SciELO - Scientific Electronic Library Online

 
vol.21 número4Terapia láser de baja frecuencia y mucositis oral en oncología pediátrica: sinopsis de revisiones sistemáticasOncología integradora y atención centrada en el paciente: ¿la cirugía paliativa tiene algún papel en estos dos conceptos? índice de autoresíndice de assuntospesquisa de artigos
Home Pagelista alfabética de periódicos  

Serviços Personalizados

Journal

Artigo

Indicadores

Links relacionados

  • Não possue artigos similaresSimilares em SciELO

Compartilhar


Gaceta mexicana de oncología

versão On-line ISSN 2565-005Xversão impressa ISSN 1665-9201

Gac. mex. oncol. vol.21 no.4 Ciudad de México Out./Dez. 2022  Epub 05-Dez-2022

https://doi.org/10.24875/j.gamo.22000110 

Review articles

New insights about organoids as model of study for breast cancer research

Nuevas perspectivas sobre organoides como modelo de estudio para investigación en cáncer de mama

Ignacio U. Macías-Paz1 

Alejandro Rivera-Arenas1 

Elizabeth Reyna-Beltrán1 

Alejandra Tavera-Tapia*  1 

1Dr. Alberto Romo Caballero´s School of Medicine, Research Division, Universidad Autónoma de Tamaulipas, Tampico, Tamps., Mexico


Abstract

Although the current advances in breast cancer (BC) research, it is still one of the leading causes of death from neoplastic diseases in women and causes millions of new cases worldwide. It has become evident that traditional methods used for BC research have serious limitations. We have reviewed the most recent findings in the application of Breast Cancer Organoids (BCOs) to explore extremely rare BC forms, BC hallmarks, biobanks, and interaction with the microenvironment. Moreover, we explored the importance of its applicability in clinical trials, drug screening, and personalized medicine.

Keywords Organoids; Breast cancer; Clinical trials; Personalized medicine

Resumen

A pesar de los avances actuales en la investigación del cáncer de mama (CM), sigue siendo una de las principales causas de muerte por enfermedades neoplásicas en mujeres y provoca millones de casos nuevos en todo el mundo. Se ha hecho evidente que los métodos tradicionales utilizados para la investigación en CM tienen serias limitaciones. Hemos revisado los hallazgos más recientes en la aplicación de organoides de cáncer de mama (OCM) para explorar formas extremadamente raras de CM, características distintivas de CM, biobancos e interacción con el microambiente. Además, exploramos la importancia de su aplicabilidad en ensayos clínicos, cribado de fármacos y medicina personalizada.

Palabras clave Organoides; Cáncer de mama; Ensayos clínicos; Medicina personalizada

Introduction

BC and traditional models of study

Among females, BC is the most commonly diagnosed cancer and the leading cause of cancer death. More than 2.1 milliaxon new cases are diagnosed every year worldwide, with an estimated incidence of 24.2% and a mortality rate of 15% in females.1 The most used methods of study for BC include 2D monolayer cells, patient-derived xenografts (PDXs), and genetically engineered mouse models (GEMMs). 2D monolayer cells have some advantages as easy management, feasible establishment, and are the most economic model for BC research2. Although, it is a model that does not bear similarity to the original tumor as it becomes immortalized; it does not reflect the interaction with the microenvironment; can acquire mutations that do not originate in the original tumor due to the 2D culture and do not acquire a constitution like an organ, therefore, the cellular hierarchy is lost. On the contrary, PDXs and GEMM models are more complex and suitable to outmatch some problems inherent to cell lines. The PDXs consist of tumor tissue or tumor cells from a patient, which are implanted into an immune compromised or humanized mouse. PDXs are models that can be used to evaluate drugs for treatments, preserve a certain degree of tumor hierarchy, heterogeneity, and functions from the original tissue. Beckhove et al., 2003, developed the first PDX using human primary BC transplants and DeRose et al., 2011, established clinically defined BC subtypes PDX models3,4. In the case of GEMMs, they are useful to study genetic pathways, therapeutic approaches, cancer progression, and metastasis. The first GEMM to model Human Epidermal Growth Factor Receptor 2 (HER2+) BC was obtained by Muller et al., 1988, and a GEMM developed to model BRCA1 BC was obtained by Behbod et al., 1999, to study the involvement of specific genes in oncogenesis5,6. Although these advantages, there are caveats that prevent its translation into clinics. Among them, the complete heterogeneity of the tumor is not preserved, its maintenance is more expensive and technically more difficult, and in the case of GEMMs, the establishment takes longer to perform. Both models are hampered by the interspecies difference, the microenvironment is not fully recapitulated and mouse stroma can interfere with therapeutic response, for instance. They show poor clinical predictability and reproducibility, specific therapies cannot be tested, and high-throughput screening cannot be performed7-9. Hence, there is still a gap in research that requires other models10.

Organoids definition

Organoids are three-dimensional (3D) structures that can be derived from pluripotent stem cells (PSCs), adult cellular tissue (stem or differentiated cells), embryonic progenitors, tissue segments, and whole organ explants11. The concept was first used in 1946 concerning a tumor case study12. Its meaning evolved to commonly refer to tissues or structures that resemble an organ; however, until the development of organoids in 2009, this concept was used specifically for self-organizing in vitro structures13. They are a useful tool to investigate organogenesis, repair, homeostasis, and disease modeling, including single-gene disorders and more complex maladies, such as cancer14,15. The advent of the “organoid era” began with the establishment and development of organoids from the intestine, until today the best characterized system13. Since then, organoids from several other tissues have been established and modified genetically for disease modeling or have been obtained from tumor tissues, leading to the development of the so-called tumor or cancer organoids16.

