (Fig. 17). However, it is limited in that it cannot be directly utilized for large-scale data analysis such as high-throughput drug discovery based on the 3D cell culture vessel.
      Fig 17
      Fig. 17SPL3DTM product that enables 3D cell cultivation by inserting a scaffold into SPL's 12-well plate, and the Spheroid Forming Unit (SFU) 15 ml tube which can suspense cell culture .

      2.6.6 Organogenix [

      Solutions for Life Science Research & Clinical Diagnostics | MBL. https://www.mblintl.com/assets/NCP_Handbook.pdf.

      ]

      Organogenix provides a nano culture plate (NCP) in a well plate (24, 96, 384) or dish. NCP is a scaffold-type 3D cell culture system without hydrogel or chemical pretreatment. Cells cultured in this system exhibit high proliferation characteristics and high uniformity, making them suitable for conventional HTS analysis. In addition, low binding/high binding allows the formation of a 3D cell monolayer (Fig. 18). Nano-Culture Plate 96-well plates are intended for the primary culture of cancer in tumor tissue. They are characterized by a shorter incubation period compared to collagen gel, the ability to experiment with the media exchange without containing gel components in an exclusive media, and the convenience of the multi-plate type to test the susceptibility to different anticancer drugs.
      Fig 18
      Fig. 18Organogenix NanoCulture Spheroid product and 3D cell culture image
      [

      Solutions for Life Science Research & Clinical Diagnostics | MBL. https://www.mblintl.com/assets/NCP_Handbook.pdf.

      ]
      .

      2.6.7 Trevigen [,,
      • Ogino T.
      • Matsunaga N.
      • Tanaka T.
      • et al.
      Post-transcriptional repression of circadian component CLOCK regulates cancer-stemness in murine breast cancer cells.
      ,
      • Ramos R.I.
      • Bustos M.A.
      • Wu J.
      • et al.
      Upregulation of cell surface GD3 ganglioside phenotype is associated with human melanoma brain metastasis.
      ]

      Trevigen commercialized the Cultrex® 3D culture spheroid cell invasion assay. The corresponding 3D cell culture model is a better representation of tumors in vivo. In particular, various physiological properties, such as similar morphology, cell-cell bond formation, decreased proliferation rate, increased tumor cell survival, tumor dormancy, and hypoxic core have been well implemented. By reflecting these characteristics in 3D culture cell invasion analysis, tumor invasion can be evaluated through a physiological approach. It also provides quantified visual analysis data through image-based analysis. The Cultrex® 3D spheroid cell invasion assay is performed using a special ECM that induces 3D cell aggregation and a 96-well plate for 3D cell culture. The use of a special ECM optimized to induce 3D cell invasion as a scaffold is an advantage of 3D cell culture model vessels. (Fig. 19).
      Fig 19
      Fig. 19Trevigen's Cultrex® 3D Culture Spheroid Cell Invasion Assay and 3D cell culture image .

      2.6.8 3D biomatrix [,]

      3D Biomatrix commercialized Perfecta3D® hanging drop plates (96, 384) easily aggregate cells into a single spoil using the hanging drop method. These products are sold commercially through Sigma-Aldrich product suppliers. The technique of making droplets by inserting a pipette tip into a hole and inserting cells and media is similar to Akura™ PLUS Hanging Drop System; however, the size and structure of the hole are different (Fig. 20). The product also has a limitation in that it cannot be cultured by mixing various ECM and 3D cultured cells.
      Fig 20
      Fig. 203D Biomatrix Perfecta3D hanging drop plates and graphs showing the change in spheroid size with cell number.

      2.7 Research applications of 3D cell culture

      2.7.1 Precision medicine

      Precision medicine refers to the analysis of multiple pieces of information, such as an individual's genomic and lifestyle information, to select patient-specific therapeutic agents. Previously, cell lines cultured in the form of a 2D monolayer were used to analyze the efficacy of cancer-related drugs [
      • Barretina J.
      • Caponigro G.
      • Stransky N.
      • et al.
      The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity.
      ,
      • Smith S.C.
      • Baras A.S.
      • Lee J.K.
      • et al.
      The COXEN principle: translating signatures of in vitro chemosensitivity into tools for clinical outcome prediction and drug discovery in cancer.
      ]. However, recently, many studies have been conducted on sophisticated drug efficacy analysis systems that can better predict drug efficacy in clinical practice by simulating the in vivo tumor microenvironment based on a 3D cell culture system and modeling real cancer patients. In addition, analyzing drug efficacy using patient-derived cells is less expensive and time-consuming compared to animal models or clinical studies, facing fewer ethical issues [
      • Sivaraman A.
      • Leach J.K.
      • Townsend S.
      • et al.
      A microscale in vitro physiological model of the liver: predictive screens for drug metabolism and enzyme induction.
      ,
      • Aparicio S.
      • Hidalgo M.
      • Kung A.L.
      Examining the utility of patient-derived xenograft mouse models.
      ]. By combining patient-derived cancer cells with a 3D cell culture model, molecular genomic profiling studies related to cancer drug discovery and tumor subtypes have been performed. In addition, these patient materials are accumulated along with data in the form of an organoid bank, which can ultimately open the door to preclinical screening of personalized drug candidate panels to improve low cancer treatment outcomes and by reducing side effects [
      • Edmondson R.
      • Broglie J.J.
      • Adcock A.F.
      • et al.
      Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors.
      ]. The 3D cell culture model is expected to provide a criterion for selecting the optimal therapeutic agent treatment by culturing patient-derived cells from the patient and analyzing the response to various drugs.

