1
|
Fatima E, Gautam Y, Thapa B, Das R, Singh A, Trivedi L, Singh P, Singh K, Bhatt D, Vasudev PG, Gupta A, Chanda D, Bawankule DU, Shanker K, Khan F, Negi AS. 5H-benzo[c]fluorene derivative exhibits antiproliferative activity via microtubule destabilization. Bioorg Chem 2024; 153:107891. [PMID: 39454495 DOI: 10.1016/j.bioorg.2024.107891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/20/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024]
Abstract
Present study aimed at a single component cyclization of 2-benzylidene-1-tetralones for the preparation of 5H-benzo[c]fluorenes and their antiproliferative activity. This ring closure reaction underwent via reductive cyclization in the presence of a sodium borohydride-aluminium chloride system. Ten diverse 5H-benzo[c]fluorene derivatives were prepared and evaluated for antiproliferative activity against three human cancer cell lines by SRB assay. Four of these benzofluorenes exhibited significant antiproliferative effect with an IC50 < 10.75 µM. The best representative compound 21, exhibited IC50 against K562 leukemic cells at 3.27 µM in SRB assay and 7.68 µM in Soft agar colony assay. It exhibited a microtubule destabilization effect in tubulin kinetics and inhibited 82.9 % microtubule polymer mass at 10 µM concentration in Protein Sedimentation assay (Microtubule). Compound 21 exerted G0/G1 phase arrest in cell division cycle analysis in K562 cells. It also induced apoptosis in K562 cells via activation of Caspase cascade pathway. Furthermore, compound 21 also possessed anti-inflammatory activity by inhibiting TNF-α and IL-6 moderately. It exhibited significant in vivo efficacy and reduced K562 tumour in xenograft mice by 47 % at an 80 mg/kg oral dose. Further, it was found to be safe and well tolerable up to 1000 mg/kg in Swiss albino mice. Compound 21 needs to be optimized for better in vivo efficacy in rodent models for further development.
Collapse
Affiliation(s)
- Eram Fatima
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India
| | - Yashveer Gautam
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow 226015, India; Department of Chemistry, Pandit Prithi Nath PG College, 96/12 Mahatma Gandhi Marg, Kanpur 208001, India
| | - Barsha Thapa
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow 226015, India
| | - Ranjana Das
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow 226015, India
| | - Amrita Singh
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India
| | - Laxmikant Trivedi
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India
| | - Palak Singh
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India
| | - Kavita Singh
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow 226015, India
| | - Divya Bhatt
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India
| | - Prema G Vasudev
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India
| | - Atul Gupta
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India
| | - Debabrata Chanda
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India
| | - Dnyaneshwar U Bawankule
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India
| | - Karuna Shanker
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India
| | - Feroz Khan
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India
| | - Arvind S Negi
- CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India.
| |
Collapse
|
2
|
Bando K, Kusakawa S, Adachi H, Yamamoto M, Iwata M, Kitanaka A, Ogimura E, Osada T, Tamura M, Terai O, Watanabe T, Yoda T, Yotsumoto T, Zaizen K, Sato Y. Protocol improvement and multisite validation of a digital soft agar colony formation assay for tumorigenic transformed cells intermingled in cell therapy products. Cytotherapy 2024; 26:769-777. [PMID: 38556961 DOI: 10.1016/j.jcyt.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND AIMS The administration of human cell-processed therapeutic products (hCTPs) is associated with a risk of tumorigenesis due to the transformed cellular contaminants. To mitigate this risk, these impurities should be detected using sensitive and validated assays. The digital soft agar colony formation (D-SAC) assay is an ultrasensitive in vitro test for detecting tumorigenic transformed cells in hCTPs. METHODS In this study, we first evaluated the colony formation efficiency (CFE) precision of tumorigenic reference cells in positive control samples according to a previously reported D-SAC assay protocol (Protocol I) from multiple laboratories. However, the CFE varied widely among laboratories. Thus, we improved and optimized the test protocol as Protocol II to reduce variability in the CFE of tumorigenic reference cells. Subsequently, the improved protocol was validated at multiple sites. Human mesenchymal stromal cells (hMSCs) were used as model cells, and positive control samples were prepared by spiking them with HeLa cells. RESULTS Based on the previously reported protocol, the CFE was estimated using an ultra-low concentration (0.0001%) of positive control samples in multiple plates. Next, we improved the protocol to reduce the CFE variability. Based on the CFE results, we estimated the sample size as the number of wells (Protocol II) and assessed the detectability of 0.0001% HeLa cells in hMSCs to validate the protocol at multiple sites. Using Protocol I yielded low CFEs (mean: 30%) and high variability between laboratories (reproducibility coefficient of variance [CV]: 72%). In contrast, Protocol II, which incorporated a relatively high concentration (0.002%) of HeLa cells in the positive control samples, resulted in higher CFE values (mean: 63%) and lower variability (reproducibility CV: 18%). Moreover, the sample sizes for testing were estimated as the number of wells per laboratory (314-570 wells) based on the laboratory-specific CFE (42-76%). Under these conditions, all laboratories achieved a detection limit of 0.0001% HeLa cells in hMSCs in a predetermined number of wells. Moreover, colony formation was not observed in the wells seeded with hMSCs alone. CONCLUSIONS The D-SAC assay is a highly sensitive and robust test for detecting malignant cells as impurities in hCTPs. In addition, optimal assay conditions were established to test tumorigenic impurities in hCTPs with high sensitivity and an arbitrary false negative rate.
Collapse
Affiliation(s)
- Kiyoko Bando
- Regenerative & Cellular Medicine Office, Sumitomo Pharma Co., Ltd., Kobe, Japan; The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan.
