
Prospects and challenges of ovarian cancer organoids in chemotherapy research (Review)
- Authors:
- Published online on: February 24, 2025 https://doi.org/10.3892/ol.2025.14944
- Article Number: 198
-
Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Ovarian cancer (OC) is a malignant tumor that poses a significant threat to women's health. Globally, OC is the third most prevalent gynecological malignancy, yet it is associated with the highest mortality rate among these types of cancer (1). In 2022, it was estimated that 324,398 new cases of OC would be diagnosed and 206,839 related deaths would occur worldwide (2). OC is often diagnosed in its advanced stages, with current screening techniques unable to facilitate an earlier diagnosis (3); thus, the overall 5-year relative survival rate for OC is 50% (4). Delayed clinical presentation and a late diagnosis also contribute to the high mortality rate associated with this disease, mainly due to a significant lack of sensitive and specific biomarkers for early-stage disease (5).
Currently, the standard chemotherapeutic regimen for OC commonly employs a combination of taxane-based (such as paclitaxel) and platinum-based drugs (6). Cisplatin was the first platinum compound used in the primary treatment of OC; however, it is associated with dose-limiting toxicity, including minor symptoms such as nausea, severe kidney impairment and peripheral neuropathy. To overcome the toxicity associated with cisplatin use, organic platinum analogs of cisplatin, such as carboplatin, have been developed and used instead of cisplatin in initial OC chemotherapy (7). In patients with advanced disease, carboplatin plus paclitaxel chemotherapy is considered an optimal choice (8). Over time, chemotherapy for OC has resulted in progressive enhancements in survival, and the use of platinum/paclitaxel combinations, particularly in combination with intraperitoneal delivery, has the potential to double or increase survival time (9). In addition, heat-intraperitoneal perfusion chemotherapy may improve the disease-free survival of patients with OC, especially when the residual tumor is ≤1 cm or invisible, but it may only improve overall survival (OS) in patients with recurrent disease with a residual tumor of ≤1 cm (10).
Several challenges remain in the treatment of OC, despite advances in chemotherapy. For example, all forms of maintenance therapy have thus far failed to prolong survival. Intraperitoneal therapy has been shown to improve survival in small-volume stage III disease but is associated with significant toxicity (11). Furthermore, a substantial proportion of patients diagnosed with advanced OC are at an increased risk of recurrence (12) and drug resistance (13), which presents a significant challenge for clinicians in terms of developing effective treatment strategies. It is estimated that 60–80% of patients with epithelial ovarian carcinoma (EOC) will achieve complete remission following a combination of surgical and postoperative chemotherapy treatments; however, ~80% of these patients face the risk of chemotherapeutic resistance and potential recurrence (14).
With advancements in technology and research, organoid models have been shown to demonstrate notable improvements over traditional models in multiple aspects, primarily benefiting from their ability to more accurately simulate the complexity and dynamics of biological systems. Traditional two-dimensional (2D) culture models often overlook the three-dimensional (3D) structure of cells and the interactions between cells (15), and xenograft models have drawbacks such as immune rejection and species differences (16). Although patient-derived xenograft (PDX) models retain the tumor characteristics of patients (17,18), they are costly and technically complex to establish and maintain (19). By contrast, organoids, which are 3D dynamic tumor models that can simulate tumor environments in vitro, can mimic the disease development process (20). Organoids can overcome the limitations of traditional models, and thus provide new perspectives for OC research and treatment. Thus, there is an urgent need to develop new in vitro cultivation techniques that can mimic the development of the disease and produce organoids, and can be successfully grown from ovarian tumors, ascites or pleural fluid (21). As a novel approach to cell culture, organoid technology offers significant potential for developing individualized treatments and screening anticancer drugs. This is due to the high degree of tumor replication, the preservation of tumor heterogeneity, the short growth cycle and the stable transmission observed in organoids, which collectively address the limitations of existing preclinical models, including cell lines and animal models (22). The efficacy and resistance of chemotherapeutic drugs can be more accurately predicted through organoid culture, providing a basis for individualized treatment; therefore, organoids may be considered a convenient and efficient platform for studying drug resistance mechanisms (23).
The objective of the present review was to present a thorough overview of the definition, formation and construction of OC organoids. Additionally, the current review considers the advancement of OC organoids in chemotherapy, and their applications, deficiencies and prospects.
Overview of OC organoids
Organoids represent a 3D in vitro cell culture system (23), which are derived from cellular or organ progenitors and, through in vitro differentiation, develop functional cell clusters that display the essential properties of their organ of origin (24). Furthermore, they can preserve the heterogeneity of the original tumor and mimic the tumor growth microenvironment (23); they are thus considered a powerful model for simulating human disease.
In a 2D cell model, cells are grown as a single cell layer in a petri dish. Notably, 2D cell culture models cannot accurately represent the heterogeneity of tumors since they lack the inherent complexity and the TME of the original tumor (25). In addition, researchers have transplanted patient-derived subcutaneous or in situ tumor cells into immunodeficient mice to simulate the heterogeneity of the original tumors; however, this PDX model is expensive, time-consuming, and has low transplantation rates (26,27) and ethical drawbacks (28), Patient-derived organoids (PDOs) have emerged as a pivotal preclinical model system in the field of cancer research and clinical studies (29,30). Organoids retain a high degree of genomic similarity to their corresponding tumors; moreover, they can exhibit the inheritance of certain diseases and the ability to epigenetically alter their phenotype, which may facilitate the development of specific interventions to promote personalized medicine (31,32). The primary advantages of in vitro organoids over other in vitro tumor models are: i) They retain a high degree of heterogeneity among tumor cells, ii) they can be cultivated in small samples, and iii) they maintain the ability for long-term expansion, cryopreservation and genetic modification (24). The advantages and disadvantages of in vitro models are shown in Table I.
In order to generate organoids, tumor cells isolated from primary tumor material, such as surgically excised tissues, biopsies, ascites, pleural fluid or circulating tumor cells, need to be capsulated in a 3D matrix and cultured in medium containing variable growth factors and hormones (33) (Fig. 1). The composition and concentration of certain substances in the medium vary depending on the specific experiment or study; however, advanced Dulbecco's Modified Eagle Medium (DMEM)/Nutrient Mixture F-12 (F-12) is widely used (34,35). DMEM and F-12 are often used in combination to obtain higher concentrations of components of DMEM and a wider variety of components in the F-12 nutrient mix. DMEM/F-12, however, is free of growth factors, hormones and lipids; therefore, to achieve optimal cell proliferation when using DMEM/F-12, it is necessary to add an appropriate combination of growth factors, hormones, proteins or peptides, depending on the specific research needs and cell type. The mixture of growth factors utilized for OC organoid propagation is not standardized and may vary between studies. For OC organoids, it has been suggested that R-spondin 1 (an activator of the Wnt pathway), Noggin (an inhibitor of BMP-dependent differentiation) and 17β-estradiol are necessary for growth, while neuromodulin-1 (NRG1) may be needed for organoid expansion (36).
The development of OC organoids from multiple stages and subtypes (56 organoid lineages) has previously been successfully achieved (37). Organoid cultivation techniques can be applied to a wide range of patient samples, including tissues, biopsies, ascites and pleural effusions (38–40). Most OC organoids generated thus far have been high-grade serous ovarian carcinoma (HGSOC), but there are also other types of cancer, such as low-grade serous OC, mucinous OC, ovarian clear cell carcinoma (OCCC) and ovarian endometrioid carcinoma (ENOC) (41). Furthermore, the proliferation of primary ovarian organoids from different histological subtypes, including HGSOC, OCCC and ENOC, has been reported to have a success rate of 80% and to retain the original mutation pattern (42). Moreover, OC organoids are suitable for constructing biobanks to further improve organoid construction efficiency. For example, a study by Hoffmann et al (43) successfully generated 15 organoids (with an efficiency of ~30%) from 13–45 patients under conditions of low Wnt expression for long-term passaging culture. The OC organoids were cryopreserved for >5 months and passaged every 10–20 days. This previous study showed that there were no significant changes in the morphology or proliferation capacity of the organoids even after repeated thawing and freezing cycles.
OC organoid model applications
Application of OC organoid models for chemotherapeutic drug screening
Drug screening, which refers to testing the effects of a variety of drugs on several cell lines, is used to identify and develop medicines (44). In cancer therapy, drug screening aims to screen numerous compounds or new compounds for specific physiological activity against tumor cells. The present review aimed to summarize the differences in drug screening methods and their application in tumor therapy. For this purpose, models such as 2D cell cultures, multicellular tumor spheroids (MTSs) and organ tissues were assessed (Table II).