      2.7.2 New drug development

      Drug development involves the search for new drug candidates, non-clinical trials, and clinical trials for new drug candidates. Both non-clinical trials (animal trials) and clinical trials require considerable time and costs. For example, out of 50 million substances, approximately 250 new drug candidates enter non-clinical and clinical trials, from which only one new drug is considered to be at the market. The probability of new drug development has decreased recently. Drug development remains a time-consuming and expensive business because the success rate of drug development that has successfully reached the market through successful clinical trials is very low. Therefore, as the success rate of drug discovery remains low, the introduction of new 3D cell culture model technologies that can increase the accuracy of drug development and discovery is urgently needed. With low success rates in clinical trials, drug discovery remains a slow and expensive business. As attrition rates in drug discovery remain high, there is an urgent need for new technologies that provide better precision. Two of the most promising areas expected to improve the success rates of drug development are the advancement of precision medicine with the prospect of new biomarkers and more precise drug targets, and the availability of new preclinical models that better recapitulate in vivo microenvironmental factors. While traditional monolayer cultures are still predominant in cellular assays used for HTS, 3D cell culture model techniques for applications in drug discovery are making rapid progress [
      • van de Wetering M.
      • Francies H.E.
      • Francis J.M.
      • et al.
      Prospective derivation of a living organoid biobank of colorectal cancer patients.
      ,
      • Ryan S.L.
      • Baird A.M.
      • Vaz G.
      • et al.
      Drug discovery approaches utilizing three-dimensional cell culture.
      ,
      • Sittampalam S.
      • Eglen R.
      • Ferguson S.
      • et al.
      Three-dimensional cell culture assays: are they more predictive of in vivo efficacy than 2D monolayer cell-based assays?.
      ,
      • Montanez-Sauri S.I.
      • Beebe D.J.
      • Sung K.E.
      Microscale screening systems for 3D cellular microenvironments: platforms, advances, and challenges.
      ]. One of the most promising areas that are expected to improve the success rate of drug development is a novel preclinical model that summarizes the in vivo and intra-tumoral microenvironment to precisely analyze target biomarkers in addition to drug efficacy. In drug efficacy analysis studies using HTS, the existing 2D cell culture method is still predominantly applied; however, recently, many efforts have been made to graft 3D cell culture models for HTS analysis as well as image-based high content screening (HCS) analysis [
      • Lee S.-.Y.
      • Doh I.
      • Nam D.-.H.
      • et al.
      3D Cell-Based High-Content Screening (HCS) using a micropillar and microwell chip platform.
      ,
      • Lukonin I.
      • Zinner M.
      • Liberali P.
      Organoids in image-based phenotypic chemical screens.
      ,
      • Lee S.Y.
      • Teng Y.
      • Son M.
      • et al.
      Three-dimensional aggregated spheroid model of hepatocellular carcinoma using a 96-pillar/well plate.
      ,
      • Lee S.Y.
      • Doh I.
      • Lee D.W.
      A high throughput apoptosis assay using 3D cultured cells.
      ,
      • Lee S.-.Y.
      • Teng Y.
      • Son M.
      • et al.
      High-dose drug heat map analysis for drug safety and efficacy in multi-spheroid brain normal cells and GBM patient-derived cells.
      ]. Therefore, by changing the conventional 2D cell culture based HTS system to an HTS system using a 3D cell culture model, it can be more effective to identify potential drug candidates which are expected to dramatically reduce the astronomical costs and time for preclinical and clinical trials. There are examples of a unique ex vivo live tissue sensitivity assay (LTSA), in which precision-cut and uniform small tissue slices derived from pancreatic ductal adenocarcinoma PDX tumors were arrayed in a 96-well plate and screened against clinically relevant regimens within 3 to 5 days. The correlation between the sensitivity of the tissue section and the patient's clinical response and outcome was statistically analyzed. The LTSA assay results were further confirmed through in vitro biochemical methods and in vivo animal PDX models. The results of these PDX and LTSA methods reflect the response of clinical patients and can be used as a personalized strategy to improve the effectiveness of systemic treatment for pancreatic cancer patients, and can also be applied to the field of drug development to select efficacious anticancer drugs. [
      • Moreira L.
      • Bakir B.
      • Chatterji P.
      • et al.
      Pancreas 3D organoids: current and future aspects as a research platform for personalized medicine in pancreatic cancer.
      ,
      • Baker L.A.
      • Tiriac H.
      • Clevers H.
      • et al.
      Modeling pancreatic cancer with organoids.
      ,
      • Porter R.L.
      • Magnus N.K.C.
      • Thapar V.
      • et al.
      Epithelial to mesenchymal plasticity and differential response to therapies in pancreatic ductal adenocarcinoma.
      ,
      • Kang Y.
      • Deng J.
      • Ling J.
      • et al.
      3D imaging analysis on an organoid-based platform guides personalized treatment in pancreatic ductal adenocarcinoma.
      ]

      2.7.3 Drug repurposing

      Drug development has a very low success rate, and astronomical costs and time remain problematic. Therefore, efforts are being made to develop new drugs using drug-repurposing strategies. Drug repurposing (also called drug repositioning, reprofiling, or re-tasking) is a strategy for identifying new drug efficacy and use by expanding the range of medical indications for which a drug was originally approved [
      • Ashburn T.T.
      • Thor K.B.
      Drug repositioning: identifying and developing new uses for existing drugs.
      ]. This drug repurposing strategy offers several advantages over the development of an entirely new drug that targets a new indication. First, repurposed drugs are less likely to be discontinued owing to safety concerns in subsequent efficacy trials, as they have already been demonstrated to be sufficiently safe drugs in preclinical models and humans. Second, as the formulation for most preclinical and clinical trials in the safety evaluation stage has already been completed, it is possible to shorten the development period for further drug optimization and improvement to ensure safety. The time and cost required for phase 3 clinical trials are similar to those for drug development, but there may be significant cost and time savings in preclinical and phase 1 and phase 2 clinical trials [
      • Breckenridge A.
      • Jacob R.
      Overcoming the legal and regulatory barriers to drug repurposing.
      ]. The cost of bringing a repurposed drug to the market is estimated at $300 million on average, compared to an estimated $2–3 billion for new chemicals [
      • Nosengo N.
      Can you teach old drugs new tricks?.
      ]. Therefore, if the 3D-HTS analysis method is applied to quickly and accurately discover new targets, routes, and indications of repurposed drugs, it is expected to increase the drug development success rate compared to new drug development and dramatically reduce cost and time.

