Open Access

Combination of tumor organoids with advanced technologies: A powerful platform for tumor evolution and treatment response (Review)

  • Authors:
    • Ying Wu
    • Fan Zhang
    • Furong Du
    • Juan Huang
    • Shuqing Wei
  • View Affiliations

  • Published online on: March 27, 2025     https://doi.org/10.3892/mmr.2025.13505
  • Article Number: 140
  • Copyright: © Wu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Malignant tumors notably decrease life expectancy. Despite advances in cancer diagnosis and treatment, the mechanisms underlying tumorigenesis, progression and drug resistance have not been fully elucidated. An emerging method to study tumors is tumor organoids, which are a three‑dimensional miniature structure. These retain the patient‑specific tumor heterogeneity while demonstrating the histological, genetic and molecular features of original tumors. Compared with conventional cancer cell lines and animal models, patient‑derived tumor organoids are more advanced at physiological and clinical levels. Their synergistic combination with other technologies, such as organ‑on‑a‑chip, 3D‑bioprinting, tissue‑engineered cell scaffolds and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR‑associated protein 9, may overcome limitations of the conventional 3D organoid culture and result in the development of more appropriate model systems that preserve the complex tumor stroma, inter‑organ and intra‑organ communications. The present review summarizes the evolution of tumor organoids and their combination with advanced technologies, as well as the application of tumor organoids in basic and clinical research. 

Introduction

Cancer, a leading cause of mortality, notably decreases life expectancy. In 2022, there were ~20.0 million new cases and ~9.7 million cancer-associated mortalities worldwide (1). Despite advances in cancer diagnosis and treatment, mechanisms underlying tumorigenesis, progression and drug resistance have not been fully elucidated. Due to tumor heterogeneity, different individuals with the same type of cancer may exhibit different responses to the same therapy; thus models need to be established that can recapitulate the tumors to study the mechanisms of tumorigenesis, progression and drug resistance.

Preclinical models include two-dimensional (2D) cell lines, patient-derived xenografts (PDXs) and organoids. Despite simple operation and culture, 2D cell lines cannot definitively predict the drug response of patients due to accumulation of gene mutations during passaging (2). Additionally, 2D cell lines are unreliable compared with in vivo models because of variations in cell phenotypical behaviors (3). PDXs, which are created by engraftment of patient tumor tissue into immunocompetent mice, recapitulate the tumor heterogeneity while preserving the biological and molecular features of original tumors (4,5), but they are time-consuming, expensive and may undergo mouse-specific tumor evolution rendering them unable to reflect the pathogenic process of patients (6,7). Therefore, application of PDXs is limited by the complex operation, duration, high cost and low success rate. Organoids, a novel type of three-dimensional (3D) miniature structure derived from adult or embryonic stem cells (SCs), not only retains in vivo tumor characteristics and heterogeneity, but also can predict the sensitivity of multiple drugs simultaneously, with the advantages of high success rate of generation, short time-frame and low cost (8) (Table I). Currently, organoids from multiple types of cancer have been established, including colorectal cancer (CRC), breast, pancreatic and lung cancer (912). These tumor organoids not only preserve the features of original tumors at genomic, molecular and epigenetic levels, but also contribute to predicting patient responses to therapies, thus offering potential for unveiling the biology of tumorigenesis, promoting drug discovery and personalized treatment in cancer.

Table I.

Advantages and limitations of cancer cell lines, PDXs and tumor organoids in cancer research.

Table I.

Advantages and limitations of cancer cell lines, PDXs and tumor organoids in cancer research.

CategoryCancer cell linesPDXsTumor organoids
AdvantagesSimple operation and culture; high throughputReplicate tumor heterogeneity while preserving the biological features of the tumor and predict the drug sensitivityHigh throughput; able to recapitulate the tumor heterogeneity while preserving the biological features of the tumor and predict the drug sensitivity; high success rate of generation, short time; low cost
LimitationsUnable to reflect the physiological status of tumors and predict drug sensitivity; difficulty in replicating tumor microenvironmentComplex operation; time-consuming; high cost; low success rate; can possibly undergo mouse-specific tumor evolutionLack of immune system; unable to model tumor-stroma interactions

[i] PDX, patient-derived xenograft.

The present review aimed to summarize evolution of tumor organoids and their combination with advanced technologies, such as organ-on-a-chip, 3D-bioprinting, tissue-engineered cell scaffolds and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (CRISPR-Cas9), as well as the application of tumor organoids in basic and clinical research.

Definition and evolution of organoids

Organoids, a type of 3D microstructure cultured in vivo under similar conditions to the human microenvironment, are primarily established based on the self-organization and differentiation of cells with stem cell characteristics (13). Organoids exhibit a natural self-organizing structure similar to native organs. In 1907, separated sponge cells were shown to self-organize into a complete organism (14). Subsequently, limbal SCs from 3T3 trophoblast cells were transplanted into damaged eyes following culture, laying the basis for the development of 3D organoid technology (15,16). Induced pluripotent SCs (iPSCs) are widely used in in vitro and in vivo preclinical studies (1719). Since the first human iPSC-derived motor neurons from patients were cultured in a petri dish, several techniques, such as somatic cell nuclear transfer approach and transcription factor-based somatic cell reprogramming, have been developed to generate iPSC lines (20). Since 2009, when the intestinal organoids were first cultured successfully, organoids have gained much attention in cancer research (21,22).

In 2011, gut and retinal organoids from SCs were generated successfully (23,24). Long-term culture protocols for primary colon adenoma/adenocarcinoma have been established (25). In 2014, prostate cancer organoids from metastatic biopsy and circulating tumor cells (CTCs) were first constructed (26) and breast cancer organoids derived from CTCs resemble the immunohistochemical features of breast tumors (27). In 2015, Boj et al (28) first generated pancreatic organoids from human neoplastic tissue; orthotopically transplanted organoids can be used to study the pathogenesis of pancreatic ductal adenocarcinoma. In the same year, Bartfeld et al (29) demonstrated the feasibility of establishing human gastric cancer organoids. Subsequently, other types of tumor organoid have been established, including glioblastoma and liver, endometrial, bladder and esophageal cancer (3039) (Fig. 1). Therefore, tumor organoids have value in cancer research. With the rapid development of biotechnology, they can not only supplement to the current evidence-based medicine, but also have advantages in clinical and translational research (40,41). Tumor organoids combined with advanced technologies, such as organ-on-a chip and 3D bio-printing, are also emerging, which not only promote the drug-testing process, but also reveal the synergistic effect in complex treatment regimens.

Combination of tumor organoids with advanced technologies

Tumor organoids reflect diverse key characteristics of tumor progression, but they lack characteristics such as vasculature, stomal components and tissue-resident immune cells (42). Moreover, multiple biophysical and biochemical factors from the tumor microenvironment (TME) are difficult to replicate accurately using conventional 3D organoid culture. Therefore, technologies, including organ-on-a-chip, 3D bio-printing, tissue-engineered cell scaffolds and CRISPR-Cas9 (Table II), are combined with tumor organoids as more precise models to study the mechanisms of tumorigenesis, progression and drug resistance (Fig. 2).

Table II.

Advantages and limitations of advanced technologies.

Table II.

Advantages and limitations of advanced technologies.

Category Organ-on-a-chip3D bio-printingTissue-engineered cell scaffoldsCRISPR-Cas9
AdvantagesSpecific stroma; a small amount of tissue required for analysis; high-resolution optical measurement; real-time tracking of organoid morphogenesis; inexpensive to manufacturePrecisely controls the spatial arrangement of cells, biomaterials and soluble factors; can scale up the organoid systems with hierarchical architectureSupports cell proliferation and attachment; simulates extracellular matrix function in vivo; can enlarge the organoid systems with vascular networkIdentifies novel targets for cancer therapy; produces genetically inhibited models for drug development
LimitationsHigh variability and lack of standardization in methods; small size of the organoid systemUnable to replicate tumor progression when monodispersed tumor cells are used as bio-printing building blocksDifficult to adjust for the unique tissue microenvironmentHigh off-target effects; difficult to validate the mutations in target genes

[i] CRISPR-Cas9, clustered regularly interspaced short palindromic repeats-associated protein 9.