Notably, 2D cell cultures are the mainstay of fundamental cellular research, and are used to predict drug activity, metabolism and toxicity in vitro (45). In cancer screening, cytotoxicity testing is often performed on cultured tumor cell lines in monolayer culture due to the rapid and uncontrolled growth of these cell lines (46). By contrast, the main disadvantage of 2D systems is their inability to reproduce complex 3D structures or to communicate with the TME (47,48). Thus, 2D culture models may differ greatly from growing tumors with regard to cell morphology, proliferation, and gene and protein expression (49). Therefore, only 10% of the drugs that pass in vitro testing positively impact the clinic or lead to approval. The proportion of anticancer drugs that demonstrate clinical efficacy is even lower, at ~5% (50). Furthermore, a number of drugs reported to have potent anticancer effects in 2D cell culture models have failed clinical trials (51). In 2011, ~900 anticancer drugs passed cell.based tests, but only 12 were approved by the Food and Drug Administration after clinical testing (52).
The cells in MTS models more closely mimic the cell shape and environment in vivo than monolayer culture models (53,54); however, the size of spheroids has a substantial influence on their survival and reaction to drugs (55). Spheroids <100 µm do not reveal the complexity of the tumor or tissue (56). On the other hand, spheroids with diameters in the range of 400–500 µm can reflect the behavior of tumors in vivo due to the presence of different cellular layers: A necrotic core surrounded by a region of quiescent cells and an outer region composed of proliferating cells. The peripheral cells may reflect the in vivo tumor situation next to the capillaries, while the distant internal cells remain quiescent or die due to apoptosis or necrosis (57). This may also be why a number of reports (58–60) have shown that antitumor drugs are less effective against 3D cultured cancer cells than against 2D cultures.
It has also been shown that the MTS model has the potential to screen for antitumor drugs. Hirst et al (61) previously grew a series of EOC cell lines in 3D cell cultures to form multicellular tumor spheres (MCTSs). These MCTSs were characterized on the basis of the molecular and cellular properties of EOC, and were screened against cells grown in 2D cell culture to identify previously underappreciated anticancer agents. The MCTSs demonstrated enhanced resistance to chemotherapy, showed symptoms of senescence and hypoxia, and expressed numerous stem-cell-related transcripts, including ALDH1A and CD133 (PROM1). This previous study identified licofidone as a candidate with more activity in the MCTSs than in the 2D-cultured cells using a clinically repurposed drug library. Licofidone has been shown to be synergized with paclitaxel in an ovarian MCTS model, as well as in a patient-derived tumor xenograft model. The combination of licofidone and paclitaxel significantly increased the median survival time (>141 days) in mice compared with paclitaxel (115 days), licofidone (37 days) or vector (30 days). In addition, the Mantel-Haenszel hazard ratio (HR) was confirmed to be superior to the vector (HR=0.037) and paclitaxel (HR=0.017). The results of this previous study identified the potential use of an underappreciated anti-inflammatory agent in OC treatment.
Although the MTS model has demonstrated its unique advantages in antitumor drug screening, most identified drugs have failed in clinical applications due to the differences between MTSs and the real in vivo environment. Therefore, researchers have begun to explore OC organoid models. Cavarzerani et al (62) derived organoids from the ascites or tissues of patients with OC. In this previous study, the development of an organoid culture model with a higher growth rate, a faster drug response and better drug penetration into the extracellular matrix (ECM) was revealed to be possible in a passive microfluidic platform that maintained the vital signs of the sample and collected data for up to 16 medicines on a single plate. Achieving successful cultivation of organoids originating from patients with HGSOC was considered feasible in Mimetas Dual Lane OrganoPlates®. Thus, OC organoids may potentially be used to assess the antitumor activity of drugs.
OC organoids in chemosensitivity testing
Organoids can also be applied to chemosensitivity testing. In vitro susceptibility and resistance tests are used to determine whether a sample of a patient's tumor tissue exhibits a response (i.e., a reduction in tumor survival) when exposed to a selected chemotherapeutic agent under laboratory conditions (63). Drug response measurements aim to assess the therapeutic effectiveness of a drug across a specified concentration range. Wherever possible, multi-drug response metrics should be used to account for possible experimental variations during the measurement, initial population and cell division count, including: Emax (maximal effect of the drug), EC50 (drug concentration that reaches Emax), IC50 (median inhibitory concentration), GI50 (concentration that decreases the overall cell growth by 50%), GR50 (50% inhibition of cell growth rate) and AUC (area under the dose-response curve, which represents the cumulative effect of the medicinal product) (64). Therefore, a multi-model perspective offers a comparative analysis of sensitivity testing in OC (Table II).
Carboplatin
Carboplatin is the most common first-line chemotherapy agent in the treatment of gynecological malignancies. The sensitivity of ovarian epithelial adenocarcinoma cell lines to carboplatin chemotherapy in vitro varies in 2D or 3D models (65). Compared with other ex vivo and in vivo models, the response of the 3D system to carboplatin has been revealed to be in best agreement with the in vivo model. To assess the current differences in treatment response in in vitro and in vivo models, and to deal with this problem, a comparative study was conducted by Maru et al (40) to distinguish the response of four different models to carboplatin chemotherapy, including 2D single layer, 3D sphere, 3D tumor ex vivo and mouse xenograft models. Six previously characterized EOC cell lines with different chemosensitivities were used and viability tests were carried out on each model. The in vivo results in the mouse model correlated with the 2D response in 3/6 cell lines, while they correlated with the 3D sphere response in 4/6 cell lines and the 3D ex vivo model in 5/5 cell lines. These results highlight the variability of treatment responses across models, and indicated that the response of carboplatin was best correlated with the response of EOC cell lines grown in a 3D ex vivo model.
Similarly, Brodeur et al (65) investigated the response of single-layer and spherical (3D) EOC cells to carboplatin, and then compared it with the in vivo response (xenografts). Mice were injected with six ovarian epithelial adenocarcinoma cell lines that received three different concentrations of carboplatin. Their responses were evaluated and classified based on tumor volume and immunofluorescence measurements. The same ovarian epithelial adenocarcinoma cell lines were seeded onto shallow adhesion plates to form spheroids and were processed. Flow cytometric analysis was performed to classify ovarian epithelial adenocarcinoma cell lines based on IC50; this was compared to previously published 2D IC50 results. The results showed that the 3D system was in the best agreement with the in vivo model; in particular, the 2D super-resistant ovarian epithelial adenocarcinoma cell lines became more sensitive in either the mouse model or the 3D system. By contrast, the ultrasensitive ovarian epithelial adenocarcinoma cell line in the 2D system was more resistant in xenografts and spherical tissues.
Additionally, Thorel et al (66) established seven models (four cell lines, two PDOs and one PDX), all derived from the same OCCC. In order to establish their relevance, a comprehensive characterization was conducted using morphological, histological and transcriptomic analyses, as well as an evaluation of the patient's response to treatment, which was compared with the patient's chemotherapeutic response and showed resistance to drugs, carboplatin, gemcitabine and doxorubicin. Only PDX and PDO models derived from tumors could replicate the heterogeneity of patients. Patients were refractory to carboplatin, doxorubicin and gemcitabine, whereas cancer cells were susceptible to these agents. The PDX and PDO models, on the other hand, were resistant to these three drugs. Transcriptome analysis was consistent with these results, as models that faithfully reproduced the clinical response differed from the classical 2D cell culture models. Subsequently, the researchers examined the potential of previously unused drugs and determined that the histone deacetylase inhibitor belinostat was a potential therapeutic agent based on the response of the PDO. These findings indicated that PDX and PDO models may be the most relevant; however, only the PDO model offered all of the necessary preconditions for prediction and could be used in conventionally refractory types of cancer, especially aggressive ones.
Paclitaxel
Paclitaxel is a commonly used medicine for the treatment of OC. In a previous study, 3D patient-derived tumor spheroid structures were tested for paclitaxel susceptibility in comparison to 2D monolayer cultures. Loessner et al (67) performed an extensive study of EOC using hydrogel as a 3D model. The two cell lines (OV-MZ-6 and SKOV-3) were revealed to exhibit similar proliferation in 2D cultures, but different in 3D cultures. Spherical cell formation could only be observed in 3D cultures when the cells were embedded in hydrogels. The proliferation of OV-MZ-6 and SK-OV-3 cells in polyethylene glycol hydrogel possessing arginyl-glycyl-aspartic acid functionalized sites with glutamine, a general nutritional addition to the cell culture process, and matrix metalloproteinase sensitive sites for 14 days, followed by paclitaxel treatment, demonstrated a higher survival rate (40–60%) 7 days post-treatment compared with the single cells (20%) in 2D culture. Thus, 2D chemosensitivity evaluation does not necessarily reflect patient pathophysiology.