      2.7.4 Basic research

      3D cell culture models mimicking the living body are expected to be applied to various basic biological studies, such as drug mechanisms and cell differentiation in the human body. But there is still remains to be done to develop systems that can accurately mimic the conditions and disease-related pathologies in vivo. [
      • Shamir E.R.
      • Ewald A.J.
      Three-dimensional organotypic culture: experimental models of mammalian biology and disease.
      ,
      • Kretzschmar K.
      • Clevers H.
      Organoids: modeling development and the stem cell niche in a dish.
      ] An ideal 3D cell culture model simulates a disease-associated tissue-specific physiological or pathophysiological disease microenvironment in which cells can proliferate, aggregate, and differentiate [
      • Griffith L.G.
      • Swartz M.A.
      Capturing complex 3D tissue physiology in vitro.
      ]. Reactivity to drugs may vary depending on the composition of the ECM, the interaction between the ECM and cells, immunomodulatory molecules, and the action of immune cells as well as disease-related target cells [
      • Turley S.J.
      • Cremasco V.
      • Astarita J.L.
      Immunological hallmarks of stromal cells in the tumour microenvironment.
      ,
      • Johansson A.
      • Hamzah J.
      • Ganss R.
      More than a scaffold: stromal modulation of tumor immunity.
      ,
      • Stock K.
      • Estrada M.F.
      • Vidic S.
      • et al.
      Capturing tumor complexity in vitro: comparative analysis of 2D and 3D tumor models for drug discovery.
      ]. Therefore, it is very important to develop a 3D cell culture model considering that responsiveness to a wide range of drugs varies not only with specific cell lines or tumor types but also with the surrounding stroma. Thus, it may be possible to conduct research for efficient drug development and discovery, enabling a new pharmacological approach and development of a useful in vitro toxicity and efficacy testing platform.

      3. Conclusions

      In this paper, we summarized 3D cell culture-related technologies and commercially available products among these models and their application in preclinical and clinical research. To date, there is no standardized method for 3D cell culture model development because unlike in 2D cell culture model development, various methods can be implemented for 3D models depending on the ECM, structure of the culture vessel, characteristics of the cells used, and composition of the culture media. Researchers are still developing newer 3D cell culture models, and many attempts have been made to commercialize them. Although many products are on the market, there are still many obstacles to 3D cell culture models forming a market for consumption by life science researchers and healthcare workers. The obstacles that exist in research using 3D cell culture models can be categorized into the following two main categories.
      • 1.
        User-friendliness: The 3D cell culture model must be cultivated while forming a cell mass; therefore, care must be taken for the replacement of the culture media, maintenance of the ECM, or cross-contamination. These problems will require effort to simplify the experimental procedure by developing a culture container and a method that is easy to handle.
      • 2.
        Standardization of analysis: In the 2D cell culture, the activity of the cell is well standardized by the MTT assay or indirect measurement using the luminescence assay by measuring ATP. However, in 3D cell culture models, diffusion and permeability of existing MTT or ATP reagents differs for each cell mass. Currently, it is common to measure the size of a cell mass (spheroid) by staining. However, it is difficult to represent the activity of cells within a cell mass. Efforts to precisely measure the activity of cells in cell clusters using organization transparency (CLARITY) or confocal microscopes have been ongoing, but standardization of the technology has not yet been achieved. If these problems are resolved and the technology standardized, a biocompatible 3D cell culture model is expected to have a significant impact in the field of precision medicine, new drug bio-industry, and basic research.
      The field of 3D cell culture-based HTS has grown exponentially over the past few years. In particular, 3D drug efficacy evaluation and toxicity tests are in progress targeting various organ diseases ranging from skin, digestive system, respiratory system, heart and central nervous system diseases. In this regard, there are efforts to develop an optimized 3D culture model that can mimic a microenvironment similar to in vivo tissue microenvironment and disease pathology. Considering that there are approximately 300 biochemical constituents that are not cellular components only in the ECM, this is still a difficult task. However, if various 3D cell culture methods that can be used are developed and knowledge of biomedical engineering is used and applied, it is expected that 3D biomimetic platform-based drug development, discovery, and precision medical research may be achieved.

      Declaration of Competing Interest

      The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

      Acknowledgements

      This work was supported by the Gachon University research fund of 2022.(GCU- 202205760001). This work also was supported by the Ministry of Science and ICT, (Project Number: 2023-22030007-11, NIDS:1711179109) and Commercialization Promotion Agency for R&D Outcomes (COMPA).