Strengths as models of study

Breast cancer organoids (BCOs) have become an option for cancer study because they offer advantages and alternatives over the traditional methods. BCO can be established from the tumor tissue in culture, even from minor sources. Other benefits include the lower generation time for a stable organoid model, the efficiency of passage, and overall, the cost is affordable in comparison with animal models17. BCOs also provide a solid and reproducible platform to perform high-precision assays, which is limited and not always reproducible in the other models. This advantage can be applied in drug screening for novel treatments, single-cell profiling for transcriptome and epigenome analysis, and whole-exome and whole-genome sequencing, among other approaches18. Furthermore, BCOs preserve the 3D structure, all the cells and interactions involved in the tumor which are lost in the 2D cultures, together with the tumor heterogeneity including immune components and intercellular interactions between tumorigenic cells, the matrix, and the tumor niche19. Furthermore, because its technology does not depend on an animal, there are no interspecies interferences20. Moreover, BCOs are amenable to translational research including creating self-personalized models of a patient tumor, which can be molecularly profiled and tested for multiple drugs to find better therapeutic and individualized options. They can be preserved in long collections known as biobanks for further investigation. In addition, their application in clinical trials with comparison purposes is increasing, as it seems to be a more accurate model for personalized medicine (Fig. 1).

Figure 1 Applications of organoids in basic and translational Breast Cancer (BC) research. 

Disadvantages

The use of BCO has been largely discussed, as they still have limitations. It has been demonstrated that BCOs resemble the primary tumor at genomic, transcriptomic, and proteomic level. Nonetheless, it is debated until which point these signals remain identical to the tumor of origin. If the culture conditions interfere and, for example, the length and number of passages can also alter the expression levels of the BCO, leading to a loss of intratumor heterogeneity21. They initially provide a platform to understand the niche, but as the culture condition is extended, specific cell types could be enriched. In particular, a report has examined that culture favors the growth of a percentage of benign cells that in the tumor would act as supporters of growth, and in consequence, the natural evolution of the tumor is masked in vitro22. When the BCOs are not “contaminated” by such epithelial supporters cells, are possible to study the effect of particular environmental factors and/or perform cocultures with stromal cells to comprehend these interactions, but as such there are authors that comment that these systems are no longer per se the original tumor23. Other concerns include that each study of BCO has differences in the culture media, time for passaging, and/or strategies for generating the organoids; as consequence, there is a strong need of international standardization in the pipelines for isolation, enrichment, and characterization techniques employed. A collective effort should be made to establish clear guidelines and ways to assess quality and validity in organoid models24. For instance, some authors use or not extracellular matrix components25 and synthetic scaffold designs, among others. For example, a technical caveat was related with the effect of frozening BCO. In a recent study, BCOs from frozen tissues matched viability and drug response from BCO from fresh tissues with an optimized slow freezing technique in dimethyl sulfoxide26, this shows how improvements in the protocols are crucial for BCO standardization. One of the arguments against them is their relative high cost in comparison with 2D monolayer cells. It is correct that this technology is more complex, so therefore, it depends on more technological and human-trained staff for its management. Although, in comparison with the PDXs and GEMMs, they are cheaper27. Nonetheless, it has been seen that due to their advantages and more accessible management, its use has increased both in laboratories and in clinical trials. There is an ongoing collective effort to standardize and/or improve protocols, which could translate into better results that are worth the investment24. In addition, recent findings using transmission electron microscopy studying ultrastructural characteristics of BCO have concluded that these models recapitulate mammary gland morphology and display specific structural features that could be used to classify and identify BC subtypes28. A 3D bioprinting platform was developed to control the 3D formation of mammary organoids, which is adjustable to diverse culturing protocols and potentially to BCO, adding efficiency and scalability29. In addition, new methods with a high success rate are being proposed for the generation of BCO from surgical and biopsy samples30. These denote that as more technology is used for the characterization/generation of BCO, current technological difficulties will be overcome.

Applications of BCO

Organoids from extremely rare BC types

BCOs allow exploration of rare forms of BC that lacked solid models due to its infrequency, which lead to limited therapeutic options. Nowadays, this technology is used to characterize rare forms of BC. Pan et al., 2020, established a BCO from a 65-year-old woman with Paget disease, which resembled the original tumor and was useful for genomic characterization finding novel copy number alterations, mutational signatures, and somatic mutations, proving its value for clinicopathological research31. Another BCO system was described for giant papillary carcinoma, an infrequent form of duct lobular BC. Furthermore, it was applied for drug sensitivity tests that included endocrine and targeted therapies and resulted quite sensitive to fulvestrant, which has important implications for personalized medicine32. Thinking about its potential, it would be interesting the future development of an organoid system from male BC, which still lacks a reliable model of study.

Exploration of BC microenvironment

BCOs offer the possibility to study the microenvironment along with its specific interactions. It has been seen that the cocultures of normal breast organoids with fibrospheres are useful for understanding epithelial-stromal interactions33. BCO cocultures with fibroblasts have been performed, proposing novel tools such as optical coherence tomography, to assess stromal-epithelial interactions to study premalignancy34 and reveal how stromal cells promote cancer invasion through regulation of basal gene expression35. In addition, novel scaffold-free platforms are being used for studying normal breast and BCO to analyze diverse stimuli from the microenvironment, as well as neoplastic progression allowing the analysis of multi-phenotypic and multi-morphologic states36.