In order to determine the optimal conditions for analyzing the drug response of SKOV-3 and OVCAR-3 spheroids with the expected properties (roundness approaching 1.0; diameter in the range of 200–500 µm) to paclitaxel, two methods were used to evaluate the spheroids in two 3D cultures, after the addition of methylcellulose (0.25 and 0.5%, w/v) to the culture medium. Compared with 2D culture, SKOV-3 and OVCAR-3 spheroids retained cell density, increased apoptosis and increased resistance to paclitaxel treatment. The results indicated that 3D cultures may provide more reliable drug response results than 2D monolayer cultures (68). Meanwhile, Bi et al (69) assessed paired tumors and adjacent normal tissues from the same patient. At tumor-killing doses, they reported minimal cell destruction with variable chemotherapeutic agents. The results indicated that, in comparison with other drug treatments, the OC organoids of the patient were more sensitive to paclitaxel, and the survival rate was 48.7% after exposure to this single agent. The patient OC organoids were also more sensitive to gemcitabine (65% decrease in viability) and topotecan (56% decrease in viability) than the previously received chemotherapy regimen carboplatin + paclitaxel. Thus, organoids were considered more sensitive to paclitaxel compared to 2D monolayers.
Other drugs
Bevacizumab (Avastin®) is a recombinant humanized monoclonal antibody against vascular endothelial growth factor (VEGF) (70), which binds to and neutralizes the biological properties of human VEGF by blocking VEGF interaction with its receptor (71). Bevacizumab is frequently used as an adjuvant treatment in gynecological tumors (72). In mice bearing HOC22 ×enografts, OS and complete remission were markedly prolonged when bevacizumab was used in combination with chemotherapy and continued at the end of chemotherapy. Bevacizumab, on its own, inhibited the formation of ascites and had a limited impact on tumor load (73). Bi et al (69) also tested the sensitivity of PDOs to either bevacizumab alone or in combination with first-line standard chemotherapy. Overall, bevacizumab as a monotherapy had a minimal effect on cell viability and, when combined with chemotherapy, had little additive effect on cell killing
Olaparib was the first approved poly (ADP-ribose) polymerase inhibitor (PARPi) used to treat patients with advanced BRCA-mutated OC (70,72,74). PARPis inhibit PARP in cells with BRCA-mutated tumors; they can cause ‘synthetic lethality’ by targeting two DNA repair pathways at the same time and do not affect normal cells (73), Hill et al (41) formulated a 3D spherical functional evaluation methodology aimed at quantifying the responsiveness of two PARPi agents, specifically niraparib and olaparib, within ascites-derived primary cell cultures (AsPCs) derived from patients diagnosed with HGSOC. An agarose-based protocol for AsPC preparation enabled efficient isolation and propagation of monolayer and 3D AsPCs. The results showed a distinct sensitivity difference to PARPis, with monolayer AsPCs exhibiting 88 and 52% sensitivity to niraparib and olaparib, respectively, compared with 66 and 38% sensitivity in 3D AsPCs. The latter aligns with prior homologous recombination deficiency (HRD) estimates in EOC (40–60%), emphasizing the relevance of 3D models.
Application of OC organoid models in individualized therapy
The application of organoids in personalized medicine has also begun to be implemented (74,75). The development of personalized medicine is about preventing, diagnosing and treating pathological conditions based on individual characteristics (38). The characteristics of the individual include the entire body, tissues and cells, including genetic, epigenetic, transcriptomic, proteomic and metabolomic markers (76). In addition, gene expression profiles can be studied and changes associated with tumors can be identified (77). Personalized medicine obtains patient-specific disease information to select drugs and the correct dosage, and to optimize therapeutic procedures (78). Personalized medicine also influences preclinical studies of drugs, not just regarding efficacy and safety, but also to reduce drug development costs (38). OC encompasses a diverse array of histotypes, each characterized by distinct origins, underlying genetic and epigenetic alterations, and varying survival outcomes. Moreover, the differing prevalence of these histotypes across various ethnicities adds another layer of intricacy (79). In light of these multifaceted variables, the use of traditional 2D cell lines, albeit immortalized and homogeneous, as in vitro models of OC, are inherently limited in their capacity to capture the complexity of the disease at any single time point or in a microenvironment that accurately mirrors the in vivo scenario. Consequently, organoid technology has emerged as a promising avenue for advancing cancer research and facilitating the development of tailored therapeutic strategies (80). In clinical practice, OC organoid models have demonstrated their unique value in personalized therapy. For example, Lui et al (81) provided prospective and retrospective evidence from ≥18 cases, which indicated that organoid drug screening can accurately predict clinical response to chemotherapy and targeted therapy. This previous study also reported on a patient with platinum-resistant plasma OC who responded to ibrutinib therapy after screening, with this drug identified as having an excellent response in their organoid. After 3 months of treatment, the CA125 levels of the patient had decreased from 250 to 125 U/ml.
Preclinical studies
It is becoming increasingly apparent that standard treatment does not apply to every patient and that it would be helpful to use pretreatments to provide a more individualized regimen. Drug testing of PDOs can be executed in a timely manner; in addition, PDOs have the potential to identify the most efficacious drug in each case, and to offer a promising preclinical platform for tailoring personalized cancer treatment strategies in patients with gynecological malignancies (69).
For example, in 2019, Phan et al (82) pioneered the use of PDOs from HGSOC and ovarian sarcoma, using an automated screening system to assess the unique responses of these organoids to a panel of 240 kinase inhibitors. The high-throughput methodology not only identified sensitive signaling pathways but also facilitated the selection of the most efficacious drugs within specific molecular classes. This previous study presented a scalable and functional precision medicine platform titled Drug Efficacy Testing in 3D Culture, which can quantify the responsiveness of patient-derived cells to various drugs and drug combinations through live-cell imaging. Employing this platform, 27 tailored drug combinations were evaluated, achieving results within 10 days post-surgery. Notably, the findings revealed that the combination of carboplatin and a Bcl-xL inhibitor exhibited synergistic effects in 4 out of 8 OC samples tested, whereas afatinib and the Bcl-xL inhibitor A-1331852 demonstrated synergism in 5 of 7 OC models. These results suggested that combining the Bcl-xL inhibitor A-1331852 with either afatinib or carboplatin may hold promise as a potential future therapeutic strategy for patients with OC (83).
Biomarkers
Predictive biomarkers of therapeutic response are vital for assessing clinical benefit in patients with OC, yet they remain an unmet need (84). Notably, the potential of RAD51 foci as predictive biomarkers in HGSOC warrants validation in clinical trials (85). RAD51 has emerged as a prime candidate biomarker for such assessments due to several reasons. Firstly, upon the occurrence of DNA double-strand breaks, the kinase ATM swiftly phosphorylates histone H2AX (ᵧH2AX), initiating a cascade that leads to the formation of single-stranded DNA with 3′ projections. These, in turn, engage replication protein A, which RAD51 subsequently displaces via BRCA1 and BRCA2, facilitating homologous recombination (HRec). Given the multitude of upstream events required for RAD51 binding and its central role in HRec, RAD51 provides a holistic readout of the intricate steps of HRec. Secondly, RAD51 binding results in the formation of nucleoprotein filaments that can be visualized as foci under a microscope, with RAD51 focus deficiency serving as a functional marker for HRD (86–88). Thirdly, RAD51 foci detection has been shown to possess predictive value for platinum chemotherapy and PARPi responses in OC xenografts (89–91), correlating with in vitro platinum responses in established and primary OC cell lines (Pearson r=0.96; P=0.01). Tumor tissue cells that did not respond to platinum have been reported to have significantly higher RAD51 scores than those that responded to platinum (P<0.001). Notably, tumors with low RAD51 scores exhibited heightened platinum sensitivity, with a significantly higher likelihood of achieving pathologic complete response [relative risk (RR) 5.28; P<0.001] and platinum sensitivity (RR, ∞; P=0.05). Furthermore, the RAD51 score could accurately predict chemotherapy response (AUC, 0.90; 95% CI, 0.78.1.0; P<0.001), with an automated quantification system mirroring manual test results at 92% accuracy. In the validation cohort, RAD51-low tumors displayed enhanced platinum sensitivity (RR, ∞; P<0.001), a 100% positive predictive value for platinum sensitivity, and improved progression-free survival (HR, 0.53; 95% CI, 0.33.0.85; P<0.001) and OS (HR, 0.43; 95% CI, 0.25.0.75; P=0.003) compared with RAD51-high tumors (85). Advancements have focused on assessing HRec proficiency through RAD51 foci formation at DNA damage sites, exemplified by the repair capability (RECAP) test (92). Initially devised for breast cancer HRD assessment, this test has been validated using tumor sections and has demonstrated applicability in OC (93), underlining the versatility of tumor sections and diagnostic/therapeutic biomarker potential. Additionally, functional HRec status in PDOs has been evaluated using the RECAP test, correlating with PDO drug sensitivity (including platinum chemotherapy and PARPis) (37,42) and clinical response (41). However, as evidenced by in vivo studies, HRD alone does not guarantee a response to platinum or PARPis (41), pointing to a more intricate landscape requiring further exploration.