| | - Shinji Kusakawa
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| | - Hideki Adachi
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Preclinical Research Unit, Sumitomo Pharma Co., Ltd., Osaka, Japan
| | - Mika Yamamoto
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Non-clinical Biomedical Science, Astellas Pharma Inc., Tsukuba, Japan
| | - Miki Iwata
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Osaka Laboratory (formerly the Technology Innovation Center), Sumika Chemical Analysis Service Ltd., Osaka, Japan
| | - Atsushi Kitanaka
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Osaka Laboratory (formerly the Technology Innovation Center), Sumika Chemical Analysis Service Ltd., Osaka, Japan
| | - Eiichiro Ogimura
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Preclinical Research Unit, Sumitomo Pharma Co., Ltd., Osaka, Japan
| | - Tomoharu Osada
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Advanced Medical Business Development Department, Mediford Corporation, Tokyo, Japan
| | - Maya Tamura
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Safety Business Unit (formerly Integrated & Translational Science), Axcelead Drug Discovery Partners Inc., Fujisawa, Japan
| | - Orie Terai
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Osaka Laboratory (formerly the Technology Innovation Center), Sumika Chemical Analysis Service Ltd., Osaka, Japan
| | - Takeshi Watanabe
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Tomomi Yoda
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Preclinical Research Unit, Sumitomo Pharma Co., Ltd., Osaka, Japan
| | - Takafumi Yotsumoto
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Medicinal Safety Research Laboratories, Daiichi Sankyo Company Limited, Tokyo, Japan
| | - Kinuko Zaizen
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Kumamoto Laboratories, Mediford Corporation, Kumamoto, Japan
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan; Division of Drugs, National Institute of Health Sciences, Kawasaki, Japan
| |
Collapse
|
3
|
Lemmens M, Dorsheimer L, Zeller A, Dietz-Baum Y. Non-clinical safety assessment of novel drug modalities: Genome safety perspectives on viral-, nuclease- and nucleotide-based gene therapies. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2024; 896:503767. [PMID: 38821669 DOI: 10.1016/j.mrgentox.2024.503767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/08/2024] [Accepted: 05/13/2024] [Indexed: 06/02/2024]
Abstract
Gene therapies have emerged as promising treatments for various conditions including inherited diseases as well as cancer. Ensuring their safe clinical application requires the development of appropriate safety testing strategies. Several guidelines have been provided by health authorities to address these concerns. These guidelines state that non-clinical testing should be carried out on a case-by-case basis depending on the modality. This review focuses on the genome safety assessment of frequently used gene therapy modalities, namely Adeno Associated Viruses (AAVs), Lentiviruses, designer nucleases and mRNAs. Important safety considerations for these modalities, amongst others, are vector integrations into the patient genome (insertional mutagenesis) and off-target editing. Taking into account the constraints of in vivo studies, health authorities endorse the development of novel approach methodologies (NAMs), which are innovative in vitro strategies for genotoxicity testing. This review provides an overview of NAMs applied to viral and CRISPR/Cas9 safety, including next generation sequencing-based methods for integration site analysis and off-target editing. Additionally, NAMs to evaluate the oncogenicity risk arising from unwanted genomic modifications are discussed. Thus, a range of promising techniques are available to support the safe development of gene therapies. Thorough validation, comparisons and correlations with clinical outcomes are essential to identify the most reliable safety testing strategies. By providing a comprehensive overview of these NAMs, this review aims to contribute to a better understanding of the genome safety perspectives of gene therapies.
Collapse
Affiliation(s)
| | - Lena Dorsheimer
- Research and Development, Preclinical Safety, Sanofi, Industriepark Hoechst, Frankfurt am Main 65926, Germany.
| | - Andreas Zeller
- Pharmaceutical Sciences, pRED Innovation Center Basel, Hoffmann-La Roche Ltd, Basel 4070, Switzerland
| | - Yasmin Dietz-Baum
- Research and Development, Preclinical Safety, Sanofi, Industriepark Hoechst, Frankfurt am Main 65926, Germany
| |
Collapse
|
4
|
Creus‐Bachiller E, Fernández‐Rodríguez J, Magallón‐Lorenz M, Ortega‐Bertran S, Navas‐Rutete S, Romagosa C, Silva TM, Pané M, Estival A, Perez Sidelnikova D, Morell M, Mazuelas H, Carrió M, Lausová T, Reuss D, Gel B, Villanueva A, Serra E, Lázaro C. Expanding a precision medicine platform for malignant peripheral nerve sheath tumors: New patient-derived orthotopic xenografts, cell lines and tumor entities. Mol Oncol 2024; 18:895-917. [PMID: 37798904 PMCID: PMC10994238 DOI: 10.1002/1878-0261.13534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/07/2023] [Accepted: 10/04/2023] [Indexed: 10/07/2023] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive soft-tissue sarcomas with a poor survival rate, presenting either sporadically or in the context of neurofibromatosis type 1 (NF1). The histological diagnosis of MPNSTs can be challenging, with different tumors exhibiting great histological and marker expression overlap. This heterogeneity could be partly responsible for the observed disparity in treatment response due to the inherent diversity of the preclinical models used. For several years, our group has been generating a large patient-derived orthotopic xenograft (PDOX) MPNST platform for identifying new precision medicine treatments. Herein, we describe the expansion of this platform using six primary tumors clinically diagnosed as MPNSTs, from which we obtained six additional PDOX mouse models and three cell lines, thus generating three pairs of in vitro-in vivo models. We extensively characterized these tumors and derived preclinical models, including genomic, epigenomic, and histological analyses. Tumors were reclassified after these analyses: three remained as MPNSTs (two being classic MPNSTs), one was a melanoma, another was a neurotrophic tyrosine receptor kinase (NTRK)-rearranged spindle cell neoplasm, and, finally, the last was an unclassifiable tumor bearing neurofibromin-2 (NF2) inactivation, a neuroblastoma RAS viral oncogene homolog (NRAS) oncogenic mutation, and a SWI/SNF-related matrix-associated actin-dependent regulator of chromatin (SMARCA4) heterozygous truncated variant. New cell lines and PDOXs faithfully recapitulated histology, marker expression, and genomic characteristics of the primary tumors. The diversity in tumor identity and their specific associated genomic alterations impacted treatment responses obtained when we used the new cell lines for testing compounds against known altered pathways in MPNSTs. In summary, we present here an extension of our MPNST precision medicine platform, with new PDOXs and cell lines, including tumor entities confounded as MPNSTs in a real clinical scenario. This platform may constitute a useful tool for obtaining correct preclinical information to guide MPNST clinical trials.
