A highly accurate in vitro high-throughput assay system was developed to evaluate anticancer drugs using patient-derived tumor organoids (PDOs), similar to cancer tissues but are unsuitable for in vitro high-throughput assay systems with 96-well and 384-well plates.
Patient-derived tumor organoids (PDOs) are expected to be a preclinical cancer model with better reproducibility of disease than traditional cell culture models. PDOs have been successfully generated from a variety of human tumors to recapitulate the architecture and function of tumor tissue accurately and efficiently. However, PDOs are unsuitable for an in vitro high-throughput assay system (HTS) or cell analysis using 96-well or 384-well plates when evaluating anticancer drugs because they are heterogeneous in size and form large clusters in culture. These cultures and assays use extracellular matrices, such as Matrigel, to create tumor tissue scaffolds. Therefore, PDOs have a low throughput and high cost, and it has been difficult to develop a suitable assay system. To address this issue, a simpler and more accurate HTS was established using PDOs to evaluate the potency of anticancer drugs and immunotherapy. An in vitro HTS was created that uses PDOs established from solid tumors cultured in 384-well plates. An HTS was also developed for assessment of antibody-dependent cellular cytotoxicity activity to represent the immune response using PDOs cultured in 96-well plates.
Human cancer cell lines are widely accepted to study the biology of cancer and evaluate anticancer agents. However, these cell lines do not necessarily preserve the original characteristics of their source tissue because their morphology, gene mutation, and gene expression profile can change during culture over long periods. Furthermore, most of these cell lines are cultured in a monolayer or used as murine xenografts, neither of which physically represent tumor tissue1,2. Thus, the clinical efficacy of anticancer agents may not be the same as that observed in cancer cell lines. Therefore, in vitro systems, such as ex vivo assays using patient-derived tumor xenografts or patient-derived tumor organoids (PDOs) and tumor spheroid models that accurately reproduce the structure and function of tumor tissues, have been developed. Increasing evidence suggests that these models predict patients' response to anticancer agents by being directly comparable to the corresponding cancer tissue. These in vitro systems have been established for different tumor tissue types, and associated high-throughput assay systems (HTS) for drug screening have also been developed3,4,5,6,7. Heterogeneous ex vivo organoid cultures of primary tumors obtained from patients or patient-derived tumor xenografts have gained considerable traction in recent years because of their ease of culture and ability to maintain the complexity of cells in the stromal tissue8,9,10. These models are expected to enhance understanding of the biology of cancer and facilitate the evaluation of drug efficacy in vitro.
A series of novel PDOs were recently created from different types of tumor tissue, designated as F-PDO, under the Fukushima Translational Research Project. The PDOs form large cell clusters with a morphology similar to that of the source tumor and can be cultured for more than six months11. The comparative histology and comprehensive gene expression analyses showed that the features of the PDOs are close to those of their source tumor tissues, even after prolonged growth under culture conditions. Furthermore, a suitable HTS was established for each type of PDO in 96-well and 384-well plates. These assays were used to evaluate several molecular targeted agents and antibodies. Here, standard chemotherapeutics (paclitaxel and carboplatin) used for endometrial cancer were evaluated using F-PDOs derived from a patient who did not respond to paclitaxel and carboplatin. Accordingly, the cell growth inhibitory activity of paclitaxel and carboplatin against this PDO was weak (IC50: >10 µM). In addition, previous research has reported that the sensitivity of some F-PDOs to chemotherapeutic agents and molecular targeted agents is consistent with the clinical efficacy11,12,13. Finally, changes in the higher-order structure of the PDOs caused by anticancer agents were analyzed using a three-dimensional cell analysis system12,13. The results of the evaluation of anticancer agents using a PDO-based HTS are comparable with the clinical results obtained for these agents. Here, a protocol is presented for a simpler and more accurate HTS that can be used to evaluate the potency of anticancer agents and immunotherapy using the PDO models.
All experiments involving human-derived materials were performed under the Declaration of Helsinki and approved in advance by the ethics committee of Fukushima Medical University (approval numbers 1953 and 2192; approval dates March 18, 2020, and May 26, 2016, respectively). Written informed consent was obtained from all patients who provided the clinical specimens used in this study.
1. Culture of PDOs
NOTE: F-PDOs form cell clusters that exhibit a variety of heterogeneous morphologies and grow in suspension culture (Figure 1). Furthermore, F-PDOs can be cultured for more than 6 months and can be cryopreserved for future use.
2. Growth inhibition HTS
NOTE: The growth inhibitory activity of anticancer agents against PDOs is evaluated by measuring the intracellular ATP content, as shown in Figure 2. This step is performed using a commercially available cell viability assay kit (see Table of Materials).