Genomics
Precision healthcare represents a strategy that tailors therapeutic interventions by comprehensively assessing the interplay of an individual's genetic makeup, environmental factors and lifestyle patterns. Since genomics alone is insufficient to determine treatment options for most patients with advanced diseases, the creation of live biological specimens of tumor-like organs can facilitate the integration of genomic profiles and drug screening within an iterative framework, enabling the identification of personalized therapeutic strategies tailored to each patient (38). In comparison to traditional 2D cultures, 3D organoid cultures exhibit a broader spectrum of drug responses that more accurately mirror genomic variations (39), rendering them an ideal platform for drug sensitivity assessments in translational medicine and precision healthcare endeavors. PDX models of OC also reflect the heterogeneity of tumor genomic profiles and serve as a valuable tool for drug evaluation (94,95). Nevertheless, organoids emerge as a superior 3D culture system, surpassing PDX in terms of i) reduced tumor sample requirements, ii) expedited engraftment timelines and iii) enhanced engraftment success rates (96). Notably, de Witte et al (97) conducted in vitro drug screening utilizing 36 genome-wide profiled PDOs from 23 patients with OC, demonstrating the ability of OC PDOs to faithfully recapitulate individual patient responses to first-line chemotherapeutics. Notably, variations in sensitivity were observed, with low responsiveness to carboplatin/paclitaxel, PARPis and specific tyrosine kinase inhibitors (afatinib, adavotinib), contrasted by high responsiveness to gemcitabine, flavopiridol (a cell cycle-dependent kinase inhibitor) and vimofenib (a BRAF V600E kinase inhibitor). Therefore, OC organoid models may be mainly applied for drug screening, sensitivity and individualized therapy (Fig. 2).
Deficiencies and prospects of OC organoid models in chemotherapy
Deficiencies
Technical limitationsPDO cultures may not be representative of tumor settings because of the absence of stroma, vasculature and immune cells (35); therefore, the organoid model cannot mimic the response to immunotherapy and antiangiogenic therapy (98). Heterogeneity within PDO culture tumors is also incomplete (97). The ability of organoids to maintain biological properties and functions similar to those of the primary tumors in an in vitro environment holds paramount significance for developing clinical applications.
In vivo, tissue development is constrained by external stimulation delivered in an exact space and time sequence. Usually, this does not occur in conventional 3D culture systems, where cells are embedded in an isotropic matrix and uniformly flooded with biochemical and ecological niche signals. This limitation can be overcome by a 3D culture matrix that can release or present biomolecules under spatiotemporal control (99,100). Achieving accurate control of the growth and differentiation of organoids is a critical task in organic research, which carries profound significance across diverse areas, such as disease modeling, pharmaceutical screening and regenerative medical research.
Individual differences
Because of the significant individual differences in patients with OC and the wide variety of tumor mutant types (e.g. Trp53, Brca1, KRAS, BRAF, PTEN, PIK3CA), it is not possible to fully simulate each patient's individual conditions. This may result in some bias in the screening and evaluation of chemotherapeutic drugs, and also increases the difficulty and cost of organoid culture.
Tumor characteristics and physiological barriers, such as the blood-brain barrier (BBB), vary from patient to patient, affecting drug penetration and metabolism in cancer tissues and thus altering therapeutic efficacy. Therefore, the use of human equivalent dose (the in vivo concentration of a drug in tumor tissue) is recommended in cancer organ tissue therapeutic trials to more accurately assess drug efficacy. The development of microfluidics and an artificial BBB based on microchip organ technology may advance this field (101,102).
Cost
Compared with monolayer cell cultures, organoid cultures are more costly because of the need for specific media, including ECM (e.g., Matrigel) and cofactors (e.g., growth factors and hormones) (103,104). In addition, the existence of individual differences and the difficulty of standardization further increases the cost of experiments, as different laboratories and research groups may need to use different methods and materials to conduct experiments, leading to higher costs of duplicating experiments and validation.
The key to improving the efficacy and comparability of organoid techniques is standardization; however, there are currently notable differences in organoid culture protocols from different investigators (34,103), which may lead to inconsistent results. To achieve standardization, key factors such as tissue manipulation procedures, culture media and growth factors need to be harmonized (103). However, standardization is challenged by individual differences, unknown composition of animal-derived scaffolds and poor tunability (105). In addition, the animal-derived Engelbreth-Holm-Swarm substrates used in organoid cultures vary widely between batches and contain unknown contaminants (106), which also limits standardization. Next-generation organoid models, such as co-culture models that incorporate different cells, may partially address these issues but have variable success rates and have also not been standardized (107).
Prospects
3D bioprinting3D bioprinting is a promising method for the construction of heterogeneous and reproducible cancer models with controlled shape and structure. The 3D-bioprinted structure can be used to simulate the TME. Different cell layers can be produced using bioprinting, including normal tissue-specific cells, connective tissue and cancer cells (108). This technology can accurately replicate the complex structure and microenvironment of cancer cells, creating organoid models that are more similar to the real-world situation of the tumor (109). Baka et al (110) described a 3D-bioprinted ovarian carcinoma model that combined cancer cells (SKOV-3) and cancer-associated fibroblasts (CAFs). The resulting tumor models demonstrated their ability to maintain cell viability and proliferation, and cells were observed to self-assemble in heterogeneous aggregates. Furthermore, it was observed that CAFs were recruited and multiplied in the surrounding tumor cells in an in vivo process occurring in the TME. Notably, this approach has also demonstrated an ability to produce a large number of reproducible tumor models, which can undergo conventional experimental methods (cell viability and metabolism assays, histology and immunology, and real-time imaging). 3D bioprinting of tumor constructs as personalized in vitro models may be a useful tool for screening for anticancer drugs and establishing accurate treatment regimens. As an example, MRC-5 fibroblasts and human OC cells (OVCAR-5) were previously printed on Matrigel by means of droplet-based bioprinting to form high-throughput, reproducible multi-cellular structures in a controlled space. This approach was shown to provide a platform for minimizing the size of 3D culture models at the macro scale; this could be considered an innovative platform for high-throughput and robust personalized drug screening, including tumor and stromal cell microenvironments (111).
Gene editing technologies
Gene editing techniques, such as CRISPR-Cas9, have the potential to introduce or modify mutations that mimic tumor-forming processes, and potentially identify and modify driver genes in cancer organoids (112). Through CRISPR-Cas9, organoids can be artificially altered by mutation of function or loss of function to induce malignancy (113). Notably, these gene editing techniques may be used to modify specific genes in organoid models to investigate the role of these genes in the occurrence, progression and treatment of OC. This could help to improve the understanding of the mechanisms of OC and to develop new therapeutic agents. Lõhmussaar et al (114) previously introduced mutations into common HGSOC genes, including Trp53, Brca1, Nf1 and Pten, using the genome editing method of CRISPR-Cas9. Organoids have also been artificially modified by gain-of-function or loss-of-function mutations to induce malignancy via CRISPR-Cas9 (115). Furthermore, Kopper et al (37) demonstrated the feasibility of this approach in OC using normal Fallopian tube organoids from high-risk OC donors that could be efficiently genome-edited via CRISPR-Cas9 and clonally amplified. In addition, in this previous study, CRISPR-Cas9 genome editing was used to introduce common HGSOC gene defects into mouse oviducts and the ovarian surface epithelium cell-like organs, showing that both organ types are potentially carcinogenic.
Genomics
Single-cell sequencing has emerged as an advanced biotechnological technique capable of decoding the OC landscape at single-cell resolution. It works at the gene, transcriptome, protein, epigenome and metabolism levels, and provides detailed information that differs from bulk sequencing methods, which only provide average data, representing the overall or average characteristics of the cell population in the entire sample or lesion area on specific lesions. Single-cell sequencing techniques provide detailed insights into the immune and molecular mechanisms that underlie tumorigenesis, progression, resistance and immune escape. These insights can be used to develop innovative diagnostic markers, treatment strategies and prognostic indicators (116). Wan et al (117) reported that HGSOC-like organoid co-cultures were treated with bispecific anti-PD-1 or PD-L1 antibodies, and then underwent single-cell RNA-sequencing. The results showed that this treatment reprogrammed inert natural killer cells and T cells into highly active cytotoxic states, thus indicating the potential benefit of bispecific antibody treatment in HGSOC. Gonzalez et al (118) performed an in-depth single-cell phenotypic characterization of HGSOC by multiparametric mass cytometry. The features of HGSOC biology were examined using a cytometry antibody panel and rare tumor subtypes of HGSOC were identified. It was shown that one of these rare subtypes was enriched in epithelial-mesenchymal transition signaling and was associated with increased tumor metastasis.