      Appendix. Supplementary materials

      References

        • Harrison R.G.
        Observations on the living developing nerve fiber.
        Proc Soc Exp Biol Med. 1906; 4: 140-143
      1. Genomic and morphological changes of neuroblastoma cells in response to three-dimensional matrices.
        Tissue Eng. 2007; 13: 1035-1047
        • Huang G.S.
        • Tseng T.C.
        • Dai N.T.
        • et al.
        Fast isolation and expansion of multipotent cells from adipose tissue based on chitosan-selected primary culture.
        Biomaterials. 2015; 65: 154-162
        • Gurski L.A.
        • Petrelli N.J.
        • Jia X.
        • et al.
        3D matrices for anti-cancer drug testing and development.
        Oncology Issues. 2010; 25: 20-25
        • Wong H.L.
        • Wang M.X.
        • Cheung P.T.
        • et al.
        A 3D collagen microsphere culture system for GDNF-secreting HEK293 cells with enhanced protein productivity.
        Biomaterials. 2007; 28: 5369-5380
        • Wu M.-.H.
        • Chang Y.-.H.
        • Liu Y.-.T.
        • et al.
        Development of high throughput microfluidic cell culture chip for perfusion 3-dimensional cell culture-based chemosensitivity assay.
        Sens Actuators, B. 2011; 155: 397-407
        • Gurski L.A.
        • Jha A.K.
        • Zhang C.
        • et al.
        Hyaluronic acid-based hydrogels as 3D matrices for in vitro evaluation of chemotherapeutic drugs using poorly adherent prostate cancer cells.
        Biomaterials. 2009; 30: 6076-6085
        • Pauli C.
        • Hopkins B.D.
        • Prandi D.
        • et al.
        Personalized in vitro and in vivo cancer models to guide precision medicine.
        Cancer Discov. 2017; 7: 462-477
        • Broutier L.
        • Mastrogiovanni G.
        • Verstegen M.M.
        • et al.
        Human primary liver cancer-derived organoid cultures for disease modeling and drug screening.
        Nat Med. 2017; 23: 1424-1435
        • Kim J.
        • Koo B.K.
        • Knoblich J.A.
        Human organoids: model systems for human biology and medicine.
        Nat Rev Mol Cell Biol. 2020; 21: 571-584
        • Fatehullah A.
        • Tan S.H.
        • Barker N.
        Organoids as an in vitro model of human development and disease.
        Nat Cell Biol. 2016; 18: 246-254
        • Sato T.
        • Vries R.G.
        • Snippert H.J.
        • et al.
        Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche.
        Nature. 2009; 459: 262-265
        • Sato T.
        • Stange D.E.
        • Ferrante M.
        • et al.
        Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett's epithelium.
        Gastroenterology. 2011; 141: 1762-1772
        • Lancaster M.A.
        • Renner M.
        • Martin C.A.
        • et al.
        Cerebral organoids model human brain development and microcephaly.
        Nature. 2013; 501: 373-379
        • Takasato M.
        • Er P.X.
        • Chiu H.S.
        • et al.
        Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis.
        Nature. 2015; 526: 564-568
        • Fujii M.
        • Matano M.
        • Toshimitsu K.
        • et al.
        Human intestinal organoids maintain self-renewal capacity and cellular diversity in niche-inspired culture condition.
        Cell Stem Cell. 2018; 23 (e6): 787-793
        • Hu H.
        • Gehart H.
        • Artegiani B.
        • et al.
        Long-Term Expansion of Functional Mouse and Human Hepatocytes as 3D Organoids.
        Cell. 2018; 175 (e19): 1591-1606
        • Sachs N.
        • Papaspyropoulos A.
        • Zomer-van Ommen D.D.
        • et al.
        Long-term expanding human airway organoids for disease modeling.
        Embo J. 2019; : 38
        • Lovitt C.J.
        • Shelper T.B.
        • Avery V.M.
        Advanced cell culture techniques for cancer drug discovery.
        Biology (Basel). 2014; 3: 345-367
        • Simian M.
        • Bissell M.J
        Organoids: a historical perspective of thinking in three dimensions.
        J Cell Biol. 2017; 216: 31-40
        • Gilazieva Z.
        • Ponomarev A.
        • Rutland C.
        • et al.
        Promising applications of tumor spheroids and organoids for personalized medicine.
        Cancers (Basel). 2020; : 12
        • Gunti S.
        • Hoke A.T.K.
        • Vu K.P.
        • et al.
        Organoid and spheroid tumor models: techniques and applications.
        Cancers (Basel). 2021; : 13
        • Zanoni M.
        • Cortesi M.
        • Zamagni A.
        • et al.
        Modeling neoplastic disease with spheroids and organoids.
        J Hematol Oncol. 2020; 13: 97
        • Nunes A.S.
        • Barros A.S.
        • Costa E.C.
        • et al.
        3D tumor spheroids as in vitro models to mimic in vivo human solid tumors resistance to therapeutic drugs.
        Biotechnol Bioeng. 2019; 116: 206-226
        • Nath S.
        • Devi G.R.
        Three-dimensional culture systems in cancer research: focus on tumor spheroid model.
        Pharmacol Ther. 2016; 163: 94-108
        • McCauley H.A.
        • Wells J.M.
        Pluripotent stem cell-derived organoids: using principles of developmental biology to grow human tissues in a dish.
        Development. 2017; 144: 958-962
        • Spence J.R.
        • Mayhew C.N.
        • Rankin S.A.
        • et al.
        Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro.
        Nature. 2011; 470: 105-109
        • Bartfeld S.
        • Bayram T.
        • van de Wetering M.
        • et al.
        In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection.
        Gastroenterology. 2015; 148 (e6): 126-136
        • Garcez P.P.
        • Loiola E.C.
        • Madeiro da Costa R.
        • et al.
        Zika virus impairs growth in human neurospheres and brain organoids.
        Science. 2016; 352: 816-818
        • Takebe T.
        • Sekine K.
        • Enomura M.
        • et al.
        Vascularized and functional human liver from an iPSC-derived organ bud transplant.
        Nature. 2013; 499: 481-484
        • Dutta D.
        • Heo I.
        • Clevers H.
        Disease modeling in stem cell-derived 3D organoid systems.
        Trends Mol Med. 2017; 23: 393-410
        • Clevers H.
        Modeling development and disease with organoids.
        Cell. 2016; 165: 1586-1597
        • Lin Y.C.
        • Murayama Y.
        • Hashimoto K.
        • et al.
        Role of tumor suppressor genes in the cancer-associated reprogramming of human induced pluripotent stem cells.
        Stem Cell Res Ther. 2014; 5: 58
        • Al-Hajj M.
        • Wicha M.S.
        • Benito-Hernandez A.
        • et al.
        Prospective identification of tumorigenic breast cancer cells.
        Proc Natl Acad Sci U S A. 2003; 100: 3983-3988
        • Singh S.K.
        • Clarke I.D.
        • Terasaki M.
        • et al.
        Identification of a cancer stem cell in human brain tumors.
        Cancer Res. 2003; 63: 5821-5828
        • Bonnet D.
        • Dick J.E.
        Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell.
        Nat Med. 1997; 3: 730-737
        • Schutgens F.
        • Clevers H.
        Human organoids: tools for understanding biology and treating diseases.
        Annu Rev Pathol: Mech Dis. 2020; 15: 211-234
        • Baillargeon P.
        • Shumate J.
        • Hou S.
        • et al.
        Automating a magnetic 3D spheroid model technology for high-throughput screening.
        SLAS Technol. 2019; 24: 420-428
        • Fernandez-Vega V.
        • Hou S.
        • Plenker D.
        • et al.
        Lead identification using 3D models of pancreatic cancer.
        SLAS Discov. 2022; 27: 159-166
        • Broutier L.
        • Andersson-Rolf A.
        • Hindley C.J.
        • et al.
        Culture and establishment of self-renewing human and mouse adult liver and pancreas 3D organoids and their genetic manipulation.
        Nat Protoc. 2016; 11: 1724-1743
        • Moreira L.
        • Bakir B.
        • Chatterji P.
        • et al.
        Pancreas 3D organoids: current and future aspects as a research platform for personalized medicine in pancreatic cancer.
        Cell Mol Gastroenterol Hepatol. 2018; 5: 289-298
        • Hou S.
        • Tiriac H.
        • Sridharan B.P.
        • et al.
        Advanced development of primary pancreatic organoid tumor models for high-throughput phenotypic drug screening.
        SLAS Discov. 2018; 23: 574-584
        • Caleffi J.T.
        • Aal M.C.E.
        • Gallindo H.O.M.
        • et al.
        Magnetic 3D cell culture: state of the art and current advances.
        Life Sci. 2021; 286120028
        • Antoni D.
        • Burckel H.
        • Josset E.
        • et al.
        Three-dimensional cell culture: a breakthrough in vivo.
        Int J Mol Sci. 2015; 16: 5517-5527
        • Khetan S.
        • Burdick J.A.
        Patterning network structure to spatially control cellular remodeling and stem cell fate within 3-dimensional hydrogels.
        Biomaterials. 2010; 31: 8228-8234
        • Kim J.B.
        Three-dimensional tissue culture models in cancer biology.
        Semin Cancer Biol. 2005; 15: 365-377
        • Yip D.
        • Cho C.H.
        A multicellular 3D heterospheroid model of liver tumor and stromal cells in collagen gel for anti-cancer drug testing.
        Biochem Biophys Res Commun. 2013; 433: 327-332
        • Lee J.
        • Cuddihy M.J.
        • Kotov N.A.
        Three-dimensional cell culture matrices: state of the art.
        Tissue Eng Part B Rev. 2008; 14: 61-86
        • Bissell M.J.
        • Rizki A.
        • Mian I.S.
        Tissue architecture: the ultimate regulator of breast epithelial function.
        Curr Opin Cell Biol. 2003; 15: 753-762
        • Chitcholtan K.
        • Asselin E.
        • Parent S.
        • et al.
        Differences in growth properties of endometrial cancer in three dimensional (3D) culture and 2D cell monolayer.
        Exp Cell Res. 2013; 319: 75-87
        • Birgersdotter A.
        • Sandberg R.
        • Ernberg I.
        Gene expression perturbation in vitro–a growing case for three-dimensional (3D) culture systems.
        Semin Cancer Biol. 2005; 15: 405-412
        • Gómez-Lechón M.J.
        • Jover R.
        • Donato T.
        • et al.
        Long-term expression of differentiated functions in hepatocytes cultured in three-dimensional collagen matrix.
        J Cell Physiol. 1998; 177: 553-562
        • Semino C.E.
        • Merok J.R.
        • Crane G.G.
        • et al.
        Functional differentiation of hepatocyte-like spheroid structures from putative liver progenitor cells in three-dimensional peptide scaffolds.
        Differentiation. 2003; 71: 262-270
        • Ghosh S.
        • Spagnoli G.C.
        • Martin I.
        • et al.
        Three-dimensional culture of melanoma cells profoundly affects gene expression profile: a high density oligonucleotide array study.
        J Cell Physiol. 2005; 204: 522-531
        • Schyschka L.
        • Sánchez J.J.
        • Wang Z.
        • et al.
        Hepatic 3D cultures but not 2D cultures preserve specific transporter activity for acetaminophen-induced hepatotoxicity.
        Arch Toxicol. 2013; 87: 1581-1593
        • Elkayam T.
        • Amitay-Shaprut S.
        • Dvir-Ginzberg M.
        • et al.
        Enhancing the drug metabolism activities of C3A–a human hepatocyte cell line–by tissue engineering within alginate scaffolds.
        Tissue Eng. 2006; 12: 1357-1368
        • Bokhari M.
        • Carnachan R.J.
        • Cameron N.R.
        • et al.
        Culture of HepG2 liver cells on three dimensional polystyrene scaffolds enhances cell structure and function during toxicological challenge.
        J Anat. 2007; 211: 567-576
        • Chopra V.
        • Dinh T.V.
        • Hannigan E.V.
        Three-dimensional endothelial-tumor epithelial cell interactions in human cervical cancers.
        In Vitro Cell Dev Biol Anim. 1997; 33: 432-442
        • Torisawa Y.S.
        • Shiku H.
        • Yasukawa T.
        • et al.
        Multi-channel 3-D cell culture device integrated on a silicon chip for anticancer drug sensitivity test.
        Biomaterials. 2005; 26: 2165-2172
        • Li C.L.
        • Tian T.
        • Nan K.J.
        • et al.
        Survival advantages of multicellular spheroids vs. monolayers of HepG2 cells in vitro.
        Oncol Rep. 2008; 20: 1465-1471
      2. SIGMA-ALDRICH. https://www.sigmaaldrich.com/technical-documents/articles/biology/3d-cell-culture-technology.html#Overview.