Study of BC hallmarks

BCOs are allowing in-depth study of challenging cancer hallmarks such as angiogenesis, invasion, metastasis, inflammation, and deregulation of cellular metabolism, among others. For instance, it has been demonstrated the influence of a stromal cell line that secretes vascular endothelial growth factor (VEGF) in angiogenesis and proliferation of BCO models from MCF-737. Furthermore, a potential relation between vessel formation and metastasis was observed in cocultures of a mouse BCO with a tissue-engineered 3D microvessel model, where tumor organoids integrated into the endothelial cell lining and facilitated intravasation of circulating tumor cells38. It was observed that human cancer cells competently invaded organoids with a microvessel network of human endothelial cells coupled to the mouse circulatory system allowing extravasation of tumor cells39. To study cancer invasion, another interesting approach used organoids derived from non-tumor MCF10A cells cocultured with tumor MDA-MB 231 which allowed monitoring invasion through epithelium and basement membrane40. Using organoid studies, it was detected a subpopulation of HER2+ early cancer cells which can intravasate, activate an epithelial-mesenchymal transition dependent on Wnt and ultimately metastasize41. Regarding inflammation, a very recent publication found a link between it and metastasis in triple-negative breast cancer (TNBC), where the axis A20/TNFAIP3-CDC20-CASP1 was strongly associated with poor prognosis and survival. Besides, TNBCO treated with inflammation and necroptotic inhibitors blocked this axis-mediated metastasis42. Deregulating energetics is of particular interest in the search for specific tumor characteristics amenable to treatment. A relation between cellular energetics and invasion was reported by Zhang et al., 2019, using BCO, where invasive cancer cells rearrange into the leader and follower positions. Leader cells exhibited higher glucose uptake than follower cells and together with other energetic features, point toward metabolic regulation in different tumor cells43. Another study using organoid model demonstrated that tumor recurrence is caused by residual cells that survive therapeutic regimens by acquiring metabolic shifts different from normal and primary tumors including altered lipid metabolism and elevated ROS44. Indeed, more studies about cellular energetics and metabolic reprogramming are needed. Xiao et al., 2022, performed a metabolic study in TNBC and using patient-derived BCO, a potential target: sphingosine-1-phosphate (S1P) was identified for luminal androgen receptor (LAR) BC subtype, proving the potential of these studies for personalized medicine45. Furthermore, it would be interesting to explore emerging cancer hallmarks such as epigenetics, phenotypic plasticity, and the role of senescent cells in BC.

BCO biobanks

A biobank is a collection that gathers and stores biological material and data associated, to perform molecular/genetic studies, to compare among specimens from the same disease or against normal specimens, etc. For the field of oncology, they are a benefit that can be used for drug design/development, treatment response analysis, and personalized medicine46. Its use has become so important that currently, there are “onco-biobanks” derived from different cancers such as gastrointestinal47, colorectal48, and glioblastoma49, among others. Sachs et al., 2018, developed a biobank of BCO, providing a protocol where primary and metastatic BCOs were obtained, recapitulating multiple distinct subtypes of BC and were used to perform drug screening concomitant with results obtained from in vivo models and patients’ response to ER inhibitor tamoxifen50. In a very complete approach, Dekkers et al., 2021, published protocols for the long-term culture and culturing conditions of 45 biobanked samples including BCO from different BC subtypes, as well as the methodology for genetic manipulation and orthotopic organoid transplantation in mice for tumor growth visualization and cancer cell behavior studies51. Another biobank of TNBC was developed by Bhatia et al., 2022, characterizing different cell types, candidate genes, and survival pathways related to BC progression52.

Personalized medicine

BCO in clinical trials

We performed a search about organoids in clinical trials (www.clinicaltrials.gov). Using the word “organoid,” we found 142 studies registered until June 2022. By adding “breast cancer” to our search, we found 22 clinical trials, representing 15.5% of all the trials employing organoids. Many of them were proposed as a platform for personalized medicine, allowing comparison of BCO against PDXs to corroborate results (e.g., NCT02732860 and NCT04703244), evaluation with one (e.g., NCT03544047), or several drugs (e.g., NCT03925233 and NCT03896958) for BC treatment based on organ-like culture. Some were applied in specific forms of BC, such as tumors with positive estrogen receptor (e.g., NCT04727632), negative HER2 (e.g., NCT04450706), positive HER2 (e.g., NCT04281641 and NCT05429684), TNBC (e.g., NCT05134779 and NCT05404321), advanced/metastatic disease (e.g., NCT04655573 and NCT04526587), and including patients with a germline pathogenic variant with a moderate to high lifetime risk of BC (e.g., NCT04531696). Additionally, clinical trials are using BCO to study, predict, prevent, and treat the metastatic recurrence of TNBC (NCT05464082). It was noted that before 2015, there were very few clinical trials including BCO (2/22) and the majority of reported clinical trials are quite recent. Thus, they have updates but no results reported. Only one of them was withdrawn (NCT04281355). Nonetheless, BCO use has increased in the last years and clinical trials now are including them primarily focused on oncology precision. The summarized characteristics of each study are found in Table S1.