Conclusion
In summary, OC organoids maintain high tumor cell heterogeneity and are cultured in small samples, which can be maintained for long-term expansion and can be cryopreserved. OC organoids have been successfully developed from multiple stages and subtypes (56 organoid lines) (37). Thus far, OC organoid models have been widely used in drug screening, biomarker identification and personalized therapy; however, there are challenges to the use of these organoid models, including technical limitations, individual differences, cost issues and issues of standardization. To overcome these shortcomings, a further exploration into the application of technologies, such as 3D bioprinting, gene editing and genomics, is recommended in the optimization of organoid models to improve the future stability and reproducibility of these models. The development of these technologies is expected to make organoid models more stable and reproducible, which will better simulate the in vivo environment, and improve the accuracy and efficiency of drug screening. Therefore, the OC organoid model has a broad potential for application in chemotherapy. Along with the development of the technique, it is hoped that this model will be a more precise and efficient method to predict the prognosis of OC, and to improve therapeutic effects and the quality of life in patients with OC. In addition, research in this area may provide valuable information for other cancer treatments.
Acknowledgements
Not applicable.
Funding
This work was supported by the Natural Science Foundation General Project of Hubei Provincial (grant no. 2024AFB1044), the Development Fund Project of Yangtze University (grant no. WJ2019-22) and the Start Fund of First Affiliated Hospital of Yangtze University (grant no. 2022DIF01).
Availability of data and materials
Not applicable.
Authors' contributions
WZ designed and conceived the study, wrote the manuscript and acquired funding. YD designed and conceived the study and wrote the manuscript. HH and KC conducted the literature analysis. QZ and XC were involved in visualization. HZ and YX revised the manuscript. Data authentication is not applicable. All authors read and approved the final version of the manuscript.
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:
OC |
ovarian cancer |
2D |
two-dimensional |
3D |
three-dimensional |
PDX |
patient-derived xenograft |
OS |
overall survival |
HGSOC |
high-grade serous ovarian carcinoma |
TME |
tumor microenvironment |
ECM |
extracellular matrix |
PDOs |
patient-derived organoids |
OCCC |
ovarian clear cell carcinoma |
ENOC |
ovarian endometrioid carcinoma |
MTSs |
multicellular tumor spheroids |
MCTSs |
multicellular tumor spheres |
EOC |
epithelial ovarian carcinoma |
IC50 |
median inhibitory concentration |
VEGF |
vascular endothelial growth factor |
PARPi |
poly(ADP-ribose) polymerase inhibitor |
AsPCs |
ascites-derived primary cell cultures |
HRec |
homologous recombination |
BBB |
blood-brain barrier |
CAFs |
cancer-associated fibroblasts |
HR |
hazard ratio |
HRD |
homologous recombination deficiency |
References
Kuroki L and Guntupalli SR: Treatment of epithelial ovarian cancer. BMJ. 371:m37732020. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Matulonis UA, Sood AK, Fallowfield L, Howitt BE, Sehouli J and Karlan BY: Ovarian cancer. Nat Rev Dis Primers. 2:160612016. View Article : Google Scholar : PubMed/NCBI | |
American Cancer Society: Cancer Facts and Figures 2023. American Cancer Society; Atlanta, GA: 2023 | |
Liberto JM, Chen SY, Shih IM, Wang TH, Wang TL and Pisanic TR II: Current and emerging methods for ovarian cancer screening and diagnostics: A comprehensive review. Cancers (Basel). 14:28852022. View Article : Google Scholar : PubMed/NCBI | |
Kikuchi Y, Kita T, Takano M, Kudoh K and Yamamoto K: Treatment options in the management of ovarian cancer. Expert Opin Pharmacother. 6:743–754. 2005. View Article : Google Scholar : PubMed/NCBI | |
Bookman MA: First-line chemotherapy in epithelial ovarian cancer. Clin Obstet Gynecol. 55:96–113. 2012. View Article : Google Scholar : PubMed/NCBI | |
Ozols RF, Bundy BN, Greer BE, Fowler JM, Clarke-Pearson D, Burger RA, Mannel RS, DeGeest K, Hartenbach EM and Baergen R: Phase III trial of carboplatin and paclitaxel compared with cisplatin and paclitaxel in patients with optimally resected stage III ovarian cancer: A gynecologic oncology group study. J Clin Oncol. 41:4077–4083. 2023. View Article : Google Scholar : PubMed/NCBI | |
Kyrgiou M, Salanti G, Pavlidis N, Paraskevaidis E and Ioannidis JP: Survival benefits with diverse chemotherapy regimens for ovarian cancer: Meta-analysis of multiple treatments. J Natl Cancer Inst. 98:1655–1663. 2006. View Article : Google Scholar : PubMed/NCBI | |
Kim SI, Cho J, Lee EJ, Park S, Park SJ, Seol A, Lee N, Yim GW, Lee M, Lim W, et al: Selection of patients with ovarian cancer who may show survival benefit from hyperthermic intraperitoneal chemotherapy: A systematic review and meta-analysis. Medicine (Baltimore). 98:e183552019. View Article : Google Scholar : PubMed/NCBI | |
Ozols RF: Challenges for chemotherapy in ovarian cancer. Ann Oncol. 17 (Suppl 5):v181–v187. 2006. View Article : Google Scholar : PubMed/NCBI | |
Fung-Kee-Fung M, Oliver T, Elit L, Oza A, Hirte HW and Bryson P: Optimal chemotherapy treatment for women with recurrent ovarian cancer. Curr Oncol. 14:195–208. 2007. View Article : Google Scholar : PubMed/NCBI | |
Pokhriyal R, Hariprasad R, Kumar L and Hariprasad G: Chemotherapy resistance in advanced ovarian cancer patients. Biomark Cancer. 11:1179299×198608152019. View Article : Google Scholar : PubMed/NCBI | |
Cornelison R, Llaneza DC and Landen CN: Emerging therapeutics to overcome chemoresistance in epithelial ovarian cancer: A mini-review. Int J Mol Sci. 18:21712017. View Article : Google Scholar : PubMed/NCBI | |
Baker BM and Chen CS: Deconstructing the third dimension: How 3D culture microenvironments alter cellular cues. J Cell Sci. 125:3015–3024. 2012.PubMed/NCBI | |
Zhang Z, Bédard E, Luo Y, Wang H, Deng S, Kelvin D and Zhong R: Animal models in xenotransplantation. Expert Opin Investig Drugs. 9:2051–2068. 2000. View Article : Google Scholar : PubMed/NCBI | |
Bertotti A, Migliardi G, Galimi F, Sassi F, Torti D, Isella C, Corà D, Di Nicolantonio F, Buscarino M, Petti C, et al: A molecularly annotated platform of patient-derived xenografts (‘xenopatients’) identifies HER2 as an effective therapeutic target in cetuximab-resistant colorectal cancer. Cancer Discov. 1:508–523. 2011. View Article : Google Scholar : PubMed/NCBI | |
DeRose YS, Wang G, Lin YC, Bernard PS, Buys SS, Ebbert MT, Factor R, Matsen C, Milash BA, Nelson E, et al: Tumor grafts derived from women with breast cancer authentically reflect tumor pathology, growth, metastasis and disease outcomes. Nat Med. 17:1514–1520. 2011. View Article : Google Scholar : PubMed/NCBI | |
Sachs N, de Ligt J, Kopper O, Gogola E, Bounova G, Weeber F, Balgobind AV, Wind K, Gracanin A, Begthel H, et al: A living biobank of breast cancer organoids captures disease heterogeneity. Cell. 172:373–386.e310. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zanoni M, Cortesi M, Zamagni A, Arienti C, Pignatta S and Tesei A: Modeling neoplastic disease with spheroids and organoids. J Hematol Oncol. 13:972020. View Article : Google Scholar : PubMed/NCBI | |
Graham O, Rodriguez J, van Biljon L, Fashemi B, Graham E, Fuh K, Khabele D and Mullen M: Generation and culturing of high-grade serous ovarian cancer patient-derived organoids. J Vis Exp. 6:1912023. | |
Yani W, Qi J, Yuchen Z and Haiyan Z: Application of organoids technology in drug sensitivity test of ovarian cancer. J Int Obstet Gynecol. 49:181–185. 2022. | |
