We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Short CommunicationOpen Accesscc iconby iconnc iconnd icon

International evaluation study of a highly efficient culture assay for detection of residual human pluripotent stem cells in cell therapies

    Takeshi Watanabe

    *Author for correspondence:

    E-mail Address: takeshi.watanabe@takeda.com

    Drug Safety Research & Evaluation, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan

    ,
    Satoshi Yasuda

    Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan

    ,
    Connie L Chen

    Health & Environmental Sciences Institute, 740 Fifteenth Street NW, Suite 600, Washington, DC 20005, USA

    ,
    Louise Delsing

    CVRM Safety, Clinical Pharmacology & Safety Science, R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden

    ,
    Mick D Fellows

    CVRM Safety, Clinical Pharmacology & Safety Science, R&D, AstraZeneca, Darwin Building 310, Milton Science Park, Cambridge, CB4 OWG, UK

    ,
    Gabor Foldes

    National Heart & Lung Institute, Imperial College London, London, W120NN, UK

    Current address, BioPharmaceuticals R&D, AstraZeneca, Milstein Building, Granta Park, Cambridge, CB21 6GH, UK

    ,
    Shinji Kusakawa

    Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan

    ,
    Lucilia Pereira Mouriès

    Health & Environmental Sciences Institute, 740 Fifteenth Street NW, Suite 600, Washington, DC 20005, USA

    &
    Yoji Sato

    Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan

    Published Online:https://doi.org/10.2217/rme-2022-0207

    Abstract

    Aim & methods: The Health and Environmental Sciences Institute Cell Therapy-TRAcking, Circulation & Safety Technical Committee launched an international, multisite study to evaluate the sensitivity and reproducibility of the highly efficient culture (HEC) assay, an in vitro assay to detect residual undifferentiated human pluripotent stem cells (hPSCs) in cell therapy products. Results: All facilities detected colonies of human induced pluripotent stem cells (hiPSCs) when five hiPSCs were spiked into 1 million hiPSC-derived cardiomyocytes. Spiking with a trace amount of hiPSCs revealed that repeatability accounts for the majority of reproducibility while the true positive rate was high. Conclusion: The results indicate that the HEC assay is highly sensitive and robust and can be generally applicable for tumorigenicity evaluation of hPSC-derived cell therapy products.

    Human pluripotent stem cells (hPSCs) have the potential to revolutionize regenerative medicine. However, the potential risk of tumorigenicity due to residual undifferentiated hPSCs persisting in the final product remains a particular concern, because hPSCs are intrinsically tumorigenic and can form teratomas [1,2]. The establishment of a robust and internationally harmonized methodology to evaluate the contamination of a cell therapy product with residual undifferentiated hPSCs is needed not only for product developers but also for regulatory authorities and patients [1].

    A minimum number of hPSCs sufficient to induce teratoma in immunodeficient animals varies and depends on the administration procedure. For instance, the 50% tumor producing dose – that is, a minimal number of transplanted cells that generated teratomas in 50% of animals – is reported to be about 100 human induced pluripotent stem cells (hiPSCs) when cells were subcutaneously injected with Matrigel into severely immunodeficient mice [3]. On the other hand, 1 × 105 human embryonic stem cells or 2 × 105 hiPSCs were reported to be needed to induce teratomas when cells were injected intramyocardially [4] or intravenously [5], respectively. In clinical applications, it is presumed that approximately 109 cells need to be transplanted into a patient to treat heart failure [6], and for chimeric antigen receptor-based cell therapies, 1010 cells may be administered [7]. The recommended sensitivity for the detection of residual undifferentiated hPSCs would be different for each product and depends on the administration route/environment of the injection site and the intended clinical dose level. However, for instance, if a higher clinical dose level is intended, leveraging sensitive in vitro assays will be required, considering the limitation of maximum feasible dose in in vivo studies.

    In response to this emerging safety issue, the Health and Environmental Sciences Institute (HESI) Cell Therapy-TRAcking, Circulation & Safety (CT-TRACS) Technical Committee launched an international project to evaluate in vitro testing methods focusing on the detection of residual undifferentiated hPSCs [1,8] in collaboration with a Japanese experimental public–private partnership initiative (Multisite Evaluation Study on Analytical Methods for Non-clinical Safety Assessment of Human-Derived Regenerative Medical Products [MEASURE]).