Organ-on-a-chip

As a microfabricated device, the organ-on-chip is designated to integrate the culture of extracellular matrix (ECM), living cells and microstructures imitating organs or tissue (43,44). Integrating living human cells into a synthetically produced microenvironment models physiological homeostasis and the process of complex diseases (Fig. 3A) (43). Demers et al (45) developed a versatile microfluidic platform that mimics in vivo spatial and temporal chemical environments during neural tube development. Using similar techniques, Wang et al (46) developed a brain organoid-on-a-chip system from human iPSCs, promoting 3D culture, in situ neural differentiation and self-organization of brain organoids under continuous perfusion of neural differentiation medium in a controlled manner.

The organ-on-a-chip has the advantages of specific stroma, requiring a small amount of tissue for analysis, high-resolution optical measurement, real-time tracking of organoid morphogenesis and inexpensive manufacture. Although tumor microsystems are used to explore the cancer-specific hallmarks, the TME complexity cannot be recapitulated due to simultaneous or successive occurrence of cancer-specific hallmarks. Shirure et al (47) designed a tumor-on-a-chip microfluidic platform and that this could simultaneously model the hallmark characteristics of tumor progression in both cell lines and patient-derived organoids (PDOs), such as proliferation, migration, angiogenesis and intravasation.

At present, the culture environment of organoids lacks vascularization, leading to decreased organoid lifespan and changeability in tissue-specific functionality and architecture (48). In a previous study, 3D vascularized liver organoids comprising induced hepatic cells and decellularized liver ECM were developed based on the microfluidic system, exhibiting improved liver functionality, biosynthetic and metabolic activity, as well as drug response; this study also confirmed the feasibility of vascularized liver organ-on-a-chip systems as a high-throughput drug screening platform (49). To study tumor angiogenesis, the human primary clear cell renal cell carcinoma (ccRCC) cells are combined with endothelial cells in a vascularized, flow-directed, 3D culture system. Under continuous flow, cRCC clusters preserve the key angiogenic signaling axis between ccRCC and endothelial cells by promoting endothelial cell sprouting. This system signifies a vascularized tumor model with adjustable perfusate, input cells and matrices (50). The PDOs established in a multicellular microfluidic chip may prolong cellular function and longevity and construct an intricate organotypic TME (Fig. 3B). Targeting stroma in a tumor-chip model notably increases response to chemotherapy in cancer cells, further verifying the application of the tumor-chip device in drug testing (51). Additionally, multiorgan models of coculture with SC-derived stomach, intestinal and liver organoids have also been established, promoting the discovery of interorgan crosstalk characteristics (52). The aforementioned findings indicate how the organoids combined with organ-on-a-chip technique replicate cell maturity.

3D bio-printing

3D bio-printing accurately controls the spatial arrangement of cells, biomaterials and soluble factors, forming intricate multicellular structures (53,54). By offering tumor-specific ECM, accurate geometric architecture and biophysical properties, bio-printing can replicate the TME, thus promoting the establishment of complicated and controllable 3D tissue models. In most studies, however, monodispersed tumor cells used as bio-printing building blocks do not effectively replicate the tumor progression due to the rare presence of volumetric tumor cells in isolation (55,56). The combination of 3D bio-printing and tumor organoids allows for the introduction of miniaturized tumor aggregations into a heterogeneous 3D niche containing stromal cells and hydrogels, which are more cell-specific for simulation of TME features and high-throughput drug screening (Fig. 4A) (57,58).

Mollica et al (59) revealed that 3D bio-printed organoids and tumoroid formation are preserved effectively using novel 3D culture substrates. Reid et al (60) applied bio-printing to analyze tumorigenesis and microenvironmental redirection in breast cancer cells and demonstrated that bio-printing could promote the formation of tumor organoids in 3D collagen gels and tumor organoid arrays. Moreover, in vivo findings are accurately simulated through bio-printed organoids, highlighting the feasibility of the 3D bio-printing technique in understanding tumorigenesis and TME control. To increase the throughput of 3D drug screening, an immersion bio-printing technique has been developed to bio-print tumor organoids in multi-well plates (Fig. 4B) (61). Additionally, a 3D-bioprinted construct is used to test the resistance of anti-cancer drugs (sorafenib, cisplatin and 5-fluorouracil) in patient-derived cholangiocarcinoma cells. Compared with 2D cultures, bio-printed cholangiocarcinoma cells exhibit stem-like properties and high resistance to the aforementioned anti-cancer drugs, indicating the potential of 3D bio-printed tumor models in the discovery of targeted drugs (62). Additionally, bio-printed organoids undergo hepatocytic differentiation, including liver-specific enzyme activity and albumin synthesis (63), creating novel possibilities for regenerative medicine and individualized drug screening.

Tissue-engineered cell scaffolds

Matrigel is key for the culture of most organoids, but it may elevate the risk of animal-derived microbial infection and batch-to-batch variability in organoids, leading to unreproducible experimental results (64,65). Tissue-engineered cell scaffolds support cell proliferation and attachment and simulate ECM function in vivo (66). For cell- and tissue-derived matrices, synthetic scaffolds, such as polyethylene glycol (PEG)-based hydrogel scaffolds, allow control over the growth conditions.

For conventional organoid cultures, generic matrices are usually applied, but they are difficult to adjust to replicate the unique TME. Ng et al (67) used gelatin-based hydrogels to demonstrate CRC organoid sensitivity to multiple drugs in vivo, and found that these hydrogels may be a promising platform for biochemically and mechanically defined matrices used in multiple types of tumor organoid. In a previous study, a fully synthetic hydrogel scaffold was constructed based on the 8-arm PEG and pancreatic cancer organoids were generated successfully (68). Through regulation of hydrogel properties, the proliferation of pancreatic cancer organoids is controlled, and the phenotypic traits of the TME in vivo are effectively replicated when stromal cells are incorporated into the hydrogels (68). These findings suggest that synthetic scaffolds replicate a pathologically remodeled TME for studying normal and pancreatic cancer cells in vitro. Another study showed that ECM hydrogels generated organoids appropriate for gastrointestinal disease modeling, tissue regeneration and drug development (Fig. 5A), which may serve as effective alternatives to Matrigel (69). Accordingly, tissue-engineered cell scaffolds are promising next-generation materials for organoid technology to understand organ-based developmental biology and predict drug response in tumor organoids (6769).

CRISPR-Cas9

CRISPR-Cas9 enables more efficient gene knockout and knock-in than other types of genome editor through introduction of DNA double-strand breaks at specific genomic loci (70). In addition to identification of novel targets for cancer therapy, CRISPR-Cas9 is also used to produce genetically inhibited animal models for drug development (71,72). CRISPR-Cas9 combined with 3D organoid systems facilitates development of precise cancer models for studying diverse mechanisms of tumor progression, metastasis, interactions and drug resistance. Organoid systems not only mimic the human disease and tailor therapeutic strategies, but also serve as an experimental platform for mechanistically studying the gene function in humans (73).

Matano et al (74) introduced mutations into organoids from normal human intestinal epithelium using CRISPR-Cas9 and demonstrated that the isogenic organoids with mutations show tumorigenicity in mice. Murine gallbladder organoids with KRAS mutations or overexpression of Erb-B2 receptor tyrosine kinase 2 cause gallbladder cancer in transplanted immunocompromised mice when CRISPR-Cas9 is used for p53 or PTEN deletion (75). In another study, specific subtypes of breast cancer organoids were established following targeted knockdown of four breast cancer-associated suppressor genes in mammary progenitor cells through CRISPR-Cas9; these organoids respond to endocrine therapy or chemotherapy (Fig. 5B and C) (76,77). Vaishnavi et al (78) used CRISPR/Cas9 technology to silence RNA binding motif, single stranded interacting protein 3 (RBMS3); this facilitated the growth of BRAFV600E lung organoids and malignant progression of lung cancer. Additionally, CRISPR-Cas9 was also used to validate CRC driver genes in mouse intestinal tumor organoids and human CRC-derived organoids (79). Notably, CRISPR-Cas9-mediated homology-independent organoid transgenesis, a genetic tool for labeling specific genes in human organoids, effectively generates genetically engineered human liver duct and fetal hepatocyte organoids within 2–3 months (80,81). This genetic tool can be used to study cell fates and differentiation and identify targets for drug development, showing promise for cancer research.

