v
Search
Advanced

Publications > Journals > Future Integrative Medicine> Article Full Text

  • OPEN ACCESS

Innovations in Organoid Engineering: Construction Methods, Model Development, and Clinical Translation

  • Xue Shen1,2,
  • Haiyan Jiang2,
  • Xiaoyu Fan2,
  • Xiaoyan Duan2,
  • Tusi Lin2,3,
  • Wanfang Li2,4,
  • Jie Bao2,4,
  • Jia Xu5,
  • Bosai He1,*  and
  • Hongtao Jin2,4,6,* 
 Author information 

Abstract

Organoids are derived from self-organizing stem cells and form three-dimensional structures that are structurally and functionally similar to in vivo tissues. With the ability to replicate the in vivo microenvironment and maintain genetic stability, organoids have become a powerful tool for elucidating developmental mechanisms, accurately modeling disease processes, and efficiently screening drug candidates, and have also demonstrated significant value in the field of traditional Chinese medicine (TCM)-including applications in screening active components of TCM, studying TCM pharmacodynamic mechanisms, evaluating TCM safety, and verifying the effects of traditional non-pharmacological therapies such as acupuncture and yoga. Organoids can be cultured using air-liquid interface systems, bioreactors, and vascularization techniques. They are widely used in drug screening, disease modeling, precision medicine, and toxicity assessment. However, current limitations include high costs, difficulty in accurately replicating the microenvironment, and ethical concerns. In this review, we systematically retrieve, synthesize, and analyze relevant literature to elucidate the culture methods of organoid technology, its diverse applications across various fields, and the challenges it faces. In the future, integration with artificial intelligence may provide new insights and strategies for drug development and disease research and the modernization of TCM.

Graphical Abstract

Keywords

Organoid, Pluripotent stem cell, PSC, Tumor microenvironment, TME, High-throughput screening, HTS, Drug development

Introduction

Organoids are collections of organ-specific cells cultured in vitro.1In vitro three-dimensional (3D) cultures of pluripotent stem cells (PSCs) and adult stem cells (ASCs) can form spatially structured cell aggregates that mimic the structure and physiological functions of organs in vivo.1 According to their origin, organoids can be categorized into those derived from ASCs and those derived from PSCs.2 PSCs include embryonic stem cells and induced pluripotent stem cells (iPSCs). In general, organoids derived from ASCs are commonly utilized to recapitulate the states of their tissue of origin under in vivo homeostatic or regenerative conditions and are primarily applied in disease modeling, including cancer and neurodegenerative diseases.1,3,4 Additionally, PSC-derived organoids, which resemble fetal tissues, can self-organize into neural epithelial structures and complex neuronal networks.1,5 These organoids are mainly used to study diseases related to developmental defects, such as microcephaly.3,4

In recent years, organoids have addressed many limitations of two-dimensional (2D) models by more accurately replicating the structure and function of in vivo organs while retaining the original tissue characteristics and genetic stability.6 Organoid models derived from various organs have gradually emerged as a research focus.7 However, systematic reviews of their construction methodologies and applications remain limited. This review discusses the construction methods and diverse applications of organoids, aiming to provide innovative insights for drug development and personalized therapeutic strategies.

Organoid construction

Organoids form 3D structures that rely on matrix materials or supports to provide attachment and structural support for cells.8 Three key factors must be considered in the construction of organoids. (1) The physical characteristics of the culture environment3: When developing 3D organoid models in vitro, it is necessary to simulate the characteristics of the extracellular matrix (ECM) in tissues.9 The ECM regulates tissue development and homeostasis by releasing growth factors, cytokines, and chemokines.9 Matrigel is the most widely used substrate in organoid cultures.9 (2) Requirements of endogenous and exogenous signals3: For most organoids, the cell system needs to be stimulated with specific exogenous signals during derivation. For example, the Wnt/β-catenin signaling pathway is crucial for the development of specific anatomical regions in the mouse stomach during gastric growth.10 This signaling pathway serves as the mechanistic basis for inducing the differentiation of human pluripotent stem cells (hPSCs) into 3D human gastric fundus organoids.10 (3) Starting cell type3: Distinct initial cell types result in varied properties in the final organoids. For instance, neural ectoderm-associated optic cups,11 brain organoids,5 and mesodermal kidney organoids can currently only be derived from PSCs.12 Additionally, organoids from somatic ectodermal lineages (especially glandular tissues) are mainly derived from ASCs or dissociated adult tissues.13 Most endodermal organoids can be derived from both PSCs and ASCs, such as colonic organoids.14,15 The selection of different components forms the foundation of organoid culture, with specific construction methods described in the following sections (Fig. 1a–c).5,17–18

Schematic diagram of organoid culture method.
Fig. 1  Schematic diagram of organoid culture method.5,16–18

(a) Experimental procedure for culturing human lung organoids by the ALI method. Cell expansion was carried out in submerged culture using medium supplemented with Y-27632. When confluence reached 100%, initial differentiation was induced by removing the apical medium and switching to air-lift culture to form pseudostratified epithelial cells. (b) Schematic of the optimized bioreactor cardiac differentiation protocol. PSCs were inoculated into Geltrex-pretreated T80 bottles and cultured in E8 medium until 90% confluence. After digestion, cells were resuspended and transferred to a stirred-tank bioreactor. Differentiation was initiated when the EB diameter reached 100–300 µm: CHIR99021 medium was applied on day 0, switched to inhibitor of Wnt response-1 (IWR-1) -endo medium on day 2, and 1:1000 insulin-containing medium was introduced from day 7. Cells were enzymatically digested and frozen on day 15. (c) Vascularized brain organoids after the construction of the human BBB process. Brain and vascular organoids were generated separately and subsequently fused to simulate neurovascular co-development, leading to the formation of BBB organoids by promoting telencephalic cell fusion with surrounding cells. ALI, air-liquid interface; BBB, blood-brain barrier; EB, embryoid body; hPSCs, human pluripotent stem cells; PDO, patient-derived organoid; PSCs, pluripotent stem cells. Some images were created with BioRender.

Air-liquid interface (ALI) culture

The ALI is a 3D culture method that mimics the luminal environment of hollow organs.19,20 In ALI culture, the permeable membrane of insert petri dish chambers is pre-coated with collagen before cell inoculation.19 The apical cell surface is exposed to air, while the basal side interacts with the culture medium through the membrane, forming a 3D model that simulates the internal environment of cavity organs.19 ALI culture can also preserve various infiltrating immune cell populations (e.g., T cells and B cells).20

ALI has been widely applied in tissue engineering and cell culture to study the effects of mechanical forces on cells.21 When cultured using this system, airway epithelial cells form pseudostratified layers with tight junctions, cilia, and mucin synthesis, closely mimicking their in vivo counterparts (Fig. 1a).21 This method is also valuable for studying organoid interactions with the tumor microenvironment (TME).22 Scientists have used ALI culture to construct patient-derived organoids (PDOs) from immune-competent patients for TME studies in lung and colorectal cancer models.22 Tumor-infiltrating lymphocytes in these organoids retained the T-cell receptor profiles of the original tumors,22 offering insight into the immune characteristics of tumors and mechanisms underlying immune responses.22 Despite its advantages, the ALI method has limitations. For instance, immune cells within ALI-cultured organoids exhibit a relatively short survival time, which limits long-term in vitro maintenance and successive expansions.23,24 Moreover, as organoids grow beyond a certain size, nutrient availability becomes restricted.23,24 To address these limitations and improve the realism of ALI organoid cultures, future research should focus on optimizing the culture environment to maintain immune cells and stromal components long-term, while more accurately replicating the cellular microenvironment.23,24

Bioreactor culture

Bioreactors enhance nutrient exchange by precisely controlling microenvironmental conditions and delivering oxygen, nutrients, and supplemental agents in a regulated manner (Fig. 1b).16,25,26 Improved aeration and nutrient distribution support cell viability and promote the formation of complex organoid structures, particularly in brain organoid cultures, resulting in more complete morphological development.16,25,26

Bioreactor culture is a relatively straightforward method for supporting organoid structures, outperforming other techniques in simplicity and efficacy. Qian’s team used 3D-printed components to develop SpinΩ, a miniaturized porous rotating bioreactor that enables the derivation of iPSCs into forebrain, midbrain, and hypothalamus organoids.27 This system enhances nutrient uptake, offers a compact design, and reduces media costs.27

Bioreactor technology also helps maintain a highly undifferentiated state in organoids during long-term culture.28 For example, suspension bioreactors enable high-throughput, scalable culture of mouse embryonic stem cells, supporting their large-scale expansion while preserving pluripotency.28 However, most current organoid cultures use suspension formats, and bioreactors incorporating mechanical compression and tension are not yet available. Therefore, engineering bioreactors capable of applying programmable mechanical and reactive forces holds significant scientific and practical potential.16

Vascularization

Organoid vascularization is a process that emulates and recreates the vascular structure of organs through cellular self-reorganization, microvascular fragmentation, in vivo transplantation into the host, and microfluidics.29,30 The central nervous system (CNS) of vertebrates develops in synchrony with the vascular system.31 Since the CNS does not generate vascular progenitor cells, avascular brain organoids fail to survive when transplanted into the host.31,32 Thus, vascularization is critical for neuronal nutrition, oxygen supply, and healthy development.31,32 Vascularized organoids are increasingly used in disease models to study vascular-neural interactions. Human blood vessel organoids (hBVOs) produce vascular cells capable of penetrating brain organoids and forming vascular-like structures (Fig. 1c).33,34 The integration of hBVOs with brain organoids has been demonstrated to generate vascularized brain organoids that model interactions between neuronal and non-neuronal components in vitro.33,34 The presence of molecular markers associated with the blood-brain barrier (BBB) in vascularized brain organoids indicates that hBVOs can establish a neural-specific vascular network.33,34

Currently, the development of vascularized organoids faces challenges related to large discrepancies in the size and density of their vascular structures compared to those observed in vivo.35 Additionally, issues of insufficient stability and lack of long-term reproducibility remain.35 The integration of advanced 3D bioprinting techniques with in vitro methods for constructing external circulatory systems is expected to enable the creation of vascularized organoids possessing functional vascular networks and achieving complete maturation.35 This advancement is poised to significantly impact pharmaceutical R&D processes.

In summary, current organoid construction methods have their advantages and limitations (Table 1).16,19,21,22,27,28,33–35 ALI culture can simulate the environment of hollow organs but has limitations in cell survival and nutrient supply. Bioreactors optimize culture conditions but lack mechanical stimulation. Vascularization technology is crucial for organoid survival but faces challenges related to structure and stability. In the future, innovative technologies need to be integrated to overcome these bottlenecks and promote organoid technology for broader roles in scientific research and clinical applications.