3. HTS with a cell picking and imaging system for growth inhibition
NOTE: If there is a large deviation (when the coefficient of variation [CV] at assay is more than 20%) in the data using protocol 2, PDOs of a selected size may be seeded into 96-well or 384-well plates using a cell picking and imaging system (Figure 2). The protocol is the same as that described in steps 2.1 and 2.2 in the previous section. This step is performed using a commercially available cell picking and imaging system (see Table of Materials).
4. HTS for antibody-dependent cellular cytotoxicity
NOTE: This step is performed using a commercially available system (see Table of Materials), which is an electrical impedance measuring instrument. It is used to evaluate cytolysis of PDOs by antibody-dependent cellular cytotoxicity (ADCC) with monoclonal antibodies and natural killer (NK) cells (Figure 3). NK cells are produced from peripheral blood mononuclear cells using the NK cell production kit (see Table of Materials), following the manufacturer's instructions.
A highly accurate HTS was developed using PDOs and 384-well microplates to evaluate anticancer agents, and the development of an HTS for each PDO was previously reported10,11,12,13. The performance of the HTS was evaluated by calculating the CVs and the Z'-factor. The Z'-factor is a widely accepted method for validation of assay quality and performance, and the assay is suitable for HTS if this value is >0.514. The control datum points in the 384-well plate assay using RLUN007 showed little variability, with CV values of 5.8% and calculated Z'-factors of 0.83, as shown in Figure 4. These results indicate that this assay has high performance for HTS. To investigate the sensitivity of PDOs to anticancer agents using HTS, growth inhibition was assessed using RLUN007 treated with eight anticancer agents, specifically, epidermal growth factor receptor (EGFR) inhibitors (afatinib, erlotinib, gefitinib, lapatinib, osimertinib, and rociletinib) and paclitaxel, which are standard clinical treatments for non-small cell lung cancer, and mitomycin C as a positive control. The IC50 and AUC values of the anticancer agents for each PDO are shown in Figure 4. The RLUN007 showed high sensitivity (IC50 < 2 µM, AUC < 282) for all EGFR inhibitors and other anticancer agents. Sigmoid curves calculated for all the data indicated that the growth inhibitory activity of the anticancer agents could be accurately measured.
The cell picking and imaging system is used when the data vary significantly using the above methodology. The cell picking and imaging system, which picks cell clusters accurately without damaging them, allows for accurate HTS assays by aligning the cell cluster size to exclude cell debris from the assay system. When the system was not used, the CV value was 26.0% and the Z'-factor value was 0.23 (data not shown). However, the CV and Z'-factor values were improved at 6.4% and 0.81, respectively, using the system.
To investigate cytolysis of PDOs with ADCC activity using the electrical impedance measuring instrument, which monitors the number, morphology, and attachment of cells for a long duration, changes in impedance signals were assessed using RLUN007 treated with the antibodies (trastuzumab and cetuximab) and NK cells as effector cells in a 96-well plate. Compared with the control consisting of only target cells, the percent cytolysis increased with time. It reached 45% or 75% after 6 h at an E:T ratio of 1:1 (Figure 5A,C) or 2:1 (Figure 5B,D) without the antibodies. NK cell-mediated cytolysis using trastuzumab was approximately 60% and 90% at a ratio of 1:1 (Figure 5A, 1 µg/mL) and 2:1 (Figure 5B, 1 µg/mL), respectively, at 6 h. In contrast, cetuximab had a dose-dependent impact on NK cell-mediated cytolysis (Figure 5C,D). At the highest concentration of cetuximab, RLUN007 were destroyed at 90% and 100% at a ratio of 1:1 and 2:1, respectively (Figure 5C,D). The effect of trastuzumab was weaker than that of cetuximab, with only 60% cytotoxicity. These results indicate that the PDO assay system can evaluate ADCC activity using real-time impedance-based technology.
Figure 1: Critical points for PDO culture. (A) Color change in the medium. (B) Measurement of the quantity of PDOs from the pellet size by lining a centrifuge tube containing the PDOs with tubes marked at levels for 50-200 µL. (C) PDO density. Please click here to view a larger version of this figure.
Figure 2: Summary of the protocol used to create a high-throughput assay system using 384-well microplates. Please click here to view a larger version of this figure.
Figure 3: Summary of the protocol for high-throughput assay of ADCC activity. ADCC, antibody-dependent cellular cytotoxicity; NK, natural killer. Please click here to view a larger version of this figure.