Drug screening (DS) and personalized therapy

With the development of new BCO models and techniques as next-generation sequencing (NGS), the future is set toward personalized medicine. In a larger BCO platform obtained in China, Pan et al., 2021, performed DS looking for novel treatment options, evaluating tamoxifen, fulvestrant, paclitaxel, palbociclib, and carboplatin on neoadjuvant BCO with diverse degree of sensitivity to these drugs which demonstrate the value of organoids in DS and individualized treatment53. Chew et al., 2021, analyzed both BCO and PDX models of triple-negative BC (TNBC) identifying aberrantly activated protein kinases, specifically FGFR4 (fibroblast growth factor receptor 4) that could be targeted with tyrosine kinase inhibitors54. Furthermore, in TNBC, Ge et al., 2021, identified a microtubule-associated complex containing tektin4 and histone deacetylase 6 (HDAC6). BCO and PDXs, which have lost tektin4, were sensitive to ACY1216, a HDAC6 inhibitor, proposed as a new therapeutic strategy55. In an important percentage of BC cases, despite the treatment, tumors tend to develop resistance. As well, there is also the undesired possibility of relapse. In both cases, the combination of novel drugs with known chemotherapy regimens is explored. This type of evaluation can be performed in BCO, as done by Whittle et al., 2020, for testing a combination of inhibitors of CDK4/6, BCL2 together with fulvestrant (for estrogen receptor-positive BC). When this triple therapy was assessed in BCO derived from patients, tumor responsiveness augmented significantly56. Shao et al., 2020, employed whole-genome-wide RNA interference screening and a drug pressure model in BCO. The mechanism associated with cisplatin resistance was identified. DS allowed identification of an important number of drugs that were useless to cisplatin-resistant models and that cotreatment with bortezomib overcame such resistance57. Li et al., 2021, worked over HER2-positive BC resistant to anti-HER2 tyrosine kinase inhibitors. Their study included this subtype of BCO and found that by combining inhibition of CDK12 and anti-HER2 drugs sensitize/resensitize tumors to treatment58. Novel methodologies that could be applied to BCO and DS are under development. Such is the case of a method established by Mukundan et al., 2022, employing cytometry assays where calcein AM and propidium iodide staining were used to analyze the dose-dependent effect of drugs in tumor spheroid models59. Another interesting study was published by Walsh et al., 2014, where optical metabolic imaging of BCO was performed to measure antitumor drug responses to select optimal drug combinations60. Up until now, there was a lack of models that could recapitulate characteristics of advanced BC, either metastatic or refractory. Thus, little information could be obtained about patient stratification or prediction of cancer treatment outcomes. Despite this, BCOs are offering options to these patients and are valuable for studying specific subtypes of cancer patients and intrinsic and/or acquired resistance pathways61,62. As an example, organoids derived from advanced BC with malignant pleural effusion were used to DS yielding sensitivity to everolimus and capecitabine, the latter was consistent with the patient’s clinical response63. Even a combination of models as PDX-derived organoids has been used for exploration of metastatic BC with either Food and Drug Administration-approved and experimental DS against recurrent tumors, where treatment was reoriented in the clinic and the patient’s metastases showed remission for 5 months64. Nikulin et al., 2021, developed an organoid model from metastatic BC and tested 3,3’-diindolylmethane, a suppressor of mir-21-5p, overcoming drug resistance by enhancing response to the combination of cyclophosphamide and methotrexate65. Another study applied to refractory BC was performed by Chen et al., 2021, where a platform of patient BCO was developed for testing microtubule-targeting DS. Also, patients who received at least one drug predicted to be sensitive by BCO achieved partial response, stable disease, or long disease-free survival61. In addition, other techniques such as tumor-on-a-chip platform and 3D scaffolds are being applied to BCO to rapidly assess drug sensitivity to tailor drug therapies66,67. In other approach, for prevention, breast organoids derived from BRCA1 mutated tissue detected that inhibition of RANKL signaling with denosumab reduced proliferation, which is a form of preventive medicine targeted specifically to BRCA1 mutation carriers68.

Conclusions

Naturally, organoids for the study of BC have hurdles, although we have presented that the benefits outweigh the limitations of other traditional models. Therefore, we consider that BCOs are of vital importance to understand advanced processes of oncogenesis, the interaction with the microenvironment, the elucidation of survival pathways used in the neoplastic transformation, and metastasis. Furthermore, its multiple applications include drug discovery and screening, exploration of novel treatment strategies, the establishment of biobanks, and improvement of personalized medicine, which demonstrate that this model of study is important to find the missing pieces of BC research.

Supplementary data

Supplementary data are available at Mexican Journal of Oncology online (doi: 10.24875/j.gamo.22000110). These data are provided by the corresponding author and published online for the benefit of the reader. The contents of supplementary data are the sole responsibility of the authors.

Supplementary material, Figure S1

REFERENCES

1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018:GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394-424. [ Links ]

2. Holliday DL, Speirs V. Choosing the right cell line for breast cancer research. Breast Cancer Res. 2011;13:215. [ Links ]

3. Beckhove P, Schütz F, Diel IJ, Solomayer EF, Bastert G, Foerster J, et al. Efficient engraftment of human primary breast cancer transplants in nonconditioned NOD/Scid mice. Int J Cancer. 2003;105:444-53. [ Links ]

4. DeRose YS, Wang G, Lin YC, Bernard PS, Buys SS, Ebbert MT, et al. Tumor grafts derived from women with breast cancer authentically reflect tumor pathology, growth, metastasis and disease outcomes. Nat Med. 2011;17:1514-20. [ Links ]

5. Muller WJ, Sinn E, Pattengale PK, Wallace R, Leder P. Single-step induction of mammary adenocarcinoma in transgenic mice bearing the activated c-neu oncogene. Cell. 1988;54:105-15. [ Links ]

