1
|
A Double Fail-Safe Approach to Prevent Tumorigenesis and Select Pancreatic β Cells from Human Embryonic Stem Cells. Stem Cell Reports 2019; 12:611-623. [PMID: 30773486 PMCID: PMC6409439 DOI: 10.1016/j.stemcr.2019.01.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 12/22/2022] Open
Abstract
The transplantation of human embryonic stem cell (hESC)-derived insulin-producing β cells for the treatment of diabetes is finally approaching the clinical stage. However, even with state-of-the-art differentiation protocols, a significant percentage of undefined non-endocrine cell types are still generated. Most importantly, there is the potential for carry-over of non-differentiated cell types that may produce teratomas. We sought to modify hESCs so that their differentiated progeny could be selectively devoid of tumorigenic cells and enriched for cells of the desired phenotype (in this case, β cells). Here we report the generation of a modified hESC line harboring two suicide gene cassettes, whose expression results in cell death in the presence of specific pro-drugs. We show the efficacy of this system at enriching for β cells and eliminating tumorigenic ones both in vitro and in vivo. Our approach is innovative inasmuch as it allows for the preservation of the desired cells while eliminating those with the potential to develop teratomas. hESCs were engineered with suicide genes for safety and differentiation efficiency One cassette is exclusively expressed in teratogenic cells (safety) Another is selectively excised out in hESC-derived pancreatic β cells (selectivity) Our strategy allows for hESC-derived tumors to be prevented or ablated in vivo
Collapse
|
2
|
Garg H, Joshi A. Conditional Cytotoxic Anti-HIV Gene Therapy for Selectable Cell Modification. Hum Gene Ther 2016; 27:400-15. [PMID: 26800572 DOI: 10.1089/hum.2015.126] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Gene therapy remains one of the potential strategies to achieve a cure for HIV infection. One of the major limitations of anti-HIV gene therapy concerns recovering an adequate number of modified cells to generate an HIV-proof immune system. Our study addresses this issue by developing a methodology that can mark conditional vector-transformed cells for selection and subsequently target HIV-infected cells for elimination by treatment with ganciclovir (GCV). We used the herpes simplex virus thymidine kinase (TK) mutant SR39, which is highly potent at killing cells at low GCV concentrations. This gene was cloned into a conditional HIV vector, pNL-GFPRRESA, which expresses the gene of interest as well as green fluorescent protein (GFP) in the presence of HIV Tat protein. We show here that TK-SR39 was more potent that wild-type TK (TK-WT) at eliminating infected cells at lower concentrations of GCV. As the vector expresses GFP in the presence of Tat, transient expression of Tat either by Tat RNA transfection or transduction by a nonintegrating lentiviral (NIL) vector marked the cells with GFP for selection. In cells selected by this strategy, TK-SR39 was more potent at limiting virus replication than TK-WT. Finally, in Jurkat cells modified and selected by this approach, infection with CXCR4-tropic Lai virus could be suppressed by treatment with GCV. GCV treatment limited the number of HIV-infected cells, virus production, as well as virus-induced cytopathic effects in this model. We provide proof of principle that TK-SR39 in a conditional HIV vector can provide a safe and effective anti-HIV strategy.
Collapse
Affiliation(s)
- Himanshu Garg
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center , El Paso, Texas
| | - Anjali Joshi
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center , El Paso, Texas
| |
Collapse
|
3
|
Wu C, Hong SG, Winkler T, Spencer DM, Jares A, Ichwan B, Nicolae A, Guo V, Larochelle A, Dunbar CE. Development of an inducible caspase-9 safety switch for pluripotent stem cell-based therapies. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:14053. [PMID: 26052521 PMCID: PMC4448736 DOI: 10.1038/mtm.2014.53] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 09/12/2014] [Indexed: 02/07/2023]
Abstract
Induced pluripotent stem cell (iPSC) therapies offer a promising path for patient-specific regenerative medicine. However, tumor formation from residual undifferentiated iPSC or transformation of iPSC or their derivatives is a risk. Inclusion of a suicide gene is one approach to risk mitigation. We introduced a dimerizable-“inducible caspase-9” (iCasp9) suicide gene into mouse iPSC (miPSC) and rhesus iPSC (RhiPSC) via a lentivirus, driving expression from either a cytomegalovirus (CMV), elongation factor-1 α (EF1α) or pluripotency-specific EOS-C(3+) promoter. Exposure of the iPSC to the synthetic chemical dimerizer, AP1903, in vitro induced effective apoptosis in EF1α-iCasp9-expressing (EF1α)-iPSC, with less effective killing of EOS-C(3+)-iPSC and CMV-iPSC, proportional to transgene expression in these cells. AP1903 treatment of EF1α-iCasp9 miPSC in vitro delayed or prevented teratomas. AP1903 administration following subcutaneous or intravenous delivery of EF1α-iPSC resulted in delayed teratoma progression but did not ablate tumors. EF1α-iCasp9 expression was downregulated during in vitro and in vivo differentiation due to DNA methylation at CpG islands within the promoter, and methylation, and thus decreased expression, could be reversed by 5-azacytidine treatment. The level and stability of suicide gene expression will be important for the development of suicide gene strategies in iPSC regenerative medicine.