Application of tumor organoids

Mechanisms of tumorigenesis and drug resistance

Tumorigenesis and progression primarily depend on the accumulation of genetic alterations. Understanding the mutational process is key to analyzing the mechanism of tumorigenesis. Multiple studies have demonstrated the feasibility of introducing pathological mutations into normal organoids using genetic modification to simulate tumorigenesis (74,76,82,83) (Fig. 6). As reported by Matano et al (74), isogenic organoids with mutations show tumorigenicity in mice when gene mutations from driver pathways are introduced into organoids derived from normal human intestinal epithelium (84). By establishing AT-rich binding domain protein 1A-deficient human gastric cancer organoids using CRISPR/Cas9 technology, modes of oncogenic transformation are revealed, including essential transcriptional forkhead box protein M1/baculoviral IAP repeat-containing 5-stimulated proliferation and non-essential Wnt-inhibited mucinous differentiation (82). As a key cause of mortality in patients with cancer, the migration and invasion of tumor cells also serve important roles in tumor progression. Through coculture with mammary tumor organoids, a tissue-engineered model with physiologically realistic microvessels was created, which allows quantitative and real-time evaluation of tumor-vessel interactions under the conditions that retain various in vivo characteristics to identify targetable mechanisms of vascular recruitment and intravasation (84). In the patient-derived breast cancer organoids, CD homophilic interactions and subsequent CD44-p21-activated kinase 2 interactions mediate tumor cluster migration and aggregation (85). Moreover, invasion could also be triggered in breast cancer subtypes by basal epithelial gene expression (86).

To investigate the role of tumor organoids in treatment resistance, Boos et al (87) modeled the acquisition of chemotherapy tolerance in metastatic CRC organoids during first-line combined chemotherapy, highlighting the potential of CRC organoids in modeling chemotherapy tolerance in vivo. Similarly, in intestinal cancer organoids, atypical expression of cyclin P facilitates stemness-like phenotypes (88), which usually results in tumor metastasis, relapse and treatment resistance. Through establishment of oxaliplatin-resistant organoids and assessment of their gene expression, myoferlin was shown to be involved in oxaliplatin resistance and tumor progression in gastric cancer (89). As commonly used anti-tumor drugs in prostate cancer, resistance to androgen receptor pathway inhibitors may develop due to epigenetic reprogramming turning castration-resistant adenocarcinoma to neuroendocrine prostate cancer. Bioengineered ECM regulates the response of prostate cancer organoids to small-molecule inhibitors of epigenetic targets and dopamine receptor D2, suggesting that synthetic organoids exert a regulatory effect on ECM in response to targeted therapy in prostate cancer and serve as a novel treatment strategy to overcome resistance (90). Overall, tumor organoids serve as a powerful platform for studying drug resistance mechanisms.

Precision medicine

Tumor organoids not only contribute to understanding the mechanism of tumorigenesis, but also predict the response to therapies, including chemotherapy, radiotherapy and targeted therapy (9193). Vlachogiannis et al (92) established organoids from metastatic gastrointestinal cancer to predict the response to targeted drugs or chemotherapy, showing sensitivity of 100 and specificity of 93%. Gastric cancer organoids faithfully reflect responses to commonly used chemotherapy drugs, such as irinotecan, oxaliplatin, docetaxel, epirubicin and 5-fluerouracil (91). Tiriac et al (94) generated a pancreatic cancer organoid library and found that PDO profiling based on the next-generation sequencing of DNA and RNA and pharmacotyping may predict responses to chemotherapy in pancreatic cancer. Ji et al (95) identified potential drug combination therapies based on pharmaco-proteogenomic profiling of liver cancer organoids, offering guidance for clinical patient selection and drug combination therapies. Ganesh et al (93) revealed the heterogeneity of rectal cancer organoids in chemoradiation and ex vivo responses to clinically relevant chemoradiation associated with clinical responses. Moreover, KRAS-wild-type rectal cancer organoids are sensitive to cetuximab, while KRAS-mutant organoids are resistant, which is consistent with the results of a clinical trial that KRAS mutations are associated with resistance to EGFR-targeted therapy (93). Importantly, in a prospective, interventional clinical trial of the last-line systemic therapy based on PDOs, improved clinical outcomes were observed in patients with CRC compared with those receiving the best supportive care alone (96). In addition, the association between tumor organoids and clinical response has been identified in other types of cancer. By establishing PDOs from different stages of bladder cancer, Minoli et al (97) demonstrated that the PDOs exhibit heterogenous drug responses to standard-of-care treatment and drug screening showed sensitivity to targeted therapy. In a real-world study, lung cancer organoids were used to validate the response to osimertinib, chemotherapy and dual-targeted therapy and a high concordance was identified between lung cancer organoids and clinical response (12). PDO pharmaco-phenotyping not only reflects previous treatment responses of patients with advanced breast cancer but also serves as a potential platform to guide personalized treatment (9). Breast cancer organoids may also be used to predict patient-specific response to drug treatment (98).

Coculture of tumor organoids with immune components may generate tumor-reactive T cells, which may promote the prediction and evaluation of tumor responses at an individual level by blocking PD-1/PD-L1 (98,99). Cattaneo et al (100) described the generation and function of tumor-reactive T cells based on the coculture of tumor organoids with peripheral blood mononuclear cells (PBMCs), demonstrating the feasibility of establishing ex vivo models of T cell immunotherapy at an individual level. Meng et al (101) developed a platform for the expansion of tumor-targeted T cells from peripheral blood and revealed that the coculture of tumor organoids with PBMCs generates tumor-reactive T cells, thereby promoting personalized immunotherapy. Moreover, tumor organoids cocultured with PBMCs are also used to enrich tumor-reactive T cells from peripheral blood of patients with mismatch repair-deficient CRC and non-small-cell lung cancer (SCLC); these T cells are useful for assessing the killing efficiency of matched tumor organoids (102). In certain organoids established from immunotherapy-responsive tumors, activation of T cells and tumor killing activity have been identified using PD-1/PD-L1 blockade (98). Meanwhile, Votanopoulos et al (103) developed an immune-enhanced tumor organoid model with a high clinical association between these organoids and response to checkpoint inhibitors. Collectively, tumor organoids can predict the response to immunotherapy in cancer.

Identification of novel treatment targets

In a longitudinal, observational co-clinical study, second mitochondria-derived activator of caspase mimetic LCL161 was shown to serve as a treatment target in the organoids derived from recurrent, KRAS-mutated liver metastases from rectal cancer (104). Compared with THZ1, YPN-005, a potent inhibitor of CDK7, shows potent antitumor effects in SCLC organoids, suggesting its treatment value in SCLC (105). Based on the interaction of breast cancer organoids and tumor-specific cytotoxic T cells, epigenetic inhibitors GSK-LSD1, CUDC-101 and BML-210 identified via high-throughput screening show antitumor activities (106). Furthermore, BML-210 promotes the efficacy of PD-1-based immune checkpoint blockage.

The tumor-suppressing function and efficient delivery of drugs have key roles in cancer treatment. In multicellular hepatocellular carcinoma organoids (MCHOs) with activated Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) signaling, there is stromal activation and damaged penetration of verteporfin; inhibiting YAP/TAZ transcriptional activity in hepatocellular carcinoma (HCC) cells may facilitate the penetration of drugs into MCHOs. These findings suggest that the treatment targeting activated tumor stroma may promote drug delivery into HCC cells with increased YAP/TAZ activity (107).

Modeling tumor vascularization

To improve understanding of angiogenic signaling pathways and investigate effective treatment strategies, tumor vasculature must be preserved in organoid cultures. For organoid vascularization, implantation of organoids into highly vascularized tissue is a frequently used method (108,109). Another method is coculture with cells including vascular smooth muscle and epithelial cells (ECs) based on gene editing or microfluidic platforms (110). By integrating mesodermal progenitor cells into organoids, Wörsdörfer et al (111) found that the vascularized organoids formed following coculture with tumor cells or neural spheroids and the vessels in tumor organoids are associated with the host vessels after transplantation. Breast cancer organoids with ECs and immune cells show an obvious angiogenic response when cultured with the vascular network (112). Similarly, in the collagen- and hyaluronic acid-enriched ECM with human fibroblasts and MCF-7 cells, vascularized breast cancer organoids have been established successfully (113). Additionally, in the coculture system of organoids and ECs, vascularization is triggered by vascular endothelial growth factors and hypoxia gradients based on compartmentalized microfluidic chips (109,114). The aforementioned findings underscore the importance of coculture models in organoid vascularization.

