Cancer research has long relied on traditional models such as two-dimensional (2D) cell cultures and animal models. However, these methods have significant limitations. 2D cultures fail to replicate the complexity of tumors, while animal models are expensive, time-consuming, and often unable to reflect human tumor characteristics accurately.
A breakthrough in this field is the development of three-dimensional (3D) tumoroid models. These in vitro constructs closely mimic the tumor microenvironment (TME) and preserve essential cell-cell and cell-extracellular matrix (ECM) interactions. They offer a promising platform for studying disease progression, drug screening, and personalized medicine.1
Image Credit: Gorodenkoff/Shutterstock.com
What Are 3D Tumoroid Models?
3D tumoroids are laboratory-grown tumor models generated from patient-derived tumor cells or engineered induced pluripotent stem cells. These models retain the histological, genetic, and molecular characteristics of the original tumor, making them a valuable tool for cancer research.
Traditional 2D cell cultures lack the spatial complexity of real tumors. Cells grown in monolayers exhibit altered polarity, shape, and signaling, failing to replicate tumor heterogeneity and the TME.
Oxygen and nutrient gradients are absent, leading to unrealistic proliferation and gene expression patterns.2 Animal models, while providing a more complex environment, do not fully replicate human tumor biology. Species-specific differences impact treatment responses, and ethical concerns and high costs limit their use.3
In contrast, 3D tumoroid models overcome these limitations by preserving tumor architecture, gene expression, and cell signaling pathways, making them more predictive of in vivo tumor behavior.3
3D Cell Models - Cancer
How Are 3D Tumoroid Models Created?
Tumoroids are derived from tumor tissue obtained through surgical resection or biopsies. The tissue is digested into small cell clusters and cultured within a 3D extracellular matrix hydrogel, such as Matrigel or basement membrane extract. Over time, these cells grow and organize into structures that closely resemble the original tumor.4
Several techniques are used to cultivate 3D tumoroids. In the scaffold-based approach, tumor cells adhere to a synthetic or natural polymer, promoting ECM secretion and proliferation. Hydrogels provide a structural framework, allowing cells to organize and function similarly to in vivo conditions.
Advanced bioprinting technology enables the precise layering of cell-laden bioink to create complex tumor structures, while microfluidic devices integrate chemical gradients and mechanical forces to simulate real tumor environments.1
3D Organoid Models in Toxicology
Advantages of 3D Tumoroid Models in Cancer Research
One of the greatest advantages of 3D tumoroids is their ability to mimic tumor biology. Unlike 2D cultures, they develop distinct proliferation zones, including an outer proliferative layer, a quiescent zone, and an inner necrotic core.
This structural complexity reflects real tumor behavior, including chemotherapy resistance. For example, in response to cisplatin treatment, proliferating cells sustain DNA damage, but quiescent cells remain protected.5
Additionally, the gene expression patterns in 3D models closely resemble those found in human tumors, enabling accurate studies of tumor progression and treatment resistance.
3D tumoroids have significantly improved drug testing and discovery. Tumor organoid biobanks have been used to screen a wide range of cancer drugs, helping researchers identify effective therapies. Liver cancer organoids, for example, have demonstrated sensitivity to FDA-approved and novel anticancer drugs. Malignant rhabdoid tumor organoids have also been used to discover promising new treatment options.2
Personalized medicine is another critical application of 3D tumoroid models. Because tumors exhibit genetic mutations that affect drug response, patient-derived tumoroids can be used to screen multiple drugs and determine the most effective treatment. Research on lung cancer organoids has shown that BRCA2-mutated tumors respond better to olaparib, while EGFR-mutated tumors respond better to erlotinib.2,4
The ability to study the tumor microenvironment is another key advantage. Tumoroids allow researchers to investigate tumor-stroma interactions, revealing how different cell types influence cancer progression.
For example, vascularized breast cancer tumoroids have shown that endothelial cells promote tumor proliferation and invasion through Notch signaling.6 Additionally, studies on immune cell infiltration have revealed how tumor histology affects immune response.6
Applications of 3D Tumoroid Models
3D tumoroids are widely used for preclinical drug screening. By combining patient-derived tumoroids with genomic sequencing, researchers can identify specific drug targets. For example, studies on uterine carcinosarcoma and endometrial adenocarcinoma revealed that even though both cancers share PIK3CA and PTEN mutations, their drug response profiles differ.
Buparlisib combined with vorinostat was found to be the optimal treatment for uterine carcinosarcoma, while buparlisib combined with olaparib was more effective for endometrial adenocarcinoma.7
Understanding drug resistance is another key application. Many cancers develop resistance to receptor tyrosine kinase inhibitors (RTKIs), a common issue in head and neck squamous cell carcinoma (HNSCC). 3D tumoroid models have shown that HER3 overexpression contributes to resistance against the RTKI lapatinib, a phenomenon not observed in 2D cultures.8
Similarly, research on colorectal cancer tumoroids has identified TYRO3-mediated drug resistance, providing potential new treatment strategies.8
3D tumoroids have also advanced immuno-oncology research by allowing scientists to study tumor-immune interactions. For example, experiments with colorectal cancer organoids and T cells demonstrated that inhibiting DKK1 enhances T-cell-mediated tumor destruction.