6. Behbod F, Kittrell FS, LaMarca H, Edwards D, Kerbawy S, Heestand JC, et al. An intraductal human-in-mouse transplantation model mimics the subtypes of ductal carcinoma in situ. Breast Cancer Res. 2009;11:R66. [ Links ]

7. Invrea F, Rovito R, Torchiaro E, Petti C, Isella C, Medico E. Patient-derived xenografts (PDXs) as model systems for human cancer. Curr Opin Biotechnol. 2020;63:151-6. [ Links ]

8. Park MK, Lee CH, Lee H. Mouse models of breast cancer in preclinical research. Lab Anim Res. 2018;34:160-5. [ Links ]

9. Holen I, Speirs V, Morrissey B, Blyth K. In vivo models in breast cancer research:progress, challenges and future directions. Dis Model Mech. 2017;10:359-71. [ Links ]

10. CorròC, Novellasdemunt L, Li VS. A brief history of organoids. Am J Physiol Cell Physiol. 2020;319:C151-65. [ Links ]

11. Kaushik G, Ponnusamy MP, Batra SK. Concise review:current status of three-dimensional organoids as preclinical models. Stem Cells. 2018;36:1329-40. [ Links ]

12. Smith E, Cochcrane WJ. Cystic organoid teratoma:(report of a case). Can Med Assoc J. 1946;55:151-2. [ Links ]

13. Sato T, Vries RG, Snippert HJ, Van de Wetering M, Barker N, Stange DE, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459:262-5. [ Links ]

14. Sasai Y. Cytosystems dynamics in self-organization of tissue architecture. Nature. 2013;493:318-26. [ Links ]

15. Huch M, Koo BK. Modeling mouse and human development using organoid cultures. Development. 2015;142:3113-25. [ Links ]

16. Drost J, Clevers H. Organoids in cancer research. Nat Rev Cancer. 2018;18:407-18. [ Links ]

17. Tuveson D, Clevers H. Cancer modeling meets human organoid technology. Science. 2019;364:952-5. [ Links ]

18. Yu J, Huang W. The Progress and clinical application of breast cancer organoids. Int J Stem Cells. 2020;13:295-304. [ Links ]

19. Xu R, Zhou X, Wang S, Trinkle C. Tumor organoid models in precision medicine and investigating cancer-stromal interactions. Pharmacol Ther. 2021;218:107668. [ Links ]

20. Smith RC, Tabar V. Constructing and deconstructing cancers using human pluripotent stem cells and organoids. Cell Stem Cell. 2019;24:12-24. [ Links ]

21. Li X, Pan B, Song X, Li N, Zhao D, Li M, et al. Breast cancer organoids from a patient with giant papillary carcinoma as a high-fidelity model. Cancer Cell Int. 2020;18;20:86. [ Links ]

22. Goldhammer N, Kim J, Timmermans-Wielenga V, Petersen OW. Characterization of organoid cultured human breast cancer. Breast Cancer Res. 2019;21:141. [ Links ]

23. Lai Y, Wei X, Lin S, Qin L, Cheng L, Li P. Current status and perspectives of patient-derived xenograft models in cancer research. J Hematol Oncol. 2017;10:106. [ Links ]

24. Kim J, Koo BK, Knoblich JA. Human organoids:systems for human biology and medicine. Nat Rev Mol Cell Biol. 2020;21:571-84. [ Links ]

25. Florian S, Iwamoto Y, Coughlin M, Weissleder R, Mitchison TJ. A human organoid system that self-organizes to recapitulate growth and differentiation of a benign mammary tumor. Proc Natl Acad Sci U S A. 2019;116:11444-53. [ Links ]

26. Walsh AJ, Cook RS, Sanders ME, Arteaga CL, Skala MC. Drug response in organoids generated from frozen primary tumor tissues. Sci Rep. 2016;6:18889. [ Links ]

27. Cantrell MA, Kuo CJ. Organoid modeling for cancer precision medicine. Genome Med. 2015;7:32. [ Links ]

28. Signati L, Allevi R, Piccotti F, Albasini S, Villani L, Sevieri M, et al. Ultrastructural analysis of breast cancer patient-derived organoids. Cancer Cell Int. 2021;21:423. [ Links ]

29. Reid JA, Mollica PA, Bruno RD, Sachs PC. Consistent and reproducible cultures of large-scale 3D mammary structures using an accessible bioprinting platform. Breast Cancer Res. 2018;20:122. [ Links ]

30. Mazzucchelli S, Piccotti F, Alevi R, Truffi M, Sorrentino L, Russo L, et al. Establishment and morphological characterization of patient-derived organoids from breast cancer. Biol Proced Online. 2019;21:12. [ Links ]

31. Pan B, Zhao D, Liu Y, Li N, Song C, Li N, et al. Establishment and characterization of breast cancer organoids from a patient with mammary Paget's disease. Cancer Cell Int. 2020;20:365. [ Links ]

32. Li Y, Tang P, Cai S, Peng J, Hua G. Organoid based personalized medicine:from bench to bedside. Cell Regen. 2020;9:21. [ Links ]

33. Koledova Z. 3D Coculture of mammary organoids with fibrospheres:a model for studying epithelial-stromal interactions during mammary branching morphogenesis. Methods Mol Biol. 2017;1612:107-24. [ Links ]

34. Chhetri RV, Phillips ZF, Troester MA, Oldenburg AL. Longitudinal study of mammary epithelial and fibroblast co-cultures using optical coherence tomography reveals morphological hallmarks of pre-malignancy. PLoS One. 2012;7:e49148. [ Links ]