Table 1

Comparison of organoid construction methods

MethodsApplicationsAdvantagesLimitations
ALICultivate epidermal and respiratory epithelial cells.21 Studying organoid-tumor microenvironment interactions clarifies tumor immune traits and immune response mechanisms.22Simulate the environments of hollow organs and tubes.19Constructing PDO for tumor microenvironment research.22
BioreactorTo build more complex brain organoids.27Maintaining organoids in a highly undifferentiated state enables large-scale stem cell expansion.28Enhance nutrient exchange to promote complex organoid formation.16 Small footprint and low cost.27
VascularizationA disease model for studying vascular-neural interactions.33,34Simulate and reconstruct the organ vascular structure.The size and density of vascular structures vary significantly in vivo.35 Issues of stability, long-term maintenance, and lack of repeatability.35

Construction of representative models

Different diseases require specific organoid models to study their pathological mechanisms in depth. By combining various types of organ models, researchers can gain a comprehensive understanding of the pathological characteristics of different tissues and organs, thereby providing robust support for precision medicine (Table 2).36–47

Table 2

Methods for constructing representative organoid models

OrganoidsSourcesMethodsEstablishmentsReferences
Kidney organoidPSCsCHIR99021-FGF pathway and BMP/FGF pathwayRenal unit-related cell types originate from the primitive streak, posterior mesoderm, and renal unit progenitor cells45
Liver organoidPSCs, ASCsMonotypic cell cultureiPSCs were reprogrammed and differentiated into endoderm to form organoids, which matured in differentiation medium (DM)36
Polytypic cell co-cultureThe cells were differentiated into hepatocytes and co-cultured with human umbilical vein endothelial cells (HUVECs) and mesenchymal stem cells (MSCs) to self-organize into 3D aggregates
Lung organoidPSCsScaffold-free system culture method carrier-based scaffolding System methodPSCs form anterior foregut spheres. They differentiate into germ layers and mature into lung organoids via FGF and HH factors37,38
ASCsAlveolar type II epithelial cells (AEC2s) were co-cultured with other cells to form alveolar-like organoids46
LCOsLung cancer cells were embedded in Matrigel with epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) to induce organoid formation39
Brain organoidPSCsUnguided methodThe gametophyte differentiates into neural ectoderm embedded in matrix gel for 3D culture, which is then transferred to a rotating bioreactor40
Guided methodsBy regulating signaling pathways (e.g., BMP and Wnt), directed cell differentiation is induced. Adjusting factor intensity promotes tissue self-organization, generating functional brain organoids via long-term culture41,42
Intestinal organoidPSCsCrypts from biopsy were embedded in matrix gel and cultured in normal colon medium with 50% Wnt3a for expansion, then generated organoids via normal colon medium culture and intestinal epithelial differentiation medium
ASCs47
Bone organoidPSCsPSCs form mesoblastic aggregates, differentiate into vascular/hematopoietic cells, and generate mature bone organs under specific conditions43
BMSCsA PCL-based 3D-printed scaffold embedded with GelMA/SFMA hydrogel and PTH@MSNs formed a porous scaffold for repairing large weight-bearing bone defects44

Kidney organoid

The kidneys play a crucial role in maintaining fluid, electrolyte, and pH balance within the body by filtering plasma, facilitating the reabsorption of essential nutrients, and secreting metabolic waste.48 Kidney organoids are 3D in vitro models derived from PSCs under 3D culture conditions.49 These organoids contain epithelial renal unit-like structures expressing markers associated with podocytes, proximal tubules, medullary collaterals, distal tubules, and collecting ducts.50 Organoids exhibit similarities to natural renal tissues in terms of cell types, morphology, and functional characteristics.50,51 The construction of renal organoids is a multistage process involving differentiation of PSCs into primitive somite mesoderm.36 The cells further differentiate into intermediate mesoderm, which then splits into two distinct embryonic tissues: the metanephric mesenchyme and the ureteric bud.36 Currently, two primary methodologies are employed in the induction of renal organoids. CHIR99021, an activator of the Wnt signaling pathway, has been demonstrated to modulate both the fibroblast growth factor (FGF) pathway and the bone morphogenetic protein (BMP)/FGF pathway (Fig. 2a).52–59 The Wnt signaling pathway plays a crucial role in maintaining the balance between nephron progenitor cell expansion and mesenchymal-epithelial transition.60–62 The FGF signaling pathway is essential for maintaining nephron progenitor cells, and BMP4 promotes ureteric bud formation.60–62

Schematic diagram of the construction of representative organoid models.
Fig. 2  Schematic diagram of the construction of representative organoid models.52,53,46,54–59

(a) Schematic diagram of kidney organoids constructed by BMP/FGF and CHIR99021-FGF pathways. BMP/FGF pathway: PSCs differentiate into PSM via BMP4/Act A, then into IM via FGF9. IM further differentiates into MM and UB via FGF9/BMP7/RA. CHIR99021-FGF pathway: PSCs differentiate into PSM via Wnt agonist CHIR99021, then into IM via FGF9, and subsequently into MM and UB. (b) Schematic diagram of constructing liver organoids by monotypic cell culture method. PSCs differentiate into endoderm cells via Act A, FGF, BMP, and Wnt signaling. Endoderm cells become liver progenitor cells when induced by BMP, FGF, and HGF. Liver progenitor cells respond to OSM signals to form hepatocyte-like cells. Alternatively, culturing liver progenitor cells in ECM with FGF, EGF, and Act A modulation generates duct-like organoids. (c) Schematic diagram of constructing liver organoids by polytypic cell co-culture. iPSCs are specified toward hepatic lineages to generate iPSC-HEs. Subsequently, iPSC-HEs, MSCs, and HUVECs are co-cultured to recapitulate intercellular interactions during organogenesis. Cells influence each other via secreted factors, mimicking signal exchange in in vivo liver development. After four to six days of culture, a 3D hepatic bud is formed. (d) Schematic diagram of constructing lung organoids by the carrier-based scaffolding system method. Human-derived PSCs are differentiated into DE via Activin A treatment. Subsequently, DE cells are further induced into FE and then LPCs using a cocktail of factors including Noggin, SB431542, Smoothened agonist, FGF4, and CHIR99021. Finally, LPCs are cultured with FGF10 for 50 days to generate lung organoids. (e) Schematic diagram of brain organoids constructed by the non-guided method. Starting from PSCs, these cells interact with microfilaments to form micropatterned EBs. The embryoid bodies then develop into reproducible neuroectoderm. Combined with Matrigel droplets and CHIR99021 supplementation, the neural epithelium expands. Finally, under the influence of ECM, these cells differentiate and self-organize over 40 days to form engineered brain organoids. (f) Schematic diagram of constructing brain organoids by guided methods. PSCs differentiate into RGCs via LDN-193189, Jagged-1, and DLL-1. Subsequently, RGCs transition to astroglia progenitors under Jagged-1, DLL-1, and Onc-M. These progenitors develop into immature astrocytes with Jagged-1, DLL-1, and LIF, and finally mature into mature astrocytes over 40 days with the same factors. (g) Schematic diagram of intestinal organogenesis. PSCs are seeded onto Matrigel-coated 24-well plates. Upon reaching 80–95% confluence, cells are treated with Activin A for three days to form definitive endoderm. Subsequently, endodermal cells are treated with posterior gut induction medium containing FGF4 and CHIR99021 to induce mid/hindgut spheroids. These spheroids are then embedded in Matrigel and cultured in intestinal growth medium supplemented with EGF to generate HIOs. (h) Schematic diagram of bone marrow organoids. iPSCs form EBs in Y-27632 medium. Mesoderm is induced with mTeSR Plus medium + BMP4, CHIR99021, and VEGF. On day 2, medium is switched to Essential 6 + VEGF. On day 4, EBs are collected and overlaid with StemPro-34 SFM + IL-3 and cytokines. Cytokines are switched on days 6 and 8. On day 10, sprouting EBs are harvested, transferred to 96-well plates, and self-assemble into spherical organoids. Act A, activin A; BMP, bone morphogenetic protein; CNTF, ciliary neurotrophic factor; DE, definitive endoderm; DLL-1, delta-like ligand 1; EB, embryoid body; ECM, extracellular matrix; EGF, epidermal growth factor; ESCs, embryonic stem cells; FE, foregut endoderm; FGF, fibroblast growth factor; Flt-3L, fms-like tyrosine kinase 3 ligand; HE, hematopoietic stem cells; HGF, human growth factor; HIOs, human intestinal organoids; hNRG1, human Neuregulin-1; HUVECs, human umbilical vein endothelial cells; IL-3, interleukin-3; IM, intermediate mesoderm; iPSC, induced pluripotent stem cells; iPSC-HEs, iPSC-derived hepatocyte-like cells; LIF, leukemia inhibitory factor; LPCs, lung progenitor cells; MM, metanephric mesenchyme; MSCs, mesenchymal stem cells; Onc-M, oncostatin M; OSM, oncostatin M; PDGF-AA, platelet-derived growth factor-AA; PMA, phorbol-12-myristate-13-acetate; PSCs, pluripotent stem cells; PSM, primitive streak mesoderm; RA, retinoic acid; RGCs, radial glial cells; SCF, stem cell factor; scRNA, single-cell RNA; TPO, thrombopoietin; UB, ureteric bud; VEGF, vascular endothelial growth factor. Some images were created with BioRender.

Liver organoid

The liver performs essential functions such as metabolism, protein synthesis, and detoxification of xenobiotics.63 It also exhibits remarkable regenerative abilities following severe injury. The primary functional cell types in the liver are hepatocytes of endodermal origin and cholangiocytes.63 The formation of liver organoids can be conceptualized through two distinct culture systems: monotypic cell culture and polytypic cell co-culture.64 In monotypic cell culture systems, liver organoids generated from iPSCs and ASCs typically further differentiate into mature hepatocytes or hepatobiliary organoids (Fig. 2b).64 These cells differentiate into a high proportion of mature hepatocytes, which generally exhibit albumin production, urea synthesis, and cytochrome P450 activity.64 The polytypic cell co-culture system is employed to create vascularized and transplantable organoids designed to preserve signaling interactions between mesenchymal and parenchymal cells, which are vital for normal tissue growth and functionality (Fig. 2c).64 Compared to monotypic cultures, polytypic cell co-cultures more realistically mimic human liver metabolic processes. In long-term culture, primary liver organoids show less genomic structural variation than PSC-derived ones,65,66 retaining and stably maintaining the genetic traits of their original donors.65,66 Currently, liver organoids are primarily generated from the foregut endoderm through activation of the Wnt, FGF, BMP, and Activin/Nodal signaling pathways.67 Hyperactivation of the Wnt signaling pathway leads to hepatic stagnation.68

Lung organoid

The lung is one of the most complex organs in the human body, consisting of more than 40 cell types that form a unique structure for its primary function of effective gas exchange.69 Lung organoids are composed of a variety of cells, including lung epithelial cells, myofibroblasts, upper airway-like epithelial cells, and mesenchymal compartments.53 The emergence of tracheal structures is a critical indicator of successful lung organoid construction.53 Currently, there are two main lung organoid culture methods.70 One is a scaffold-free system used in ALI cultures, designed to enhance cell-to-cell contact, leading to the formation of lung spheroids, which subsequently develop into lung organoids. The other approach involves a carrier-based scaffolding system, primarily utilizing Matrigel. The ECM within Matrigel provides essential nutrients and channel proteins, facilitating the growth and development of lung organoids (Fig. 2d). Lung organoids can be categorized into mature lung epithelial stem cell-derived organoids, PSC-derived lung organoids, and lung cancer organoids (LCOs). Mature lung epithelial stem cells are directly cultured in vitro and induced to generate lung organoids.37,38 PSC-derived lung organoids induce PSCs to form anterior foregut spheroids, which then differentiate into specific germ layers via modulation of growth factors such as FGF and hedgehog signaling.37,38 Further differentiation directs these germ layers towards mature lung organoids.37,38 PSC-derived lung organoids provide a solution to the challenge of isolating lung stem/progenitor cells, especially in diseased lungs, and are currently the most commonly employed method for lung organoid cultivation. LCOs are created by transplanting lung cancer stem cells from patient tissues into Matrigel, with growth factors added to promote proliferation and differentiation.39

Brain organoid

Brain organoids are self-assembled 3D aggregates generated from PSCs, containing cell types and cellular structures similar to those of the embryonic human brain.71 The construction of brain organoids can be categorized into two approaches: unguided and guided methods.71 The unguided method, which aims to form the whole brain, does not utilize pattern-based growth factors (Fig. 2e). Instead, it focuses on enhancing growth conditions and creating an environment conducive to intrinsic signaling pathways that guide development.72 Neural ectoderm is generated from embryoid bodies, then embedded in Matrigel for 3D culture, after which the Matrigel droplets are transferred to a rotating bioreactor.40 This method allows rapid brain tissue development and can be used to study interactions between various brain regions. However, it can result in contamination by non-neural lineages such as mesoderm and endoderm and leads to high variability among organoids in terms of structure and cell composition.73 In contrast, guided methods use exogenous patterning factors to direct neuroepithelial development, guiding hPSCs to recapitulate the structural organization of specific brain regions.40 Examples include organoids modeling the cerebral cortex,41 hippocampus,42 and midbrain.74 The organoid differentiation protocol, known as the bootstrap method, can be customized by adding external patterning factors exclusively at the early differentiation stage to allow hPSCs to become progenitors characteristic of specific brain regions (Fig. 2f).71 Once successful patterning occurs, these factors are removed, and subsequent differentiation mirrors in vivo neural patterning.71 Directed organoid cultures produce a relatively uniform mixture of cell types with minimal variation between batches and cell lines.75