Collapse
Affiliation(s)
- Edgar Creus‐Bachiller
- Hereditary Cancer ProgramCatalan Institute of Oncology, ICO‐IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de LlobregatBarcelonaSpain
| | - Juana Fernández‐Rodríguez
- Hereditary Cancer ProgramCatalan Institute of Oncology, ICO‐IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Mouse Lab, IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
| | | | - Sara Ortega‐Bertran
- Hereditary Cancer ProgramCatalan Institute of Oncology, ICO‐IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de LlobregatBarcelonaSpain
| | - Susana Navas‐Rutete
- Hereditary Cancer ProgramCatalan Institute of Oncology, ICO‐IDIBELL, Hospitalet de LlobregatBarcelonaSpain
| | | | - Tulio M. Silva
- Department of PathologyHospital Vall d'HebronBarcelonaSpain
| | - Maria Pané
- Department of PathologyHUB‐IDIBELL, L'Hospitalet de LlobregatBarcelonaSpain
| | - Anna Estival
- Department of Medical OncologyCatalan Institute of OncologyBarcelonaSpain
| | | | - Mireia Morell
- Hereditary Cancer ProgramCatalan Institute of Oncology, ICO‐IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Mouse Lab, IDIBELL, Hospitalet de LlobregatBarcelonaSpain
| | - Helena Mazuelas
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP)BarcelonaSpain
| | - Meritxell Carrió
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP)BarcelonaSpain
| | - Tereza Lausová
- Department of NeuropathologyInstitute of Pathology, Heidelberg University HospitalHeidelbergGermany
- Clinical Cooperation Unit NeuropathologyGerman Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK)HeidelbergGermany
| | - David Reuss
- Department of NeuropathologyInstitute of Pathology, Heidelberg University HospitalHeidelbergGermany
- Clinical Cooperation Unit NeuropathologyGerman Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK)HeidelbergGermany
| | - Bernat Gel
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP)BarcelonaSpain
| | - Alberto Villanueva
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Procure ProgramCatalan Institute of OncologyBarcelonaSpain
| | - Eduard Serra
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP)BarcelonaSpain
| | - Conxi Lázaro
- Hereditary Cancer ProgramCatalan Institute of Oncology, ICO‐IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
| |
Collapse
|
5
|
Gantier M, Rispal R, Fourrier A, Ménoret S, Delbos F, Anegon I, Nguyen TH. Cryopreserved cGMP-compliant human pluripotent stem cell-derived hepatic progenitors rescue mice from acute liver failure through rapid paracrine effects on liver cells. Stem Cell Res Ther 2024; 15:71. [PMID: 38475825 DOI: 10.1186/s13287-024-03673-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Liver transplantation remains the only curative treatment for end-stage liver diseases. Unfortunately, there is a drastic organ donor shortage. Hepatocyte transplantation emerged as a viable alternative to liver transplantation. Considering their unique expansion capabilities and their potency to be driven toward a chosen cell fate, pluripotent stem cells are extensively studied as an unlimited cell source of hepatocytes for cell therapy. It has been previously shown that freshly prepared hepatocyte-like cells can cure mice from acute and chronic liver failure and restore liver function. METHODS Human PSC-derived immature hepatic progenitors (GStemHep) were generated using a new protocol with current good manufacturing practice compliant conditions from PSC amplification and hepatic differentiation to cell cryopreservation. The therapeutic potential of these cryopreserved cells was assessed in two clinically relevant models of acute liver failure, and the mode of action was studied by several analytical methods, including unbiased proteomic analyses. RESULTS GStemHep cells present an immature hepatic phenotype (alpha-fetoprotein positive, albumin negative), secrete hepatocyte growth factor and do not express major histocompatibility complex. A single dose of thawed GStemHep rescue mice from sudden death caused by acetaminophen and thioacetamide-induced acute liver failure, both in immunodeficient and immunocompetent animals in the absence of immunosuppression. Therapeutic biological effects were observed as soon as 3 h post-cell transplantation with a reduction in serum transaminases and in liver necrosis. The swiftness of the therapeutic effect suggests a paracrine mechanism of action of GStemHep leading to a rapid reduction of inflammation as well as a rapid cytoprotective effect with as a result a proteome reprograming of the host hepatocytes. The mode of action of GStemHep relie on the alleviation of inhibitory factors of liver regeneration, an increase in proliferation-promoting factors and a decrease in liver inflammation. CONCLUSIONS We generated cryopreserved and current good manufacturing practice-compliant human pluripotent stem cell-derived immature hepatic progenitors that were highly effective in treating acute liver failure through rapid paracrine effects reprogramming endogenous hepatocytes. This is also the first report highlighting that human allogeneic cells could be used as cryopreserved cells and in the absence of immunosuppression for human PSC-based regenerative medicine for acute liver failure.
Collapse
Affiliation(s)
- Malika Gantier
- GoLiver Therapeutics, 44007, Nantes, France.
- Nantes Université, Inserm, Center for Research in Transplantation and Translational Immunology, UMR 1064, 44000, Nantes, France.
| | - Raphaël Rispal
- Nantes Université, Inserm, Center for Research in Transplantation and Translational Immunology, UMR 1064, 44000, Nantes, France
| | | | - Séverine Ménoret
- Nantes Université, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016 CNRS UMS 3556, 44000, Nantes, France
| | | | - Ignacio Anegon
- Nantes Université, Inserm, Center for Research in Transplantation and Translational Immunology, UMR 1064, 44000, Nantes, France
| | | |
Collapse
|
6
|
Guerrieri AN, Bellotti C, Penzo M, Columbaro M, Pannella M, De Vita A, Gambarotti M, Mercatali L, Laranga R, Dozza B, Vanni S, Corsini S, Frisoni T, Miserocchi G, Ibrahim T, Lucarelli E. A novel patient-derived immortalised cell line of myxofibrosarcoma: a tool for preclinical drugs testing and the generation of near-patient models. BMC Cancer 2023; 23:1194. [PMID: 38057796 DOI: 10.1186/s12885-023-11658-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Myxofibrosarcoma is a rare malignant soft tissue sarcoma characterised by multiple local recurrence and can become of higher grade with each recurrence. Consequently, myxofibrosarcoma represents a burden for patients, a challenge for clinicians, and an interesting disease to study tumour progression. Currently, few myxofibrosarcoma preclinical models are available. METHODS In this paper, we present a spontaneously immortalised myxofibrosarcoma patient-derived cell line (MF-R 3). We performed phenotypic characterization through multiple biological assays and analyses: proliferation, clonogenic potential, anchorage-independent growth and colony formation, migration, invasion, AgNOR staining, and ultrastructural evaluation. RESULTS MF-R 3 cells match morphologic and phenotypic characteristics of the original tumour as 2D cultures, 3D aggregates, and on the chorioallantoic membrane of chick embryos. Overall results show a clear neoplastic potential of this cell line. Finally, we tested MF-R 3 sensitivity to anthracyclines in 2D and 3D conditions finding a good response to these drugs. CONCLUSIONS In conclusion, we established a novel patient-derived myxofibrosarcoma cell line that, together with the few others available, could serve as an important model for studying the molecular pathogenesis of myxofibrosarcoma and for testing new drugs and therapeutic strategies in diverse experimental settings.
Collapse
Affiliation(s)
- Ania Naila Guerrieri
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano 1/10, 40136, Bologna, Italy
| | - Chiara Bellotti
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano 1/10, 40136, Bologna, Italy.