Yujie S, Hong Y, Jia L and Ying X: Application prospects on organoid culture system in drug screening and treatment target for ovarian cancer. J Chin Oncol. 28:1042–1045. 2022.(In Chinese). | |
Jianjun G, Wei Q, Hao W and Xiangyu Z: Application and prospect of organoid technique in cancer research. Chin J Tissue Engineering Res. 23:1136–1141. 2019.(In Chinese). | |
Aihara A, Abe N, Saruhashi K, Kanaki T and Nishino T: Novel 3-D cell culture system for in vitro evaluation of anticancer drugs under anchorage-independent conditions. Cancer Sci. 107:1858–1866. 2016. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Byrne AT, Alférez DG, Amant F, Annibali D, Arribas J, Biankin AV, Bruna A, Budinská E, Caldas C, Chang DK, et al: Interrogating open issues in cancer medicine with patient-derived xenografts. Nat Rev Cancer. 17:6322017. View Article : Google Scholar : PubMed/NCBI | |
Sachs N and Clevers H: Organoid cultures the analysis of cancer phenotypes. Curr Opin Genet Dev. 24:68–73. 2014. View Article : Google Scholar : PubMed/NCBI | |
Bleijs M, van de Wetering M, Clevers H and Drost J: Xenograft and organoid model systems in cancer research. EMBO J. 38:e1016542019. View Article : Google Scholar : PubMed/NCBI | |
Wensink GE, Elias SG, Mullenders J, Koopman M, Boj SF, Kranenburg OW and Roodhart JML: Patient-derived organoids as a predictive biomarker for treatment response in cancer patients. NPJ Precis Oncol. 5:302021. View Article : Google Scholar : PubMed/NCBI | |
Perkhofer L, Frappart PO, Müller M and Kleger A: Importance of organoids for personalized medicine. Per Med. 15:461–465. 2018. View Article : Google Scholar : PubMed/NCBI | |
Rossi G, Manfrin A and Lutolf MP: Progress and potential in organoid research. Nat Rev Genet. 19:671–687. 2018. View Article : Google Scholar : PubMed/NCBI | |
Tsang SI, Hassan AA, To SKY and Wong AST: Experimental models for ovarian cancer research. Exp Cell Res. 416:1131502022. View Article : Google Scholar : PubMed/NCBI | |
Yang J, Huang S, Cheng S, Jin Y, Zhang N and Wang Y: Application of ovarian cancer organoids in precision medicine: Key challenges and current opportunities. Front Cell Dev Biol. 9:7014292021. View Article : Google Scholar : PubMed/NCBI | |
Aboulkheyr Es H, Montazeri L, Aref AR, Vosough M and Baharvand H: Personalized cancer medicine: An organoid approach. Trends Biotechnol. 36:358–371. 2018. View Article : Google Scholar : PubMed/NCBI | |
Maenhoudt N, Defraye C, Boretto M, Jan Z, Heremans R, Boeckx B, Hermans F, Arijs I, Cox B, Van Nieuwenhuysen E, et al: developing organoids from ovarian cancer as experimental and preclinical models. Stem Cell Reports. 14:717–729. 2020. View Article : Google Scholar : PubMed/NCBI | |
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 | |
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 | |
Jabs J, Zickgraf FM, Park J, Wagner S, Jiang X, Jechow K, Kleinheinz K, Toprak UH, Schneider MA, Meister M, et al: Screening drug effects in patient-derived cancer cells links organoid responses to genome alterations. Mol Syst Biol. 13:9552017. View Article : Google Scholar : PubMed/NCBI | |
Maru Y, Tanaka N, Itami M and Hippo Y: Efficient use of patient-derived organoids as a preclinical model for gynecologic tumors. Gynecol Oncol. 154:189–198. 2019. View Article : Google Scholar : PubMed/NCBI | |
Hill SJ, Decker B, Roberts EA, Horowitz NS, Muto MG, Worley MJ Jr, Feltmate CM, Nucci MR, Swisher EM, Nguyen H, et al: Prediction of DNA repair inhibitor response in short-term patient-derived ovarian cancer organoids. Cancer Discov. 8:1404–1421. 2018. View Article : Google Scholar : PubMed/NCBI | |
Nanki Y, Chiyoda T, Hirasawa A, Ookubo A, Itoh M, Ueno M, Akahane T, Kameyama K, Yamagami W, Kataoka F and Aoki D: Patient-derived ovarian cancer organoids capture the genomic profiles of primary tumours applicable for drug sensitivity and resistance testing. Sci Rep. 10:125812020. View Article : Google Scholar : PubMed/NCBI | |
Hoffmann K, Berger H, Kulbe H, Thillainadarasan S, Mollenkopf HJ, Zemojtel T, Taube E, Darb-Esfahani S, Mangler M, Sehouli J, et al: Stable expansion of high-grade serous ovarian cancer organoids requires a low-Wnt environment. EMBO J. 39:e1040132020. View Article : Google Scholar : PubMed/NCBI | |
Kondo J and Inoue M: application of cancer organoid model for drug screening and personalized therapy. Cells. 8:4702019. View Article : Google Scholar : PubMed/NCBI | |
Antoni D, Burckel H, Josset E and Noel G: Three-dimensional cell culture: A breakthrough in vivo. Int J Mol Sci. 16:5517–5527. 2015. View Article : Google Scholar : PubMed/NCBI | |
Kimlin LC, Casagrande G and Virador VM: In vitro three-dimensional (3D) models in cancer research: An update. Mol Carcinog. 52:167–182. 2013. View Article : Google Scholar : PubMed/NCBI | |
Shoemaker RH: The NCI60 human tumour cell line anticancer drug screen. Nat Rev Cancer. 6:813–823. 2006. View Article : Google Scholar : PubMed/NCBI | |
Rizvanov AA, Yalvaç ME, Shafigullina AK, Salafutdinov II, Blatt NL, Sahin F, Kiyasov AP and Palotás A: Interaction and self-organization of human mesenchymal stem cells and neuro-blastoma SH-SY5Y cells under co-culture conditions: A novel system for modeling cancer cell micro-environment. Eur J Pharm Biopharm. 76:253–259. 2010. View Article : Google Scholar : PubMed/NCBI | |
Enmon RM Jr, O'Connor KC, Lacks DJ, Schwartz DK and Dotson RS: Dynamics of spheroid self-assembly in liquid-overlay culture of DU 145 human prostate cancer cells. Biotechnol Bioeng. 72:579–591. 2001. View Article : Google Scholar : PubMed/NCBI | |
Westhouse RA: Safety assessment considerations and strategies for targeted small molecule cancer therapeutics in drug discovery. Toxicol Pathol. 38:165–168. 2010. View Article : Google Scholar : PubMed/NCBI | |
Wong CC, Cheng KW and Rigas B: Preclinical predictors of anticancer drug efficacy: Critical assessment with emphasis on whether nanomolar potency should be required of candidate agents. J Pharmacol Exp Ther. 341:572–578. 2012. View Article : Google Scholar : PubMed/NCBI | |
Ravi M, Paramesh V, Kaviya SR, Anuradha E and Solomon FD: 3D cell culture systems: Advantages and applications. J Cell Physiol. 230:16–26. 2015. View Article : Google Scholar : PubMed/NCBI | |
Jamieson LE, Harrison DJ and Campbell CJ: Chemical analysis of multicellular tumour spheroids. Analyst. 140:3910–3920. 2015. View Article : Google Scholar : PubMed/NCBI | |
Beningo KA, Dembo M and Wang Yl: Responses of fibroblasts to anchorage of dorsal extracellular matrix receptors. Proc Natl Acad Sci USA. 101:18024–18029. 2004. View Article : Google Scholar : PubMed/NCBI | |
Sambale F, Lavrentieva A, Stahl F, Blume C, Stiesch M, Kasper C, Bahnemann D and Scheper T: Three dimensional spheroid cell culture for nanoparticle safety testing. J Biotechnol. 205:120–129. 2015. View Article : Google Scholar : PubMed/NCBI | |
Jarockyte G, Dapkute D, Karabanovas V, Daugmaudis JV, Ivanauskas F and Rotomskis R: 3D cellular spheroids as tools for understanding carboxylated quantum dot behavior in tumors. Biochim Biophys Acta Gen Subj. 1862:914–923. 2018. View Article : Google Scholar : PubMed/NCBI | |
Mehta G, Hsiao AY, Ingram M, Luker GD and Takayama S: Opportunities and challenges for use of tumor spheroids as models to test drug delivery and efficacy. J Control Release. 164:192–204. 2012. View Article : Google Scholar : PubMed/NCBI | |
Souza AG, Silva IBB, Campos-Fernandez E, Barcelos LS, Souza JB, Marangoni K, Goulart LR and Alonso-Goulart V: Comparative assay of 2D and 3D cell culture models: Proliferation, gene expression and anticancer drug response. Curr Pharm Des. 24:1689–1694. 2018. View Article : Google Scholar : PubMed/NCBI | |