Figure 4: High-throughput assay system for growth inhibition with anticancer agents. Dose-response curve of RLUN007 to anticancer agents. The minced PDOs were seeded in 384-well plates. These were treated for 6 days with ten different concentrations of anticancer agents (between 10 µM and 1.5 nM). The data represent the mean ± standard deviation of triplicate experiments. Please click here to view a larger version of this figure.
Figure 5: High-throughput assay for ADCC activity. (A,B) Trastuzumab. (C,D) Cetuximab. (A,C) A 1:1 ratio of RLUN007 to effector cells. (B,D) Cytolysis with a ratio of RLUN007: effector cells of 1:2. The activity was measured 12 h after the addition of the effector cells. The data are presented as the mean ± standard deviation of three replicate samples. ADCC, antibody-dependent cellular cytotoxicity. Please click here to view a larger version of this figure.
The unique characteristic of PDOs is that they are not enzymatically separated into single cells during culture or assay and maintain cell clusters in culture. Therefore, the number of cells cannot be counted accurately under a microscope. To solve this problem, the number of cells is determined visually by lining a centrifuge tube containing the cells with tubes marked with levels for 50-200 µL (Figure 1B). Furthermore, because it is difficult to measure the pellet volume of cell clusters cultured in a 25 cm2 flask visually, the time of passage was determined using the medium color change from red to yellow and the noticeable increase in single cells or debriscompared with the time of passaging as indicators (Figure 1A,C). This is the point of passaging for PDOs. The quantity of PDO pellets is measured visually after centrifugation at each medium change. When the pellet volume stops increasing, and the medium turns yellow on the day after medium replacement, the medium is considered to be saturated with density, and passaging is performed. The pellet volume is defined for each PDO. If the PDOs do not proliferate, the quantity of medium is changed from 80% to 50% at the time of medium exchange, and the density of PDOs is increased in the culture.
An HTS suitable for PDOs was developed. Its throughput is at least ten to twenty 384-well plates performed using one 75 cm2 flask of PDO, and the number of plates processed per day is at least 50. In addition, the results of the evaluation of various anticancer drugs by HTS using PDOs have already been reported.
When performing HTS, clogging of the mesh filter caused by mincing of the F-PDOs using the cell fragmentation and dispersion equipment is addressed initially by changing the mesh size of the filter to 100 µm. The next step is to reduce the volume of the PDO suspension applied to the glass vessel. When preparing test substance solutions for HTS, low-molecular-weight compounds are usually dissolved in dimethyl sulfoxide, and the antibodies are dissolved in phosphate-buffered saline. The appropriate solvent is used as the test substance, and the control data are obtained from the solvent used.
The following is a description of how to deal with variability in the assay data. If there is large variation in the data in the test using 384-well plates, the assay plate should be changed to a 96-well plate format. The PDO dilution factor (number of seeded cell clusters) is also examined after seeding the plate. Finally, the cell picking and imaging system can be used to select the size of PDOs for the assay. Before adding PDOs to the chamber, single cells and small cell clusters should be removed by low-speed centrifugation to be able to correctly recognize PDOs. If single cells or small cell clusters are visible after adding PDOs to the chamber, multiple dispersions can be performed to remove the single cells. Next, although the cell picking and imaging system has a function that allows the plate to be kept warm, this function is not used because of the evaporation of the culture medium when the system is working for a long period of time. Lastly, the volume of cell clusters is unknown because it is recognized by a planar image. Furthermore, if two or more PDOs overlap, a single PDO cannot be recognized correctly.However, it is possible to remove unwanted PDOs using the remove function by checking them on the scanned image after the move.
The electrical impedance measuring instrument is generally used for adherent target cancer cells to monitor changes in impedance during cell proliferation. Therefore, a change in the cell index of non-adherent PDOs is not detected. In an attempt to solve this problem, it is necessary to investigate the seeding conditions such as PDO density and enzymatic treatment (cell dissociation enzyme and treatment time) depending on the type of PDO. The wells in the plate must also be coated with an appropriate extracellular matrix for seeding PDOs. PDOs are seeded without enzymatic treatment, depending on the type of PDO. RLUN007 was used to measure the impedance by seeding the PDOs on a 96-well plate after dispersing them by enzymatic treatment. RLUN007 was treated with the cell culture dissociation reagent for 20 min at 37 °C to disperse the cells and attach them to the wells of a 96-well plate. Given that dissociated RLUN007 cells immediately form aggregates, it is desirable to seed on the plates just after filtration using a strainer. After transferring the cell suspension from the tube to a reservoir, the reservoir was gently moved from right to left two to three times and pipetted up and down five times before seeding onto the plate. The suspension was also mixed with each addition to the well. The plate was then placed in a biological safety cabinet for 30 min (for PDOs) or 15 min (for NK cells) to allow the cells to distribute evenly in the well. The second important point is that treatment with antibodies and NK cells should be timed to occur before the cell index reaches a plateau and the value is not less than 0.5. In the case of RLUN007, the optimal time to start the assay is 20-22 h after plating, and the cell number for seeding is 5 x 104 cells/well.