Modeling tumor-immune interactions

Coculture of tumor organoids with immune components, such as fibroblasts, stroma, ECs and immune cells, models the tumor-immune interactions, which provides insights into cancer immunotherapy (115). Using the tumor organoid culture for expansion and characterization of tumor-reactive T cells, Dijkstra et al (102) developed a multifunctional platform to study tumor-immune interactions and concluded that CD8+ T cells in PBMCs of the same patient are activated in half of the CRC organoids, with similar results in non-SCLC organoids. Meanwhile, a platform for expanding tumor-targeted T cells has been reported in patients with pancreatic cancer (116). By coculturing PBMCs and autologous tumor organoids, this platform enables recognition and expansion of tumor-targeted cytotoxic T cells (99). Based on the coculture of tumor organoids with PBMCs, the establishment and functional assessment of tumor-reactive T cells has also been described (98).

By generating organoids from surgically resected types of cancer based on the air-liquid interface, Neal et al (98) demonstrated that these organoid cultures retain various endogenous immune cell types and non-immune matrix components; immune checkpoint blockade with anti-PD-1 and/or anti-PD-L1 kills the tumor cells through induction of the expansion and activation of tumor antigen-specific T cells in organoid cultures. Moreover, a previous study suggested the potential of the organoid culture system in predicting adoptive immunotherapy responses following incorporation of patient-specific mature lymph node antigen-presenting cells into organoids (103).

Challenges

There are limitations to the application of tumor organoids that need to be addressed. First, there are no standardized evaluation criteria and culture protocols. Currently, the culture conditions of tumor organoids are diverse, leading to large differences in results between laboratories and teams. These differences may arise from inconsistent tissue dissociation, undefined formulation of culture medium and different matrices. To promote the standardization and reproducibility of tumor organoid cultures, culture conditions and laboratory operations should be unified as much as possible. Organoid culture is also affected by tumor cell composition, cell activity and tumor heterogeneity. For certain types of cancer, such as prostate cancer (26), the low success rate hinders repeatability and reproducibility, thereby affecting high-throughput screening. Hence, development of standard culture and evaluation protocols and application of well-defined materials are required for improving the success rate of organoid generation. Second, due to potential inclusion of normal cells, TME reconstruction is challenging. Tumor organoid models lack certain in vivo components, such as endothelial and immune cells and fibroblasts. Although it is challenging to establish the organoids comprising immune and vascular cells, this limitation may be resolved in the future with the development of organoid technology. Third, the currently established tumor organoids are primarily from ECs. In the future, studies should establish organoids from non-ECs, which may further optimize the treatment of tumors such as CRC and lung cancer (117,118). Additionally, during the long-term culture and passage of tumor organoids epigenetic drift may occur (43). To avoid normal cells being contaminated and make organoids more mature, investigating the mechanisms underlying epigenetic drift is needed. Shi et al assessed the tumor purity in long-term cultures and found that none were contaminated with normal or non-human cells (119). In addition to recapitulating the biology that drives histologic appearance of original tumors, their organoid models had not drifted at the molecular level. More importantly, tumor organoids should be improved to model the interactions between cells, tissue and organs. Although TME can be replicated through coculture with stromal cells and ECM, the role of peripheral immune systems is not evaluated (77). Combination of tumor organoids with advanced technologies allows modeling of a more complex and realistic state, which may overcome the aforementioned challenges and create more appropriate model systems for cancer treatment.

Conclusion

Patient-derived tumor organoids are more advanced at physiological and clinical levels compared with conventional cancer cell lines and PDXs. Despite challenges, tumor organoids show potential in the treatment of cancer. Tumor organoids may be combined with advanced technologies, such as organ-on-a-chip, 3D-bioprinting, tissue-engineered cell scaffolds and CRISPR-Cas9, which may not only overcome defects of conventional culture methods, but also expand the application range, offering insights into the treatment strategies in cancer. Combined application of tumor organoids and advanced technologies allows accurate simulation of tumor heterogeneity, vascularization and tumor-immune interactions, facilitating comprehensive high-throughput drug screening to predict drug responses and optimize treatment options to promote personalized treatment in cancer.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

YW, FZ, JH and SW conceived and designed the study and wrote the manuscript. FD acquired data and revised the manuscript critially. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

CRC

colorectal cancer

2D

two-dimensional

PDX

patient-derived xenograft

CRISPR-Cas9

clustered regularly interspaced short palindromic repeats-associated protein

iPSC

induced pluripotent stem cell

CTC

circulating tumor cell

TME

tumor microenvironment

ECM

extracellular matrix

PDO

patient-derived organoid

ccRCC

clear cell renal cell carcinoma

PEG

polyethylene glycol

MCHO

multicellular hepatocellular carcinoma organoid

HCC

hepatocellular carcinoma

EC

epithelial cell

PBMC

peripheral blood mononuclear cell

RBMS3

RNA binding motif, single stranded interacting protein 3

SCLC

small cell lung cancer

YAP

Yes-associated protein

TAZ

transcriptional coactivator with PDZ binding motif

References

1 

Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024. View Article : Google Scholar : PubMed/NCBI

2 

Liu C, Qin T, Huang Y, Li Y, Chen G and Sun C: Drug screening model meets cancer organoid technology. Transl Oncol. 13:1008402020. View Article : Google Scholar : PubMed/NCBI

3 

Magré L, Verstegen MMA, Buschow S, van der Laan LJW, Peppelenbosch M and Desai J: Emerging organoid-immune co-culture models for cancer research: From oncoimmunology to personalized immunotherapies. J Immunother Cancer. 11:e0062902023. View Article : Google Scholar : PubMed/NCBI

4 

Drapkin BJ, George J, Christensen CL, Mino-Kenudson M, Dries R, Sundaresan T, Phat S, Myers DT, Zhong J, Igo P, et al: Genomic and functional fidelity of small cell lung cancer patient-derived xenografts. Cancer Discov. 8:600–615. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Izumchenko E, Paz K, Ciznadija D, Sloma I, Katz A, Vasquez-Dunddel D, Ben-Zvi I, Stebbing J, McGuire W, Harris W, et al: Patient-derived xenografts effectively capture responses to oncology therapy in a heterogeneous cohort of patients with solid tumors. Ann Oncol. 28:2595–2605. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Saito R, Kobayashi T, Kashima S, Matsumoto K and Ogawa O: Faithful preclinical mouse models for better translation to bedside in the field of immuno-oncology. Int J Clin Oncol. 25:831–841. 2020. View Article : Google Scholar : PubMed/NCBI

7 

Ben-David U, Ha G, Tseng YY, Greenwald NF, Oh C, Shih J, McFarland JM, Wong B, Boehm JS, Beroukhim R and Golub TR: Patient-derived xenografts undergo mouse-specific tumor evolution. Nat Genet. 49:1567–1575. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Tang XY, Wu S, Wang D, Chu C, Hong Y, Tao M, Hu H, Xu M, Guo X and Liu Y: Human organoids in basic research and clinical applications. Signal Transduct Target Ther. 7:1682022. View Article : Google Scholar : PubMed/NCBI

9 

Chen P, Zhang X, Ding R, Yang L, Lyu X, Zeng J, Lei JH, Wang L, Bi J, Shao N, et al: Patient-derived organoids can guide personalized-therapies for patients with advanced breast cancer. Adv Sci (Weinh). 8:e21011762021. View Article : Google Scholar : PubMed/NCBI

10 

Hsu KS, Adileh M, Martin ML, Makarov V, Chen J, Wu C, Bodo S, Klingler S, Sauvé CG, Szeglin BC, et al: Colorectal cancer develops inherent radiosensitivity that can be predicted using patient-derived organoids. Cancer Res. 82:2298–2312. 2022. View Article : Google Scholar : PubMed/NCBI

11 

Seppälä TT, Zimmerman JW, Suri R, Zlomke H, Ivey GD, Szabolcs A, Shubert CR, Cameron JL, Burns WR, Lafaro KJ, et al: Precision medicine in pancreatic cancer: Patient-derived organoid pharmacotyping is a predictive biomarker of clinical treatment response. Clin Cancer Res. 28:3296–3307. 2022. View Article : Google Scholar : PubMed/NCBI