Intestinal organoid

The intestines are a pivotal component of the digestive system, playing critical roles in food digestion, nutrient absorption, and metabolism.76 Intestinal organoids are derived from crypt-like units containing Lgr5+ stem cells,76 which can originate from adult intestinal stem cells and PSCs. These organoids retain stem cell characteristics, reflecting the activity of intestinal epithelial stem cells with the capacity to proliferate and differentiate (Fig. 2g).76–78 They can also be stably cryopreserved for long-term self-renewal.76–78 The primary method for constructing intestinal organoids involves isolating crypts from human intestinal tissues, preparing a buffer, and formulating a culture medium.79 This medium is supplemented with growth factors such as Wnt, R-spondin1, and Noggin. The addition of insulin-like growth factor 1 and FGF-2 facilitates long-term passaging and multilineage differentiation.80 Renewal and differentiation processes in intestinal organoids are regulated by the Wnt and Notch signaling pathways.81 Administration of CHIR99021 (a Wnt activator) and valproic acid (a Notch activator) has a synergistic effect that promotes self-renewal.82 Compared to intestinal stem cell-derived intestinal organoids, PSC-derived organoids retain more pronounced fetal-like characteristics.82 Furthermore, co-culture of PSC-derived intestinal organoids with endothelial cells promotes maturation.83

Bone organoid

Bone organoids are in vitro tissue models prepared using stem or progenitor cells, featuring a 3D, miniaturized, and simplified structure. They mimic the structure, function, and cellular complexity of natural bone tissue, including crucial aspects such as mineralization and angiogenesis (Fig. 2h).84,85 PSCs can be reprogrammed from adult cells to restore pluripotency,86 enabling the formation of specific cell types and their self-assembly into 3D structures that mimic the bone marrow microenvironment, allowing development of osteochondral organoids.43,86,87

Researchers have constructed bone organoids with self-mineralization ability and pluripotent differentiation potential by integrating bone mesenchymal stem cells with bone matrix-mimicking bioinks. Bone mesenchymal stem cells-embedded gelatin methacrylate microspheres were prepared and subjected to chondrogenic induction, resulting in bone callus organoids.44 In bone tissue engineering, bone organoids have various applications: they can be used to fabricate composite scaffolds that repair large weight-bearing bone defects, enhance osteogenesis and angiogenesis, and mimic natural bone tissue.44 Additionally, bone organoids serve as models for drug screening and evaluation in disease research and drug development, helping researchers study disease mechanisms.

Organoid applications in biomedicine

Organoids are pivotal in drug discovery and development. By emulating the complexity of the human environment and establishing in vitro models, organoids facilitate the rapid screening of pharmaceuticals. Moreover, they offer more authentic and dependable frameworks for toxicity assessment. Beyond their role in drug discovery, organoids hold immense potential in disease modeling. They provide substantial support for the thorough examination of disease mechanisms, thereby catalyzing novel advancements in medical research and clinical practice.

Disease modeling

Organoid technology generates 3D tissues in vitro that accurately reproduce the structure, function, molecular features, genomic alterations, expression profiles, and TME of primary tumors (Table 3).88–96 Kazama et al.89 developed patient-derived tumor organoids from renal cell carcinoma tumor samples. They demonstrated that renal cell carcinoma tumor organoids can replicate the histological features of clinical tumors in vitro while preserving their genomic integrity during long-term culture.89 Organoid technology can also reveal viral pathogenesis and host cell responses. In a study on norovirus, researchers cultivated norovirus strains using intestinal organoids and found that bile was a pivotal factor in strain-dependent norovirus replication.90 Zika virus is a flavivirus transmitted through mosquitoes and sexually.97,98 In a seminal study, Patricia P. Garcez and colleagues exposed hiPSC-derived brain organoids to the Zika virus.91 Published in the journal Nature, the study revealed that Zika virus-induced neuronal cell death occurs in the early stages of brain development. Furthermore, organoid technology plays a pivotal role in studying the pathogenesis and clinical characteristics of complex multi-system diseases. Richards’ team developed an in vitro 3D model of post-myocardial infarction myocardial tissue.92 This model was created using a combination of an oxygen diffusion gradient and chronic adrenergic stimulation to mimic the human heart after myocardial infarction. The model closely resembles transcriptome data from both an animal model of myocardial infarction and human ischemic cardiomyopathy tissue following acute infarction. Fotios Sampaziotis and colleagues successfully constructed disease models of polycystic liver disease and cystic fibrosis (CF)-associated liver disease using patient-derived iPSCs.93 They found that the experimental CF drug VX809 rescued the disease phenotype of CF cholangiopathy in vitro. Organoid disease modeling offers significant potential to enhance our understanding of organ development, maintain homeostasis, and identify disease mechanisms, thereby paving the way for innovative advancements in diagnostic and therapeutic strategies.

Table 3

Applications of organoids in disease modeling

OrgansSourcesDisease modelsApplicationsReferences
KidneyRCC tumor samplesRCCTo determine effective treatments for individual RCC patients89
HiPSCsCisplatin-induced acute kidney injury (AKI) modelValidation of the feasibility of using human kidney organoids to mimic AKI from chemotherapeutic agents in vitro94
IntestinalASCsIntestinal organoids infected with norovirusUncovering the replication regulation mechanism of norovirus90
Colorectal cancer organoidsRevealing the regulatory role of DKK2 (Wnt signaling pathway antagonist) in colorectal cancer95
BrainHiPSCsBrain organoids infected with the Zika virusStudying the effects of Zika virus infection during neurogenesis and growth91
HeartHiPSCsMI organoidOrganotypic features that summarize the short-term injury state after acute MI92
LiverHiPSCsPolycystic liver disease and CF organoidReproducing the effects of the therapeutic compounds Verapamil, Octreotide, and VX80993
LungLung tissue from adult C57B/L6 miceSmall cell lung cancer (SCLC) modelRevealing the role of KMT2C gene in SCLC96

Drug screening

Organoids provide a more accurate model of physiological conditions for drug development, making them valuable tools for testing drug efficacy and safety, as well as screening potential drug candidates (Table 4).99–106 Recent reports show that patient-derived cancer organoids have great accuracy in predicting cancer patients’ responses to various drugs.99,107,108 These organoids not only provide closer models of real physiological conditions but also serve to test drug efficacy and safety and screen drug candidates.99,107,108 Mao and colleagues developed an organ-based drug screening system to evaluate the anticancer effects of 34 drugs on human colorectal cancer.99 Through this screening, they identified fedratinib, trametinib, and bortezomib as anticancer agents with notable efficacy.99 Saito et al.100 developed a drug compound library to inhibit organoids derived from biliary tract carcinomas. Their findings demonstrated that the antifungal drugs amorolfine and fenticonazole significantly inhibited the growth of biliary tract carcinoma-derived organoids with minimal toxicity to normal biliary epithelial cells.100 The development of organoid-based high-throughput screening (HTS) marked a significant advancement in drug screening applications.101 Zhou and colleagues established a biobank of human pancreatic ductal adenocarcinoma organoid models.101 They discovered that irbesartan reversed resistance in these cells via the Hippo/YAP1/c-Jun/stemness/iron metabolism axis, as observed through HTS.101 Han et al.102 performed HTS of Food and Drug Administration (FDA)-approved drugs using SARS-CoV-2-infected lung organoids and identified potential clinical candidates, including imatinib and articaine hydrochloride. Organoids have thus provided a novel technological tool for large-scale drug screening, highlighting their ability to maintain histological characteristics long-term.

Table 4

Applications of organoids in drug screening

OrganoidsSourcesScreening drugsReferences
CRC organoidsPatient-derived colonic tissueFedratinib, trametinib, bortezomib99
BTC organoidsPatient-derived biliary tissueAmorolfine, fenticonazole100
Pancreatic ductal adenocarcinoma organoidsCell lines of pancreatic ductal adenocarcinomairbesartan101
Lung organoidshPSCs in patients with new crownsImatinib, articaine hydrochloride102
Cervical cancer organoidsPatient-derived cervical tumor tissueBortezomib, MLN2238, MLN9708, carfilzomib, panobinostat, romidepsin, homoharringtonine103
Brain organoidsAlzheimer’s disease patients’ iPSCsFlibanserin, ripasudil, everolimus104
Kidney organoidshiPSCsGFB-887105
Breast cancer organoidsPatient-derived xenograftsErbaxane, fluorouracil106

Biobank

A biobank is defined as a comprehensive repository of biospecimen entities, bioinformatic specimen phenotypic data, and specimen research information.109,110 The primary purpose of a biobank is to support scientific research, clinical diagnostics, and drug discovery and development.109,110 Additionally, biobanks facilitate the long-term preservation of biospecimens.109,110 In recent years, biobanks of organoids originating from various types have been established, enabling the preservation of histological characteristics and increasingly used in disease modeling (Table 5).39,111–116 These include intestinal,111 breast cancer,112 lung,117 ovarian,113 kidney,118 liver,114 and other organoids. Van de Wetering’s team was the first to establish a living colorectal cancer organoid biobank, highlighting their potential in individual patient-level genomic and functional studies through organoid culture platforms.111 Fadi Jacob and colleagues established a biobank composed of glioblastoma organoids consisting of 70 biological samples from a variety of patients.115 These glioblastoma organoids reflect substantial genomic alterations associated with glioblastoma pathogenesis and maintain numerous critical characteristics of their respective parental tumors.115 This biobank offers substantial support for future research in glioblastoma biology and the assessment of therapeutic approaches.115 Minsuh Kim and colleagues have established a biobank of LCOs, derived from 95 subtypes of lung cancer and covering more than 56% of lung cancer patients, providing significant data to support drug screening, genomic research, and immunotherapy-related studies.39

Table 5

Application of the organoid biobank

OrganoidsSourcesNamesApplicationsReferences
Intestinal organoidsPatient-derived colorectal cancer tissueBiobank of living colorectal cancer organoidsDemonstrating that organoid culture platforms can be used for genomic and functional studies at the individual patient level111
GBOsPatient-derived glioblastoma tissueBiobank of GBOsReflects major genomic changes associated with glioblastoma pathogenesis115
Lung organoidsPatient-derived lung cancer tissueBiobank of LCOsTo summarize the histological and genetic features of lung cancer subtypes while maintaining genomic heterogeneity with the original patient genomes39
Breast cancer organoidsPatient-derived breast cancer tissueBiobank of breast cancer organoidsGenetic characterization of breast cancer organoids confirmed and the development of drugs targeting breast cancer characteristics112,116
Ovarian organoidsPatient-derived ovarian cancer tissueBiobank of high-grade serous ovarian cancer (HGSC)Addressing the limited availability of fresh tumor material for HGSC organoid studies113
Liver organoidsPrimary liver cancer tissueBiobank of liver cancer organoidsReproducing the proliferative and metabolic subtypes of hepatocellular carcinoma and the worst/best prognostic subtypes of intrahepatic cholangiocarcinoma114

Precision medicine

Van de Wetering’s team was the first to establish a living colorectal cancer organoid biobank, demonstrating its potential for genomic and functional studies at the individual patient level.119 Consequently, selecting a more targeted and individualized treatment regimen remains a significant challenge in improving efficacy. Organoid models derived from patients’ autologous cells have emerged as a promising solution to address this critical gap.119 These models offer a unique opportunity to assess the efficacy and potential toxicity of multiple drugs in patients, thereby facilitating the development of personalized intervention strategies.119 Berkers’ team quantified the response to CF transmembrane conductance regulator modulators in vitro by evaluating in vitro forskolin-induced swelling in rectal organoids from patients.120 Their findings demonstrated that CF transmembrane conductance regulator modulation correlates with two independent indicators of in vivo treatment response (percent predicted forced expiratory volume in 1 second and sputum cell count).120 Furthermore, the team showed that biobanked stem cell resources can be correlated with in vivo therapeutic response.120 This suggests that the biobank’s stem cell resources can be utilized to personalize treatments for individual patients.120 Organoid technology has been particularly used in precision medicine for tumors. PDOs have been demonstrated to preserve the gene expression and histopathological characteristics of tumor tissues. Furthermore, they accurately mimic the tumor’s response to drugs in the patient’s body.8 Using organoids derived from a patient’s tissue, HTS can help clinicians understand the patient’s disease characteristics, drug response, and therapeutic efficacy, enabling the development of personalized treatment plans.8 Ji Shuyi and colleagues established a biobank housing patient-derived hepatocellular carcinoma organoids. They demonstrated the synergistic inhibitory effect of tesirolimus combined with lenvatinib in both organoid models and patient-derived xenograft experiments.114 This research highlights the potential for personalized treatment strategies based on individual tumor characteristics.114