Breslin S and O'Driscoll L: The relevance of using 3D cell cultures, in addition to 2D monolayer cultures, when evaluating breast cancer drug sensitivity and resistance. Oncotarget. 7:45745–45756. 2016. View Article : Google Scholar : PubMed/NCBI | |
Verjans ET, Doijen J, Luyten W, Landuyt B and Schoofs L: Three-dimensional cell culture models for anticancer drug screening: Worth the effort? J Cell Physiol. 233:2993–3003. 2018. View Article : Google Scholar : PubMed/NCBI | |
Hirst J, Pathak HB, Hyter S, Pessetto ZY, Ly T, Graw S, Koestler DC, Krieg AJ, Roby KF and Godwin AK: Licofelone enhances the efficacy of paclitaxel in ovarian cancer by reversing drug resistance and tumor stem-like properties. Cancer Res. 78:4370–4385. 2018. View Article : Google Scholar : PubMed/NCBI | |
Cavarzerani E, Caligiuri I, Bartoletti M, Canzonieri V and Rizzolio F: 3D dynamic cultures of HGSOC organoids to model innovative and standard therapies. Front Bioeng Biotechnol. 11:11353742023. View Article : Google Scholar : PubMed/NCBI | |
Samson DJ, Seidenfeld J, Ziegler K and Aronson N: Chemotherapy sensitivity and resistance assays: A systematic review. J Clin Oncol. 22:3618–3630. 2004. View Article : Google Scholar : PubMed/NCBI | |
Brooks EA, Galarza S, Gencoglu MF, Cornelison RC, Munson JM and Peyton SR: Applicability of drug response metrics for cancer studies using biomaterials. Philos Trans R Soc Lond B Biol Sci. 374:201802262019. View Article : Google Scholar : PubMed/NCBI | |
Brodeur MN, Simeone K, Leclerc-Deslauniers K, Fleury H, Carmona E, Provencher DM and Mes-Masson AM: Carboplatin response in preclinical models for ovarian cancer: Comparison of 2D monolayers, spheroids, ex vivo tumors and in vivo models. Sci Rep. 11:181832021. View Article : Google Scholar : PubMed/NCBI | |
Thorel L, Morice PM, Paysant H, Florent R, Babin G, Thomine C, Perréard M, Abeilard E, Giffard F, Brotin E, et al: Comparative analysis of response to treatments and molecular features of tumor-derived organoids versus cell lines and PDX derived from the same ovarian clear cell carcinoma. J Exp Clin Cancer Res. 42:2602023. View Article : Google Scholar : PubMed/NCBI | |
Loessner D, Stok KS, Lutolf MP, Hutmacher DW, Clements JA and Rizzi SC: Bioengineered 3D platform to explore cell-ECM interactions and drug resistance of epithelial ovarian cancer cells. Biomaterials. 31:8494–8506. 2010. View Article : Google Scholar : PubMed/NCBI | |
Tofani LB, Abriata JP, Luiz MT, Marchetti JM and Swiech K: Establishment and characterization of an in vitro 3D ovarian cancer model for drug screening assays. Biotechnol Prog. 36:e30342020. View Article : Google Scholar : PubMed/NCBI | |
Bi J, Newtson AM, Zhang Y, Devor EJ, Samuelson MI, Thiel KW and Leslie KK: Successful patient-derived organoid culture of gynecologic cancers for disease modeling and drug sensitivity testing. Cancers (Basel). 13:29012021. View Article : Google Scholar : PubMed/NCBI | |
Garcia J, Hurwitz HI, Sandler AB, Miles D, Coleman RL, Deurloo R and Chinot OL: Bevacizumab (Avastin®) in cancer treatment: A review of 15 years of clinical experience and future outlook. Cancer Treat Rev. 86:1020172020. View Article : Google Scholar : PubMed/NCBI | |
Cohen MH, Gootenberg J, Keegan P and Pazdur R: FDA drug approval summary: Bevacizumab plus FOLFOX4 as second-line treatment of colorectal cancer. Oncologist. 12:356–361. 2007. View Article : Google Scholar : PubMed/NCBI | |
Govindaraju S and Yun K: Synthesis of gold nanomaterials and their cancer-related biomedical applications: An update 3. Biotech. 8:1132018.PubMed/NCBI | |
Oliva P, Decio A, Castiglioni V, Bassi A, Pesenti E, Cesca M, Scanziani E, Belotti D and Giavazzi R: Cisplatin plus paclitaxel and maintenance of bevacizumab on tumour progression, dissemination, and survival of ovarian carcinoma xenograft models. Br J Cancer. 107:360–369. 2012. View Article : Google Scholar : PubMed/NCBI | |
Yang H, Wang Y and Wang P, Zhang N and Wang P: Tumor organoids for cancer research and personalized medicine. Cancer Biol Med. 19:319–332. 2021.PubMed/NCBI | |
Seidlitz T, Koo BK and Stange DE: Gastric organoids-an in vitro model system for the study of gastric development and road to personalized medicine. Cell Death Differ. 28:68–83. 2021. View Article : Google Scholar : PubMed/NCBI | |
Rivenbark AG, O'Connor SM and Coleman WB: Molecular and cellular heterogeneity in breast cancer: Challenges for personalized medicine. Am J Pathol. 183:1113–1124. 2013. View Article : Google Scholar : PubMed/NCBI | |
Verma M: Personalized medicine and cancer. J Pers Med. 2:1–14. 2012. View Article : Google Scholar : PubMed/NCBI | |
Offit K: Personalized medicine: New genomics, old lessons. Hum Genet. 130:3–14. 2011. View Article : Google Scholar : PubMed/NCBI | |
Peres LC, Risch H, Terry KL, Webb PM, Goodman MT, Wu AH, Alberg AJ, Bandera EV, Barnholtz-Sloan J, Bondy ML, et al: Racial/ethnic differences in the epidemiology of ovarian cancer: A pooled analysis of 12 case-control studies. Int J Epidemiol. 47:10112018. View Article : Google Scholar : PubMed/NCBI | |
Nero C, Vizzielli G, Lorusso D, Cesari E, Daniele G, Loverro M, Scambia G and Sette C: Patient-derived organoids and high grade serous ovarian cancer: From disease modeling to personalized medicine. J Exp Clin Cancer Res. 40:1162021. View Article : Google Scholar : PubMed/NCBI | |
Lui G, Richardson A, Chatterjee P, Pollastro M, Lints M, Peretti D, Rosati R, Appleyard L, Durenberger G, Diaz R, et al: Functional drug screening of organoids from ovarian cancer patients demonstrates clinical and genomic concordance and identifies novel therapeutic vulnerabilities. Cancer Res. 81:534. 2021. View Article : Google Scholar | |
Phan N, Hong JJ, Tofig B, Mapua M, Elashoff D, Moatamed NA, Huang J, Memarzadeh S, Damoiseaux R and Soragni A: A simple high-throughput approach identifies actionable drug sensitivities in patient-derived tumor organoids. Commun Biol. 2:782019. View Article : Google Scholar : PubMed/NCBI | |
Åkerlund E, Gudoityte G, Moussaud-Lamodière E, Lind O, Bwanika HC, Lehti K, Salehi S, Carlson J, Wallin E, Fernebro J, et al: The drug efficacy testing in 3D cultures platform identifies effective drugs for ovarian cancer patients. NPJ Precis Oncol. 7:1112023. View Article : Google Scholar : PubMed/NCBI | |
Clark J, Fotopoulou C, Cunnea P and Krell J: Novel ex vivo models of epithelial ovarian cancer: The future of biomarker and therapeutic research. Front Oncol. 12:8372332022. View Article : Google Scholar : PubMed/NCBI | |
Compadre AJ, van Biljon LN, Valentine MC, Llop-Guevara A, Graham E, Fashemi B, Herencia-Ropero A, Kotnik EN, Cooper I, Harrington SP, et al: RAD51 foci as a biomarker predictive of platinum chemotherapy response in ovarian cancer. Clin Cancer Res. 29:2466–2479. 2023. View Article : Google Scholar : PubMed/NCBI | |
Ceccaldi R, Rondinelli B and D'Andrea AD: Repair pathway choices and consequences at the double-strand break. Trends Cell Biol. 26:52–64. 2016. View Article : Google Scholar : PubMed/NCBI | |
Ito K, Murayama Y, Kurokawa Y, Kanamaru S, Kokabu Y, Maki T, Mikawa T, Argunhan B, Tsubouchi H, Ikeguchi M, et al: Real-time tracking reveals catalytic roles for the two DNA binding sites of Rad51. Nat Commun. 11:29502020. View Article : Google Scholar : PubMed/NCBI | |
Wilson AJ, Stubbs M, Liu P, Ruggeri B and Khabele D: The BET inhibitor INCB054329 reduces homologous recombination efficiency and augments PARP inhibitor activity in ovarian cancer. Gynecol Oncol. 149:575–584. 2018. View Article : Google Scholar : PubMed/NCBI | |
Pellegrino B, Herencia-Ropero A, Llop-Guevara A, Pedretti F, Moles-Fernández A, Viaplana C, Villacampa G, Guzmán M, Rodríguez O, Grueso J, et al: Preclinical in vivo validation of the RAD51 test for identification of homologous recombination-deficient tumors and patient stratification. Cancer Res. 82:1646–1657. 2022. View Article : Google Scholar : PubMed/NCBI | |