| | - Marianna Penzo
- Department of Medical and Surgical Sciences and Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum-University of Bologna, 40138, Bologna, Italy
| | - Marta Columbaro
- Electron Microscopy Platform, IRCCS Istituto Ortopedico Rizzoli, 40136, Bologna, Italy
| | - Micaela Pannella
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano 1/10, 40136, Bologna, Italy
| | - Alessandro De Vita
- Preclinic and Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo Per Lo Studio Dei Tumori (IRST) "Dino Amadori", 47014, Meldola, Italy
| | - Marco Gambarotti
- Department of Pathology, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano 1/10, 40136, Bologna, Italy
| | - Laura Mercatali
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano 1/10, 40136, Bologna, Italy
| | - Roberta Laranga
- 3rd Orthopaedic and Traumatologic Clinic prevalently Oncologic, IRCCS Istituto Ortopedico Rizzoli, Bologna IT, Via Pupilli 1, Bologna, 40136, Italy
| | - Barbara Dozza
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum-University of Bologna, Via Di Barbiano 1/10, 40136, Bologna, Italy
| | - Silvia Vanni
- Preclinic and Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo Per Lo Studio Dei Tumori (IRST) "Dino Amadori", 47014, Meldola, Italy
| | - Serena Corsini
- Department of Rare Skeletal Disorders, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano 1/10, 40136, Bologna, Italy
| | - Tommaso Frisoni
- 3rd Orthopaedic and Traumatologic Clinic prevalently Oncologic, IRCCS Istituto Ortopedico Rizzoli, Bologna IT, Via Pupilli 1, Bologna, 40136, Italy
| | - Giacomo Miserocchi
- Preclinic and Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo Per Lo Studio Dei Tumori (IRST) "Dino Amadori", 47014, Meldola, Italy
| | - Toni Ibrahim
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano 1/10, 40136, Bologna, Italy
| | - Enrico Lucarelli
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano 1/10, 40136, Bologna, Italy
| |
Collapse
|
7
|
Wang Z. Assessing Tumorigenicity in Stem Cell-Derived Therapeutic Products: A Critical Step in Safeguarding Regenerative Medicine. Bioengineering (Basel) 2023; 10:857. [PMID: 37508884 PMCID: PMC10376867 DOI: 10.3390/bioengineering10070857] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/08/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Stem cells hold promise in regenerative medicine due to their ability to proliferate and differentiate into various cell types. However, their self-renewal and multipotency also raise concerns about their tumorigenicity during and post-therapy. Indeed, multiple studies have reported the presence of stem cell-derived tumors in animal models and clinical administrations. Therefore, the assessment of tumorigenicity is crucial in evaluating the safety of stem cell-derived therapeutic products. Ideally, the assessment needs to be performed rapidly, sensitively, cost-effectively, and scalable. This article reviews various approaches for assessing tumorigenicity, including animal models, soft agar culture, PCR, flow cytometry, and microfluidics. Each method has its advantages and limitations. The selection of the assay depends on the specific needs of the study and the stage of development of the stem cell-derived therapeutic product. Combining multiple assays may provide a more comprehensive evaluation of tumorigenicity. Future developments should focus on the optimization and standardization of microfluidics-based methods, as well as the integration of multiple assays into a single platform for efficient and comprehensive evaluation of tumorigenicity.
Collapse
Affiliation(s)
- Zongjie Wang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
- Chan Zuckerberg Biohub Chicago, Chicago, IL 60607, USA
| |
Collapse
|
8
|
Nakamura D. The evaluation of tumorigenicity and characterization of colonies in a soft agar colony formation assay using polymerase chain reaction. Sci Rep 2023; 13:5405. [PMID: 37012331 PMCID: PMC10070612 DOI: 10.1038/s41598-023-32442-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 03/28/2023] [Indexed: 04/05/2023] Open
Abstract
In regenerative medicine, the tumorigenic potency of cells in cellular therapy products (CTPs) is a major concern for their application to patients. This study presents a method-the soft agar colony formation assay using polymerase chain reaction (PCR)-to evaluate tumorigenicity. MRC-5 cells, contaminated with HeLa cells, were cultured for up to 4 weeks in soft agar medium. Cell-proliferation-related mRNAs, Ki-67 and cyclin B, could be detected in 0.01% of HeLa cells after 5 days of culture, whereas cyclin-dependent kinase 1 (CDK1) could be detected after 2 weeks. On the other hand, CDK2, proliferating cell nuclear antigen (PCNA), and minichromosome maintenance protein 7 (MCM7) were not useful to detect HeLa cells even after 4 weeks of culture. The cancer stem cell (CSC) markers, aldehyde dehydrogenase 1 (ALDH1) and CD133 in 0.01% of HeLa cells, could be detected 2 and 4 weeks after culture, respectively. However, another CSC marker CD44 was not useful because its expression was also detected in MRC-5 cells alone. This study suggests that the application of the PCR method to the soft agar colony formation assay could evaluate not only the tumorigenic potency in the short-term but also characterize the colonies, eventually improving the safety of CTPs.
Collapse
Affiliation(s)
- Daichi Nakamura
- BoZo Research Center Inc., Tsukuba Research Institute, 8 Okubo, Tsukuba, Ibaraki, 300-2611, Japan.
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan.
| |
Collapse
|
9
|
Dashnau JL, Xue Q, Nelson M, Law E, Cao L, Hei D. A risk-based approach for cell line development, manufacturing and characterization of genetically engineered, induced pluripotent stem cell-derived allogeneic cell therapies. Cytotherapy 2023; 25:1-13. [PMID: 36109321 DOI: 10.1016/j.jcyt.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 12/27/2022]
Abstract
Advances in cellular reprogramming and gene-editing approaches have opened up the potential for a new class of ex vivo cell therapies based on genetically engineered, induced pluripotent stem cell (iPSC)-derived allogeneic cells. While these new therapies share some similarities with their primary cell-derived autologous and allogeneic cell therapy predecessors, key differences exist in the processes used for generating genetically engineered, iPSC-derived allogeneic therapies. Specifically, in iPSC-derived allogeneic therapies, donor selection and gene-editing are performed once over the lifetime of the product as opposed to as part of the manufacturing of each product batch. The introduction of a well-characterized, fully modified, clonally derived master cell bank reduces risks that have been inherent to primary-cell derived autologous and allogeneic therapies. Current regulatory guidance, which was largely developed based on the learnings gained from earlier generation therapies, leaves open questions around considerations for donor eligibility, starting materials and critical components, cell banking and genetic stability. Here, a risk-based approach is proposed to address these considerations, while regulatory guidance continues to evolve.
Collapse
Affiliation(s)
| | - Qiong Xue
- Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Monica Nelson
- Century Therapeutics, Inc., Philadelphia, Pennsylvania, USA
| | - Eric Law
- Century Therapeutics, Inc., Philadelphia, Pennsylvania, USA
| | - Lan Cao
- Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Derek Hei
- Clade Therapeutics, One Kendall Square, Cambridge, Massachusetts, USA
| |
Collapse
|
10
|
Identification of marker genes to monitor residual iPSCs in iPSC-derived products. Cytotherapy 2023; 25:59-67. [PMID: 36319564 DOI: 10.1016/j.jcyt.2022.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 09/08/2022] [Accepted: 09/27/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Engineered tissues and cell therapies based on human induced pluripotent stem cells (iPSCs) represent a promising approach for novel medicines. However, iPSC-derived cells and tissues may contain residual undifferentiated iPSCs that could lead to teratoma formation after implantation into patients. As a consequence, highly sensitive and specific methods for detecting residual undifferentiated iPSCs are indispensable for safety evaluations of iPSC-based therapies. The present study provides an approach for identifying potential marker genes for iPSC impurities in iPSC-derived cells using RNA sequencing data from iPSCs and various differentiated cell types. METHODS Identifying iPSC marker genes for each cell type individually provided a larger and more specific set of potential marker genes than considering all cell types in the analysis. Thus, the authors focused on identifying markers for iPSC impurities in iPSC-derived cardiomyocytes (iCMs) and validated the selected genes by reverse transcription quantitative polymerase chain reaction. The sensitivity of the candidate genes was determined by spiking different amounts of iPSCs into iCMs and their performance was compared with the previously suggested marker lin-28 homolog A (LIN28A). RESULTS Embryonic stem cell-related gene (ESRG), long intergenic non-protein coding RNA 678 (LINC00678), CaM kinase-like vesicle-associated (CAMKV), indoleamine 2,3-dioxygenase 1 (IDO1), chondromodulin (CNMD), LINE1-type transposase domain containing 1 (L1DT1), LIN28A, lymphocyte-specific protein tyrosine kinase (LCK), vertebrae development-associated (VRTN) and zinc finger and SCAN domain containing 10 (ZSCAN10) detected contaminant iPSCs among iCMs with a limit of detection that ranged from 0.001% to 0.1% depending on the gene and iCM batch used. CONCLUSIONS Using the example of iCMs, the authors provide a strategy for identifying a set of highly specific and sensitive markers that can be used for quality assessment of iPSC-derived products.