Toxicity assessment

Stem cells exhibit varying sensitivities to exogenous chemicals depending on whether they are undergoing proliferation or differentiation.121 Conducting toxicological studies based on organoid formation processes helps evaluate the developmental toxicity of chemicals, identify the sensitive periods during which chemical action occurs, and pinpoint molecular targets related to toxicity pathways.121 This approach facilitates a deeper understanding of how external substances affect cells at different stages (Table 6).121–128 Leite’s team developed a functional human liver organoid capable of identifying acetaminophen as a substance inducing hepatic fibrosis and activating hepatic stellate cells.122 This liver organoid represents the first model able to detect compound-induced hepatic stellate cell activation.122 Li’s team developed an ovarian organoid model derived from germline stem cells.123 This model was utilized to ascertain that salinomycin impedes ovarian organoid formation and germ cell populations by inducing apoptosis.123 Koichiro Susa and his team investigated the effects of three drugs—tenofovir, aristolochic acid, and cisplatin—on kidney cells.124 They discovered that at low concentrations, these drugs caused injury in the proximal renal tubular area without damaging podocytes.124 Using a specialized kidney organoid model, they evaluated the toxicity of these drugs.124 In their study, Wang et al.125 employed cholangiocellular organoids to model chlorpromazine-induced intrahepatic bile duct injury. Their findings showed that chlorpromazine itself did not induce an inflammatory response, but when combined with TNF-α, it resulted in a synergistic effect.125 The application of organoids in the assessment of environmental pollutants, including heavy metals and pesticides, is a promising field of research. These organoids can replicate the effects of pollutants on human organs, thereby facilitating the evaluation of health risks posed by environmental toxins. Chen’s team methodically examined the neurotoxicity of arsenic and lead by utilizing optic vesicle brain organoids.126 Their findings revealed that concurrent exposure to arsenic and lead impeded neural and retinal development, while also triggering proteins linked with carcinogenesis in optic vesicle brain organoids.126

Table 6

Application of organoids in toxicity assessment

OrgansSourcesApplicationsReferences
LiverHepaRG cells and primary human HSCsAPAP induces activation of HSCs122
Human liver tissueCPZ and TNF-α synergistically induce intrahepatic bile duct injury125
OvarianFemale germ-line stem cell (FGSC)Salinomycin damages ovarian organoids by inducing apoptosis123
KidneyPSCsTenofovir, aristolochic acid, and cisplatin induce proximal renal tubular injury124
iPSCsAccelerated tacrolimus nephrotoxicity by rapamycin (autophagy inducer) treatment127
LungiPSCsTwo drugs, GNE7915 and amiodarone, were studied, and amiodarone was found to cause a more severe lung injury phenotype128
BrainESCsCo-exposure of As and Pb interferes with neural and retinal development and activates proteins associated with carcinogenesis126

Application of organoids in TCM

Organoids have unique advantages. They reproduce organ morphology and function through a three-dimensional structure, simulate the in vivo microenvironment with multiple cell types, and allow real-time dynamic observation.129 With these advantages, organoids integrate the holistic view of TCM and modern molecular biology across scales. They provide technical support for drug efficacy screening, target identification, and safety evaluation in the field of TCM.129

Screening of active components in TCM

TCM has diverse chemical components and complex interactions between these components. This makes the accurate identification and screening of its pharmacologically active substances a bottleneck in the modernization research of TCM. Organoid technology simulates the complex molecular network regulation process mediated by TCM components after they enter the human body, thus solving the problem of screening active components of TCM. Take the classic TCM formula Pien Tze Huang (PZH) as an example. Its main ingredients include Moschus (musk), Calculus Bovis (bezoar), Fel Serpentis (snake bile), and Radix Notoginseng (notoginseng root).130,131 Researchers constructed a colorectal cancer organoids model to screen for active components in PZH. The results showed that ginsenoside F2 and ginsenoside Re, two components contained in PZH, can inhibit the growth of colorectal cancer organoids.132 In the field of liver cancer research, the natural compound Omuralide A can directly target transketolase and activate p53 signaling, thereby inhibiting the growth of hepatocellular carcinoma organoids.133

Research on the pharmacological mechanism of TCM

TCM compound prescriptions contain a variety of active components, which exert their effects through multiple pathways and targets.134 Organoid technology evaluates the effects of each component on specific cells or tissues via high-throughput screening, and thus becomes an important tool for studying the multi-target mechanism of action of TCM.134 In the research on TCM against Alzheimer’s disease, brain organoids are used. They confirm that ginseng polysaccharide (GP) can do two things: inhibit the accumulation of amyloid-β and downregulate neuroinflammation.135 Amyloid-β is the main component of amyloid plaques. The mechanism of GP is related to promoting neuronal mitophagy. This provides support for GP to become a candidate drug for anti-Alzheimer’s disease.135 In anti-tumor research, researchers established a gastric cancer organoid model. They found that decursin can inhibit the growth of gastric organoids and regulate the expression of cathepsin C and autophagy-related proteins.136 This study reveals a new mechanism by which decursin regulates cell growth and autophagy. It also indicates that decursin may become a potential anti-cancer drug that inhibits both cell growth and autophagy.136

Safety assessment of TCM

TCM has complex components. Natural toxic components (such as alkaloids and glycosides), improper processing or compatibility, and excessive use may all cause toxicity risks.137 Therefore, scientific evaluation is needed. It can identify safe doses, toxic target organs and mechanisms of action. This provides a basis for medication in special populations.137 In the research on TCM toxicity evaluation, organoid technology has shown unique value. For example, esculentoside A is the main toxic component of Phytolaccae Radix (pokeweed root).138 Researchers conducted toxicity evaluation using kidney organoids. They confirmed two things: esculentoside A has nephrotoxicity. And its nephrotoxicity is related to epithelial-mesenchymal transition mediated by the STING signaling pathway.138 In the research on the protective effects of TCM, Haiyang Chen and his team constructed a brain organoid model. They studied the intervention effect of Codonopsis pilosula polysaccharides (CPPs) on rotenone-induced neuronal toxicity.139 The results showed two things: first, CPPs can effectively improve the cytotoxicity of brain organoids caused by rotenone; second, CPPs can correct the abnormal DNA methylation induced by rotenone.139 This provides reliable experimental support for the research on the toxicity protection mechanism of TCM components.

Drugs need to be absorbed by the intestines and metabolized by the liver before acting on target organs. In vitro single-organ models cannot simulate the complex metabolic process of TCM in the human body. Traditional animal models also have this limitation.140 In the future, the integration of organoids and TCM can be promoted in three aspects. First, optimize models. Strengthen the simulation of specific pathological organs (such as liver injury and intestinal barrier dysfunction), improve the functional maturity of intestinal and liver organoids, and replace animal models to reduce species interference.134 Second, build “multi-organ organoid chips” through interdisciplinary cooperation. These chips can reproduce the complete action path of TCM—from entering the human body to exerting therapeutic effects.134 Third, innovative applications. Assist in TCM syndrome differentiation and typing as well as individualized medication, and accelerate the translation of TCM.134 Its value can also be explored in the research of traditional non-pharmacological therapies. These include analyzing the mechanism of acupuncture effects and conducting in vitro verification of yoga’s regulation on the function of specific organs.

Frontier technologies of organoids

As a cutting-edge field in 3D cell culture, organoid technology provides revolutionary tools for life science and medical research by mimicking organ development and physiological functions. Its rapid advancement stems from the interdisciplinary integration of gene editing, single-cell sequencing, 3D bioprinting, and artificial intelligence (AI).

Gene editing technology

Organoids simulate in vitro organ development and physiological functions, while gene editing technologies knock in, knock out, or modify specific genes to precisely explore gene function mechanisms in development, disease, and other processes.141 For example, in testicular organoids, researchers knocked out outer dense fiber 2 and intraflagellar transport 88 genes to inhibit primary cilia formation.142 Results showed reduced cilia numbers, shortened cilia lengths, and affected in vitro tubule morphogenesis.142 Beyond labeling cell types to study migration, growth, and development, gene editing technologies can model diseases. For example, researchers introduced the LRRK2-G2019S mutation via gene editing to generate midbrain-specific organoids from Parkinson’s disease patients.143 These studies demonstrate the potential of combining genome engineering with organoid technology, enabling in-depth exploration of developmental processes in different organs and the roles of specific molecular pathways.

Single-cell RNA sequencing (scRNA-seq)

scRNA-seq enables high-throughput transcriptomic profiling of individual cells. It compares genetic information, reveals intercellular heterogeneity, and identifies distinct cell subpopulations to explore disease progression.144 It is primarily used to analyze interactions between cell subpopulations and gene regulatory processes.145 Zhao et al.146 established seven hepatobiliary tumor organoids and used scRNA-seq to detect cancer stem cell markers. They found that cluster of differentiation 44-positive cell populations may induce drug resistance in hepatocellular carcinoma 272.146 Additionally, while integrating scRNA-seq into routine clinical diagnosis and personalized medicine, most current scRNA-seq-based clinical studies remain exploratory, mainly focusing on re-evaluating and better understanding disease processes, as well as identifying diagnostic and therapeutic markers.147

3D printing technology

Bioprinting techniques use bioinks made of biocompatible non-living materials and cells to build 3D constructs in a controlled manner and with micrometric resolution.148 Compared to 2D cell cultures and animal models, 3D bioprinted organoid models better recapitulate native tissue architecture, intercellular interactions, and cell-ECM crosstalk.149 In bone tissue engineering, researchers combined 3D printing with polycaprolactone scaffolds, parathyroid hormone-loaded mesoporous silica nanoparticles, and gelatin methacrylate/methacrylated silk fibroin composite hydrogels to create porous PM@GS/polycaprolactone scaffolds for repairing large load-bearing bone defects.44 A study used 3D bioprinting to create 3D structures from iPSC-derived cortical neurons and glial cells, which correctly express neuronal and astrocyte markers.148 This approach offers possibilities for generating more complex human neural 3D structures.148 Additionally, 3D bioprinting can mimic the structure of neuromuscular tissue (NMT). The Jeong Sik Kong team developed a CNS-decellularized ECM bioink.150 Using 3D printing, they created NMT with high contractility for long-term culture.150 With the development of 3D bioprinting technology, it is expected to play a key role in disease research, drug development, tissue engineering, and regenerative medicine in the future.

AI technology

AI is a field of computer science that aims to develop computer systems capable of performing tasks that typically require human intelligence, such as visual perception, speech recognition, decision-making, and language translation.151 The use of AI algorithms enables detailed analysis of complex organoid behavior, diverse cellular interactions, and dynamic responses to external stimuli.152 Deep intelligent perception of images is an emerging technology in AI research, training on large datasets for image prediction, classification, and other complex image perception tasks.153 Chen’s team utilizes various 2D depth characteristics to approximate features in 3D medical images.154 Researchers use an automatic detection method to detect cerebral microbleeds, employing a support vector machine classifier and magnetization rate-weighted imaging for this purpose.154 The use of AI in analyzing organoid omic data helps researchers identify various cell types and track cell state changes, revealing natural processes of cellular growth and disease development.155,156 This framework can identify conserved and specific developmental trajectories, as well as genes and functions expressed during development.156 The integration of AI technology improves precision, objectivity, and the ability to handle complex data in organoid research, driving progress toward precision therapies with organoids.

Discussion

As a frontier in 3D cell culture, organoid technology provides revolutionary tools for life science and medical research by mimicking organ development and physiological functions. As a major research model for 3D cell culture in recent years, organoids have overcome the shortcomings of traditional 2D models in simulating cell growth, microenvironment, and genetic stability.6 For instance, organoids retain properties of the original tumor tissue, including similar pathological features, gene expression patterns, and drug responses to the parental tumor.157,158 Additionally, they can restore the ductal response in the fibrotic microenvironment.157,158 PSC-derived organoids exhibit high stability at the chromosomal and structural levels, maintaining genetic integrity throughout months of culture.65 Conventional mouse models fail to mimic human-specific microbial interactions, whereas organoids can create in vitro models closer to human physiology.159 For example, while mouse models assess immune cell interactions and gut microbiota changes, they struggle to establish causality and cannot replicate human-specific microbial crosstalk.159 In contrast, organoids construct physiologically relevant in vitro models to accurately study human microbial interactions, addressing the complexity of the gut microenvironment and challenges in real-time parameter evaluation. This enables more effective deduction of causal relationships between hosts and microbiota.159 In salivary gland organoid culture, single-cell dissociation is avoided to maintain cellular diversity, as traditional methods may cause cell transformation through dissociation.160 Organoid culture can also induce cell differentiation by adjusting culture conditions and adding specific substances (e.g., DAPT) to study cell fate changes, which are difficult to accurately model and investigate in mouse models.160

Although organoid technology bridges the gap between cell lines and in vivo models, current systems still face challenges. The difficulty of accurately replicating the natural TME, ethical concerns related to controversial issues, and the high costs of research impede the advancement of organoid technology.