    Several in vitro testing methods have been reported for the detection of residual hPSCs: flow cytometry [9], the Glycostem test [10], quantitative reverse transcription PCR [9], droplet digital PCR [7,11,12] and highly efficient culture (HEC) assay [13,14]. In regard to the sensitivity of testing methods, the limit of detection (LOD) of assays is relatively high for flow cytometry and the Glycostem test (0.05–0.1%) [9,10]. On the other hand, PCR-based assays are highly sensitive (LOD: 0.0003–0.001%) [7,9,11,12], and recently an even more sensitive methodology based on reverse transcription loop-mediated isothermal amplification was reported (LOD: 0.00002%) [15]. However, some types of hiPSC-derived cells are known to retain the expression of pluripotency marker genes during the differentiation process [12,13]; therefore, the marker genes for these molecular biology-based assays need to be carefully validated prior to the assay for each cell therapy product (CTP) to avoid false positive results. The HEC assay is a simple, culture-based assay that can directly detect hPSCs by identifying hPSC-derived colonies under culture condition that favor the growth of hPSCs [13]. Its high sensitivity and robustness were confirmed and compared with PCR-based assays in a multisite study previously organized by MEASURE (LOD: 0.001%) [14]. That study further demonstrated that a detection sensitivity of 0.00002% could be achieved by adding a step consisting of enriching the targeted undifferentiated hPSCs by utilizing a magnetic-activated cell sorting system [14].

    Considering the wide variety of hPSC-derived CTPs (e.g., direction and degree of differentiation), it is important that several sensitive methodologies for detecting residual hPSCs be validated at the international level and available to be leveraged by product developers based on which will best serve their specific use, context and needs. Therefore, CT-TRACS launched an international collaborative project for assay validation focusing on two methods – the HEC assay and ddPCR-based assay. In the present study, the authors evaluated the sensitivity and interlaboratory variability of the HEC assay at multiple facilities according to International Organization for Standardization (ISO) standards. This international, multisite study serves to confirm previous results published by the MEASURE program in Japan. The performance of the HEC assay was evaluated in the previous MEASURE study by spiking hiPSCs into somatic cells such as human mesenchymal stromal cells and human peripheral blood mononuclear cell-derived T cells, but not into hPSC-derived cells. Therefore, hiPSC-derived cardiomyocytes were selected in the present study, since they more closely mimic hPSC-derived CTPs.

    Materials & methods

    Cells

    For hiPSCs, Cellartis human iPS cell line 18 (ChiPSC18) cells were purchased from Takara Bio, Inc., and were maintained with the Cellartis DEF-CS culture system (Takara Bio, Inc.) according to the manufacturer's instructions. The cultured ChiPSC18 (passages 19–22) were frozen in Stem-Cellbanker (Zenoaq Resource Co. Ltd) and cryopreserved in liquid nitrogen. The frozen hiPSCs were thawed and used for experiments after two to four passages. For hiPSC-derived cardiomyocytes, iCell cardiomyocytes (#C1106, Lot#103700) were purchased from FUJIFILM Cellular Dynamics, Inc., and were used for experiments immediately after thawing using iCell cardiomyocyte plating medium (#M1001; FUJIFILM Cellular Dynamics, Inc.) according to the manufacturer's instructions.