        • Lv D.
        • Hu Z.
        • Lu L.
        • et al.
        Three-dimensional cell culture: a powerful tool in tumor research and drug discovery.
        Oncol Lett. 2017; 14: 6999-7010
      3. NanoShuttle™-PL. https://cdn.thomassci.com/FetchFile.ashx?id=aa252cf0-dec2-44c9-a253-c009cf593eb5

      4. Elveflow: High performance – Elvesys. https://www.elveflow.com/organs-on-chip/3d-cell-culture-methods-and-applications-a-short-review/.

      5. genetic engineering & biotechnology news. https://www.genengnews.com/gen-articles/supporting-cells-with-scaffold-technology/3812?page=2.

        • Lee D.W.
        • Choi Y.S.
        • Seo Y.J.
        • et al.
        High-throughput screening (HTS) of anticancer drug efficacy on a micropillar/microwell chip platform.
        Anal Chem. 2014; 86: 535-542
        • Giobbe G.G.
        • Crowley C.
        • Luni C.
        • et al.
        Extracellular matrix hydrogel derived from decellularized tissues enables endodermal organoid culture.
        Nat Commun. 2019; 10: 5658
        • Jee J.H.
        • Lee D.H.
        • Ko J.
        • et al.
        Development of collagen-based 3D matrix for gastrointestinal tract-derived organoid culture.
        Stem Cells Int. 2019; 20198472712
        • Cruz-Acuña R.
        • Quirós M.
        • Huang S.
        • et al.
        PEG-4MAL hydrogels for human organoid generation, culture, and in vivo delivery.
        Nat Protoc. 2018; 13: 2102-2119
        • Ng S.
        • Tan W.J.
        • Pek M.M.X.
        • et al.
        Mechanically and chemically defined hydrogel matrices for patient-derived colorectal tumor organoid culture.
        Biomaterials. 2019; 219119400
        • Gjorevski N.
        • Sachs N.
        • Manfrin A.
        • et al.
        Designer matrices for intestinal stem cell and organoid culture.
        Nature. 2016; 539: 560-564
        • Broguiere N.
        • Isenmann L.
        • Hirt C.
        • et al.
        Growth of epithelial organoids in a defined hydrogel.
        Adv Mater. 2018; 30e1801621
        • Cushing M.C.
        • Anseth K.S.
        Hydrogel cell cultures.
        Science. 2007; 316: 1133-1134
        • Dawson E.
        • Mapili G.
        • Erickson K.
        • et al.
        Biomaterials for stem cell differentiation.
        Adv Drug Deliv Rev. 2008; 60: 215-228
      6. Trevigen Joins the Bio-Techne Family. https://www.bio-techne.com/reagents/cell-culture-reagents-trevigen.