Collapse
|
11
|
Hirai T, Kono K, Kusakawa S, Yasuda S, Sawada R, Morishita A, Hata S, Wakita A, Kageyama T, Takahashi R, Watanabe S, Shiraishi N, Sato Y. Evaluation of the reproducibility and positive controls of cellular immortality test for the detection of immortalized cellular impurities in human cell-processed therapeutic products. Regen Ther 2022; 21:540-546. [PMID: 36382135 PMCID: PMC9634468 DOI: 10.1016/j.reth.2022.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Contamination of human cell-processed therapeutic products (hCTPs) with tumorigenic/immortalized cellular impurities is a major concern in the manufacturing and quality control of hCTPs. The cellular immortality test based on cell growth analysis is a method for detecting tumorigenic/immortalized cellular impurities in hCTPs. However, the performance of the cellular immortality test has not yet been well characterized. In this study, we examined the reproducibility of the cellular immortality test in detecting HeLa cells as a model of tumorigenic cellular impurities, as well as the applicability of other models of cellular impurities with different tumorigenicity to the cellular immortality test. METHODS Using HeLa cells as a model for cellular impurities, we measured the growth rate of human mesenchymal stem cells (hMSCs) supplemented with HeLa cells at concentrations ranging from 0.01 to 0.0001% at each passage in three laboratories and evaluated the reproducibility of the detection of immortalized cellular impurities. In addition, HEK293 cells (another immortalized cell line) and MRC-5 cells (a non-immortalized cell line) were employed as cellular impurity models that exhibit different growth characteristics from HeLa cells, and the ability of the cellular immortality test to detect these different impurities when mixed with hMSCs was examined. RESULTS In the multisite study, the growth rate of hMSCs supplemented with 1 and 10 HeLa cells (0.0001% and 0.001%) significantly increased and reached a plateau in all three laboratories, whereas those of hMSCs alone eventually decreased. Moreover, when hMSCs were supplemented with 10 and 100 HEK293 and MRC-5 cells (0.001% and 0.01%), the growth rate significantly increased. The growth rate of hMSCs supplemented with HEK293 cells increased with passage and remained high, whereas that of hMSCs supplemented with MRC-5 cells eventually decreased, as in the case of hMSCs alone. CONCLUSIONS These results indicate that the cellular immortality test is reproducible and can detect immortalized (i.e., potentially tumorigenic) cells such as HEK293 cells with a lower growth rate than HeLa cells by discriminating against normal cells, which could contribute to ensuring the safety and quality of hCTPs.
Collapse
Affiliation(s)
- Takamasa Hirai
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
| | - Ken Kono
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
| | - Shinji Kusakawa
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
| | - Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan,Department of Quality Assurance Science for Pharmaceuticals, Graduate School of Pharmaceutical Sciences, Nagoya City University, Aichi, Japan
| | - Rumi Sawada
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
| | | | | | - Atsushi Wakita
- Clinical Pathology Division, Tsukuba Research Institute, BoZo Research Center Inc., Ibaraki, Japan
| | - Takayasu Kageyama
- Clinical Pathology Division, Tsukuba Research Institute, BoZo Research Center Inc., Ibaraki, Japan
| | - Ryo Takahashi
- Clinical Pathology Division, Tsukuba Research Institute, BoZo Research Center Inc., Ibaraki, Japan
| | - Sono Watanabe
- Analytical Research Group, Research Division, HEALIOS K.K., Hyogo, Japan
| | - Norihiko Shiraishi
- New Healthcare Solutions, Corporate Strategy Department, Strategy Division, Kyowakirin Co., Ltd., Tokyo, Japan
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan,Next Generation Life Science Technology Development Project, Kanagawa Institute of Industrial Science and Technology, Kanagawa, Japan,Department of Cellular and Gene Therapy Products, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan,Corresponding author. Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki Ward, Kawasaki City, Kanagawa 210-9501, Japan. Fax: +81-44-270-6526.
| |
Collapse
|
12
|
Chan AML, Ng AMH, Mohd Yunus MH, Hj Idrus RB, Law JX, Yazid MD, Chin KY, Shamsuddin SA, Mohd Yusof MR, Razali RA, Mat Afandi MA, Hassan MNF, Ng SN, Koh B, Lokanathan Y. Safety study of allogeneic mesenchymal stem cell therapy in animal model. Regen Ther 2022; 19:158-165. [PMID: 35252487 PMCID: PMC8861582 DOI: 10.1016/j.reth.2022.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/01/2021] [Accepted: 01/27/2022] [Indexed: 02/05/2023] Open
Abstract
Intravenous (IV) infusion of mesenchymal stem cells (MSCs) from nascent tissues like Wharton's Jelly of the umbilical cord is reported to offer therapeutic effects against chronic diseases. However, toxicological data essential for the clinical application of these cells are limited. Thus, this study aimed to determine the safety of IV infusion of Wharton's Jelly derived MSCs (WJ-MSCs) in rats. Fifteen male Sprague–Dawley rats were randomised into the control or treatment group. Each group received an equal volume of saline or WJ-MSC (10 × 106 cell/kg) respectively. The animals were evaluated for physical, biochemical and haematological changes at Week 0, 2, 4, 8 and 12 during the 12-week study. Acute toxicity was performed during Week 2 and sub-chronic toxicity during Week 12. At the end of the study, the relative weight of organs was calculated and histology was performed for lung, liver, spleen and kidney. The findings from physical, serum biochemistry and complete blood count demonstrated no statistically significant differences between groups. However, pathological evaluation reported minor inflammation in the lungs for all groups, but visible healing and resolution of inflammation were observed in the treatment group only. Additionally, the histological images of the treatment group had significantly improved pulmonary structures compared to the control group. In summary, the IV administration of WJ-MSC was safe in the rats. Further studies are needed to determine the long-term safety of the WJ-MSC in both healthy and diseased animal models. Intravenous infusion of high-dose WJ-MSC in rats is safe. No physical, biochemical and haematological adverse side effects were observed from the treatment. WJ-MSC successfully suppressed inflammation and stimulated regeneration in histopathological analysis.