Collapse
Affiliation(s)
- Chuanfeng Wu
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH) , Bethesda, Maryland, USA
| | - So Gun Hong
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH) , Bethesda, Maryland, USA
| | - Thomas Winkler
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH) , Bethesda, Maryland, USA
| | | | - Alexander Jares
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH) , Bethesda, Maryland, USA
| | - Brian Ichwan
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH) , Bethesda, Maryland, USA
| | - Alina Nicolae
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, Maryland, USA
| | - Vicky Guo
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH) , Bethesda, Maryland, USA
| | - Andre Larochelle
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH) , Bethesda, Maryland, USA
| | - Cynthia E Dunbar
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH) , Bethesda, Maryland, USA
| |
Collapse
|
4
|
Barese CN, Krouse AE, Metzger ME, King CA, Traversari C, Marini FC, Donahue RE, Dunbar CE. Thymidine kinase suicide gene-mediated ganciclovir ablation of autologous gene-modified rhesus hematopoiesis. Mol Ther 2012; 20:1932-43. [PMID: 22910293 DOI: 10.1038/mt.2012.166] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Despite the genotoxic complications encountered in clinical gene therapy trials for primary immunodeficiency diseases targeting hematopoietic cells with integrating vectors; this strategy holds promise for the cure of several monogenic blood, metabolic and neurodegenerative diseases. In this study, we asked whether the inclusion of a suicide gene in a standard retrovirus vector would allow elimination of vector-containing stem and progenitor cells and their progeny in vivo following transplantation, using our rhesus macaque transplantation model. Following stable engraftment with autologous CD34(+) cells transduced with a retrovirus vector encoding a highly sensitive modified Herpes simplex virus thymidine kinase SR39, the administration of the antiviral prodrug ganciclovir (GCV) was effective in completely eliminating vector-containing cells in all hematopoietic lineages in vivo. The sustained absence of vector-containing cells over time, without additional GCV administration, suggests that the ablation of TkSR39 GCV-sensitive cells occurred in the most primitive hematopoietic long-term repopulating stem or progenitor cell compartment. These results are a proof-of-concept that the inclusion of a suicide gene in integrating vectors, in addition to a therapeutic gene, can provide a mechanism for later elimination of vector-containing cells, thereby increasing the safety of gene transfer.
Collapse
Affiliation(s)
- Cecilia N Barese
- Hematology Branch, The National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Fung RKF, Kerridge IH, Skene LLC, Munsie MJ. Tempering hope with realism: induced pluripotent stem cells in regenerative medicine. Med J Aust 2012; 196:622-5. [PMID: 22676869 DOI: 10.5694/mja11.11039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Ronald K F Fung
- Centre for Values, Ethics and the Law in Medicine, Sydney Medical School, University of Sydney, Sydney, NSW, Australia.
| | | | | | | |
Collapse
|
6
|
Rosser AE, Kelly CM, Dunnett SB. Cell transplantation for Huntington’s disease: practical and clinical considerations. FUTURE NEUROLOGY 2011. [DOI: 10.2217/fnl.10.78] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Huntington’s disease is a dominantly inherited neurodegenerative disorder, usually starting in mid-life and leading to progressive disability and early death. There are currently no disease-modifying treatments available. Cell transplantation is being considered as a potential therapy, following proof of principle that cell transplantation can improve outcomes in another basal ganglia disorder, namely Parkinson’s disease. The principle aim is to replace the striatal medium spiny neurons lost in Huntington’s disease with new cells that are able to take over their function and reconnect the circuitry. This article reviews the experimental background and evidence from clinical studies that suggest that cell transplantation may improve function in Huntington’s disease, reviews the current status of the field and considers the current challenges to taking this experimental strategy forward to becoming a reliable therapeutic option.