The ECM is a dynamic structure that continuously reshapes and controls tissue homeostasis.11 Its components act as ligands for cell receptors like integrins, constantly interacting with epithelial cells to transmit signals that regulate adhesion, migration, proliferation, apoptosis, survival, or differentiation.11 Additionally, cells continuously rebuild and reshape the ECM through synthesis, degradation, recombination, and chemical modification.12 Therefore, different batches of ECM differ in spatial structure and cytokine composition, making it difficult to mimic the immune components and blood vessels in the natural TME.161 If the lack of TME in PDOs prevents tumor cells from forming clones, it leads to discrepancies between in vitro and in vivo models.162 The future inclusion of PDOs with immune components from lymph nodes and blood has the potential to create more comprehensive models.163 To address this challenge, co-culture organoid models incorporating immune cells, vascular systems, and cancer-associated fibroblasts have been developed.163 These models can mimic tumor infiltration and peripheral immune cell populations in the TME and co-culture with immune cells from peripheral blood mononuclear cells or lymph nodes to simulate tumor immune processes, including T cell proliferation, activation, and tumor cell recognition.163 Additionally, decellularization technology can bridge the gap between 3D culture assays and in vivo tumor models.164 By analyzing decellularized normal and colon tumor tissues, scientists confirmed that the composition and arrangement of the ECM remain largely intact after decellularization.165 Combining PDO models with decellularization technology and co-culture assays in the future may provide a more powerful platform for cancer research and anticancer drug screening.164

There are also concerns that when PDOs are transplanted into animals for culturing, it may lead to the entry of animal genes into the human genome, posing a genetic risk.166 PDOs also address the human rights of donors, which must be distinguished from tissue organs derived directly from the human body.167 However, there are no binding principles or legal norms that define the rights and obligations of donors and biobanks.167 In the future, different types of organoids will likely be ethically ranked and evaluated, while also accounting for diverse cultural and social contexts.168 Furthermore, sophisticated ethical value frameworks can be developed and implemented in a timely manner, taking into account both scientific and technological advancements as well as shifts in ethical concepts.168

The cost of organoids is currently a significant limiting factor in their widespread use. Acquiring high-quality, high-viability stem cells or primary cells is challenging, and cell processing and preservation require meticulous handling. Emerging organoid models, such as 3D bioprinted organoids, microfluidic organoid models, and genetically engineered organoids, further complicate material procurement and cost-effectiveness.169,170 Additionally, determining whether organoids have functions similar to in vivo organs and validating their drug response consistency with in vivo conditions require integrating multiple detection technologies and analytical methods, which demand high expertise and experience from technicians.169,170 To address this challenge, biobanks and shared sample repository systems are being established to provide standardized patient-derived cells, effectively reducing sample acquisition difficulties and costs.169,170 In the future, organoid technology is expected to integrate gene editing and data-sharing platforms, enabling researchers to optimize material selection strategies and reduce resource waste. This will accelerate their transition from laboratory research to clinical applications.

The limitations of this review include insufficient discussion of emerging technical details (e.g., 3D bioprinting parameters, single-cell spatial transcriptomics) in organoid culture, inadequate coverage of clinical regenerative medicine applications (e.g., organ transplantation immunity, personalized cell therapy) and tissue-specific organoids (ophthalmic/cardiovascular), and a lack of systematic cross-species comparisons between human and animal organoids in disease modeling. These gaps suggest opportunities to deepen technical insights, integrate cross-disciplinary applications, and update research perspectives.

Conclusions

As an emerging 3D cell culture system, organoid technology has demonstrated substantial potential in basic research and translational medicine by recapitulating in vivo organ structures and functions. Generated through methods like ALI culture, bioreactor systems, and vascularization strategies, organoids create representative models of kidneys, livers, lungs, and brains for multi-dimensional simulations of organ development, disease pathogenesis, and drug responses. By mimicking the in vivo microenvironment, this technology plays a pivotal role in biomedical research, facilitating HTS, establishing physiologically relevant toxicity assessment models, and advancing disease modeling and biobanking for precision medicine.

This review also explores emerging organoid technologies, such as 3D bioprinting for scalable model fabrication, microfluidic systems for dynamic microenvironment control, and genetically engineered organoids for gene-disease association studies. These innovations address traditional limitations in model consistency and complexity, opening new frontiers for mechanistic research and clinical applications, as well as offering novel technical support for accelerating the modernization and translational application of TCM.

Declarations

Acknowledgement

None.

Funding

This study was supported by the National Natural Science Foundation of China (Grant No. 82074104), the National Natural Science Foundation of China (Grant No. 82474169), and the Research Project of Clinical Toxicology Transformation from the Chinese Society of Toxicology (No. CST2021CT101).

Conflict of interest

Dr. Hongtao Jin has been an Associate Editor of Future Integrative Medicine since November 2021. Dr. Hongtao Jin is the chairman of Beijing Union-Genius Pharmaceutical Technology Development Co., Ltd., and Dr. Wanfang Li and Dr. Jie Bao are employees of the company. The authors have no other conflict of interest to note.

Authors’ contributions

Writing–review & editing (XS, HYJ, XYF, XYD, HTJ), writing–original draft (XS, TSL), validation (XS), data curation (XS, HYJ, XYD, WFL, JB, BSH), investigation (XS, XYF, XYD, TSL, HTJ), conceptualization (XS, HYJ, XYF, WFL, JB, BSH, HTJ), methodology (HYJ), supervision (XYF, WFL, JB, BSH, HTJ), resources (WFL, JX, BSH, HTJ), and funding acquisition (HTJ). All authors have approved the final version and publication of the manuscript.