    HEC assay

    After thawing, the iCell cardiomyocytes were suspended with Essential 8 Flex medium (Thermo Fisher Scientific) containing 50 nM Chroman 1 (#HY-15392; MedChem Express), 5 μM emricasan (#S7775; Selleckchem), polyamine supplement (#P8483, 1:1000 dilution; Sigma-Aldrich) and 0.7 μM trans-ISRIB (#5284, Tocris; E8F+CEPT [16] medium). These cells were seeded in six-well plates coated with laminin-521 (BioLamina AB) [13] at a cell density of 1 × 106 per well after checking the viability by cell counter, Countess automated cell counter (Thermo Fisher Scientific) or LUNA-FL dual fluorescence cell counter (Logos Biosystems) or manually using a counting chamber. The ChiPSC18 cells were dissociated into single cells with ×1 TrypLE Express and were suspended with E8F+CEPT medium. After confirming the viability (>∼80%) in the same manner, 5 × 101 cells/ml of the ChiPSC18 cell suspension was carefully prepared by serial dilution from 5 × 105 cells/ml of the ChiPSC18 cell suspension at a ratio of 10. The 0.1 or 0.3 ml of 5 × 101 cells/ml of the ChiPSC18 cell suspension was added to the wells previously seeded with iCell cardiomyocytes to spike 5 (0.0005%) or 15 (0.0015%) hiPSCs into 1 × 106 iCell cardiomyocytes. Four wells were prepared for each concentration, and wells in which only 1 × 106 iCell cardiomyocytes were seeded (nonspiked sample) were prepared for each experiment (n = 1 or 2). Three days after seeding, the media were replaced with Essential 8 Flex medium, and after a total of 7 days of incubation at 37°C in 5% CO2, the wells were fixed and stained using a Vector Blue alkaline phosphatase (ALP) substrate kit (Vector Laboratories, Inc.) according to a previously reported method [14]. 7 days in culture was sufficient to detect the hiPSC colonies, considering that the doubling time of hiPSC when cultured with Essential 8 medium on laminin-521-coated wells was approximately 21.5 h [13]. The ALP-positive hiPSC colonies were counted manually by two operators under a microscope, and the colony formation rate (the ratio of the total number of colonies to the number of spiked hiPSCs) was calculated. The experiment was conducted at four facilities (facilities A, B, C and D) and was repeated three-times at each facility except for facility A, where the experiment was repeated two times.

    Statistical analysis

    The true positive rate (TPR) for detection of hiPSC colonies with the HEC assay was calculated based on the assumption that the number of hiPSC colonies formed on wells follows a Poisson distribution, as previously reported [14]. The probability of no hiPSC colony formation (k = 0; i.e., the false negative rate [FNR]) in a single experiment was expressed using the mean number of actual hiPSC colonies obtained by repeated experiments (λ = n):

    Psgl (X=0)=en×n0/0!=en

    The FNR of a set of two experiments was calculated as the probability of no hiPSC colony formation in a single experiment (e-n) raised to the second power:

    Pdbl (X=0)=en×2=e2n

    The probability of hiPSC colony formation (k >0; i.e., TPR) in a single experiment and of a set of three experiments led to the calculation 1 - FNR:

    Psgl (X>0)=1Psgl (X=0)Pdbl (X>0)=1Pdbl (X=0)

    In this study, use of the term “TPR” was identical to “clinical/diagnostic sensitivity” as defined in ISO/TS 17822-1:2014 [17], whereas the term ‘sensitivity’ was used in a broad sense.

    The repeatability and reproducibility of the number of hiPSC colonies were statistically analyzed, according to ISO 5725 [18,19]. The repeatability is defined as precision under conditions where independent test results are obtained with the same method on identical test items in the same laboratory by the same operator using the same equipment within short intervals of time, and reproducibility is defined as precision under conditions where test results are obtained with the same method on identical test items in different laboratories with different operators using different equipment [20]. Variability from a collaborative validation study can be modeled as:

    SR2=SL2+Sr2

    where sR2 is the reproducibility variance, sL2 is the between-laboratory variance and sr2 is the repeatability variance.

    The value of sr2 is calculated by

    Sr2=i=1p(ni1)Si2i=1p(ni1)

    where p is the number of laboratories, ni is the number of test results in the i-th laboratory and si is the standard deviation of the test results in the i-th laboratory.

    The value of sL2 is calculated by:

    SL2=Sm2Sr2/n^,Sm2>Sr2/n^0,Sm2Sr2/n^

    where:

    Sm2=1p1i=1pni(y¯im^)2n^
    n^=1p1[i=1pnii=1pni2i=1pni]
    m^=i=1pniy¯ii=1pni
    and y¯i is the mean of results in the i-th laboratory.

    To identify outliers, Mandel's k and h statistics, Cochran's statistics and Grubbs' statistics were used as consistency tests. The critical values of less than 5% were used as criteria for outliers.