Collapse
Affiliation(s)
- Alvin Man Lung Chan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Kuala Lumpur, Malaysia
- Ming Medical Sdn Bhd, D3-3 (2nd Floor), Block D3 Dana 1 Commercial Centre, Jalan PJU 1a/46, 47301, Petaling Jaya, Selangor, Malaysia
| | - Angela Min Hwei Ng
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Kuala Lumpur, Malaysia
| | - Mohd Heikal Mohd Yunus
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Kuala Lumpur, Malaysia
| | - Ruszymah Bt Hj Idrus
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Kuala Lumpur, Malaysia
| | - Jia Xian Law
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Kuala Lumpur, Malaysia
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Kuala Lumpur, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Kuala Lumpur, Malaysia
| | - Sharen Aini Shamsuddin
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Kuala Lumpur, Malaysia
| | - Mohd Rafizul Mohd Yusof
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Kuala Lumpur, Malaysia
| | - Rabiatul Adawiyah Razali
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Kuala Lumpur, Malaysia
| | - Mohd Asyraf Mat Afandi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Kuala Lumpur, Malaysia
| | - Muhammad Najib Fathi Hassan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Kuala Lumpur, Malaysia
| | - See Nguan Ng
- Ming Medical Sdn Bhd, D3-3 (2nd Floor), Block D3 Dana 1 Commercial Centre, Jalan PJU 1a/46, 47301, Petaling Jaya, Selangor, Malaysia
| | - Benson Koh
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Kuala Lumpur, Malaysia
- Ming Medical Sdn Bhd, D3-3 (2nd Floor), Block D3 Dana 1 Commercial Centre, Jalan PJU 1a/46, 47301, Petaling Jaya, Selangor, Malaysia
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Kuala Lumpur, Malaysia
- Corresponding author.
| |
Collapse
|
13
|
Inagaki E, Arai E, Hatou S, Sayano T, Taniguchi H, Negishi K, Kanai Y, Sato Y, Okano H, Tsubota K, Shimmura S. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:841-849. [PMID: 35666752 PMCID: PMC9397653 DOI: 10.1093/stcltm/szac036] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 04/09/2022] [Indexed: 11/25/2022] Open
Abstract
Pluripotent stem cell (PSC)-based cell therapies have increased steadily over the past few years, and assessing the risk of tumor formation is a high priority for clinical studies. Current in vivo tumorigenesis studies require several months and depend strongly on the site of grafting. In this study, we report that the anterior eye chamber is preferable to the subcutaneous space for in vivo tumorigenesis studies for several reasons. First, cells can easily be transplanted into the anterior chamber and monitored in real-time without sacrificing the animals due to the transparency of the cornea. Second, tumor formation is faster than with the conventional subcutaneous method. The median tumor formation time in the subcutaneous area was 18.50 weeks (95% CI 10.20-26.29), vs. 4.0 weeks (95% CI 3.34-.67) in the anterior chamber (P = .0089). When hiPSCs were spiked with fibroblasts, the log10TPD50 was 3.26, compared with 4.99 when hiPSCs were transplanted without fibroblasts. There was more than a 40-fold difference in the log10TPD50 values with fibroblasts. Furthermore, the log10TPD50 for HeLa cells was 1.45 and 100% of animals formed tumors at a concentration greater than 0.1%, indicating that the anterior chamber tumorigenesis assays can be applied for cancer cell lines as well. Thus, our method has the potential to become a powerful tool in all areas of tumorigenesis studies and cancer research.
Collapse
Affiliation(s)
- Emi Inagaki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Japanese Society for the Promotion of Science (JSPS), Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Eri Arai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Shin Hatou
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Cellusion Inc., Tokyo, Japan
| | - Tomoko Sayano
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Cellusion Inc., Tokyo, Japan
| | - Hiroko Taniguchi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Yae Kanai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Yasunori Sato
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Shigeto Shimmura
- Corresponding author: Shigeto Shimmura, MD, Department of Ophthalmology, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan. Tel: +81 3 3358 5962; Fax: +81 3 3359 8302;
| |
Collapse
|
14
|
Evaluation of two in vitro assays for tumorigenicity assessment of CRISPR-Cas9 genome-edited cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 23:241-253. [PMID: 34703845 PMCID: PMC8505356 DOI: 10.1016/j.omtm.2021.09.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 09/03/2021] [Indexed: 12/26/2022]
Abstract
Off-target editing is one of the main safety concerns for the use of CRISPR-Cas9 genome editing in gene therapy. These unwanted modifications could lead to malignant transformation, which renders tumorigenicity assessment of gene therapy products indispensable. In this study, we established two in vitro transformation assays, the soft agar colony-forming assay (SACF) and the growth in low attachment assay (GILA) as alternative methods for tumorigenicity evaluation of genome-edited cells. Using a CRISPR-Cas9-based approach to transform immortalized MCF10A cells, we identified PTPN12, a known tumor suppressor, as a valid positive control in GILA and SACF. Next, we measured the limit of detection for both assays and proved that SACF is more sensitive than GILA (0.8% versus 3.1% transformed cells). We further validated SACF and GILA by identifying a set of positive and negative controls and by testing the suitability of another cell line (THLE-2). Moreover, in contrast to SACF and GILA, an in vivo tumorigenicity study failed to detect the known tumorigenic potential of PTPN12 deletion, demonstrating the relevance of GILA and SACF in tumorigenicity testing. In conclusion, SACF and GILA are both attractive and valuable additions to preclinical safety assessment of gene therapy products.
Collapse
|
15
|
Mizuno M, Endo K, Katano H, Amano N, Nomura M, Hasegawa Y, Ozeki N, Koga H, Takasu N, Ohara O, Morio T, Sekiya I. Transplantation of human autologous synovial mesenchymal stem cells with trisomy 7 into the knee joint and 5 years of follow-up. Stem Cells Transl Med 2021; 10:1530-1543. [PMID: 34342383 PMCID: PMC8550709 DOI: 10.1002/sctm.20-0491] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 05/20/2021] [Accepted: 06/16/2021] [Indexed: 01/16/2023] Open
Abstract
Mesenchymal stem cells (MSCs) can show trisomy 7; however, the safety of these cells has not been fully investigated. The purposes of this study were to determine the ratio of patients whose synovial MSCs were transplanted clinically, to intensively investigate MSCs with trisomy 7 from a safety perspective, and to follow up the patients for 5 years after transplantation. Synovial MSCs at passage 0 were transplanted into a knee for degenerative meniscus tears in 10 patients, and the patients were checked at 5 years. The synovial MSCs were evaluated at passages 0 to 15 by G‐bands and digital karyotyping, and trisomy 7 was found in 3 of 10 patients. In those three patients, 5% to 10% of the synovial MSCs showed trisomy 7. The mRNA expressions of representative oncogenes and genes on chromosome 7 did not differ between MSCs with and without trisomy 7. Whole‐genome sequencing and DNA methylation analysis showed similar results for MSCs with and without trisomy 7. Transplantation of human synovial MSCs with trisomy 7 into eight mouse knees did not result in tumor formation under the skin or in the knees after 8 weeks in any mouse, whereas transplanted HT1080 cells formed tumors. In vitro chondrogenic potentials were similar between MSCs with and without trisomy 7. Five‐year follow‐ups revealed no serious adverse events in all 10 human patients, including 3 who had received MSCs with trisomy 7. Overall, our findings indicated that synovial MSCs with trisomy 7 were comparable with MSCs without trisomy 7 from a safety perspective.