12 

Wang HM, Zhang CY, Peng KC, Chen ZX, Su JW, Li YF, Li WF, Gao QY, Zhang SL, Chen YQ, et al: Using patient-derived organoids to predict locally advanced or metastatic lung cancer tumor response: A real-world study. Cell Rep Med. 4:1009112023. View Article : Google Scholar : PubMed/NCBI

13 

Drost J and Clevers H: Organoids in cancer research. Nat Rev Cancer. 18:407–418. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Wilson HV: A new method by which sponges may be artificially reared. Science. 25:912–915. 1907. View Article : Google Scholar : PubMed/NCBI

15 

Lindberg K, Brown ME, Chaves HV, Kenyon KR and Rheinwald JG: In vitro propagation of human ocular surface epithelial cells for transplantation. Invest Ophthalmol Vis Sci. 34:2672–2679. 1993.PubMed/NCBI

16 

Pellegrini G, Traverso CE, Franzi AT, Zingirian M, Cancedda R and De Luca M: Long-term restoration of damaged corneal surfaces with autologous cultivated corneal epithelium. Lancet. 349:990–993. 1997. View Article : Google Scholar : PubMed/NCBI

17 

Kim JY, Nam Y, Rim YA and Ju JH: Review of the current trends in clinical trials involving induced pluripotent stem cells. Stem Cell Rev Rep. 18:142–154. 2022. View Article : Google Scholar : PubMed/NCBI

18 

Cheng H, Liu C, Cai X, Lu Y, Xu Y and Yu X: iPSCs derived from malignant tumor cells: Potential application for cancer research. Curr Stem Cell Res Ther. 11:444–450. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Orqueda AJ, Giménez CA and Pereyra-Bonnet F: iPSCs: A minireview from bench to bed, including organoids and the crispr system. Stem Cells Int. 2016:59347822016. View Article : Google Scholar : PubMed/NCBI

20 

Dimos JT, Rodolfa KT, Niakan KK, Weisenthal LM, Mitsumoto H, Chung W, Croft GF, Saphier G, Leibel R, Goland R, et al: Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science. 321:1218–1221. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, Stange DE, van Es JH, Abo A, Kujala P, Peters PJ and Clevers H: Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 459:262–265. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Kuratnik A and Giardina C: Intestinal organoids as tissue surrogates for toxicological and pharmacological studies. Biochem Pharmacol. 85:1721–1726. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Spence JR, Mayhew CN, Rankin SA, Kuhar MF, Vallance JE, Tolle K, Hoskins EE, Kalinichenko VV, Wells SI, Zorn AM, et al: Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature. 470:105–109. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Eiraku M, Takata N, Ishibashi H, Kawada M, Sakakura E, Okuda S, Sekiguchi K, Adachi T and Sasai Y: Self-organizing optic-cup morphogenesis in three-dimensional culture. Nature. 472:51–56. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Sato T, Stange DE, Ferrante M, Vries RG, Van Es JH, Van den Brink S, Van Houdt WJ, Pronk A, Van Gorp J, Siersema PD and Clevers H: Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett's epithelium. Gastroenterology. 141:1762–1772. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Gao D, Vela I, Sboner A, Iaquinta PJ, Karthaus WR, Gopalan A, Dowling C, Wanjala JN, Undvall EA, Arora VK, et al: Organoid cultures derived from patients with advanced prostate cancer. Cell. 159:176–187. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Yu M, Bardia A, Aceto N, Bersani F, Madden MW, Donaldson MC, Desai R, Zhu H, Comaills V, Zheng Z, et al: Cancer therapy. Ex vivo culture of circulating breast tumor cells for individualized testing of drug susceptibility. Science. 345:216–220. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Boj SF, Hwang CI, Baker LA, Chio II, Engle DD, Corbo V, Jager M, Ponz-Sarvise M, Tiriac H, Spector MS, et al: Organoid models of human and mouse ductal pancreatic cancer. Cell. 160:324–338. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Bartfeld S, Bayram T, van de Wetering M, Huch M, Begthel H, Kujala P, Vries R, Peters PJ and Clevers H: In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection. Gastroenterology. 148:126–136.e6. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Hubert CG, Rivera M, Spangler LC, Wu Q, Mack SC, Prager BC, Couce M, McLendon RE, Sloan AE and Rich JN: A three-dimensional organoid culture system derived from human glioblastomas recapitulates the hypoxic gradients and cancer stem cell heterogeneity of tumors found in vivo. Cancer Res. 76:2465–2477. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Broutier L, Mastrogiovanni G, Verstegen MM, Francies HE, Gavarró LM, Bradshaw CR, Allen GE, Arnes-Benito R, Sidorova O, Gaspersz MP, et al: Human primary liver cancer-derived organoid cultures for disease modeling and drug screening. Nat Med. 23:1424–1435. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Girda E, Huang EC, Leiserowitz GS and Smith LH: The use of endometrial cancer patient-derived organoid culture for drug sensitivity testing is feasible. Int J Gynecol Cancer. 27:1701–1707. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Pauli C, Hopkins BD, Prandi D, Shaw R, Fedrizzi T, Sboner A, Sailer V, Augello M, Puca L, Rosati R, et al: Personalized in vitro and in vivo cancer models to guide precision medicine. Cancer Discov. 7:462–477. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Kijima T, Nakagawa H, Shimonosono M, Chandramouleeswaran PM, Hara T, Sahu V, Kasagi Y, Kikuchi O, Tanaka K, Giroux V, et al: Three-Dimensional organoids reveal therapy resistance of esophageal and oropharyngeal squamous cell carcinoma cells. Cell Mol Gastroenterol Hepatol. 7:73–91. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Tanaka N, Osman AA, Takahashi Y, Lindemann A, Patel AA, Zhao M, Takahashi H and Myers JN: Head and neck cancer organoids established by modification of the CTOS method can be used to predict in vivo drug sensitivity. Oral Oncol. 87:49–57. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Kopper O, de Witte CJ, Lõhmussaar K, Valle-Inclan JE, Hami N, Kester L, Balgobind AV, Korving J, Proost N, Begthel H, et al: An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity. Nat Med. 25:838–849. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Kim M, Mun H, Sung CO, Cho EJ, Jeon HJ, Chun SM, Jung DJ, Shin TH, Jeong GS, Kim DK, et al: Patient-derived lung cancer organoids as in vitro cancer models for therapeutic screening. Nat Commun. 10:39912019. View Article : Google Scholar : PubMed/NCBI

38 

Grassi L, Alfonsi R, Francescangeli F, Signore M, De Angelis ML, Addario A, Costantini M, Flex E, Ciolfi A, Pizzi S, et al: Organoids as a new model for improving regenerative medicine and cancer personalized therapy in renal diseases. Cell Death Dis. 10:2012019. View Article : Google Scholar : PubMed/NCBI

39 

Chen D, Tan Y, Li Z, Li W, Yu L, Chen W, Liu Y, Liu L, Guo L, Huang W and Zhao Y: Organoid cultures derived from patients with papillary thyroid cancer. J Clin Endocrinol Metab. 106:1410–1426. 2021. View Article : Google Scholar : PubMed/NCBI

40 

Wörsdörfer P, Takashi I, Asahina I, Sumita Y and Ergün S: Do not keep it simple: Recent advances in the generation of complex organoids. J Neural Transm (Vienna). 127:1569–1577. 2020. View Article : Google Scholar : PubMed/NCBI

41 

Demyan L, Habowski AN, Plenker D, King DA, Standring OJ, Tsang C, St Surin L, Rishi A, Crawford JM, Boyd J, et al: Pancreatic cancer patient-derived organoids can predict response to neoadjuvant chemotherapy. Ann Surg. 276:450–462. 2022. View Article : Google Scholar : PubMed/NCBI

42 

Beato F, Reverón D, Dezsi KB, Ortiz A, Johnson JO, Chen DT, Ali K, Yoder SJ, Jeong D, Malafa M, et al: Establishing a living biobank of patient-derived organoids of intraductal papillary mucinous neoplasms of the pancreas. Lab Invest. 101:204–217. 2021. View Article : Google Scholar : PubMed/NCBI

43 

Ma C, Peng Y, Li H and Chen W: Organ-on-a-Chip: A new paradigm for drug development. Trends Pharmacol Sci. 42:119–133. 2021. View Article : Google Scholar : PubMed/NCBI