35. Hanley CJ, Henriet E, Sirka OK, Thomas GJ, Ewald AJ. Tumor-resident stromal cells promote breast cancer invasion through regulation of the basal phenotype. Mol Cancer Res. 2020;18:1615-22. [ Links ]

36. Djomehri SI, Burman B, Gonzalez ME, Takayama S, Kleer CG. A reproducible scaffold-free 3D organoid model to study neoplastic progression in breast cancer. J Cell Commun Signal. 2019;13:129-43. [ Links ]

37. Pinto MP, Badtke MM, Dudevoir ML, Harrell JC, Jacobsen BM, Horwitz KB. Vascular endothelial growth factor secreted by activated stroma enhances angiogenesis and hormone-independent growth of estrogen receptor-positive breast cancer. Cancer Res. 2010;70:2655-64. [ Links ]

38. Silvestri VL, Henriet E, Linville RM, Wong AD, Searson PC, Ewald AJ. A tissue-engineered 3D microvessel model reveals the dynamics of mosaic vessel formation in breast cancer. Cancer Res. 2020;80:4288-301. [ Links ]

39. Fernández-Periáñez R, Molina-Privado I, Rojo F, Guijarro-Muñoz I, Alonso-Camino V, Zazo S, et al. Basement membrane-rich organoids with functional human blood vessels are permissive niches for human breast cancer metastasis. PLoS One. 2013;8:e72957. [ Links ]

40. Parigoris E, Lee S, Mertz D, Turner M, Liu AY, Sentosa J, et al. Cancer cell invasion of mammary organoids with basal-in phenotype. Adv Health Mater. 2021;10:e2000810. [ Links ]

41. Harper KL, Sosa MS, Entenberg D, Hosseini H, Cheung JF, Nobre R, et al. Mechanism of early dissemination and metastasis in Her2+ mammary cancer. Mature. 2016;540:588-92. [ Links ]

42. Song C, Kendi AT, Lowe VJ, Lee S. The A20/TNFAIP3-CDC20-CASP1 axis promotes inflammation-mediated metastatic disease in triple-negative breast cancer. Anticancer Res. 2022;42:681-95. [ Links ]

43. Zhang J, Goliwas KF, Wang W, Raufalele PV, Bordeleau F, Reinhart-King CA. Energetic regulation of coordinated leader-follower dynamics during collective invasion of breast cancer cells. Proc Natl Acad Sci U S A. 2019;116:7867-72. [ Links ]

44. Havas KM, Milchevskaya V, Radic K, Alladin A, Kafkia E, Garcia M, et al. Metabolic shifts in residual breast cancer drive tumor recurrence. J Clin Invest. 2017;127:2091-105. [ Links ]

45. Xiao Y, Ma D, Yang YS, Yang F Ding JH, Gong Y, et al. Comprehensive metabolomics expands precision medicine for triple-negative breast cancer. Cell Res. 2022;32:477-90. [ Links ]

46. Schutgens F, Clevers H. Human organoids:tools for understanding biology and treating diseases. Annu Rev Pathol. 2020;15:211-34. [ Links ]

47. 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;359:920-6. [ Links ]

48. 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;161:933-45. [ Links ]

49. Jacob F, Salinas RD, Zhang DY, Nguyen PT, Schnoll JG, Wong SZ, et al. A patient-derived glioblastoma organoid model and biobank recapitulates inter-and intra-tumoral heterogeneity. Cell. 2020;180:188-204.e22. [ Links ]

50. Sachs N, De Ligt J, Kopper O, Gogola E, Bounova G, Weeber F, et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell. 2018;172:373-86.e10. [ Links ]

51. Dekkers JF, Van Vliet EJ, Sachs N, Rosenbluth JM, Kopper O, Rebel HG, et al. Long-term culture, genetic manipulation and xenotransplantation of human normal and breast cancer organoids. Nat Protoc. 2021;16:1936-65. [ Links ]

52. Bhatia S, Kramer M, Russo S, Naik P, Arun G, Brophy K, et al. Patient-derived triple-negative breast cancer organoids provide robust model systems that recapitulate tumor intrinsic characteristics. Cancer Res. 2022;82:1174-92. [ Links ]

53. Pan B, Li X, Zhao D, Li N, Wang K, Li M, et al. Optimizing individualized treatment strategy based on breast cancer organoid model. Clin Transl Med. 2021;11:e380. [ Links ]

54. Chew NJ, Sian TC, Nguyen EV, Shin SY, Yang J, Hui MN, et al. Evaluation of FGFR targeting in BC through interrogation of patient-derived models. Breast Cancer Res. 2021;23:82. [ Links ]

55. Ge LP, Jin X, Yanh YS, Liu XY, Shao ZM, Di GH, et al. Tektin4 loss promotes triple-negative breast cancer metastasis through HDAC6-mediated tubulin deacetylation and increases sensitivity to HDAC6 inhibitor. Oncogene. 2021;40:2323-34. [ Links ]

56. Whittle JR, Vaillant F, Surgenor E, Policheni AN, Giner G, Capaldo BD, et al. Dual targeting of CDK4/6 and BCL2 pathways augments tumor response in estrogen receptor-positive breast cancer. Clin Cancer Res. 2020;26:4120-34. [ Links ]

57. Shao F, Lyu X, Miao K, Xie L, Wang H, Xiao H, et al. Enhanced protein damage clearance induces broad drug resistance in multitype of cancers revealed by an evolution drug-resistant model and genome-wide siRNA screening. Adv Sci (Weinh). 2020;7:2001914. [ Links ]