Studies on gastric cancer organoids revealed that HER2 knockdown increases cytotoxic T lymphocyte infiltration, reducing PD-L1 expression and enhancing the immune response.9
Exploring Dementia in 3D: Techniques and Applications
Challenges and Current Limitations
Despite their many advantages, 3D tumoroids face several challenges. Standardized protocols for tumoroid establishment are lacking, leading to variations in results and reproducibility issues.
The time required to culture these models limits their rapid clinical application, and their cultivation requires specialized conditions, making them costly and technically demanding. Additionally, most extracellular matrices used in tumoroid culture are derived from animals, raising safety concerns for clinical applications.2,10
The Future of 3D Tumoroid Models in Cancer Research
Emerging technologies are enhancing the potential of 3D tumoroids. Single-cell RNA sequencing is being used to analyze tumor heterogeneity, while machine learning is helping to interpret large-scale drug response datasets.10,11
Automation is improving high-throughput screening, as demonstrated by recent studies on triple-negative breast cancer tumoroids.12 The development of human-derived ECMs is reducing cross-species interactions, improving clinical relevance.3
Another promising innovation is the use of multiorgan chips, which simulate systemic drug effects on healthy tissue, reducing reliance on animal models.3 Additionally, tumoroid-based studies on epigenetic drug targeting have identified new therapeutic options, such as menin-MLL inhibitors for endometrial cancer.13
Conclusion
3D tumoroid models have revolutionized cancer research by accurately replicating tumor biology, drug resistance mechanisms, and treatment responses.
Their ability to preserve tumor heterogeneity and mimic the tumor microenvironment makes them an invaluable tool for drug screening and personalized medicine.
While challenges remain, ongoing advancements in automation, biomaterials, and artificial intelligence will continue to refine these models, solidifying their role in the future of cancer treatment.
Beyond Animal Testing: The Rise of Ethical Alternatives in Clinical Research
References
- Rodrigues J, Heinrich MA, Teixeira LM, Prakash J. 3D in vitro model (R) evolution: unveiling tumor–stroma interactions. (2021). Trends in cancer, 7(3):249-64. Available at: https://doi.org/10.1016/j.trecan.2020.10.009
- Jiang X, Oyang L, Peng Q, Liu Q, Xu X, Wu N, et al. (2023). Organoids: opportunities and challenges of cancer therapy. Frontiers in Cell and Developmental Biology, 11:1232528. Available at: https://doi.org/10.3389/fcell.2023.1232528
- Kalla J, Pfneissl J, Mair T, Tran L, Egger G. (2024). A systematic review on the culture methods and applications of 3D tumoroids for cancer research and personalized medicine. Cellular Oncology, 28:1-26. Available at: https://doi.org/10.1007/s13402-024-00960-8
- Lv J, Du X, Wang M, Su J, Wei Y, Xu C.(2024). Construction of tumor organoids and their application to cancer research and therapy. Theranostics, 14(3):1101. Available at: https://doi.org/10.7150/thno.91362
- 5 reasons cancer researchers adopt 3d cell culture: a review of recent literature. Merck. [Online] Available at: https://www.sigmaaldrich.com/IN/en/technical-documents/technical-article/cell-culture-and-cell-culture-analysis/3d-cell-culture/5-reasons-cancer-researchers-adopt-3d-cell-culture-white-paper?srsltid=AfmBOooFfWCMjgJ9gE1s7dyws-S2WLe5fMmuPTZCPkv147Gs10dpoG9O (Accessed on 6 Jan 2025).
- Fontana F, Marzagalli M, Sommariva M, Gagliano N, Limonta P. (2021). In vitro 3D cultures to model the tumor microenvironment. Cancers, 13(12):2970. Available at: https://doi-org/10.3390/cancers13122970
- Bleijs M, van de Wetering M, Clevers H, Drost J. (2019). Xenograft and organoid model systems in cancer research. The EMBO journal, 38(15):e101654. Available at: https://doi.org/10.15252/embj.2019101654
- Nikdouz A, Orso F. (2023). Emerging roles of 3D-culture systems in tackling tumor drug resistance. Cancer Drug Resistance, 6(4):788. Available at: https://doi.org/10.20517/cdr.2023.93
- Ma X, Wang Q, Li G, Li H, Xu S, Pang D. (2024). Cancer organoids: A platform in basic and translational research. Genes & diseases, 11(2):614-32. Available at: https://doi.org/10.1016/j.gendis.2023.02.052
- Luo Z, Zhou X, Mandal K, He N, Wennerberg W, Qu M, et al. (2021). Reconstructing the tumor architecture into organoids. Advanced drug delivery reviews, 176:113839. Available at: https://doi.org/10.1016/j.addr.2021.113839
- Bai L, Wu Y, Li G, Zhang W, Zhang H, Su J. (2024). AI-enabled organoids: construction, analysis, and application. Bioactive Materials, 31:525-48. Available at: https://doi.org/10.1016/j.bioactmat.2023.09.005
- Sirenko O, Lim A, Brock CK, Nikolov K, Olsen C, Cromwell EF, et al. (2022). Abstract 180: Automation and high content imaging of 3D triple negative breast cancer patient-derived tumoroids assay for compound screening. Cancer Research, 82(12_Supplement):180–180. Available at: https://aacrjournals.org/cancerres/article/82/12_Supplement/180/702958/Abstract-180-Automation-and-high-content-imaging
- Veninga V, Voest EE. (2021). Tumor organoids: Opportunities and challenges to guide precision medicine. Cancer Cell, 39(9):1190-201. Available at: https://doi.org/10.1016/j.ccell.2021.07.020
Further Reading