44 

Kutluk H, Bastounis EE and Constantinou I: Integration of extracellular matrices into organ-on-chip systems. Adv Healthc Mater. 12:e22032562023. View Article : Google Scholar : PubMed/NCBI

45 

Demers CJ, Soundararajan P, Chennampally P, Cox GA, Briscoe J, Collins SD and Smith RL: Development-on-chip: In vitro neural tube patterning with a microfluidic device. Development. 143:1884–1892. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Wang Y, Wang L, Zhu Y and Qin J: Human brain organoid-on-a-chip to model prenatal nicotine exposure. Lab Chip. 18:851–860. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Shirure VS, Bi Y, Curtis MB, Lezia A, Goedegebuure MM, Goedegebuure SP, Aft R, Fields RC and George SC: Tumor-on-a-chip platform to investigate progression and drug sensitivity in cell lines and patient-derived organoids. Lab Chip. 18:3687–3702. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Garreta E, Kamm RD, de Sousa Lopes SM, Lancaster MA, Weiss R, Trepat X, Hyun I and Montserrat N: Rethinking organoid technology through bioengineering. Nat Mater. 20:145–155. 2021. View Article : Google Scholar : PubMed/NCBI

49 

Jin Y, Kim J, Lee JS, Min S, Kim S, Ahn DH, Kim YG and Cho SW: Vascularized liver organoids generated using induced hepatic tissue and dynamic liver-specific microenvironment as a drug testing platform. Adv Funct Mater. 28:18019542018. View Article : Google Scholar

50 

Miller CP, Tsuchida C, Zheng Y, Himmelfarb J and Akilesh S: A 3D human renal cell carcinoma-on-a-chip for the study of tumor angiogenesis. Neoplasia. 20:610–620. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Haque MR, Wessel CR, Leary DD, Wang C, Bhushan A and Bishehsari F: Patient-derived pancreatic cancer-on-a-chip recapitulates the tumor microenvironment. Microsyst Nanoeng. 8:362022. View Article : Google Scholar : PubMed/NCBI

52 

Picollet-D'hahan N, Zuchowska A, Lemeunier I and Le Gac S: Multiorgan-on-a-Chip: A systemic approach to model and decipher inter-organ communication. Trends Biotechnol. 39:788–810. 2021. View Article : Google Scholar : PubMed/NCBI

53 

Mandrycky C, Wang Z, Kim K and Kim DH: 3D bioprinting for engineering complex tissues. Biotechnol Adv. 34:422–434. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Datta P, Barui A, Wu Y, Ozbolat V, Moncal KK and Ozbolat IT: Essential steps in bioprinting: From pre- to post-bioprinting. Biotechnol Adv. 36:1481–1504. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Wang X, Luo Y, Ma Y, Wang P and Yao R: Converging bioprinting and organoids to better recapitulate the tumor microenvironment. Trends Biotechnol. 42:648–663. 2024. View Article : Google Scholar : PubMed/NCBI

56 

Ding S, Feng L, Wu J, Zhu F, Tan Z and Yao R: Bioprinting of stem cells: Interplay of bioprinting process, bioinks, and stem cell properties. ACS Biomater Sci Eng. 4:3108–3124. 2018. View Article : Google Scholar : PubMed/NCBI

57 

Gaspar VM, Lavrador P, Borges J, Oliveira MB and Mano JF: Advanced bottom-up engineering of living architectures. Adv Mater. 32:e19039752020. View Article : Google Scholar : PubMed/NCBI

58 

Yoon WH, Lee HR, Kim S, Kim E, Ku JH, Shin K and Jung S: Use of inkjet-printed single cells to quantify intratumoral heterogeneity. Biofabrication. 12:0350302020. View Article : Google Scholar : PubMed/NCBI

59 

Mollica PA, Booth-Creech EN, Reid JA, Zamponi M, Sullivan SM, Palmer XL, Sachs PC and Bruno RD: 3D bioprinted mammary organoids and tumoroids in human mammary derived ECM hydrogels. Acta Biomater. 95:201–213. 2019. View Article : Google Scholar : PubMed/NCBI

60 

Reid JA, Palmer XL, Mollica PA, Northam N, Sachs PC and Bruno RD: A 3D bioprinter platform for mechanistic analysis of tumoroids and chimeric mammary organoids. Sci Rep. 9:74662019. View Article : Google Scholar : PubMed/NCBI

61 

Maloney E, Clark C, Sivakumar H, Yoo K, Aleman J, Rajan SAP, Forsythe S, Mazzocchi A, Laxton AW, Tatter SB, et al: Immersion bioprinting of tumor organoids in multi-well plates for increasing chemotherapy screening throughput. Micromachines (Basel). 11:2082020. View Article : Google Scholar : PubMed/NCBI

62 

Mao S, He J, Zhao Y, Liu T, Xie F, Yang H, Mao Y, Pang Y and Sun W: Bioprinting of patient-derived in vitro intrahepatic cholangiocarcinoma tumor model: Establishment, evaluation and anti-cancer drug testing. Biofabrication. 12:0450142020. View Article : Google Scholar : PubMed/NCBI

63 

Bernal PN, Bouwmeester M, Madrid-Wolff J, Falandt M, Florczak S, Rodriguez NG, Li Y, Größbacher G, Samsom RA, van Wolferen M, et al: Volumetric bioprinting of organoids and optically tuned hydrogels to build liver-like metabolic biofactories. Adv Mater. 34:e21100542022. View Article : Google Scholar : PubMed/NCBI

64 

Kleinman HK and Martin GR: Matrigel: Basement membrane matrix with biological activity. Semin Cancer Biol. 15:378–386. 2005. View Article : Google Scholar : PubMed/NCBI

65 

Talbot NC and Caperna TJ: Proteome array identification of bioactive soluble proteins/peptides in Matrigel: Relevance to stem cell responses. Cytotechnology. 67:873–883. 2015. View Article : Google Scholar : PubMed/NCBI

66 

Aisenbrey EA and Murphy WL: Synthetic alternatives to Matrigel. Nat Rev Mater. 5:539–551. 2020. View Article : Google Scholar : PubMed/NCBI

67 

Ng S, Tan WJ, Pek MMX, Tan MH and Kurisawa M: Mechanically and chemically defined hydrogel matrices for patient-derived colorectal tumor organoid culture. Biomaterials. 219:1194002019. View Article : Google Scholar : PubMed/NCBI

68 

Below CR, Kelly J, Brown A, Humphries JD, Hutton C, Xu J, Lee BY, Cintas C, Zhang X, Hernandez-Gordillo V, et al: A microenvironment-inspired synthetic three-dimensional model for pancreatic ductal adenocarcinoma organoids. Nat Mater. 21:110–119. 2022. View Article : Google Scholar : PubMed/NCBI

69 

Kim S, Min S, Choi YS, Jo SH, Jung JH, Han K, Kim J, An S, Ji YW, Kim YG and Cho SW: Tissue extracellular matrix hydrogels as alternatives to Matrigel for culturing gastrointestinal organoids. Nat Commun. 13:16922022. View Article : Google Scholar : PubMed/NCBI

70 

Komor AC, Badran AH and Liu DR: CRISPR-Based technologies for the manipulation of eukaryotic genomes. Cell. 169:5592017. View Article : Google Scholar : PubMed/NCBI

71 

Aguirre AJ, Meyers RM, Weir BA, Vazquez F, Zhang CZ, Ben-David U, Cook A, Ha G, Harrington WF, Doshi MB, et al: Genomic copy number dictates a gene-independent cell response to CRISPR/Cas9 targeting. Cancer Discov. 6:914–929. 2016. View Article : Google Scholar : PubMed/NCBI

72 

Sharma G, Sharma AR, Bhattacharya M, Lee SS and Chakraborty C: CRISPR-Cas9: A preclinical and clinical perspective for the treatment of human diseases. Mol Ther. 29:571–586. 2021. View Article : Google Scholar : PubMed/NCBI

73 

Artegiani B, van Voorthuijsen L, Lindeboom RGH, Seinstra D, Heo I, Tapia P, López-Iglesias C, Postrach D, Dayton T, Oka R, et al: Probing the tumor suppressor function of BAP1 in CRISPR-engineered human liver organoids. Cell Stem Cell. 24:927–943.e6. 2019. View Article : Google Scholar : PubMed/NCBI