References

  1. Clevers H. Modeling Development and Disease with Organoids. Cell 2016;165(7):1586-1597 View Article PubMed/NCBI
  2. Lee SW, Jung DJ, Jeong GS. Gaining New Biological and Therapeutic Applications into the Liver with 3D In Vitro Liver Models. Tissue Eng Regen Med 2020;17(6):731-745 View Article PubMed/NCBI
  3. Rossi G, Manfrin A, Lutolf MP. Progress and potential in organoid research. Nat Rev Genet 2018;19(11):671-687 View Article PubMed/NCBI
  4. Kaushik G, Ponnusamy MP, Batra SK. Concise Review: Current Status of Three-Dimensional Organoids as Preclinical Models. Stem Cells 2018;36(9):1329-1340 View Article PubMed/NCBI
  5. Dao L, You Z, Lu L, Xu T, Sarkar AK, Zhu H, et al. Modeling blood-brain barrier formation and cerebral cavernous malformations in human PSC-derived organoids. Cell Stem Cell 2024;31(6):818-833.e11 View Article PubMed/NCBI
  6. Liu Q, Yao X, Wei Z, Xie Y. Action mechanism and potential function on main components of organoid culture medium. Chin J Tissue Eng Res 2021;25(31):5072-5078
  7. Liu Y. Organoids and chips: biomedical and drug’s new models. Basic and Clinical Medicine 2024;44(9):1201
  8. Zhang Y. Organoids: an ideal model for frontier biomedical research. Frontier Science and Technology 2024;14(12):1-2
  9. Kim S, Min S, Choi YS, Jo SH, Jung JH, Han K, et al. Tissue extracellular matrix hydrogels as alternatives to Matrigel for culturing gastrointestinal organoids. Nat Commun 2022;13(1):1692 View Article PubMed/NCBI
  10. McCracken KW, Aihara E, Martin B, Crawford CM, Broda T, Treguier J, et al. Wnt/β-catenin promotes gastric fundus specification in mice and humans. Nature 2017;541(7636):182-187 View Article PubMed/NCBI
  11. Eiraku M, Takata N, Ishibashi H, Kawada M, Sakakura E, Okuda S, et al. Self-organizing optic-cup morphogenesis in three-dimensional culture. Nature 2011;472(7341):51-56 View Article PubMed/NCBI
  12. Low JH, Li P, Chew EGY, Zhou B, Suzuki K, Zhang T, et al. Generation of Human PSC-Derived Kidney Organoids with Patterned Nephron Segments and a De Novo Vascular Network. Cell Stem Cell 2019;25(3):373-387.e9 View Article PubMed/NCBI
  13. Tsai KK, Huang SS, Northey JJ, Liao WY, Hsu CC, Cheng LH, et al. Screening of organoids derived from patients with breast cancer implicates the repressor NCOR2 in cytotoxic stress response and antitumor immunity. Nat Cancer 2022;3(6):734-752 View Article PubMed/NCBI
  14. Múnera JO, Kechele DO, Bouffi C, Qu N, Jing R, Maity P, et al. Development of functional resident macrophages in human pluripotent stem cell-derived colonic organoids and human fetal colon. Cell Stem Cell 2023;30(11):1434-1451.e9 View Article PubMed/NCBI
  15. Yui S, Nakamura T, Sato T, Nemoto Y, Mizutani T, Zheng X, et al. Functional engraftment of colon epithelium expanded in vitro from a single adult Lgr5+ stem cell. Nat Med 2012;18(4):618-623 View Article PubMed/NCBI
  16. Licata JP, Schwab KH, Har-El YE, Gerstenhaber JA, Lelkes PI. Bioreactor Technologies for Enhanced Organoid Culture. Int J Mol Sci 2023;24(14):11427 View Article PubMed/NCBI
  17. Castaneda DC, Jangra S, Yurieva M, Martinek J, Callender M, Coxe M, et al. Protocol for establishing primary human lung organoid-derived air-liquid interface cultures from cryopreserved human lung tissue. STAR Protoc 2023;4(4):102735 View Article PubMed/NCBI
  18. Prondzynski M, Berkson P, Trembley MA, Tharani Y, Shani K, Bortolin RH, et al. Efficient and reproducible generation of human iPSC-derived cardiomyocytes and cardiac organoids in stirred suspension systems. Nat Commun 2024;15(1):5929 View Article PubMed/NCBI
  19. Cao X, Coyle JP, Xiong R, Wang Y, Heflich RH, Ren B, et al. Invited review: human air-liquid-interface organotypic airway tissue models derived from primary tracheobronchial epithelial cells-overview and perspectives. In Vitro Cell Dev Biol Anim 2021;57(2):104-132 View Article PubMed/NCBI
  20. Santos AJM, van Unen V, Lin Z, Chirieleison SM, Ha N, Batish A, et al. A human autoimmune organoid model reveals IL-7 function in coeliac disease. Nature 2024;632(8024):401-410 View Article PubMed/NCBI
  21. Whitcutt MJ, Adler KB, Wu R. A biphasic chamber system for maintaining polarity of differentiation of cultured respiratory tract epithelial cells. In Vitro Cell Dev Biol 1988;24(5):420-428 View Article PubMed/NCBI
  22. Neal JT, Li X, Zhu J, Giangarra V, Grzeskowiak CL, Ju J, et al. Organoid Modeling of the Tumor Immune Microenvironment. Cell 2018;175(7):1972-1988.e16 View Article PubMed/NCBI
  23. Rauth S, Karmakar S, Batra SK, Ponnusamy MP. Recent advances in organoid development and applications in disease modeling. Biochim Biophys Acta Rev Cancer 2021;1875(2):188527 View Article PubMed/NCBI
  24. Zhang J, Qin T, Mao Z, Wang G. Application and prospect of air-liquid culture in organoid research. Chin J Geriatr Care 2024;22(3):102-106 View Article
  25. Ovando-Roche P, West EL, Branch MJ, Sampson RD, Fernando M, Munro P, et al. Use of bioreactors for culturing human retinal organoids improves photoreceptor yields. Stem Cell Res Ther 2018;9(1):156 View Article PubMed/NCBI
  26. Phelan MA, Gianforcaro AL, Gerstenhaber JA, Lelkes PI. An Air Bubble-Isolating Rotating Wall Vessel Bioreactor for Improved Spheroid/Organoid Formation. Tissue Eng Part C Methods 2019;25(8):479-488 View Article PubMed/NCBI
  27. Qian X, Jacob F, Song MM, Nguyen HN, Song H, Ming GL. Generation of human brain region-specific organoids using a miniaturized spinning bioreactor. Nat Protoc 2018;13(3):565-580 View Article PubMed/NCBI
  28. zur Nieden NI, Cormier JT, Rancourt DE, Kallos MS. Embryonic stem cells remain highly pluripotent following long term expansion as aggregates in suspension bioreactors. J Biotechnol 2007;129(3):421-432 View Article PubMed/NCBI
  29. Liu M, Fan W. Construction strategy for vascularization of organoid. Chin J Tissue Eng Res 2025;29(13):2774-2783 View Article
  30. Liu H, Yue L, Liu C, Tan Y. Research progress in vascularization of renal organoids. Chin J Pharmacol Toxicol 2024;38(8):633-640 View Article
  31. Paredes I, Himmels P, Ruiz de Almodóvar C. Neurovascular Communication during CNS Development. Dev Cell 2018;45(1):10-32 View Article PubMed/NCBI
  32. Bautch VL, James JM. Neurovascular development: The beginning of a beautiful friendship. Cell Adh Migr 2009;3(2):199-204 View Article PubMed/NCBI
  33. Sun XY, Ju XC, Li Y, Zeng PM, Wu J, Zhou YY, et al. Generation of vascularized brain organoids to study neurovascular interactions. Elife 2022;11:e76707 View Article PubMed/NCBI
  34. Ahn Y, An JH, Yang HJ, Lee DG, Kim J, Koh H, et al. Human Blood Vessel Organoids Penetrate Human Cerebral Organoids and Form a Vessel-Like System. Cells 2021;10(8):2036 View Article PubMed/NCBI
  35. Sun K, Wang T, Li J, Li H. Ideas and technical challenges of vascularized organoid construction. Chin J Comp Med 2023;33(2):126-133 View Article
  36. Jiang X, Yang T, Liang N, Wang Y, Teng J. Research advances in renal organoids and its applications for evaluating drug nephrotoxicity. Journal of Clinical Nephrology 2024;24(8):687-691 View Article
  37. Nikolić MZ, Caritg O, Jeng Q, Johnson JA, Sun D, Howell KJ, et al. Human embryonic lung epithelial tips are multipotent progenitors that can be expanded in vitro as long-term self-renewing organoids. Elife 2017;6:e26575 View Article PubMed/NCBI
  38. Yang T, Wang X, Bai N, Dong L, Che H, Cai Y. Lung organoids: A new way to study human lung development and disease. Acad J Chin PLA Med Sch 2021;42(6):658-664 View Article
  39. Kim M, Mun H, Sung CO, Cho EJ, Jeon HJ, Chun SM, et al. Patient-derived lung cancer organoids as in vitro cancer models for therapeutic screening. Nat Commun 2019;10(1):3991 View Article PubMed/NCBI
  40. Xia X, Zhang SC. Differentiation of neuroepithelia from human embryonic stem cells. Methods Mol Biol 2009;549:51-58 View Article PubMed/NCBI
  41. Xiang Y, Tanaka Y, Patterson B, Kang YJ, Govindaiah G, Roselaar N, et al. Fusion of Regionally Specified hPSC-Derived Organoids Models Human Brain Development and Interneuron Migration. Cell Stem Cell 2017;21(3):383-398.e7 View Article PubMed/NCBI
  42. Sakaguchi H, Kadoshima T, Soen M, Narii N, Ishida Y, Ohgushi M, et al. Generation of functional hippocampal neurons from self-organizing human embryonic stem cell-derived dorsomedial telencephalic tissue. Nat Commun 2015;6:8896 View Article PubMed/NCBI
  43. Khan AO, Rodriguez-Romera A, Reyat JS, Olijnik AA, Colombo M, Wang G, et al. Human Bone Marrow Organoids for Disease Modeling, Discovery, and Validation of Therapeutic Targets in Hematologic Malignancies. Cancer Discov 2023;13(2):364-385 View Article PubMed/NCBI
  44. Yang J, Fatima K, Zhou X, He C. Meticulously engineered three-dimensional-printed scaffold with microarchitecture and controlled peptide release for enhanced bone regeneration. Biomater Transl 2024;5(1):69-83 View Article PubMed/NCBI
  45. Wang G, Wang Y, Liang J, Liu Z. Research and application of kidney organoids. J Nephrol Dialy Transplant 2024;33(3):245-253 View Article
  46. Vazquez-Armendariz AI, Tata PR. Recent advances in lung organoid development and applications in disease modeling. J Clin Invest 2023;133(22):e170500 View Article PubMed/NCBI
  47. Jelinsky SA, Derksen M, Bauman E, Verissimo CS, van Dooremalen WTM, Roos JL, et al. Molecular and Functional Characterization of Human Intestinal Organoids and Monolayers for Modeling Epithelial Barrier. Inflamm Bowel Dis 2023;29(2):195-206 View Article PubMed/NCBI
  48. Takasato M, Er PX, Chiu HS, Maier B, Baillie GJ, Ferguson C, et al. Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis. Nature 2015;526(7574):564-568 View Article PubMed/NCBI
  49. Parigoris E, Lee JH, Liu AY, Zhao X, Takayama S. Extended longevity geometrically-inverted proximal tubule organoids. Biomaterials 2022;290:121828 View Article PubMed/NCBI
  50. Schutgens F, Rookmaaker MB, Margaritis T, Rios A, Ammerlaan C, Jansen J, et al. Tubuloids derived from human adult kidney and urine for personalized disease modeling. Nat Biotechnol 2019;37(3):303-313 View Article PubMed/NCBI
  51. Garreta E, Prado P, Tarantino C, Oria R, Fanlo L, Martí E, et al. Fine tuning the extracellular environment accelerates the derivation of kidney organoids from human pluripotent stem cells. Nat Mater 2019;18(4):397-405 View Article PubMed/NCBI
  52. Zhang D, Du X, Kong F, Zhao S. Molecular mechanism and application of kidney organoids from pluripotent cells. Chin J Cell Stem Cell (Electronic Edition) 2018;8(1):49-52 View Article
  53. Miller AJ, Dye BR, Ferrer-Torres D, Hill DR, Overeem AW, Shea LD, et al. Generation of lung organoids from human pluripotent stem cells in vitro. Nat Protoc 2019;14(2):518-540 View Article PubMed/NCBI
  54. Prior N, Inacio P, Huch M. Liver organoids: from basic research to therapeutic applications. Gut 2019;68(12):2228-2237 View Article PubMed/NCBI
  55. Takebe T, Sekine K, Enomura M, Koike H, Kimura M, Ogaeri T, et al. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature 2013;499(7459):481-484 View Article PubMed/NCBI
  56. Lancaster MA, Corsini NS, Wolfinger S, Gustafson EH, Phillips AW, Burkard TR, et al. Guided self-organization and cortical plate formation in human brain organoids. Nat Biotechnol 2017;35(7):659-666 View Article PubMed/NCBI
  57. Jovanovic VM, Weber C, Slamecka J, Ryu S, Chu PH, Sen C, et al. A defined roadmap of radial glia and astrocyte differentiation from human pluripotent stem cells. Stem Cell Reports 2023;18(8):1701-1720 View Article PubMed/NCBI
  58. Bouffi C, Wikenheiser-Brokamp KA, Chaturvedi P, Sundaram N, Goddard GR, Wunderlich M, et al. In vivo development of immune tissue in human intestinal organoids transplanted into humanized mice. Nat Biotechnol 2023;41(6):824-831 View Article PubMed/NCBI
  59. Frenz-Wiessner S, Fairley SD, Buser M, Goek I, Salewskij K, Jonsson G, et al. Generation of complex bone marrow organoids from human induced pluripotent stem cells. Nat Methods 2024;21(5):868-881 View Article PubMed/NCBI