      7. Sigma-Aldrich Products - Life Science. https://www.sigmaaldrich.com/KR/ko/technical-documents/protocol/cell-culture-and-cell-culture-analysis/3d-cell-culture/3d-culture-matrix-rat-collagen-i.

        • Park D.W.
        • Choi D.S.
        • Ryu H.S.
        • et al.
        A well-defined in vitro three-dimensional culture of human endometrium and its applicability to endometrial cancer invasion.
        Cancer Lett. 2003; 195: 185-192
        • Kokenyesi R.
        • Murray K.P.
        • Benshushan A.
        • et al.
        Invasion of interstitial matrix by a novel cell line from primary peritoneal carcinosarcoma, and by established ovarian carcinoma cell lines: role of cell-matrix adhesion molecules, proteinases, and E-cadherin expression.
        Gynecol Oncol. 2003; 89: 60-72
      8. Corning. https://ecatalog.corning.com/life-sciences/b2c/US/en/Surfaces/Extracellular-Matrices-ECMs/Corning%C2%AE-Matrigel%C2%AE-Matrix/p/356234.

      9. Corning. https://www.corning.com/worldwide/en/products/life-sciences/resources/stories/at-the-bench/organoid-vs-spheroid-what-is-the-difference.html.

      10. Cultrex BME & ECM Products From R&D Systems. https://www.rndsystems.com/products/cultrex-basement-membrane-extract-bme-and-extracellular-matrices-ecm.

        • Chi H.C.
        • Tsai C.Y.
        • Wang C.S.
        • et al.
        DOCK6 promotes chemo- and radioresistance of gastric cancer by modulating WNT/β-catenin signaling and cancer stem cell traits.
        Oncogene. 2020; 39: 5933-5949
        • Green Y.S.
        • Sargis T.
        • Reichert E.C.
        • et al.
        Hypoxia-Associated Factor (HAF) mediates neurofibromin ubiquitination and degradation leading to Ras-ERK pathway activation in hypoxia.
        Mol Cancer Res. 2019; 17: 1220-1232
      11. Advanced BioMatrix. https://advancedbiomatrix.com/glycosil.html.

      12. ESI BIO. https://esibio.com/.

        • Maloney E.
        • Clark C.
        • Sivakumar H.
        • et al.
        Immersion bioprinting of tumor organoids in multi-well plates for increasing chemotherapy screening throughput.
        Micromachines (Basel). 2020; : 11
        • Gaetani R.
        • Feyen D.A.
        • Verhage V.
        • et al.
        Epicardial application of cardiac progenitor cells in a 3D-printed gelatin/hyaluronic acid patch preserves cardiac function after myocardial infarction.
        Biomaterials. 2015; 61: 339-348
      13. Promocell. https://www.promocell.com/product/3d-cell-culture-matrix-kit-alginate-hydrogel/.

      14. Cambridge Bioscience. https://www.bioscience.co.uk/product~786282.

        • Valenti F.
        • Sacconi A.
        • Ganci F.
        • et al.
        The miR-205-5p/BRCA1/RAD17 axis promotes genomic instability in head and neck squamous cell carcinomas.
        Cancers (Basel). 2019; : 11
      15. Cosmo Bio. https://www.cosmobio.com/en/.

      16. Advanced BioMatrix. https://advancedbiomatrix.com/mebiol-thermoreversible.html.

        • Aeby E.A.
        • Misun P.M.
        • Hierlemann A.
        • et al.
        Microfluidic hydrogel hanging-drop network for long-term culturing of 3D microtissues and simultaneous high-resolution imaging.
        Adv Biosyst. 2018; 21800054
        • Kanda K.
        • Sugama K.
        • Hayashida H.
        • et al.
        Eccentric exercise-induced delayed-onset muscle soreness and changes in markers of muscle damage and inflammation.
        Exerc Immunol Rev. 2013; 19: 72-85
      17. Sigma-Aldrich Products - Life Science. https://www.sigmaaldrich.com/catalog/product/sigma/f5386?lang=ko&region=KR.