74 

Matano M, Date S, Shimokawa M, Takano A, Fujii M, Ohta Y, Watanabe T, Kanai T and Sato T: Modeling colorectal cancer using CRISPR-Cas9-mediated engineering of human intestinal organoids. Nat Med. 21:256–262. 2015. View Article : Google Scholar : PubMed/NCBI

75 

Erlangga Z, Wolff K, Poth T, Peltzer A, Nahnsen S, Spielberg S, Timrott K, Woller N, Kühnel F, Manns MP, et al: Potent antitumor activity of liposomal irinotecan in an organoid- and CRISPR-Cas9-based murine model of gallbladder cancer. Cancers (Basel). 11:19042019. View Article : Google Scholar : PubMed/NCBI

76 

Dekkers JF, Whittle JR, Vaillant F, Chen HR, Dawson C, Liu K, Geurts MH, Herold MJ, Clevers H, Lindeman GJ and Visvader JE: Modeling breast cancer using CRISPR-Cas9-mediated engineering of human breast organoids. J Natl Cancer Inst. 112:540–544. 2020. View Article : Google Scholar : PubMed/NCBI

77 

Zhan T, Rindtorff N, Betge J, Ebert MP and Boutros M: CRISPR/Cas9 for cancer research and therapy. Semin Cancer Biol. 55:106–119. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Vaishnavi A, Juan J, Jacob M, Stehn C, Gardner EE, Scherzer MT, Schuman S, Van Veen JE, Murphy B, Hackett CS, et al: Transposon mutagenesis reveals RBMS3 silencing as a promoter of malignant progression of BRAFV600E-driven lung tumorigenesis. Cancer Res. 82:4261–4273. 2022. View Article : Google Scholar : PubMed/NCBI

79 

Takeda H, Kataoka S, Nakayama M, Ali MAE, Oshima H, Yamamoto D, Park JW, Takegami Y, An T, Jenkins NA, et al: CRISPR-Cas9-mediated gene knockout in intestinal tumor organoids provides functional validation for colorectal cancer driver genes. Proc Natl Acad Sci USA. 116:15635–15644. 2019. View Article : Google Scholar : PubMed/NCBI

80 

Artegiani B, Hendriks D, Beumer J, Kok R, Zheng X, Joore I, de Sousa Lopes SC, van Zon J, Tans S and Clevers H: Fast and efficient generation of knock-in human organoids using homology-independent CRISPR-Cas9 precision genome editing. Nat Cell Biol. 22:321–331. 2020. View Article : Google Scholar : PubMed/NCBI

81 

Hendriks D, Artegiani B, Hu H, de Sousa Lopes SC and Clevers H: Establishment of human fetal hepatocyte organoids and CRISPR-Cas9-based gene knockin and knockout in organoid cultures from human liver. Nat Protoc. 16:182–217. 2021. View Article : Google Scholar : PubMed/NCBI

82 

Lo YH, Kolahi KS, Du Y, Chang CY, Krokhotin A, Nair A, Sobba WD, Karlsson K, Jones SJ, Longacre TA, et al: A CRISPR/Cas9-engineered ARID1A-deficient human gastric cancer organoid model reveals essential and nonessential modes of oncogenic transformation. Cancer Discov. 11:1562–1581. 2021. View Article : Google Scholar : PubMed/NCBI

83 

Yao Q, Cheng S, Pan Q, Yu J, Cao G, Li L and Cao H: Organoids: Development and applications in disease models, drug discovery, precision medicine, and regenerative medicine. MedComm (2020). 5:e7352024. View Article : Google Scholar : PubMed/NCBI

84 

Silvestri VL, Henriet E, Linville RM, Wong AD, Searson PC and Ewald AJ: A tissue-engineered 3D microvessel model reveals the dynamics of mosaic vessel formation in breast cancer. Cancer Res. 80:4288–4301. 2020. View Article : Google Scholar : PubMed/NCBI

85 

Liu X, Taftaf R, Kawaguchi M, Chang YF, Chen W, Entenberg D, Zhang Y, Gerratana L, Huang S, Patel DB, et al: Homophilic CD44 interactions mediate tumor cell aggregation and polyclonal metastasis in patient-derived breast cancer models. Cancer Discov. 9:96–113. 2019. View Article : Google Scholar : PubMed/NCBI

86 

Cheung KJ, Gabrielson E, Werb Z and Ewald AJ: Collective invasion in breast cancer requires a conserved basal epithelial program. Cell. 155:1639–1651. 2013. View Article : Google Scholar : PubMed/NCBI

87 

Boos SL, Loevenich LP, Vosberg S, Engleitner T, Öllinger R, Kumbrink J, Rokavec M, Michl M, Greif PA, Jung A, et al: Disease modeling on tumor organoids implicates AURKA as a therapeutic target in liver metastatic colorectal cancer. Cell Mol Gastroenterol Hepatol. 13:517–540. 2022. View Article : Google Scholar : PubMed/NCBI

88 

Sánchez-Botet A, Quandt E, Masip N, Escribá R, Novellasdemunt L, Gasa L, Li VSW, Raya Á, Clotet J and Ribeiro MPC: Atypical cyclin P regulates cancer cell stemness through activation of the WNT pathway. Cell Oncol (Dordr). 44:1273–1286. 2021. View Article : Google Scholar : PubMed/NCBI

89 

Harada K, Sakamoto N, Ukai S, Yamamoto Y, Pham QT, Taniyama D, Honma R, Maruyama R, Takashima T, Ota H, et al: Establishment of oxaliplatin-resistant gastric cancer organoids: Importance of myoferlin in the acquisition of oxaliplatin resistance. Gastric Cancer. 24:1264–1277. 2021. View Article : Google Scholar : PubMed/NCBI

90 

Mosquera MJ, Kim S, Bareja R, Fang Z, Cai S, Pan H, Asad M, Martin ML, Sigouros M, Rowdo FM, et al: Extracellular matrix in synthetic hydrogel-based prostate cancer organoids regulate therapeutic Response to EZH2 and DRD2 Inhibitors. Adv Mater. 34:e21000962022. View Article : Google Scholar : PubMed/NCBI

91 

Yao Y, Xu X, Yang L, Zhu J, Wan J, Shen L, Xia F, Fu G, Deng Y, Pan M, et al: Patient-Derived organoids predict chemoradiation responses of locally advanced rectal cancer. Cell Stem Cell. 26:17–26.e6. 2020. View Article : Google Scholar : PubMed/NCBI

92 

Vlachogiannis G, Hedayat S, Vatsiou A, Jamin Y, Fernández-Mateos J, Khan K, Lampis A, Eason K, Huntingford I, Burke R, et al: Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 359:920–926. 2018. View Article : Google Scholar : PubMed/NCBI

93 

Ganesh K, Wu C, O'Rourke KP, Szeglin BC, Zheng Y, Sauvé CG, Adileh M, Wasserman I, Marco MR, Kim AS, et al: A rectal cancer organoid platform to study individual responses to chemoradiation. Nat Med. 25:1607–1614. 2019. View Article : Google Scholar : PubMed/NCBI

94 

Tiriac H, Belleau P, Engle DD, Plenker D, Deschênes A, Somerville TDD, Froeling FEM, Burkhart RA, Denroche RE, Jang GH, et al: Organoid profiling identifies common responders to chemotherapy in pancreatic cancer. Cancer Discov. 8:1112–1129. 2018. View Article : Google Scholar : PubMed/NCBI

95 

Ji S, Feng L, Fu Z, Wu G, Wu Y, Lin Y, Lu D, Song Y, Cui P, Yang Z, et al: Pharmaco-proteogenomic characterization of liver cancer organoids for precision oncology. Sci Transl Med. 15:eadg33582023. View Article : Google Scholar : PubMed/NCBI

96 

Jensen LH, Rogatto SR, Lindebjerg J, Havelund B, Abildgaard C, do Canto LM, Vagn-Hansen C, Dam C, Rafaelsen S and Hansen TF: Precision medicine applied to metastatic colorectal cancer using tumor-derived organoids and in-vitro sensitivity testing: A phase 2, single-center, open-label, and non-comparative study. J Exp Clin Cancer Res. 42:1152023. View Article : Google Scholar : PubMed/NCBI