Collapse
Affiliation(s)
- Mitsuru Mizuno
- Center for Stem Cell and Regenerative MedicineTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Kentaro Endo
- Center for Stem Cell and Regenerative MedicineTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Hisako Katano
- Center for Stem Cell and Regenerative MedicineTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Naoki Amano
- Department of Fundamental Cell TechnologyCenter for iPS Cell Research and Application, Kyoto UniversityKyotoJapan
| | - Masaki Nomura
- Department of Fundamental Cell TechnologyCenter for iPS Cell Research and Application, Kyoto UniversityKyotoJapan
| | | | - Nobutake Ozeki
- Center for Stem Cell and Regenerative MedicineTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Hideyuki Koga
- Department of Joint Surgery and Sports MedicineGraduate School, Tokyo Medical and Dental University (TMDU)TokyoJapan
| | - Naoko Takasu
- Department of Fundamental Cell TechnologyCenter for iPS Cell Research and Application, Kyoto UniversityKyotoJapan
| | - Osamu Ohara
- Department of Applied GenomicsKazusa DNA Research InstituteChibaJapan
| | - Tomohiro Morio
- Department of Pediatrics and Developmental BiologyGraduate School, Tokyo Medical and Dental University (TMDU)TokyoJapan
| | - Ichiro Sekiya
- Center for Stem Cell and Regenerative MedicineTokyo Medical and Dental University (TMDU)TokyoJapan
| |
Collapse
|
16
|
Wang Z, Sargent EH, Kelley SO. Ultrasensitive Detection and Depletion of Rare Leukemic B Cells in T Cell Populations via Immunomagnetic Cell Ranking. Anal Chem 2021; 93:2327-2335. [PMID: 33432815 DOI: 10.1021/acs.analchem.0c04202] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Rare CD19+ leukemic B cells present in purified T cell populations can cause disease relapse and even the failure of CD19-targeting CAR-T therapy as these rare cells have the ability to self-mask their surface CD19 and escape from the recognition of T cells. It is therefore critical to efficiently detect and robustly deplete rare leukemic B cells in samples of therapeutic T cells. Here, we present a novel microfluidic approach to address the challenges specific to quality control of therapeutic T cells - CAR-QC. CAR-QC utilizes immunomagnetic labeling with a highly selective microfluidic device to rank and isolate rare leukemic B cells in T cell populations. CAR-QC offers ultrasensitive detection of leukemic B cells at single-cell resolution and robust depletion efficiency up to 99.985%. We demonstrate that CAR-QC outperforms flow cytometry and magnetic-activated cell sorting for detecting or purifying spiked samples. In addition, we prove that the improved performance of CAR-QC helps to avoid the occurrence and possibly relapse of rare leukemic B cells in vitro.
Collapse
Affiliation(s)
- Zongjie Wang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto M5S 3G4, Canada.,Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto M5S 3G9, Canada
| | - Edward H Sargent
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto M5S 3G4, Canada
| | - Shana O Kelley
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto M5S 3G9, Canada.,Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada.,Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto M5S 1A8, Canada.,Department of Chemistry, Faculty of Arts and Science, University of Toronto, Toronto M5S 3H6, Canada
| |
Collapse
|
17
|
Tumorigenicity assessment of cell therapy products: The need for global consensus and points to consider. Cytotherapy 2019; 21:1095-1111. [DOI: 10.1016/j.jcyt.2019.10.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 10/01/2019] [Indexed: 12/11/2022]
|
18
|
Transient induction of telomerase expression mediates senescence and reduces tumorigenesis in primary fibroblasts. Proc Natl Acad Sci U S A 2019; 116:18983-18993. [PMID: 31481614 PMCID: PMC6754593 DOI: 10.1073/pnas.1907199116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Telomerase is an enzymatic ribonucleoprotein complex that acts as a reverse transcriptase in the elongation of telomeres. Telomerase activity is well documented in embryonic stem cells and the vast majority of tumor cells, but its role in somatic cells remains to be understood. Here, we report an unexpected function of telomerase during cellular senescence and tumorigenesis. We crossed Tert heterozygous knockout mice (mTert +/- ) for 26 generations, during which time there was progressive shortening of telomeres, and obtained primary skin fibroblasts from mTert +/+ and mTert -/- progeny of the 26th cross. As a consequence of insufficient telomerase activities in prior generations, both mTert +/+ and mTert -/- fibroblasts showed comparable and extremely short telomere length. However, mTert -/- cells approached cellular senescence faster and exhibited a significantly higher rate of malignant transformation than mTert +/+ cells. Furthermore, an evident up-regulation of telomerase reverse-transcriptase (TERT) expression was detected in mTert +/+ cells at the presenescence stage. Moreover, removal or down-regulation of TERT expression in mTert +/+ and human primary fibroblast cells via CRISPR/Cas9 or shRNA recapitulated mTert -/- phenotypes of accelerated senescence and transformation, and overexpression of TERT in mTert -/- cells rescued these phenotypes. Taking these data together, this study suggests that TERT has a previously underappreciated, protective role in buffering senescence stresses due to short, dysfunctional telomeres, and preventing malignant transformation.
Collapse
|
19
|
Ito E, Miyagawa S, Takeda M, Kawamura A, Harada A, Iseoka H, Yajima S, Sougawa N, Mochizuki-Oda N, Yasuda S, Sato Y, Sawa Y. Tumorigenicity assay essential for facilitating safety studies of hiPSC-derived cardiomyocytes for clinical application. Sci Rep 2019; 9:1881. [PMID: 30760836 PMCID: PMC6374479 DOI: 10.1038/s41598-018-38325-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/26/2018] [Indexed: 11/09/2022] Open
Abstract
Transplantation of cardiomyocytes (CMs) derived from human induced pluripotent stem cells (hiPSC-CMs) is a promising treatment for heart failure, but residual undifferentiated hiPSCs and malignant transformed cells may lead to tumor formation. Here we describe a highly sensitive tumorigenicity assay for the detection of these cells in hiPSC-CMs. The soft agar colony formation assay and cell growth analysis were unable to detect malignantly transformed cells in hiPSC-CMs. There were no karyotypic abnormalities during hiPSCs subculture and differentiation. The hiPSC markers TRA1-60 and LIN28 showed the highest sensitivity for detecting undifferentiated hiPSCs among primary cardiomyocytes. Transplantation of hiPSC-CMs with a LIN28-positive fraction > 0.33% resulted in tumor formation in nude rats, whereas no tumors were formed when the fraction was < 0.1%. These findings suggested that combination of these in vitro and in vivo tumorigenecity assays can verify the safety of hiPSC-CMs for cell transplantation therapy.