    Results

    For all facilities, well-defined ALP-positive hiPSC colonies could be detected in both 5 (0.0005%) and 15 (0.0015%) hiPSC-spiked conditions (Figure 1). The mean number of hiPSC colonies showed a slight variation among the facilities. Colonies for 5 and 15 hiPSC-spiked conditions were 2.7 (2.1–3.3) and 7.9 (5.1–12.1), respectively (Table 1), and revealed a good dependency on the number of spiked hiPSCs. The colony formation rate calculated as the ratio of the total number of colonies to the number of spiked hiPSCs was similar between the 5 and 15 hiPSC-spiked conditions, and the overall colony formation rate was confirmed to be 54% (42–67%) for the 5 hiPSC-spiked condition and 53% (34–81%) for the 15 hiPSC-spiked condition (Table 1). For nonspiked samples, no colonies were observed in any wells at any of the facilities.

    Figure 1. Representative images of alkaline phosphatase-positive human induced pluripotent stem cell colonies from each facility.

    Facility A (A). Facility B (B). Facility C (C). Facility D (D). Scale bars, 250 μm.

    Table 1. Results of the highly efficient culture assay.
    Number of spiked hiPSCs (%)5 (0.0005)
     Number of hiPSC coloniesColony formation rate (%)True positive rate for detecting hiPSC colonies (%)
       SingleTwo – repeated
    Facility A2.6 ± 0.253 ± 49399
    Facility B2.1 ± 0.942 ± 198898
    Facility C3.3 ± 0.967 ± 1896100
    Facility D2.7 ± 1.253 ± 2593100
    Overall2.7 ± 0.954 ± 1992 ± 499 ± 1
    Number of spiked hiPSCs (%)15 (0.0015)
     Number of hiPSC coloniesColony formation rate (%)True positive rate for detecting hiPSC colonies (%)
       SingleTwo – repeated
    Facility A6.9 ± 3.046 ± 20100100
    Facility B5.1 ± 1.234 ± 899100
    Facility C12.1 ± 2.581 ± 17100100
    Facility D7.3 ± 0.948 ± 6100100
    Overall7.9 ± 3.353 ± 22100 ± 0100 ± 0

    Five or 15 hiPSCs were spiked into 1 × 106 iCell cardiomyocytes and were cultured in E8F+CEPT/LN521. After 7 days of incubation, the alkaline phosphatase-positive hiPSC colonies were counted manually under a microscope, and the colony formation rate was calculated as the ratio of the total number of colonies to the number of spiked hiPSCs. These experiments were conducted at four facilities (Facilities A, B, C and D), and the values (mean ± standard deviation) were obtained from two (Facility A) or three (other facilities) quadruplicate experiments. The true positive rate for detecting hiPSC colonies at four facilities (mean ± standard deviation) was calculated with a single and two repeated experiments.

    hiPSC: Human induced pluripotent stem cell.

    To confirm the TPR needed to detect spiked hiPSCs under the experimental conditions, the authors used the Poisson distribution formula and calculated the probability of detecting hiPSC colonies, with one or more colony present. The TPR of a single experiment varied slightly between facilities when spiking with five (0.0005%) hiPSCs (88–96%), but the mean value at all the facilities was sufficiently high (92%), and spiking with 15 (0.0015%) hiPSCs gave a TPR of 99% or higher at all facilities (Table 1). Two repeated experiments increased the TPR for detection of hiPSC colonies, and a TPR of 100% was also calculated in most facilities when spiking with five cells (Table 1).

    The statistical analysis on repeatability and reproducibility of the number of hiPSC colonies revealed that the standard deviation of repeatability (sr) and reproducibility (sR) was 1.0 and 1.0 for the 5 hiPSC-spiked condition and 1.9 and 3.5 for the 15 hiPSC-spiked condition (Table 2). The coefficients of variation (CV) of repeatability and reproducibility were both 36% for the 5 hiPSC-spiked condition and 24% and 44% for the 15 hiPSC-spiked condition, and the ratio of sR/sr was 1.00 and 1.80 for each condition, respectively (Table 2). There were no excluded outliers from the consistency test for both the 5 and 15 hiPSC-spiked conditions.