97 

Minoli M, Cantore T, Hanhart D, Kiener M, Fedrizzi T, La Manna F, Karkampouna S, Chouvardas P, Genitsch V, Rodriguez-Calero A, et al: Bladder cancer organoids as a functional system to model different disease stages and therapy response. Nat Commun. 14:22142023. View Article : Google Scholar : PubMed/NCBI

98 

Neal JT, Li X, Zhu J, Giangarra V, Grzeskowiak CL, Ju J, Liu IH, Chiou SH, Salahudeen AA, Smith AR, et al: Organoid Modeling of the Tumor Immune Microenvironment. Cell. 175:1972–1988.e16. 2018. View Article : Google Scholar : PubMed/NCBI

99 

Bozkus CC and Bhardwaj N: Tumor organoid-originated biomarkers predict immune response to PD-1 blockade. Cancer Cell. 39:1187–1189. 2021. View Article : Google Scholar : PubMed/NCBI

100 

Cattaneo CM, Dijkstra KK, Fanchi LF, Kelderman S, Kaing S, van Rooij N, van den Brink S, Schumacher TN and Voest EE: Tumor organoid-T-cell coculture systems. Nat Protoc. 15:15–39. 2020. View Article : Google Scholar : PubMed/NCBI

101 

Meng Q, Xie S, Gray GK, Dezfulian MH, Li W, Huang L, Akshinthala D, Ferrer E, Conahan C, Perea Del Pino S, et al: Empirical identification and validation of tumor-targeting T cell receptors from circulation using autologous pancreatic tumor organoids. J Immunother Cancer. 9:e0032132021. View Article : Google Scholar : PubMed/NCBI

102 

Dijkstra KK, Cattaneo CM, Weeber F, Chalabi M, van de Haar J, Fanchi LF, Slagter M, van der Velden DL, Kaing S, Kelderman S, et al: Generation of tumor-reactive T cells by co-culture of peripheral blood lymphocytes and tumor organoids. Cell. 174:1586–1598.e12. 2018. View Article : Google Scholar : PubMed/NCBI

103 

Votanopoulos KI, Forsythe S, Sivakumar H, Mazzocchi A, Aleman J, Miller L, Levine E, Triozzi P and Skardal A: Model of patient-specific immune-enhanced organoids for immunotherapy screening: Feasibility study. Ann Surg Oncol. 27:1956–1967. 2020. View Article : Google Scholar : PubMed/NCBI

104 

Kryeziu K, Moosavi SH, Bergsland CH, Guren MG, Eide PW, Totland MZ, Lassen K, Abildgaard A, Nesbakken A, Sveen A and Lothe RA: Increased sensitivity to SMAC mimetic LCL161 identified by longitudinal ex vivo pharmacogenomics of recurrent, KRAS mutated rectal cancer liver metastases. J Transl Med. 19:3842021. View Article : Google Scholar : PubMed/NCBI

105 

Choi YJ, Lee H, Kim DS, Kim DH, Kang MH, Cho YH, Choi CM, Yoo J, Lee KO, Choi EK, et al: Discovery of a novel CDK7 inhibitor YPN-005 in small cell lung cancer. Eur J Pharmacol. 907:1742982021. View Article : Google Scholar : PubMed/NCBI

106 

Zhou Z, Van der Jeught K, Fang Y, Yu T, Li Y, Ao Z, Liu S, Zhang L, Yang Y, Eyvani H, et al: An organoid-based screen for epigenetic inhibitors that stimulate antigen presentation and potentiate T-cell-mediated cytotoxicity. Nat Biomed Eng. 5:1320–1335. 2021. View Article : Google Scholar : PubMed/NCBI

107 

Cho K, Ro SW, Lee HW, Moon H, Han S, Kim HR, Ahn SH, Park JY and Kim DY: YAP/TAZ suppress drug penetration into hepatocellular carcinoma through stromal activation. Hepatology. 74:2605–2621. 2021. View Article : Google Scholar : PubMed/NCBI

108 

Grebenyuk S and Ranga A: Engineering organoid vascularization. Front Bioeng Biotechnol. 7:392019. View Article : Google Scholar : PubMed/NCBI

109 

Strobel HA, Moss SM and Hoying JB: Vascularized tissue organoids. Bioengineering (Basel). 10:1242023. View Article : Google Scholar : PubMed/NCBI

110 

Bhat SM, Badiger VA, Vasishta S, Chakraborty J, Prasad S, Ghosh S and Joshi MB: 3D tumor angiogenesis models: Recent advances and challenges. J Cancer Res Clin Oncol. 147:3477–3494. 2021. View Article : Google Scholar : PubMed/NCBI

111 

Wörsdörfer P, Dalda N, Kern A, Krüger S, Wagner N, Kwok CK, Henke E and Ergün S: Generation of complex human organoid models including vascular networks by incorporation of mesodermal progenitor cells. Sci Rep. 9:156632019. View Article : Google Scholar : PubMed/NCBI

112 

Shirure VS, Bi Y, Curtis MB, Lezia A, Goedegebuure MM, Goedegebuure SP, Aft R, Fields RC and George SC: Tumor-on-a-chip platform to investigate progression and drug sensitivity in cell lines and patient-derived organoids. Lab Chip. 18:3687–3702. 2018. View Article : Google Scholar : PubMed/NCBI

113 

Mazio C, Casale C, Imparato G, Urciuolo F and Netti PA: Recapitulating spatiotemporal tumor heterogeneity in vitro through engineered breast cancer microtissues. Acta Biomater. 73:236–249. 2018. View Article : Google Scholar : PubMed/NCBI

114 

Nashimoto Y, Hayashi T, Kunita I, Nakamasu A, Torisawa YS, Nakayama M, Takigawa-Imamura H, Kotera H, Nishiyama K, Miura T and Yokokawa R: Integrating perfusable vascular networks with a three-dimensional tissue in a microfluidic device. Integr Biol (Camb). 9:506–518. 2017. View Article : Google Scholar : PubMed/NCBI

115 

Grönholm M, Feodoroff M, Antignani G, Martins B, Hamdan F and Cerullo V: Patient-Derived organoids for precision cancer immunotherapy. Cancer Res. 81:3149–3155. 2021. View Article : Google Scholar : PubMed/NCBI

116 

Meng Q, Liu Z, Rangelova E, Poiret T, Ambati A, Rane L, Xie S, Verbeke C, Dodoo E, Del Chiaro M, et al: Expansion of tumor-reactive T cells from patients with pancreatic cancer. J Immunother. 39:81–89. 2016. View Article : Google Scholar : PubMed/NCBI

117 

Almeqdadi M, Mana MD, Roper J and Yilmaz ÖH: Gut organoids: Mini-tissues in culture to study intestinal physiology and disease. Am J Physiol Cell Physiol. 317:C405–C419. 2019. View Article : Google Scholar : PubMed/NCBI

118 

Vazquez-Armendariz AI and Tata PR: Recent advances in lung organoid development and applications in disease modeling. J Clin Invest. 133:e1705002023. View Article : Google Scholar : PubMed/NCBI

119 

Shi R, Radulovich N, Ng C, Liu N, Notsuda H, Cabanero M, Martins-Filho SN, Raghavan V, Li Q, Mer AS, et al: Organoid cultures as preclinical models of non-small cell lung cancer. Clin Cancer Res. 26:1162–1174. 2020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2025
Volume 31 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu Y, Zhang F, Du F, Huang J and Wei S: Combination of tumor organoids with advanced technologies: A powerful platform for tumor evolution and treatment response (Review). Mol Med Rep 31: 140, 2025.
APA
Wu, Y., Zhang, F., Du, F., Huang, J., & Wei, S. (2025). Combination of tumor organoids with advanced technologies: A powerful platform for tumor evolution and treatment response (Review). Molecular Medicine Reports, 31, 140. https://doi.org/10.3892/mmr.2025.13505
MLA
Wu, Y., Zhang, F., Du, F., Huang, J., Wei, S."Combination of tumor organoids with advanced technologies: A powerful platform for tumor evolution and treatment response (Review)". Molecular Medicine Reports 31.6 (2025): 140.
Chicago
Wu, Y., Zhang, F., Du, F., Huang, J., Wei, S."Combination of tumor organoids with advanced technologies: A powerful platform for tumor evolution and treatment response (Review)". Molecular Medicine Reports 31, no. 6 (2025): 140. https://doi.org/10.3892/mmr.2025.13505