        • Dixon B.
        • Smith R.
        • Campbell D.J.
        • et al.
        The effect of etanercept on lung leukocyte margination and fibrin deposition after cardiac surgery.
        Am J Respir Crit Care Med. 2013; 188: 751-754
        • Fischer C.P.
        • Bochicchio G.
        • Shen J.
        • et al.
        A prospective, randomized, controlled trial of the efficacy and safety of fibrin pad as an adjunct to control soft tissue bleeding during abdominal, retroperitoneal, pelvic, and thoracic surgery.
        J Am Coll Surg. 2013; 217: 385-393
        • Murphy K.C.
        • Hoch A.I.
        • Harvestine J.N.
        • et al.
        Mesenchymal stem cell spheroids retain osteogenic phenotype through α2β1 signaling.
        Stem Cells Transl Med. 2016; 5: 1229-1237
        • Sutherland R.M.
        • Inch W.R.
        • McCredie J.A.
        • et al.
        A multi-component radiation survival curve using an in vitro tumour model.
        Int J Radiat Biol Relat Stud Phys Chem Med. 1970; 18: 491-495
        • Sutherland R.M.
        • McCredie J.A.
        • Inch W.R.
        Growth of multicell spheroids in tissue culture as a model of nodular carcinomas.
        J Natl Cancer Inst. 1971; 46: 113-120
        • Zhang Q.
        • Nguyen A.L.
        • Shi S.
        • et al.
        Three-dimensional spheroid culture of human gingiva-derived mesenchymal stem cells enhances mitigation of chemotherapy-induced oral mucositis.
        Stem Cells Dev. 2012; 21: 937-947
        • Cheng N.C.
        • Chen S.Y.
        • Li J.R.
        • et al.
        Short-term spheroid formation enhances the regenerative capacity of adipose-derived stem cells by promoting stemness, angiogenesis, and chemotaxis.
        Stem Cells Transl Med. 2013; 2: 584-594
        • Zhang B.L.
        • Qin D.Y.
        • Mo Z.M.
        • et al.
        Hurdles of CAR-T cell-based cancer immunotherapy directed against solid tumors.
        Sci China Life Sci. 2016; 59: 340-348
        • Liu X.
        • Jiang S.
        • Fang C.
        • et al.
        Affinity-tuned ErbB2 or EGFR chimeric antigen receptor T cells exhibit an increased therapeutic index against tumors in mice.
        Cancer Res. 2015; 75: 3596-3607
        • Zhao Z.
        • Chen X.
        • Dowbaj A.M.
        • et al.
        Organoids.
        Nat Rev Methods Primers. 2022; 2: 94
        • Huch M.
        • Gehart H.
        • van Boxtel R.
        • et al.
        Long-term culture of genome-stable bipotent stem cells from adult human liver.
        Cell. 2015; 160: 299-312
        • Lee H.J.
        Trends and prospects of 3D bioprinting technology, information and communication technology.
        Promotion Center. 2017; (10.13140Working Paper (PDF Available))
        • Schneeberger K.
        • Spee B.
        • Costa P.
        • et al.
        Converging biofabrication and organoid technologies: the next frontier in hepatic and intestinal tissue engineering?.
        Biofabrication. 2017; 9013001
        • Mironov V.
        • Visconti R.P.
        • Kasyanov V.
        • et al.
        Organ printing: tissue spheroids as building blocks.
        Biomaterials. 2009; 30: 2164-2174
        • Kim S.T.
        • Kim J.
        • Shin S.
        • et al.
        3-Dimensional micropillar drug screening identifies FGFR2-IIIC overexpression as a potential target in metastatic giant cell tumor.
        Oncotarget. 2017; 8: 36484-36491
        • Lee D.W.
        • Choi S.Y.
        • Kim S.Y.
        • et al.
        A novel 3D pillar/well array platform using patient-derived head and neck tumor to predict the individual radioresponse.
        Transl Oncol. 2022; 24101483
        • Lee D.W.
        • Kim J.E.
        • Lee G.H.
        • et al.
        High-throughput 3D tumor spheroid array platform for evaluating sensitivity of proton-drug combinations.
        Int J Mol Sci. 2022; : 23
        • Ratliff M.
        • Kim H.
        • Qi H.
        • et al.
        Patient-derived tumor organoids for guidance of personalized drug therapies in recurrent glioblastoma.
        Int J Mol Sci. 2022; 23: 6572
        • Kim H.
        • El-Khoury V.
        • Schulte N.
        • et al.
        Personalized functional profiling using ex-vivo patient-derived spheroids points out the potential of an antiangiogenic treatment in a patient with a metastatic lung atypical carcinoid.
        Cancer Biol Ther. 2022; 23: 96-102
      18. MarketResearch. https://www.marketresearch.com/MarketsandMarkets-v3719/3D-Cell-Culture-Type-Hydrogel-31623529/.

      19. PRWeb. https://www.prweb.com/releases/2022/04/prweb18612869.htm.

      20. Grand View Research. https://www.grandviewresearch.com/industry-analysis/3d-cell-culture-market.

      21. Corning. http://www.corning.com/catalog/cls/documents/protocols/CLS-AN-235.pdf.

      22. InSphero - Unrivaled Biological InSight. https://insphero.com/?gclid=CjwKCAjwpKyYBhB7EiwAU2Hn2TkXAtwHE0l9rsaz6bhmWFLXY9212YmHn1xp7DATPdrflrFPjEtYWRoCoagQAvD_BwE.

      23. https://www.corning.com/worldwide/en/products/life-sciences/products/surfaces/matrigel-matrix-3d-plates.html.

      24. https://www.corning.com/catalog/cls/documents/application-notes/CLS-AN-572-A4.pdf.

      25. https://www.corning.com/worldwide/en/products/life-sciences/products/microplates/elplasia-plates.html.

      26. https://www.corning.com/worldwide/en/products/life-sciences/products/cell-Culture/corning-elplasia-12k-flask.html.

      27. https://www.corning.com/catalog/cls/documents/product-information-sheets/CLS-AC-035.pdf.

      28. https://insphero.com/wp-content/uploads/2022/02/Akura-PLUS-Hanging-Drop-System-Technical-Specifications_FEB22.pdf.

      29. InSphero - Unrivaled Biological InSight. https://insphero.com/?gclid=CjwKCAjwpKyYBhB7EiwAU2Hn2TkXAtwHE0l9rsaz6bhmWFLXY9212YmHn1xp7DATPdrflrFPjEtYWRoCoagQAvD_BwE.

      30. Imperial College London. https://www.imperial.ac.uk/media/imperial-college/medicine/facilities/film/CellAsics-Onix-Microfluidic-Platform-Brochure.pdf.

      31. Merck Millipore. https://www.merckmillipore.com/KR/ko/life-science-research/cell-culture-systems/5EOb.qB.8DAAAAE_YkJ3.Mmg,nav?ReferrerURL=https%3A%2F%2Fwww.google.com%2F&bd=1.

      32. MBD. http://www.mbdbiotech.com/.

      33. SPL Life Sciences. http://www.spllifesciences.com/en/.

      34. SPL Life Sciences. http://ispl.co.kr/m21.php?cate=1&idx=347.

      35. Solutions for Life Science Research & Clinical Diagnostics | MBL. https://www.mblintl.com/assets/NCP_Handbook.pdf.

      36. TheWell Bioscience. https://www.thewellbio.com/applications/invasion-assay/?gclid=EAIaIQobChMIy5WUl73r-QIVAa6WCh0oeAdIEAAYASAAEgJ4pfD_BwE.