Collapse
Affiliation(s)
- Emiko Ito
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Maki Takeda
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Ai Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Akima Harada
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Hiroko Iseoka
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Shin Yajima
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Nagako Sougawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Noriko Mochizuki-Oda
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Kanagawa, 210-9501, Japan
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Kanagawa, 210-9501, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
20
|
Shigeto J, Ichiki T, Nii T, Konno K, Nakanishi Y, Sugiyama D. Preclinical Toxicity Studies for Regenerative Medicine in Japan. Clin Ther 2018; 40:1813-1822. [DOI: 10.1016/j.clinthera.2018.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/05/2018] [Accepted: 09/07/2018] [Indexed: 01/14/2023]
|
21
|
Kuroda T, Yasuda S, Nakashima H, Takada N, Matsuyama S, Kusakawa S, Umezawa A, Matsuyama A, Kawamata S, Sato Y. Identification of a Gene Encoding Slow Skeletal Muscle Troponin T as a Novel Marker for Immortalization of Retinal Pigment Epithelial Cells. Sci Rep 2017; 7:8163. [PMID: 28811571 PMCID: PMC5557831 DOI: 10.1038/s41598-017-08014-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/05/2017] [Indexed: 12/26/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) are leading candidate raw materials for cell-based therapeutic products (CTPs). In the development of hPSC-derived CTPs, it is imperative to ensure that they do not form tumors after transplantation for safety reasons. Because cellular immortalization is a landmark of malignant transformation and a common feature of cancer cells, we aimed to develop an in vitro assay for detecting immortalized cells in CTPs. We employed retinal pigment epithelial (RPE) cells as a model of hPSC-derived products and identified a gene encoding slow skeletal muscle troponin T (TNNT1) as a novel marker of immortalized RPE cells by comprehensive microarray analysis. TNNT1 mRNA was commonly upregulated in immortalized RPE cells and human induced pluripotent stem cells (hiPSCs), which have self-renewal ability. Additionally, we demonstrated that TNNT1 mRNA expression is higher in several cancer tissues than in normal tissues. Furthermore, stable expression of TNNT1 in ARPE-19 cells affected actin filament organization and enhanced their migration ability. Finally, we established a simple and rapid qRT-PCR assay targeting TNNT1 transcripts that detected as low as 3% of ARPE-19 cells contained in normal primary RPE cells. Purified hiPSC-derived RPE cells showed TNNT1 expression levels below the detection limit determined with primary RPE cells. Our qRT-PCR method is expected to greatly contribute to process validation and quality control of CTPs.
Collapse
Affiliation(s)
- Takuya Kuroda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Tokyo, Japan
- Foundation for Biomedical Research and Innovation, Kobe, Japan
| | - Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Tokyo, Japan
- Foundation for Biomedical Research and Innovation, Kobe, Japan
| | - Hiroyuki Nakashima
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Tokyo, Japan
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Nozomi Takada
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Tokyo, Japan
- Platform of Therapeutics for Rare Disease, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Satoko Matsuyama
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Tokyo, Japan
- Platform of Therapeutics for Rare Disease, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Shinji Kusakawa
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Tokyo, Japan
- Foundation for Biomedical Research and Innovation, Kobe, Japan
| | - Akihiro Umezawa
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Akifumi Matsuyama
- Platform of Therapeutics for Rare Disease, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Shin Kawamata
- Foundation for Biomedical Research and Innovation, Kobe, Japan
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Tokyo, Japan.
- Foundation for Biomedical Research and Innovation, Kobe, Japan.
- Department of Quality Assurance Science for Pharmaceuticals, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
- Department of Cellular & Gene Therapy Products, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.
- Department of Drug Discovery and Evolution, Graduated School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
22
|
Hasebe-Takada N, Kono K, Yasuda S, Sawada R, Matsuyama A, Sato Y. Application of cell growth analysis to the quality assessment of human cell-processed therapeutic products as a testing method for immortalized cellular impurities. Regen Ther 2016; 5:49-54. [PMID: 31245501 PMCID: PMC6581850 DOI: 10.1016/j.reth.2016.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 06/06/2016] [Accepted: 06/29/2016] [Indexed: 01/31/2023] Open
Abstract
In human cell-processed therapeutic products (hCTPs) for clinical application, tumorigenic cellular impurities in the manufacturing process are a major concern. Because cellular immortalization is one of the prerequisite steps in tumorigenesis, we tested whether cell growth analysis can be employed to check for immortalized (and potentially tumorigenic) cellular impurities in hCTPs. We monitored the growth of human bone marrow-derived mesenchymal stem cells (BMSCs) mixed with HeLa cells at a ratio of 1/106 or more and compared their growth rates with that of BMSCs alone. The cell growth analysis detected a significant increase in the growth rate of the BMSCs spiked with 0.0001% HeLa within 30 days at a probability of 47%. When human adipose-derived stem cells (ADSCs) were spiked with ASC52telo cells, a human telomerase reverse transcriptase (hTERT)-immortalized adipose-derived mesenchymal stem cell line, at a ratio of 0.001% or more, their growth rates were significantly increased within 15 passages, compared with that of ADSCs alone. These results indicate that cell growth analysis for the detection of immortalized cellular impurities in human somatic stem cells is simple and can be useful for the quality assessment of hCTPs in the manufacturing process.
Collapse
Key Words
- ADSC, adipose-derived stem cell
- Adipose-derived stem cell
- BMSC, bone marrow-derived mesenchymal stem cell
- Bone marrow-derived mesenchymal stem cell
- Cellular therapy
- DMEM, Dulbecco's modified Eagle's medium
- FBS, fetal bovine serum
- PBS, phosphate buffered saline
- P = n, passage n
- Quality assessment
- Regenerative medicine
- Tumorigenicity
- hCTP, human cell-processed therapeutic product
- hMSC, human mesenchymal stem cell
- hTERT, human telomerase reverse transcriptase
- qRT-PCR, quantitative reverse transcription polymerase chain reaction
Collapse
Affiliation(s)
- Nozomi Hasebe-Takada
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Tokyo, Japan
- Platform of Therapeutics for Rare Disease, National Institutes of Biomedical Innovation, Health and Nutrition, Amagasaki, Japan
| | - Ken Kono
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Tokyo, Japan
| | - Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Tokyo, Japan
| | - Rumi Sawada
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Tokyo, Japan
| | - Akifumi Matsuyama
- Platform of Therapeutics for Rare Disease, National Institutes of Biomedical Innovation, Health and Nutrition, Amagasaki, Japan
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Tokyo, Japan
- Department of Quality Assurance Science for Pharmaceuticals, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
- Department of Cellular and Gene Therapy Products, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Department of Translational Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
23
|
Chen Y, Zhou Q, Zhang L, Wang R, Jin M, Qiu Y, Kong D. Idelalisib induces G1 arrest and apoptosis in chronic myeloid leukemia K562 cells. Oncol Rep 2016; 36:3643-3650. [DOI: 10.3892/or.2016.5176] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 06/11/2016] [Indexed: 11/06/2022] Open
|