    Table 2. The results of statistical analysis on repeatability and reproducibility.
    Number of spiked human induced pluripotent stem cells (%)5 (0.0005)15 (0.0015)
    General mean of colony number m^2.77.9
    Repeatability standard deviation Sr1.01.9
    Reproducibility standard deviation SR1.03.5
    Repeatability coefficient of variation (Sr)36%24%
    Reproducibility coefficient of variation (SR)36%44%
    γ = SR/Sr1.001.80
    Number of excluded outliers00
    Number of measurement values1111
    Number of valid laboratories44

    Discussion

    During the manufacturing process of hPSC-derived CTPs, most undifferentiated hPSCs are likely eliminated because most differentiation protocols are not optimal for the survival of undifferentiated hPSCs [21]. However, efforts to purify the final product and to minimize its contamination with undifferentiated hPSCs are critical to mitigate the potential tumorigenicity risks of hPSC-derived CTPs [1]. The HEC assay is a simple and sensitive culture-based assay and has, in fact, been employed in the tumorigenicity assessment of hPSC-derived CTPs, under the requirements of the regulatory agencies [22].

    In the present study, the authors evaluated the sensitivity of the HEC assay at international and multisite levels and confirmed that all four facilities in the study could detect hiPSC colonies in wells in which 5 hiPSCs were spiked into 1 × 106 iCell cardiomyocytes (LOD: 0.0005%). The mean number of hiPSC colonies showed an increase depending on the number of spiked hiPSCs between the 5 and 15 hiPSC-spiked conditions. The overall colony formation rate was constant among the facilities and was higher than 50% regardless of the number of spiked hiPSCs. Residual undifferentiated hPSCs in the final product are considered ‘contaminants’ or ‘impurities’ to the final differentiated CTP [1]. Therefore, it is important to assess both the LOD and the specificity of the assay methods to detect such residual cells, as outlined in International Conference on Harmonisation (ICH) Q2 (R1) [23]. In terms of the specificity of the assay, the basis of the HEC assay is to directly detect the residual hPSCs by identifying the colony derived from each residual hPSC with ALP activities. Actually, no colonies stained with an ALP substrate were detected in nonspiked samples at the four facilities, which indicates no false positive signals in this study and results in high specificity of the HEC assay. Additionally, its specificity was previously secured by visually identifying the colonies positively stained with antibodies and a lectin against several hPSC markers [14] and will also be confirmed with spiked samples prepared at each assay for the determination of the LOD [22]. For confirmation of the validity of LOD in the present study, the authors calculated the TPR based on the FNR, which is the probability of no colony formation in hiPSC-spiked samples. As a result, the mean value of TPR for detecting 0.0005% hiPSC contamination was 92% even with a single experiment and approached 100% with two repeated experiments in all facilities. These results indicate the high sensitivity and the robustness of the HEC assay to detect residual hiPSCs and that a negative result obtained from a product with an unknown contamination rate means that the hiPSC content is less than 0.0005% at a probability of about 90% or higher even with a single experiment. If the experiment were repeated, the probability would be almost 100%. This suggests that the assay appears to be capable of an even lower detection limit, but that would have to be experimentally verified.

    Interestingly, the sensitivity with which the authors could detect undifferentiated cells in a differentiated population is highly superior to the sensitivity reported from in vivo teratoma assays. The results show that at least 5 hiPSC cells in a 1 million dose can be detected. While there are conflicting reports on how many hPSCs are needed to form a teratoma in vivo, the lowest reported numbers from Kanemura et al. [3] and Yasuda et al. [24] showed that the 50% tumor producing dose was 132 and 631, respectively, when 201B7 hiPSC line was subcutaneously transplanted with Matrigel into NOD/Shi-scid IL2Rγnull mice. The authors of the present study speculate that with further validation and evaluation of the HEC assay, it will be possible to take full advantage of this increased sensitivity, providing more precise safety predictions of hPSC-derived CTPs and reduced use of animals in preclinical testing.

    The statistical analysis on the number of hiPSC colonies to estimate the precision of HEC methodology revealed that the CV values of repeatability (CV [sr]) and reproducibility (CV [sR]) were 36% and 36% for the five hiPSC-spiked condition and 24% and 44% for the 15 hiPSC-spiked condition, respectively. These results show the precision of the HEC assay within an international, multisite study for the first time. Repeatability is used to evaluate the ability of the method to generate similar results with the successive measurements. As expected, a higher number of hiPSCs spiked into iCell cardiomyocytes lowered the CV value of repeatability. Although the CV value of reproducibility with 15-cell spiking was higher than that of 5-cell spiking, the sR/sr ratio was quite improved with 15-cell spiking. It should be noted that sR was calculated as the same value of sr (i.e., sR/sr is 1) based on the formula just because repeatability variance was very high in the 5-cell spiking condition. These results would help clarify the value and limitations of the HEC assay for evaluating CTPs. The CV (sr) and CV (sR) values were the same for the 5 hiPSC-spiked conditions; therefore, the main part of the reproducibility was considered to be repeatability. In addition, an increase in the ratio of sR to sr showed a lower contribution of repeatability to reproducibility when more hiPSCs (15 cells) were spiked. This indicates that possible variation in the actual number of spiked hiPSCs may cause the variances of repeatability and reproducibility, especially when a smaller number of hiPSCs is spiked. Thus, accurate execution of both preparation of hiPSC cell suspension and spiking of these cells into the samples is essential for the sensitivity and precision of the HEC assay.