58. Li H, Wang J, Yi Z, Li C, Wang H, Zhang J, et al. CDK12 inhibition enhances sensitivity of HER2+breast cancers to HER2-tyrosine kinase inhibitor via suppressing PI3K/AKT. Eur J Cancer. 2021;145:92-108. [ Links ]

59. Mukundan S, Bell J, Teryek M, Hernandez C, Love AC, Parekkadan B, et al. Automated assessment of cancer drug efficacy on breast tumor spheroids in aggrewell™400 plates using image cytometry. J Fluoresc. 2022;32:521-31. [ Links ]

60. Walsh AJ, Cook RS, Sanders ME, Aurisicchio L, Ciliberto G, Arteaga CL, et al. Quantitative optical imaging of primary tumor organoid metabolism predicts drug response in breast cancer. Cancer Res. 2014;74:5184-94. [ Links ]

61. 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:e2101176. [ Links ]

62. Vivarelli S, Candido S, Caruso G, Falzone L, Libra M. Patient-derived tumor organoids for drug repositioning in cancer care:a promising approach in the era of tailored treatment. Cancers (Basel). 2020;12:3636. [ Links ]

63. Pan B, Zhao D, Liu Y, Li N, Song C, Li N, et al. Breast cancer organoids from malignant pleural effusion-derived tumor cells as an individualized medicine platform. In Vitro Cell Dev Biol Anim. 2021;57:510-8. [ Links ]

64. 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;3:232-50. [ Links ]

65. Nikulin SV, Alekseev BY, Sergeeva NS, Karalkin PA, Nezhurina EK, Kirsanova VA, et al. Breast cancer organoid model allowed to reveal potentially beneficial combinations of 3, 3'-diindolylmethane and chemotherapy drugs. Biochimie. 2020;179:217-27. [ Links ]

66. Shirure VS, Bi Y, Curtis MB, Lezia A, Goedegebuure MM, Goedegebuure SP, et al. Tumor-on-a-chip platform to investigate progression and drug sensitivity in cell lines and patient-derived organoids. Lab Chip. 2018;18:3687-702. [ Links ]

67. Nayak B, Balancher GM, Manjunath S, Rangarajan A, Chatterjee K. Tissue mimetic 3D scaffold for breast tumor-derived organoid culture toward personalized chemotherapy. Colloids Surf B Biointerfaces. 2019;180:334-43. [ Links ]

68. Nolan E, Vaillant F, Branstetter D, Pal B, Giner G, Whitehead L, et al. RANK ligand as a potential target for breast cancer prevention in BRCA1-mutation carriers. Nat Med. 2016;22:933-9. [ Links ]