  60. Finer G, Maezawa Y, Ide S, Onay T, Souma T, Scott R, et al. Stromal Transcription Factor 21 Regulates Development of the Renal Stroma via Interaction with Wnt/β-Catenin Signaling. Kidney360 2022;3(7):1228-1241 View Article PubMed/NCBI
  61. Huh SH, Ha L, Jang HS. Nephron Progenitor Maintenance Is Controlled through Fibroblast Growth Factors and Sprouty1 Interaction. J Am Soc Nephrol 2020;31(11):2559-2572 View Article PubMed/NCBI
  62. Mills CG, Lawrence ML, Munro DAD, Elhendawi M, Mullins JJ, Davies JA. Asymmetric BMP4 signalling improves the realism of kidney organoids. Sci Rep 2017;7(1):14824 View Article PubMed/NCBI
  63. Tang XY, Wu S, Wang D, Chu C, Hong Y, Tao M, et al. Human organoids in basic research and clinical applications. Signal Transduct Target Ther 2022;7(1):168 View Article PubMed/NCBI
  64. Zhu X, Zhang B, He Y, Bao J. Liver Organoids: Formation Strategies and Biomedical Applications. Tissue Eng Regen Med 2021;18(4):573-585 View Article PubMed/NCBI
  65. Huch M, Gehart H, van Boxtel R, Hamer K, Blokzijl F, Verstegen MM, et al. Long-term culture of genome-stable bipotent stem cells from adult human liver. Cell 2015;160(1-2):299-312 View Article PubMed/NCBI
  66. Liang G, Zhang Y. Genetic and epigenetic variations in iPSCs: potential causes and implications for application. Cell Stem Cell 2013;13(2):149-159 View Article PubMed/NCBI
  67. Ober EA, Lemaigre FP. Development of the liver: Insights into organ and tissue morphogenesis. J Hepatol 2018;68(5):1049-1062 View Article PubMed/NCBI
  68. McLin VA, Rankin SA, Zorn AM. Repression of Wnt/beta-catenin signaling in the anterior endoderm is essential for liver and pancreas development. Development 2007;134(12):2207-2217 View Article PubMed/NCBI
  69. Franks TJ, Colby TV, Travis WD, Tuder RM, Reynolds HY, Brody AR, et al. Resident cellular components of the human lung: current knowledge and goals for research on cell phenotyping and function. Proc Am Thorac Soc 2008;5(7):763-766 View Article PubMed/NCBI
  70. Yang G, Fang H, Liu X, Fu Z, Zhu F. Progress in research and application of lung organoids. Chin J Pathophysiol 2024;40(6):1122-1127 View Article
  71. Qian X, Song H, Ming GL. Brain organoids: advances, applications and challenges. Development 2019;146(8):dev166074 View Article PubMed/NCBI
  72. Lancaster MA, Renner M, Martin CA, Wenzel D, Bicknell LS, Hurles ME, et al. Cerebral organoids model human brain development and microcephaly. Nature 2013;501(7467):373-379 View Article PubMed/NCBI
  73. Jacob F, Schnoll JG, Song H, Ming GL. Building the brain from scratch: Engineering region-specific brain organoids from human stem cells to study neural development and disease. Curr Top Dev Biol 2021;142:477-530 View Article PubMed/NCBI
  74. Jo J, Xiao Y, Sun AX, Cukuroglu E, Tran HD, Göke J, et al. Midbrain-like Organoids from Human Pluripotent Stem Cells Contain Functional Dopaminergic and Neuromelanin-Producing Neurons. Cell Stem Cell 2016;19(2):248-257 View Article PubMed/NCBI
  75. Sloan SA, Darmanis S, Huber N, Khan TA, Birey F, Caneda C, et al. Human Astrocyte Maturation Captured in 3D Cerebral Cortical Spheroids Derived from Pluripotent Stem Cells. Neuron 2017;95(4):779-790.e6 View Article PubMed/NCBI
  76. Yu D, Cao H, Wang X. Advances and applications of organoids: a review. Sheng Wu Gong Cheng Xue Bao 2021;37(11):3961-3974 View Article PubMed/NCBI
  77. Carvalho MR, Yan LP, Li B, Zhang CH, He YL, Reis RL, et al. Gastrointestinal organs and organoids-on-a-chip: advances and translation into the clinics. Biofabrication 2023;15(4):042004 View Article PubMed/NCBI
  78. Derricott H, Luu L, Fong WY, Hartley CS, Johnston LJ, Armstrong SD, et al. Developing a 3D intestinal epithelium model for livestock species. Cell Tissue Res 2019;375(2):409-424 View Article PubMed/NCBI
  79. Qiu G, Yu L, Zhang C, Dong L. Progress in Construction and Application of Intestinal Organoid Models. Chinese Journal of Animal Nutrition 2023;35(7):4231-4237 View Article
  80. Fujii M, Matano M, Toshimitsu K, Takano A, Mikami Y, Nishikori S, et al. Human Intestinal Organoids Maintain Self-Renewal Capacity and Cellular Diversity in Niche-Inspired Culture Condition. Cell Stem Cell 2018;23(6):787-793.e6 View Article PubMed/NCBI
  81. Almeqdadi M, Mana MD, Roper J, Yilmaz ÖH. Gut organoids: mini-tissues in culture to study intestinal physiology and disease. Am J Physiol Cell Physiol 2019;317(3):C405-C419 View Article PubMed/NCBI
  82. Taelman J, Diaz M, Guiu J. Human Intestinal Organoids: Promise and Challenge. Front Cell Dev Biol 2022;10:854740 View Article PubMed/NCBI
  83. Palikuqi B, Nguyen DT, Li G, Schreiner R, Pellegata AF, Liu Y, et al. Adaptable haemodynamic endothelial cells for organogenesis and tumorigenesis. Nature 2020;585(7825):426-432 View Article PubMed/NCBI
  84. Chen S, Chen X, Geng Z, Su J. The horizon of bone organoid: A perspective on construction and application. Bioact Mater 2022;18:15-25 View Article PubMed/NCBI
  85. Wang J, Chen X, Li R, Wang S, Geng Z, Shi Z, et al. Standardization and consensus in the development and application of bone organoids. Theranostics 2025;15(2):682-706 View Article PubMed/NCBI
  86. Al Abbar A, Ngai SC, Nograles N, Alhaji SY, Abdullah S. Induced Pluripotent Stem Cells: Reprogramming Platforms and Applications in Cell Replacement Therapy. Biores Open Access 2020;9(1):121-136 View Article PubMed/NCBI
  87. O’Connor SK, Katz DB, Oswald SJ, Groneck L, Guilak F. Formation of Osteochondral Organoids from Murine Induced Pluripotent Stem Cells. Tissue Eng Part A 2021;27(15-16):1099-1109 View Article PubMed/NCBI
  88. Xu H, Jiao D, Liu A, Wu K. Tumor organoids: applications in cancer modeling and potentials in precision medicine. J Hematol Oncol 2022;15(1):58 View Article PubMed/NCBI
  89. Kazama A, Anraku T, Kuroki H, Shirono Y, Murata M, Bilim V, et al. Development of patient-derived tumor organoids and a drug testing model for renal cell carcinoma. Oncol Rep 2021;46(4):226 View Article PubMed/NCBI
  90. Ettayebi K, Crawford SE, Murakami K, Broughman JR, Karandikar U, Tenge VR, et al. Replication of human noroviruses in stem cell-derived human enteroids. Science 2016;353(6306):1387-1393 View Article PubMed/NCBI
  91. Garcez PP, Loiola EC, Madeiro da Costa R, Higa LM, Trindade P, Delvecchio R, et al. Zika virus impairs growth in human neurospheres and brain organoids. Science 2016;352(6287):816-818 View Article PubMed/NCBI
  92. Richards DJ, Li Y, Kerr CM, Yao J, Beeson GC, Coyle RC, et al. Human cardiac organoids for the modelling of myocardial infarction and drug cardiotoxicity. Nat Biomed Eng 2020;4(4):446-462 View Article PubMed/NCBI
  93. Sampaziotis F, de Brito MC, Madrigal P, Bertero A, Saeb-Parsy K, Soares FAC, et al. Cholangiocytes derived from human induced pluripotent stem cells for disease modeling and drug validation. Nat Biotechnol 2015;33(8):845-852 View Article PubMed/NCBI
  94. Ma R, Yue L, Yun Z, Lou Z, Liu Q, Cui H, et al. Modeling acute kidney injury induced by cisplatin in human kidney organoids. Chinese Journal of Pharmacology and Toxicology 2024;38(4):279-285 View Article
  95. Shin JH, Jeong J, Choi J, Lim J, Dinesh RK, Braverman J, et al. Dickkopf-2 regulates the stem cell marker LGR5 in colorectal cancer via HNF4α1. iScience 2021;24(5):102411 View Article PubMed/NCBI
  96. Na F, Pan X, Chen J, Chen X, Wang M, Chi P, et al. KMT2C deficiency promotes small cell lung cancer metastasis through DNMT3A-mediated epigenetic reprogramming. Nat Cancer 2022;3(6):753-767 View Article PubMed/NCBI
  97. Hayes EB. Zika virus outside Africa. Emerg Infect Dis 2009;15(9):1347-1350 View Article PubMed/NCBI
  98. Musso D, Roche C, Robin E, Nhan T, Teissier A, Cao-Lormeau VM. Potential sexual transmission of Zika virus. Emerg Infect Dis 2015;21(2):359-361 View Article PubMed/NCBI
  99. Mao Y, Wang W, Yang J, Zhou X, Lu Y, Gao J, et al. Drug repurposing screening and mechanism analysis based on human colorectal cancer organoids. Protein Cell 2024;15(4):285-304 View Article PubMed/NCBI
  100. Saito Y, Muramatsu T, Kanai Y, Ojima H, Sukeda A, Hiraoka N, et al. Establishment of Patient-Derived Organoids and Drug Screening for Biliary Tract Carcinoma. Cell Rep 2019;27(4):1265-1276.e4 View Article PubMed/NCBI
  101. Zhou T, Xie Y, Hou X, Bai W, Li X, Liu Z, et al. Irbesartan overcomes gemcitabine resistance in pancreatic cancer by suppressing stemness and iron metabolism via inhibition of the Hippo/YAP1/c-Jun axis. J Exp Clin Cancer Res 2023;42(1):111 View Article PubMed/NCBI
  102. Han Y, Duan X, Yang L, Nilsson-Payant BE, Wang P, Duan F, et al. Identification of SARS-CoV-2 inhibitors using lung and colonic organoids. Nature 2021;589(7841):270-275 View Article PubMed/NCBI
  103. Seol HS, Oh JH, Choi E, Kim S, Kim H, Nam EJ. Preclinical investigation of patient-derived cervical cancer organoids for precision medicine. J Gynecol Oncol 2023;34(3):e35 View Article PubMed/NCBI
  104. Park JC, Jang SY, Lee D, Lee J, Kang U, Chang H, et al. A logical network-based drug-screening platform for Alzheimer’s disease representing pathological features of human brain organoids. Nat Commun 2021;12(1):280 View Article PubMed/NCBI
  105. Westerling-Bui AD, Fast EM, Soare TW, Venkatachalan S, DeRan M, Fanelli AB, et al. Transplanted organoids empower human preclinical assessment of drug candidate for the clinic. Sci Adv 2022;8(27):eabj5633 View Article PubMed/NCBI
  106. Guillen KP, Fujita M, Butterfield AJ, Scherer SD, Bailey MH, Chu Z, et al. A human breast cancer-derived xenograft and organoid platform for drug discovery and precision oncology. Nat Cancer 2022;3(2):232-250 View Article PubMed/NCBI
  107. Vlachogiannis G, Hedayat S, Vatsiou A, Jamin Y, Fernández-Mateos J, Khan K, et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science 2018;359(6378):920-926 View Article PubMed/NCBI
  108. Yao Y, Xu X, Yang L, Zhu J, Wan J, Shen L, et al. Patient-Derived Organoids Predict Chemoradiation Responses of Locally Advanced Rectal Cancer. Cell Stem Cell 2020;26(1):17-26.e6 View Article PubMed/NCBI
  109. Vaught J. Biobanking Comes of Age: The Transition to Biospecimen Science. Annu Rev Pharmacol Toxicol 2016;56:211-228 View Article PubMed/NCBI
  110. Huang T, Chen L, Wang H. Advances in organoid and related biospecimen repository research. Scientia Sinica (Vitae) 2024;54(6):1029-1040 View Article
  111. van de Wetering M, Francies HE, Francis JM, Bounova G, Iorio F, Pronk A, et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell 2015;161(4):933-945 View Article PubMed/NCBI
  112. Chen P, Zhang X, Ding R, Yang L, Lyu X, Zeng J, et al. Patient-Derived Organoids Can Guide Personalized-Therapies for Patients with Advanced Breast Cancer. Adv Sci (Weinh) 2021;8(22):e2101176 View Article PubMed/NCBI
  113. Senkowski W, Gall-Mas L, Falco MM, Li Y, Lavikka K, Kriegbaum MC, et al. A platform for efficient establishment and drug-response profiling of high-grade serous ovarian cancer organoids. Dev Cell 2023;58(12):1106-1121.e7 View Article PubMed/NCBI
  114. Ji S, Feng L, Fu Z, Wu G, Wu Y, Lin Y, et al. Pharmaco-proteogenomic characterization of liver cancer organoids for precision oncology. Sci Transl Med 2023;15(706):eadg3358 View Article PubMed/NCBI
  115. Jacob F, Salinas RD, Zhang DY, Nguyen PTT, Schnoll JG, Wong SZH, et al. A Patient-Derived Glioblastoma Organoid Model and Biobank Recapitulates Inter- and Intra-tumoral Heterogeneity. Cell 2020;180(1):188-204.e22 View Article PubMed/NCBI
  116. Granat LM, Kambhampati O, Klosek S, Niedzwecki B, Parsa K, Zhang D. The promises and challenges of patient-derived tumor organoids in drug development and precision oncology. Animal Model Exp Med 2019;2(3):150-161 View Article PubMed/NCBI
  117. Li H, Chen Z, Chen N, Fan Y, Xu Y, Xu X. Applications of lung cancer organoids in precision medicine: from bench to bedside. Cell Commun Signal 2023;21(1):350 View Article PubMed/NCBI