    Since the sensitivity of the HEC assay is determined by the hPSC-spiked sample, the efficiency of the colony formation from each spiked hPSC is highly important to lower the detection limit. Additionally, insufficient efficiency of colony formation may cause false negative results when hPSC-derived CTPs are evaluated; therefore, securing a higher colony formation rate is essential for this assay. It is well known that enzymatic dissociation causes cell death by apoptosis and anoikis [25–27], and a landmark paper has reported that the ρ-associated coiled coil-forming protein serine/threonine kinase inhibitor Y-27632 improves the survival of hPSC [28]. Recently, Chen et al. reported that the combination of chroman 1, emricasan, polyamines and trans-ISRIB (CEPT) enhanced cell survival of hPSCs by simultaneously blocking several stress mechanisms, and its effects on cytoprotection of hPSCs were superior to that of Y-27632 [16]. Therefore, CEPT, instead of Y-27632, was used in the present study. The mean values of colony formation rate (54% ± 19%, mean ± standard deviation, under the five hiPSC-spiked condition) was higher than that in the MEASURE study using Y-27632 (31% ± 21%, mean ± standard deviation, under the 10 hiPSC [ChiPSC18]-spiked condition [14]), indicating that CEPT treatment has beneficial effects on the efficiency of colony formation in the HEC assay, though further experiments, especially experiments using a head-to-head comparison, are necessary.

    In the present study, the experiments were performed using hiPSC-derived cardiomyocytes only. However, it has been reported that the HEC assay can be applied for various cell types, including human mesenchymal stromal cells [13,14], human neurons [13] and hPSC-derived neural stem cells [22]. Furthermore, the MEASURE study using human peripheral blood mononuclear cell-derived T cells indicates that the HEC assay can also be applied for suspension cell products [14] with some modifications (the assay uses adherent cultures and should be optimized for cell suspensions). Therefore, it is expected that the HEC assay can be broadly applied for hPSC-derived CTPs with, perhaps, the exception of the CTPs that inhibit survival of undifferentiated hPSCs such as hiPSC-derived differentiated retinal pigment epithelial cells that secret pigment epithelium-derived factor and induce apoptotic cell death of hPSCs [29].

    Conclusion

    The HESI CT-TRACS international, multisite study using hiPSC-derived cardiomyocytes was able to prove the high sensitivity of the HEC assay (LOD: 0.0005%) and confirm that the mean TPR for detecting 0.0005% hiPSC contamination was sufficiently high (92%) even when looking at partial data from a single experiment. The findings indicate the robustness of the HEC assay for the evaluation of product contamination with residual undifferentiated hPSCs and that this in vitro methodology can be generally applicable for the evaluation of the potential tumorigenicity risk of hPSC-derived CTPs. It is considered to be a valuable in vitro approach to be included in a ‘toolbox’ for tumorigenicity assessment, which will contribute to the future standardization of tumorigenicity risk assessment of hPSC-derived CTPs.

    Summary points
    • Human pluripotent stem cells (hPSCs) are intrinsically tumorigenic and can form teratomas.

    • The potential risk of residual undifferentiated hPSCs remains a particular concern of hPSC-derived products.

    • Highly efficient culture (HEC) assay is an in vitro assay to detect residual undifferentiated hPSCs.

    • The Health and Environmental Sciences Institute, Cell Therapy-TRAcking, Circulation & Safety Technical Committee launched an international, multisite study to evaluate the HEC assay.

    • The study confirmed that the HEC assay is highly sensitive and robust.