In general, the culture and assays for tumor organoids use extracellular matrices such as Matrigel to create tumor tissue scaffolds or enzymes such as trypsin and collagenase to disrupt the organoids3,4,5,6,7. The advantage of this method is that no extracellular matrix or enzymatic treatment is required during culture and assay (except for assays using the electrical impedance measuring instrument), which significantly reduces labor requirements and costs. Furthermore, this method is relatively easy to adapt to HTS assay systems and various measurement systems. However, the use of an extracellular matrix is desirable for some research purposes because it can act as a scaffold for cells and affect morphogenesis, differentiation, and homeostasis in tissues.
In this study, PDOs (RLUN007) with an EGFR mutation (L858R) that is clinically sensitive to EGFR inhibitors and high expression of the EGFR gene (data not shown) were used to evaluate EGFR inhibitors. It was demonstrated that the sensitivity of RLUN007 to EGFR inhibitors was higher than those of other lung cancer-derived F-PDOs13 (Figure 4). Thus, an HTS using PDOs, which retain the characteristics of tumor tissue, is superior for the evaluation of potential anticancer agents and presents opportunities for drug assessment and advances in personalized medicine. Although HTS is suitable for the initial screening of agents, it does not reproduce the tumor microenvironment and thus cannot evaluate the efficacy of drugs in vivo. Therefore, an in vitro system that can mimic human tumor tissue in vivo by co-culture with vascular endothelial cells and other stromal cells or organ-on-a-chip technology in the absence of animal models is now under development.
The authors have nothing to disclose.
We would like to thank the patients who provided the clinical specimens used in this research. This research is supported by grants from the Translational Research Program of Fukushima Prefecture.
384-well Ultra-Low Attachment Spheroid Microplate | Corning | 4516 | Plates for HTS |
40-µm Cell Strainer | Corning | 352340 | |
AdoptCell-NK kit | Kohjin Bio | 16030400 | Kit for NK cell production |
Cancer Cell Expansion Media plus | Fujifilm Wako Pure Chemical | 032-25745 | Medium for F-PDO |
ALyS505N-175 | Cell Science & Technology institute | 10217P10 | Medium for NK cells |
CELL HANDLER | Yamaha Motor | – | Cell picking and imaging system |
CellPet FT | JTEC | – | Cell fragmentation and dispersion equipment |
CellTiter-Glo 3D Cell Viability Assay | Promega | G9683 | Cell viability luminescent assay, intracellular ATP measuring reagent |
Echo 555 | Labcyte | – | Liquid handler |
EnSpire | PerkinElmer | – | Plate reader |
E-plate VIEW 96 | Agilent | 300601020 | Plates are specifically designed to perform cell-based assays with the xCELLigence RTCA System |
Fibronectin Solution | Fujifilm Wako Pure Chemical | 063-05591 | Plate coating for xCELLigence RTCA System |
F-PDO | Fujifilm Wako Pure Chemical or Summit Pharmaceuticals International | – | The F-PDO can be purchased from Fujifilm Wako Pure Chemicals or Summit Pharmaceuticals International |
Morphit software, version 6.0 | The Edge Software Consultancy | Biological data analysis software | |
Multidrop Combi | ThermoFisher Scientific | 5840300 | Cell suspension dispenser |
Precision Chamber | Yamaha Motor | JLE9M65W230 | Chamber for picking cell clusters using CELL HANDLER |
Precision Tip | Yamaha Motor | JLE9M65W300 | Micro tip for picking cell clusters using CELL HANDLER |
RLUN007 | Fujifilm Wako Pure Chemical or Summit Pharmaceuticals International | Lung tumor derived F-PDO | |
TrypLE Express | ThermoFisher Scientific | 12604021 | Cell culture dissociation reagent |
Ultra-Low Attachment 25 cm² Flask | Corning | 4616 | Culture flask for PDO |
Ultra-Low Attachment 75 cm² Flask | Corning | 3814 | Culture flask for PDO |
Vi-CELL XR Cell Viability Analyzer System | Beckman coulter | – | Cell viability analyzer |
xCELLigence immunotherapy software, version 2.3 | ACEA Bioscience | – | Analysis software for xCELLigence RTCA System |
xCELLigence RTCA System | ACEA Bioscience | – | Electrical impedance measuring instrument for cytolysis |