  118. Yousef Yengej FA, Jansen J, Rookmaaker MB, Verhaar MC, Clevers H. Kidney Organoids and Tubuloids. Cells 2020;9(6):1326 View Article PubMed/NCBI
  119. Bartfeld S, Clevers H. Stem cell-derived organoids and their application for medical research and patient treatment. J Mol Med (Berl) 2017;95(7):729-738 View Article PubMed/NCBI
  120. Berkers G, van Mourik P, Vonk AM, Kruisselbrink E, Dekkers JF, de Winter-de Groot KM, et al. Rectal Organoids Enable Personalized Treatment of Cystic Fibrosis. Cell Rep 2019;26(7):1701-1708.e3 View Article PubMed/NCBI
  121. Liu W, Mei X, Chen Y, Xiang Z, Gao Z, Xing L. Progress in 3D Organoid Models and Application Perspectives in Toxicity Test of Chemicals. Asian J Ecotoxicol 2021;16(4):32-42 View Article
  122. Leite SB, Roosens T, El Taghdouini A, Mannaerts I, Smout AJ, Najimi M, et al. Novel human hepatic organoid model enables testing of drug-induced liver fibrosis in vitro. Biomaterials 2016;78:1-10 View Article PubMed/NCBI
  123. Li X, Zheng M, Xu B, Li D, Shen Y, Nie Y, et al. Generation of offspring-producing 3D ovarian organoids derived from female germline stem cells and their application in toxicological detection. Biomaterials 2021;279:121213 View Article PubMed/NCBI
  124. Susa K, Kobayashi K, Galichon P, Matsumoto T, Tamura A, Hiratsuka K, et al. ATP/ADP biosensor organoids for drug nephrotoxicity assessment. Front Cell Dev Biol 2023;11:1138504 View Article PubMed/NCBI
  125. Wang Z, Xing C, van der Laan LJW, Verstegen MMA, Spee B, Masereeuw R. Cholangiocyte organoids to study drug-induced injury. Stem Cell Res Ther 2024;15(1):78 View Article PubMed/NCBI
  126. Chen S, Abdulla A, Yan H, Mi Q, Ding X, He J, et al. Proteome signatures of joint toxicity to arsenic (As) and lead (Pb) in human brain organoids with optic vesicles. Environ Res 2024;243:117875 View Article PubMed/NCBI
  127. Kim JW, Nam SA, Seo E, Lee JY, Kim D, Ju JH, et al. Human kidney organoids model the tacrolimus nephrotoxicity and elucidate the role of autophagy. Korean J Intern Med 2021;36(6):1420-1436 View Article PubMed/NCBI
  128. Yamamoto Y, Gotoh S, Korogi Y, Seki M, Konishi S, Ikeo S, et al. Long-term expansion of alveolar stem cells derived from human iPS cells in organoids. Nat Methods 2017;14(11):1097-1106 View Article PubMed/NCBI
  129. Han X, Shen PL, Ma MY, Jia YY. Organoids and Organ-on-Chips: Cutting-Edge Strategies Chinese Medicine. J Nanjing Univ Tradit Chin Med 2025;41(7):847-855 View Article
  130. Chen Z. Pien Tze Huang (PZH) as a Multifunction Medicinal Agent in Traditional Chinese Medicine (TCM): a review on cellular, molecular and physiological mechanisms. Cancer Cell Int 2021;21(1):146 View Article PubMed/NCBI
  131. Huang L, Zhang Y, Zhang X, Chen X, Wang Y, Lu J, et al. Therapeutic Potential of Pien-Tze-Huang: A Review on Its Chemical Composition, Pharmacology, and Clinical Application. Molecules 2019;24(18):3274 View Article PubMed/NCBI
  132. Gou H, Su H, Liu D, Wong CC, Shang H, Fang Y, et al. Traditional Medicine Pien Tze Huang Suppresses Colorectal Tumorigenesis Through Restoring Gut Microbiota and Metabolites. Gastroenterology 2023;165(6):1404-1419 View Article PubMed/NCBI
  133. Jia D, Liu C, Zhu Z, Cao Y, Wen W, Hong Z, et al. Novel transketolase inhibitor oroxylin A suppresses the non-oxidative pentose phosphate pathway and hepatocellular carcinoma tumour growth in mice and patient-derived organoids. Clin Transl Med 2022;12(11):e1095 View Article PubMed/NCBI
  134. Huang R, Su LL, Ji D, Yin FZ, Lu TL, Shao Y, et al. Panorama of Organoid Technologies in Chinese Medicine: Opportunities and Challenges from Basie Mechanisms to Clinical Practice. J Nanjing Univ Tradit Chin Med 2025;41(7):865-868 View Article
  135. Zhang S, Liu F, Li J, Jing C, Lu J, Chen X, et al. A 4.7-kDa polysaccharide from Panax ginseng suppresses Aβ pathology via mitophagy activation in cross-species Alzheimer’s disease models. Biomed Pharmacother 2023;167:115442 View Article PubMed/NCBI
  136. Kim S, Lee SI, Kim N, Joo M, Lee KH, Lee MW, et al. Decursin inhibits cell growth and autophagic flux in gastric cancer via suppression of cathepsin C. Am J Cancer Res 2021;11(4):1304-1320 PubMed/NCBI
  137. Yuan M, Gao JP, Wang XB, Xu HX. Research Progress in Toxicology of Chinese Medicines. Asia Pac Tradit Med 2024;20(1):225-232
  138. Gu S, Wu G, Lu D, Wang Y, Tang L, Zhang W. Human kidney organoids model of Esculentoside A nephrotoxicity to investigate the role of epithelial-mesenchymal transition via STING signaling. Toxicol Lett 2023;373:172-183 View Article PubMed/NCBI
  139. Chen H, Wen Y, Yu Z, Du X, Pan W, Liu T. Codonopsis pilosula polysaccharide alleviates rotenone-induced murine brain organoids death through downregulation of gene body DNA methylation modification in the ZIC4/PGM5/CAMTA1 axis. Biochem Biophys Rep 2024;37:101593 View Article PubMed/NCBI
  140. Yang J, Jiang Y, Li M, Wu K, Wei S, Zhao Y, et al. Organoid, organ-on-a-chip and traditional Chinese medicine. Chin Med 2025;20(1):22 View Article PubMed/NCBI
  141. Yi SA, Zhang Y, Rathnam C, Pongkulapa T, Lee KB. Bioengineering Approaches for the Advanced Organoid Research. Adv Mater 2021;33(45):e2007949 View Article PubMed/NCBI
  142. Goldsmith TM, Sakib S, Webster D, Carlson DF, Van der Hoorn F, Dobrinski I. A reduction of primary cilia but not hedgehog signaling disrupts morphogenesis in testicular organoids. Cell Tissue Res 2020;380(1):191-200 View Article PubMed/NCBI
  143. Smits LM, Reinhardt L, Reinhardt P, Glatza M, Monzel AS, Stanslowsky N, et al. Modeling Parkinson’s disease in midbrain-like organoids. NPJ Parkinsons Dis 2019;5:5 View Article PubMed/NCBI
  144. Tan W, Chen J, Wang Y, Xiang K, Lu X, Han Q, et al. Single-cell RNA sequencing in diabetic kidney disease: a literature review. Ren Fail 2024;46(2):2387428 View Article PubMed/NCBI
  145. Wang S, Sun ST, Zhang XY, Ding HR, Yuan Y, He JJ, et al. The Evolution of Single-Cell RNA Sequencing Technology and Application: Progress and Perspectives. Int J Mol Sci 2023;24(3):2943 View Article PubMed/NCBI
  146. Zhao Y, Li ZX, Zhu YJ, Fu J, Zhao XF, Zhang YN, et al. Single-Cell Transcriptome Analysis Uncovers Intratumoral Heterogeneity and Underlying Mechanisms for Drug Resistance in Hepatobiliary Tumor Organoids. Adv Sci (Weinh) 2021;8(11):e2003897 View Article PubMed/NCBI
  147. Jovic D, Liang X, Zeng H, Lin L, Xu F, Luo Y. Single-cell RNA sequencing technologies and applications: A brief overview. Clin Transl Med 2022;12(3):e694 View Article PubMed/NCBI
  148. Salaris F, Colosi C, Brighi C, Soloperto A, Turris V, Benedetti MC, et al. 3D Bioprinted Human Cortical Neural Constructs Derived from Induced Pluripotent Stem Cells. J Clin Med 2019;8(10):1595 View Article PubMed/NCBI
  149. Juraski AC, Sharma S, Sparanese S, da Silva VA, Wong J, Laksman Z, et al. 3D bioprinting for organ and organoid models and disease modeling. Expert Opin Drug Discov 2023;18(9):1043-1059 View Article PubMed/NCBI
  150. Kong JS, Huang X, Choi YJ, Yi HG, Kang J, Kim S, et al. Promoting Long-Term Cultivation of Motor Neurons for 3D Neuromuscular Junction Formation of 3D In Vitro Using Central-Nervous-Tissue-Derived Bioink. Adv Healthc Mater 2021;10(18):e2100581 View Article PubMed/NCBI
  151. Hamet P, Tremblay J. Artificial intelligence in medicine. Metabolism 2017;69S:S36-S40 View Article PubMed/NCBI
  152. Shi H, Kowalczewski A, Vu D, Liu X, Salekin A, Yang H, et al. Organoid intelligence: Integration of organoid technology and artificial intelligence in the new era of in vitro models. Med Nov Technol Devices 2024;21:100276 View Article PubMed/NCBI
  153. Sun Y, Huang F, Zhang H, Jiang H, Luo G. [A review on depth perception techniques in organoid images]. Sheng Wu Yi Xue Gong Cheng Xue Za Zhi 2024;41(5):1053-1061 View Article PubMed/NCBI
  154. Chen H, Yu L, Dou Q, Shi L, Mok VCT, Heng PA. Automatic detection of cerebral microbleeds via deep learning based 3D feature representation. 2015 IEEE 12th International Symposium on Biomedical Imaging (ISBI). Brooklyn, NY, USA 2015:764-767 View Article
  155. Wu H, Wang H, Wang L, Luo X, Zou D. Application progress and challenges of artificial intelligence in organoid research. China Oncology 2024;34(2):210-219 View Article
  156. He C, Kalafut NC, Sandoval SO, Risgaard R, Sirois CL, Yang C, et al. BOMA, a machine-learning framework for comparative gene expression analysis across brains and organoids. Cell Rep Methods 2023;3(2):100409 View Article PubMed/NCBI
  157. Prince E, Cruickshank J, Ba-Alawi W, Hodgson K, Haight J, Tobin C, et al. Biomimetic hydrogel supports initiation and growth of patient-derived breast tumor organoids. Nat Commun 2022;13(1):1466 View Article PubMed/NCBI
  158. Zhu M, Song C, Du Y, Long M. Fibrotic microenvironment regulates chronic liver injury repair based on liver organoid microarray studie. Journal of Medical Biomechanics 2024;39(S1):675
  159. Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol 2009;9(5):313-323 View Article PubMed/NCBI
  160. Yoon YJ, Kim D, Tak KY, Hwang S, Kim J, Sim NS, et al. Salivary gland organoid culture maintains distinct glandular properties of murine and human major salivary glands. Nat Commun 2022;13(1):3291 View Article PubMed/NCBI
  161. Chen J, Hong C, Wang X. Research progress and challenges in the application of gastrointestinal cancer organoids. J Dig Oncol (Electronic Version) 2021;13(4):294-297
  162. Li Y, Liao W, Sun L. Application of tumor organoids simulating the tumor microenvironment in basic and clinical research of tumor immunotherapy. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2024;49(8):1316-1326 View Article PubMed/NCBI
  163. Yang R, Yu Y. Patient-derived organoids in translational oncology and drug screening. Cancer Lett 2023;562:216180 View Article PubMed/NCBI
  164. Xu R, Zhou X, Wang S, Trinkle C. Tumor organoid models in precision medicine and investigating cancer-stromal interactions. Pharmacol Ther 2021;218:107668 View Article PubMed/NCBI
  165. Romero-López M, Trinh AL, Sobrino A, Hatch MM, Keating MT, Fimbres C, et al. Recapitulating the human tumor microenvironment: Colon tumor-derived extracellular matrix promotes angiogenesis and tumor cell growth. Biomaterials 2017;116:118-129 View Article PubMed/NCBI
  166. Lan L, Huang H. Progress in organoid research and ethical consideration. Biology Teaching 2023;48(9):4-7 View Article
  167. Zhou Z, Cong L, Cong X. Patient-Derived Organoids in Precision Medicine: Drug Screening, Organoid-on-a-Chip and Living Organoid Biobank. Front Oncol 2021;11:762184 View Article PubMed/NCBI
  168. Chen Q, Zhao S, Peng Y. Organoids: technological innovation and ethical controversies. Synthetic Biology Journal 2024;5(4):898-907 View Article
  169. Yang H, Cheng J, Zhuang H, Xu H, Wang Y, Zhang T, et al. Pharmacogenomic profiling of intra-tumor heterogeneity using a large organoid biobank of liver cancer. Cancer Cell 2024;42(4):535-551.e8 View Article PubMed/NCBI
  170. Farin HF, Mosa MH, Ndreshkjana B, Grebbin BM, Ritter B, Menche C, et al. Colorectal Cancer Organoid-Stroma Biobank Allows Subtype-Specific Assessment of Individualized Therapy Responses. Cancer Discov 2023;13(10):2192-2211 View Article PubMed/NCBI

About this Article

Cite this article
Shen X, Jiang H, Fan X, Duan X, Lin T, Li W, et al. Innovations in Organoid Engineering: Construction Methods, Model Development, and Clinical Translation. Future Integr Med. Published online: Sep 19, 2025. doi: 10.14218/FIM.2025.00023.
Copy        Export to RIS        Export to EndNote
Article History
Received Revised Accepted Published
March 31, 2025 May 21, 2025 July 8, 2025 September 19, 2025
DOI http://dx.doi.org/10.14218/FIM.2025.00023
  • Future Integrative Medicine
  • pISSN 2993-5253
  • eISSN 2835-6357
Back to Top

Innovations in Organoid Engineering: Construction Methods, Model Development, and Clinical Translation

Xue Shen, Haiyan Jiang, Xiaoyu Fan, Xiaoyan Duan, Tusi Lin, Wanfang Li, Jie Bao, Jia Xu, Bosai He, Hongtao Jin
  • Reset Zoom
  • Download TIFF