    Acknowledgments

    The authors gratefully acknowledge the continuous support of HESI's Cell-Therapy: TRAcking, Circulation & Safety Technical Committee and the Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-derived Product, the Forum for Innovative Regenerative Medicine and Thusharika Kodagoda and Hatsue Furukawa for their expert technical support of the project.

    Financial & competing interests disclosure

    This work was partly supported by the Japan Agency for Medical Research and Development (AMED) under grant number JP20mk0104176 and 20mk0104177 and the Health and Environmental Sciences Institute (HESI) Cell Therapy-TRAcking, Circulation, and Safety (CT-TRACS) Technical Committee. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    No writing assistance was utilized in the production of this manuscript.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Sato Y, Bando H, Di Piazza M et al. Tumorigenicity assessment of cell therapy products: the need for global consensus and points to consider. Cytotherapy 21(11), 1095–1111 (2019). •• Excellent review paper for currently available in vivo and in vitro testing methods for tumorigenicity evaluation of cell therapy products.
    • 2. Yamanaka S. Pluripotent stem cell-based cell therapy – promise and challenges. Cell. Stem Cell. 27(4), 523–531 (2020).
    • 3. Kanemura H, Go MJ, Shikamura M et al. Tumorigenicity studies of induced pluripotent stem cell (iPSC)-derived retinal pigment epithelium (RPE) for the treatment of age-related macular degeneration. PLOS ONE 9(1), e85336 (2014).
    • 4. Kooreman NG, Wu JC. Tumorigenicity of pluripotent stem cells: biological insights from molecular imaging. J. R. Soc. Interface. 7(Suppl. 6), S753–S763 (2010).
    • 5. Bedel A, Beliveau F, Lamrissi-Garcia I et al. Preventing pluripotent cell teratoma in regenerative medicine applied to hematology disorders. Stem Cells Transl. Med. 6(2), 382–393 (2016).
    • 6. Tohyama S, Fukuda K. Future treatment of heart failure using human iPSC-derived cardiomyocytes. In: Etiology and Morphogenesis of Congenital Heart Disease: From Gene Function and Cellular Interaction to Morphology. Nakanishi TMarkwald RRBaldwin HSKeller BBSrivastava DYamagishi H (Eds). Springer, Tokyo, Japan (2016).
    • 7. Chung L, Cogburn LA, Sui L, Dashnau JL. Development of an induced pluripotent stem cell-specific microRNA assay for detection of residual undifferentiated cells in natural killer cell therapy products. Cytotherapy 24(7), 733–741 (2022). • Excellent paper that introduces sensitive in vitro methodology for the detection of residual undifferentiated pluripotent stem cells in the product.
    • 8. Bando H, Di Piazza M, Gowing G et al. Safety of cell therapy products: in-vitro methods to assess the tumorigenicity of human cell-based therapeutic products. Cytotherapy 22(5), S42 (2022).
    • 9. Kuroda T, Yasuda S, Kusakawa S et al. Highly sensitive in vitro methods for detection of residual undifferentiated cells in retinal pigment epithelial cells derived from human iPS cells. PLOS ONE 7(5), e37342 (2012).
    • 10. Tateno H, Onuma Y, Ito Y et al. A medium hyperglycosylated podocalyxin enables noninvasive and quantitative detection of tumorigenic human pluripotent stem cells. Sci. Rep. 4, 4069 (2014).
    • 11. Kuroda T, Yasuda S, Matsuyama S et al. Highly sensitive droplet digital PCR method for detection of residual undifferentiated cells in cardiomyocytes derived from human pluripotent stem cells. Regen. Ther. 2, 17–23 (2015). • Excellent paper that introduces sensitive in vitro methodology for the detection of residual undifferentiated pluripotent stem cells in the product.
    • 12. Sekine K, Tsuzuki S, Yasui R et al. Robust detection of undifferentiated iPSC among differentiated cells. Sci. Rep. 10, 10293 (2020).
    • 13. Tano K, Yasuda S, Kuroda T, Saito H, Umezawa A, Sato Y. A novel in vitro method for detecting undifferentiated human pluripotent stem cells as impurities in cell therapy products using a highly efficient culture system. PlOS ONE 9(10), e110496 (2014). • Excellent paper that introduces sensitive in vitro methodology for the detection of residual undifferentiated pluripotent stem cells in the product.