        • Ogino T.
        • Matsunaga N.
        • Tanaka T.
        • et al.
        Post-transcriptional repression of circadian component CLOCK regulates cancer-stemness in murine breast cancer cells.
        Elife. 2021; : 10
        • Ramos R.I.
        • Bustos M.A.
        • Wu J.
        • et al.
        Upregulation of cell surface GD3 ganglioside phenotype is associated with human melanoma brain metastasis.
        Mol Oncol. 2020; 14: 1760-1778
      37. Volt Tecnologia: Citometria de Fluxo e Elispot. http://www.volttecnologia.com.br/pdfs/3D/Hanging%20Drop%20Plates.pdf.

      38. [[1D Biomatrix. http://www.3dbiomatrix.com/.

        • Barretina J.
        • Caponigro G.
        • Stransky N.
        • et al.
        The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity.
        Nature. 2012; 483: 603-607
        • Smith S.C.
        • Baras A.S.
        • Lee J.K.
        • et al.
        The COXEN principle: translating signatures of in vitro chemosensitivity into tools for clinical outcome prediction and drug discovery in cancer.
        Cancer Res. 2010; 70: 1753-1758
        • Sivaraman A.
        • Leach J.K.
        • Townsend S.
        • et al.
        A microscale in vitro physiological model of the liver: predictive screens for drug metabolism and enzyme induction.
        Curr Drug Metab. 2005; 6: 569-591
        • Aparicio S.
        • Hidalgo M.
        • Kung A.L.
        Examining the utility of patient-derived xenograft mouse models.
        Nat Rev Cancer. 2015; 15: 311-316
        • Edmondson R.
        • Broglie J.J.
        • Adcock A.F.
        • et al.
        Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors.
        Assay Drug Dev Technol. 2014; 12: 207-218
        • van de Wetering M.
        • Francies H.E.
        • Francis J.M.
        • et al.
        Prospective derivation of a living organoid biobank of colorectal cancer patients.
        Cell. 2015; 161: 933-945
        • Ryan S.L.
        • Baird A.M.
        • Vaz G.
        • et al.
        Drug discovery approaches utilizing three-dimensional cell culture.
        Assay Drug Dev Technol. 2016; 14: 19-28
        • Sittampalam S.
        • Eglen R.
        • Ferguson S.
        • et al.
        Three-dimensional cell culture assays: are they more predictive of in vivo efficacy than 2D monolayer cell-based assays?.
        Assay Drug Dev Technol. 2015; 13: 254-261
        • Montanez-Sauri S.I.
        • Beebe D.J.
        • Sung K.E.
        Microscale screening systems for 3D cellular microenvironments: platforms, advances, and challenges.
        Cell Mol Life Sci. 2015; 72: 237-249
        • Lee S.-.Y.
        • Doh I.
        • Nam D.-.H.
        • et al.
        3D Cell-Based High-Content Screening (HCS) using a micropillar and microwell chip platform.
        Anal Chem. 2018; 90: 8354-8361
        • Lukonin I.
        • Zinner M.
        • Liberali P.
        Organoids in image-based phenotypic chemical screens.
        Exp Mol Med. 2021; 53: 1495-1502
        • Lee S.Y.
        • Teng Y.
        • Son M.
        • et al.
        Three-dimensional aggregated spheroid model of hepatocellular carcinoma using a 96-pillar/well plate.
        Molecules. 2021; : 26
        • Lee S.Y.
        • Doh I.
        • Lee D.W.
        A high throughput apoptosis assay using 3D cultured cells.
        Molecules. 2019; : 24
        • Lee S.-.Y.
        • Teng Y.
        • Son M.
        • et al.
        High-dose drug heat map analysis for drug safety and efficacy in multi-spheroid brain normal cells and GBM patient-derived cells.
        PLoS One. 2021; 16e0251998
        • Baker L.A.
        • Tiriac H.
        • Clevers H.
        • et al.
        Modeling pancreatic cancer with organoids.
        Trends Cancer. 2016; 2: 176-190
        • Porter R.L.
        • Magnus N.K.C.
        • Thapar V.
        • et al.
        Epithelial to mesenchymal plasticity and differential response to therapies in pancreatic ductal adenocarcinoma.
        Proc Natl Acad Sci U S A. 2019; 116: 26835-26845
        • Kang Y.
        • Deng J.
        • Ling J.
        • et al.
        3D imaging analysis on an organoid-based platform guides personalized treatment in pancreatic ductal adenocarcinoma.
        J Clin Invest. 2022; : 132
        • Ashburn T.T.
        • Thor K.B.
        Drug repositioning: identifying and developing new uses for existing drugs.
        Nat Rev Drug Discov. 2004; 3: 673-683
        • Breckenridge A.
        • Jacob R.
        Overcoming the legal and regulatory barriers to drug repurposing.
        Nat Rev Drug Discov. 2019; 18: 1-2
        • Nosengo N.
        Can you teach old drugs new tricks?.
        Nature. 2016; 534: 314-316
        • Shamir E.R.
        • Ewald A.J.
        Three-dimensional organotypic culture: experimental models of mammalian biology and disease.
        Nat Rev Mol Cell Biol. 2014; 15: 647-664
        • Kretzschmar K.
        • Clevers H.
        Organoids: modeling development and the stem cell niche in a dish.
        Dev Cell. 2016; 38: 590-600
        • Griffith L.G.
        • Swartz M.A.
        Capturing complex 3D tissue physiology in vitro.
        Nat Rev Mol Cell Biol. 2006; 7: 211-224
        • Turley S.J.
        • Cremasco V.
        • Astarita J.L.
        Immunological hallmarks of stromal cells in the tumour microenvironment.
        Nat Rev Immunol. 2015; 15: 669-682
        • Johansson A.
        • Hamzah J.
        • Ganss R.
        More than a scaffold: stromal modulation of tumor immunity.
        Biochim Biophys Acta. 2016; 1865: 3-13
        • Stock K.
        • Estrada M.F.
        • Vidic S.
        • et al.
        Capturing tumor complexity in vitro: comparative analysis of 2D and 3D tumor models for drug discovery.
        Sci Rep. 2016; 6: 28951