Supplementary Material

Supplementary Table 1 Current clinical trials considering BC organoids models 

Number of clinical trial Status Title/aim* Interventions and organoid application Study type Enrollment Country Study start-study completion Main finding
NCT03544047 Unknown Drug sensitivity verification or prediction of therapy for BC by patient derived organoid model To evaluate the consistency and accuracy of patient-derived organoid model of BC to predict the clinical efficacy of the drug paclitaxel Interventional 50 F China January 2019-July 2020 NR
NCT02732860 Recruiting Personalized patient-derived xenograft (pPDX) modeling to test drug response in matching host (REFLECT) Molecular profiling and in vivo drug testing in pPDX and organoid cultures. Study includes triple-negative BC and other types of cancer as ovarian and colorectal Observational 120 F/M Canada December 2015-November 2023 NR
NCT04450706 Recruiting Precision oncology for metastatic BC Clinically actionable outcomes identified by the use of organoids and drug screening in HER2 negative patients Interventional 15 F/M USA February 2021-August 2025 NR
NCT03925233 Unknown BC treatment based on organ-like culture Establish a drug sensitivity test method by evaluating several drugs: trastuzumab hydrochloride, epirubicin hydrochloride, fluorouracil, paclitaxel, gemcitabine, cisplatin, recombinant human endostatin, pirarubicin, pyrrolidine, and other in BCO Observational 300 F China January 2019-December 2021 NR
NCT04727632 Recruiting Concordance between positive fluoroestradiol-PET/CT scan and response to hormonal therapies from drug profiling results using patient-derived organoid models [18 F]Fluoroestradiol (FES) PET/CT in estrogen receptor positive BC patients Interventional 6 F/M USA March 2021-April 2025 NR
NCT04703244 Recruiting Generate patient-derived xenografts (PDX) and organoids from BC patients with residual disease after neoadjuvant therapy Chemotherapy or endocrine therapy for BC Observational 100 F/M USA January 2021-January 2042 NR
NCT04504747 Not yet recruiting Real-time molecular analysis of BC receiving neoadjuvant chemotherapy (NAC) Establish patient derived organoids exposed to NAC Observational 150 F France January 2021-January 2030 NR
NCT03896958 Recruiting Enable best in class functional precision testing of a patient’s tumor tissue to help guide optimal therapy (PIONEER initiative) Organoid drug screening approaches in addition to traditional genomic profiling. This study includes several types of cancer including BC Observational 1000 F/M USA March 2019-March 2024 NR
NCT04281355 Withdrawn Individualized locoregional treatment of initially biopsy proven node-positive BC after primary systemic therapy Drug tests in whole-tumor organoid cultures, algorithm-based digital image analysis, and gene expression analysis to improve response prediction, facilitate tailoring of pre-operative systematic therapy, and increase eradication rates. Interventional 0 Sweden January 2021-December 2029 NR
NCT01287468 Unknown Investigate the tumor suppressive effect of anti-inflammatory phytomedicines on regulation of stromal immune cells and fibroblasts in BC When potent and specific anti-tumor effects are detected, the investigators will extend the study to a three-dimensional collagen/Matrigel culture system for ex vivo study focusing on the stromal cell-mediated or -associated anti-tumor effects using organoid: tissue culture systems Observational 415 F Taiwan June 2010-December 2014 NR
NCT04531696 Recruiting Tissue donation program for metastatic BC patients or patients with a germline pathogenic variants (Leuven Program for Postmortem Tissue Donation to Enhance Research) BC and hereditary diseases; unravel metastatic BC evolution, biology, heterogeneity, and treatment resistance including sub-studies with patient-derived organoids Interventional 100 F/M Belgium November 2020-September 2035 NR
NCT04281641 Recruiting Explore markers in early prediction of the efficacy of pre-operative pertuzumab plus trastuzumab (PH) combined with chemotherapy for early stage or locally advanced human epidermal growth factor receptor-2 (HER-2)-positive primary BC HER2-positive BC, organoids will be used for drug sensitivity assay Interventional 94 F/M China April 2020-April 2030 NR
NCT05177432 Recruiting Quadratic phenotypic optimization platform (QPOP) To investigate the feasibility of QPOP as a clinical decision support platform to identify Interventional 26 F Singapore December 2021-December 2025 NR
NCT05007379 Not yet recruiting Cohort study to determine the antitumor activity of new CAR macrophages in BC patients’ derived organoids To collect tumor samples to develop patients’ derived organoids to test the antitumor activity of newly developed CAR macrophages. Observational 100 F/M France September 2021-September 2023 NR
NCT04655573 Recruiting Assess the feasibility of generating patient-derived micro-organospheres from patients with advanced BC to determine sensitivity to the most common forms of chemotherapy Advanced BC (ER+, HER2-, ER+/HER2+, ER-/HER2+), derive micro-organospheres from biopsies from patients. After biopsy, treatment will be personalized based in the micro-organospheres Observational 15 F/M USA April 2022-October 2024 NR
NCT05134779 Recruiting Deconvoluting interactions between genes, the cancer environment, and the immune system to develop therapies Designed to build a live tissue biobank of patient-derived tumor organoids (PDOs) derived from tumor at surgery, preceded or not by neoadjuvant therapy (NAT), and at recurrence/metastasis in specifically triple negative BC patients Observational 400 F USA January 2022-December 2028 NR
NCT05404321 Not yet recruiting Establishment of an ex vivo tumor collection of triple-negative BC to validate the interest of innovative therapies and the search for predictive biomarkers of response to treatment Establishment of ex vivo BC organoid models (TNBC) Observational 163 F France June 2022-December 2026 NR
NCT05317221 Not yet recruiting Developing BC organoids To develop a living biobank from prospective patient-derived BC tissue to explore genetic profile of the patients with the organoids and the patient biopsy. Cultivate BCOs to predict the treatment response to existing and novel combination treatments Observational 60 F Netherlands May 2022-May 2028 NR
NCT05464082 Not yet recruiting Functional precision oncology to predict, prevent, and treat To develop patient derived models (PDMs), comprising patient-derived xenografts (PDXs) and organoids (PDO) and perform genomic Interventional 80 F/M USA September 2022-September 2027 NR
NCT05429684 Recruiting Precise therapy for refractory HER2-positive advanced BC HER2-positive advanced BC. Evaluate several drugs as trastuzumab, pertuzumab, nab paclitaxel, pyrotinib, capecitabine, everolimus, CDK4/6 inhibitor, and anti-PD-1 monoclonal Interventional 120 F China January 2021-February 2024 NR
NCT05381038 Not yet recruiting Optimizing and personalizing azacitidine combination therapy for treating solid tumors QPOP and CURATE (artificial intelligence) Includes BC and gastrointestinal cancer. Set the foundation to investigate the applicability of QPOP drug selection followed by CURATE AI-guided dose optimization of the selected azacitidine combination therapy. In the QPOP drug selection stage, participants will undergo a baseline biopsy for organoid generation. Interventional 10 F/M Singapore June 2022-April 2027 NR
NCT04526587 Recruiting Biomarkers and clinical features of metastatic BC in patients treated with CDK4/6 inhibitors Stage IV BC, metastatic carcinoma, organoid, and patient-derived xenograft (PDX) models will be used to functionally assess the mechanisms occurring with resistance Observational 300 F/M USA July 2020-July 2025 NR

F: female; M: male; BC: breast cancer; BCO: breast cancer organoid.

*Summarized information has been included; NR: not reported. Information was retrieved from www.clinicaltrials.gov with information available until June 2022. Readers are encouraged to visit the webpage, as data are continually updated with novel advances.

FundingThis research has not received any specific grant from agencies in the public, commercial, or for-profit sectors.

Ethical disclosures

Protection of human and animal subjects. The authors declare that no experiments were performed on humans or animals for this study.

Confidentiality of data. The authors declare that no patient data appear in this article.

Right to privacy and informed consent. The authors declare that no patient data appear in this article.

Received: August 18, 2022; Accepted: September 21, 2022

* Correspondence: Alejandra Tavera-Tapia E-mail: ataveratapia@gmail.com

Conflicts of interest

The authors declare no conflicts of interest.

Creative Commons License Sociedad Mexicana de Oncología. Published by Permanyer. This is an open ccess article under the CC BY-NC-ND license