    • 14. Watanabe T, Yasuda S, Kusakawa S et al. Multisite studies for validation and improvement of a highly efficient culture assay for detection of undifferentiated human pluripotent stem cells intermingled in cell therapy products. Cytotherapy 23(2), 176–183 (2021). • Excellent paper that introduces sensitive in vitro methodology for the detection of residual undifferentiated pluripotent stem cells in the product.
    • 15. Yasui R, Matsui A, Sekine K, Okamoto S, Taniguchi H. Highly sensitive detection of human pluripotent stem cells by loop-mediated isothermal amplification. Stem. Cell. Rev. Rep. 18(8), 2995–3007 (2022). • Excellent paper that introduces sensitive in vitro methodology for the detection of residual undifferentiated pluripotent stem cells in the product.
    • 16. Chen Y, Tristan CA, Chen L et al. A versatile polypharmacology platform promotes cytoprotection and viability of human pluripotent and differentiated cells. Nat. Methods. 18, 528–541 (2021). • Excellent paper that introduces novel methodology to enhance the viability of human pluripotent stem cells. This new methodology is considered to be highly significant to improve the robustness/sensitivity of the highly efficient culture assay.
    • 17. ISO/TS 17822-1:2014. In vitro diagnostic test systems – qualitative nucleic acid-based in vitro examination procedures for detection and identification of microbial pathogens – part 1: general requirements, terms and definitions (December 2014) (2014). (Accessed 30 June 2020). www.iso.org/standard/60601.html
    • 18. International Organization for Standardization (ISO). Accuracy (trueness and precision) of measurement methods and results – part 2: basic method for the determination of repeatability and reproducibility of a standard measurement method. ISO 5725-2:2019; Geneva, Switzerland (2019). www.iso.org/standard/69419.html
    • 19. Luping T, Schouenborg B. Methodology of inter-comparison tests and statistical analysis of the test results – Nordtest project no. 1483-1499. www.diva-portal.org/smash/get/diva2:962159/FULLTEXT01.pdf
    • 20. International Organization for Standardization (ISO). Statistics – vocabulary and symbols – part 1: general statistical terms and terms used in probability. ISO, Geneva, Switzerland, 3534–1 (2006). www.iso.org/standard/40145.html
    • 21. Peterson SE, Garitaonandia I, Loring JF. The tumorigenic potential of pluripotent stem cells: what can we do to minimize it? BioEssays 38(Suppl. 1), S86–S95 (2016).
    • 22. Garitaonandia I, Gonzalez R, Christiansen-Weber T, Abramihina T, Poustovoitov M, Noskov A et al. Neural stem cell tumorigenicity and biodistribution assessment for phase I clinical trial in Parkinson's disease. Sci. Rep. 6, 34478 (2016).
    • 23. International Conference on Harmonization. Q2(R1): Validation of analytical procedures: text and methodology. (Accessed 26 December 2022). https://database.ich.org/sites/default/files/Q2%28R1%29%20Guideline.pdf
    • 24. Yasuda S, Kusakawa S, Kuroda T et al. Tumorigenicity-associated characteristics of human iPS cell lines. PL0S ONE 13(10), e0205022 (2018).
    • 25. Chen G, Hou Z, Gulbranson DR, Thomson JA. Actin–myosin contractility is responsible for the reduced viability of dissociated human embryonic stem cells. Cell. Stem. Cell. 7(2), 240–248 (2010).
    • 26. Ohgushi M, Matsumura M, Eiraku M et al. Molecular pathway and cell state responsible for dissociation-induced apoptosis in human pluripotent stem cells. Cell. Stem. Cell. 7(2), 225–239 (2010).
    • 27. Xu Y, Zhu X, Hahm HS et al. Revealing a core signaling regulatory mechanism for pluripotent stem cell survival and self-renewal by small molecules. Proc. Natl Acad. Sci. USA 107(18), 8129–8134 (2010).
    • 28. Watanabe K, Ueno M, Kamiya D et al. A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat. Biotechnol. 25, 681–686 (2007).
    • 29. Kawamata S, Kanemura H, Sakai N, Takahashi M, Go MJ. Design of a tumorigenicity test for induced pluripotent stem cell (iPSC)-derived cell products. J. Clin. Med. 4(1), 159–171 (2015).