Mukhopadhyay A, Elattar A, Cerbinskaite A, Wilkinson SJ, Drew Y, Kyle S, Los G, Hostomsky Z, Edmondson RJ and Curtin NJ: Development of a functional assay for homologous recombination status in primary cultures of epithelial ovarian tumor and correlation with sensitivity to poly(ADP-ribose) polymerase inhibitors. Clin Cancer Res. 16:2344–2351. 2010. View Article : Google Scholar : PubMed/NCBI | |
Shah MM, Dobbin ZC, Nowsheen S, Wielgos M, Katre AA, Alvarez RD, Konstantinopoulos PA, Yang ES and Landen CN: An ex vivo assay of XRT-induced Rad51 foci formation predicts response to PARP-inhibition in ovarian cancer. Gynecol Oncol. 134:331–337. 2014. View Article : Google Scholar : PubMed/NCBI | |
Meijer TG, Verkaik NS, Sieuwerts AM, van Riet J, Naipal KAT, van Deurzen CHM, den Bakker MA, Sleddens HFBM, Dubbink HJ, den Toom TD, et al: Functional ex vivo assay reveals homologous recombination deficiency in breast cancer beyond BRCA gene defects. Clin Cancer Res. 24:6277–6287. 2018. View Article : Google Scholar : PubMed/NCBI | |
van Wijk LM, Vermeulen S, Meijers M, van Diest MF, Ter Haar NT, de Jonge MM, Solleveld-Westerink N, van Wezel T, van Gent DC, Kroep JR, et al: The RECAP test rapidly and reliably identifies homologous recombination-deficient ovarian carcinomas. Cancers (Basel). 12:28052020. View Article : Google Scholar : PubMed/NCBI | |
Liu JF, Palakurthi S, Zeng Q, Zhou S, Ivanova E, Huang W, Zervantonakis IK, Selfors LM, Shen Y, Pritchard CC, et al: Establishment of patient-derived tumor xenograft models of epithelial ovarian cancer for preclinical evaluation of novel therapeutics. Clin Cancer Res. 23:1263–1273. 2017. View Article : Google Scholar : PubMed/NCBI | |
Cybulska P, Stewart JM, Sayad A, Virtanen C, Shaw PA, Clarke B, Stickle N, Bernardini MQ and Neel B: A genomically characterized collection of high-grade serous ovarian cancer xenografts for preclinical testing. Am J Pathol. 188:1120–1131. 2018. View Article : Google Scholar : PubMed/NCBI | |
Hidalgo M, Amant F, Biankin AV, Budinská E, Byrne AT, Caldas C, Clarke RB, de Jong S, Jonkers J, Mælandsmo GM, et al: Patient-derived xenograft models: An emerging platform for translational cancer research. Cancer Discov. 4:998–1013. 2014. View Article : Google Scholar : PubMed/NCBI | |
de Witte CJ, Valle-Inclan JE, Hami N, Lõhmussaar K, Kopper O, Vreuls CPH, Jonges GN, van Diest P, Nguyen L, Clevers H, et al: Patient-Derived ovarian cancer organoids mimic clinical response and exhibit heterogeneous inter- and intrapatient drug responses. Cell Rep. 31:1077622020. View Article : Google Scholar : PubMed/NCBI | |
Zhou Z, Cong L and Cong X: Patient-Derived organoids in precision medicine: Drug screening, organoid-on-a-chip and living organoid biobank. Front Oncol. 11:7621842021. View Article : Google Scholar : PubMed/NCBI | |
Narita Y, Kitazoe Y, Kurihara Y, Okuhara Y, Takamatsu K, Saito N and Doi Y: Increase or decrease of HDL-cholesterol concentrations during pravastatin treatment depending on the pre-treatment HDL cholesterol levels. Eur J Clin Pharmacol. 52:461–463. 1997. View Article : Google Scholar : PubMed/NCBI | |
Mosiewicz KA, Kolb L, van der Vlies AJ, Martino MM, Lienemann PS, Hubbell JA, Ehrbar M and Lutolf MP: In situ cell manipulation through enzymatic hydrogel photopatterning. Nat Mater. 12:1072–1078. 2013. View Article : Google Scholar : PubMed/NCBI | |
Verduin M, Hoeben A, De Ruysscher D and Vooijs M: Patient-derived cancer organoids as predictors of treatment response. Front Oncol. 11:6419802021. View Article : Google Scholar : PubMed/NCBI | |
Ahn SI, Sei YJ, Park HJ, Kim J, Ryu Y, Choi JJ, Sung HJ, MacDonald TJ, Levey AI and Kim YT: Microengineered human blood-brain barrier platform for understanding nanoparticle transport mechanisms. Nat Commun. 11:1752020. View Article : Google Scholar : PubMed/NCBI | |
Nagle PW, Plukker JTM, Muijs CT, van Luijk P and Coppes RP: Patient-derived tumor organoids for prediction of cancer treatment response. Semin Cancer Biol. 53:258–264. 2018. View Article : Google Scholar : PubMed/NCBI | |
Psilopatis I, Sykaras AG, Mandrakis G, Vrettou K and Theocharis S: Patient-derived organoids: The beginning of a new era in ovarian cancer disease modeling and drug sensitivity testing. Biomedicines. 11:12022. View Article : Google Scholar : PubMed/NCBI | |
Kenny PA, Lee GY, Myers CA, Neve RM, Semeiks JR, Spellman PT, Lorenz K, Lee EH, Barcellos-Hoff MH, Petersen OW, et al: The morphologies of breast cancer cell lines in three-dimensional assays correlate with their profiles of gene expression. Mol Oncol. 1:84–96. 2007. View Article : Google Scholar : PubMed/NCBI | |
Hughes CS, Postovit LM and Lajoie GA: Matrigel: A complex protein mixture required for optimal growth of cell culture. Proteomics. 10:1886–1890. 2010. View Article : Google Scholar : PubMed/NCBI | |
Chan WS, Mo X, Ip PPC and Tse KY: Patient-derived organoid culture in epithelial ovarian cancers-Techniques, applications, and future perspectives. Cancer Med. 12:19714–19731. 2023. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Annett S, Moore G, Short A, Marshall A, McCrudden C, Yakkundi A, Das S, McCluggage WG, Nelson L, Harley I, et al: FKBPL-based peptide, ALM201, targets angiogenesis and cancer stem cells in ovarian cancer. Br J Cancer. 122:361–371. 2020. View Article : Google Scholar : PubMed/NCBI | |
Baka Z, Godier C, Lamy L, Mallick A, Gribova V, Figarol A, Bezdetnaya L, Chateau A, Magne Z, Stiefel M, et al: A coculture based, 3D bioprinted ovarian tumor model combining cancer cells and cancer associated fibroblasts. Macromol Biosci. 23:e22004342023. View Article : Google Scholar : PubMed/NCBI | |
Xu F, Celli J, Rizvi I, Moon S, Hasan T and Demirci U: A three-dimensional in vitro ovarian cancer coculture model using a high-throughput cell patterning platform. Biotechnol J. 6:204–212. 2011. View Article : Google Scholar : PubMed/NCBI | |
Driehuis E and Clevers H: CRISPR/Cas 9 genome editing and its applications in organoids. Am J Physiol Gastrointest Liver Physiol. 312:G257–G265. 2017. View Article : Google Scholar : PubMed/NCBI | |
Neal JT and Kuo CJ: Organoids as models for neoplastic transformation. Ann Rev Pathol. 11:199–220. 2016. View Article : Google Scholar : PubMed/NCBI | |
Lõhmussaar K, Kopper O, Korving J, Begthel H, Vreuls CPH, van Es JH and Clevers H: Assessing the origin of high-grade serous ovarian cancer using CRISPR-modification of mouse organoids. Nat Commun. 11:26602020. View Article : Google Scholar : PubMed/NCBI | |
Dumont S, Jan Z, Heremans R, Van Gorp T, Vergote I and Timmerman D: Organoids of epithelial ovarian cancer as an emerging preclinical in vitro tool: A review. J Ovarian Res. 12:1052019. View Article : Google Scholar : PubMed/NCBI | |
Li Z, Gu H, Xu X, Tian Y, Huang X and Du Y: Unveiling the novel immune and molecular signatures of ovarian cancer: Insights and innovations from single-cell sequencing. Front Immunol. 14:12880272023. View Article : Google Scholar : PubMed/NCBI | |
Wan C, Keany MP, Dong H, Al-Alem LF, Pandya UM, Lazo S, Boehnke K, Lynch KN, Xu R, Zarrella DT, et al: Enhanced efficacy of simultaneous PD-1 and PD-L1 immune checkpoint blockade in high-grade serous ovarian cancer. Cancer Res. 81:158–173. 2021. View Article : Google Scholar : PubMed/NCBI | |
Gonzalez VD, Samusik N, Chen TJ, Savig ES, Aghaeepour N, Quigley DA, Huang YW, Giangarrà V, Borowsky AD, Hubbard NE, et al: Commonly occurring cell subsets in high-grade serous ovarian tumors identified by single-cell mass cytometry. Cell Rep. 22:1875–1888. 2018. View Article : Google Scholar : PubMed/NCBI |