Collapse
Affiliation(s)
- Anne E Rosser
- Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Claire M Kelly
- The Brain Repair Group, Cardiff University School of Biosciences, Museum Av., Cardiff CF10 3AX, UK
| | - Stephen B Dunnett
- The Brain Repair Group, Cardiff University School of Biosciences, Museum Av., Cardiff CF10 3AX, UK
| |
Collapse
|
7
|
HSV-tk expressing mesenchymal stem cells exert bystander effect on human glioblastoma cells. Cancer Lett 2009; 290:58-67. [PMID: 19765892 DOI: 10.1016/j.canlet.2009.08.028] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 07/23/2009] [Accepted: 08/24/2009] [Indexed: 01/14/2023]
Abstract
Previously we have reported adipose-tissue derived human mesenchymal stem cells (AT-MSC) as cellular delivery vehicles for tumor-targeted cancer gene therapy. In this report we aimed to determine whether Herpes simplex virus - thymidine kinase (HSV-tk) expressing AT-MSC (TK-MSC) could exert cytotoxic effect on tumor cells upon treatment with prodrug ganciclovir (GCV). Direct co-cultures of human glioblastoma cells 8-MG-BA, 42-MG-BA and U-118 MG with TK-MSC/GCV resulted in substantial viability decrease in vitro. This therapeutic paradigm was most efficient against 8-MG-BA glioblastoma cells exhibiting cytotoxicity (>50%) in the presence of TK-MSC and 0.1microM GCV. Rapid apoptosis induction in three glioblastoma cell lines and TK-MSC demonstrated both bystander cytotoxic effect on tumor cells and GCV conversion-mediated suicide effect on TK-MSC. Furthermore, we were able to demonstrate formation of gap junctions between AT-MSC and human glioblastoma cells as a mechanism contributing to bystander cytotoxicity. Inability of human HeLa and MCF7 to form gap junctions with AT-MSC rendered these cell refractory to the TK-MSC/GCV mediated cytotoxicity. Gap junction intercellular communication (GJIC) capability of AT-MSC with tumor cells further supports the exploitation of mesenchymal stem cells for approaches relying on the bystander effect. Biological consequences of these capabilities remain to be further explored.
Collapse
|
8
|
Hmadcha A, Abdelkrim H, Domínguez-Bendala J, Juan DB, Wakeman J, Jane W, Arredouani M, Mohamed A, Soria B, Bernat S. The immune boundaries for stem cell based therapies: problems and prospective solutions. J Cell Mol Med 2009; 13:1464-75. [PMID: 19583810 PMCID: PMC3828859 DOI: 10.1111/j.1582-4934.2009.00837.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Stem cells have fascinated the scientific and clinical communities for over a century. Despite the controversy that surrounds this field, it is clear that stem cells have the potential to revolutionize medicine. However, a number of significant hurdles still stand in the way of the realization of this potential. Chiefly among these are safety concerns, differentiation efficiency and overcoming immune rejection. Here we review current progress made in this field to optimize the safe use of stem cells with particular emphasis on prospective interventions to deal with challenges generated by immune rejection.
Collapse
Affiliation(s)
- Abdelkrim Hmadcha
- Department of Cell Therapy and Regenerative Medicine, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Sevilla, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Abstract
The lack of disease-modifying treatments currently available for not just some but most neurodegenerative diseases, including Parkinson's disease, Huntington's disease, and even stroke, helps explain increasing interest in cell-based therapies. One key aim of such treatment is to replace neurons or glia lost as a result of the disease, with a view to the cells integrating functionally within the host tissue in order to reconstruct neural circuitry. Clinical trials using primary human fetal tissue as a cell source commenced in Parkinson's disease (PD) in the 1980s; currently, comparable neural transplantation trials in Huntington's disease are underway. Disappointing results of later controlled trials in PD illustrated not least the vital importance of methodological issues relating to the structure and implementation of clinical trials, and these issues will be considered here in more depth.
Collapse
|
10
|
Vilalta M, Dégano IR, Bagó J, Aguilar E, Gambhir SS, Rubio N, Blanco J. Human adipose tissue-derived mesenchymal stromal cells as vehicles for tumor bystander effect: a model based on bioluminescence imaging. Gene Ther 2008; 16:547-57. [PMID: 19092860 DOI: 10.1038/gt.2008.176] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Human adipose tissue mesenchymal stromal cells (AMSCs) share common traits, including similar differentiation potential and cell surface markers, with their bone marrow counterparts. Owing to their general availability, higher abundance and ease of isolation AMSCs may be convenient autologous delivery vehicles for localized tumor therapy. We demonstrate a model for tumor therapy development based on the use of AMSCs expressing renilla luciferase and thymidine kinase, as cellular vehicles for ganciclovir-mediated bystander killing of firefly luciferase expressing tumors, and noninvasive bioluminescence imaging to continuously monitor both, tumor cells and AMSCs. We show that the therapy delivering AMSCs survive long time within tumors, optimize the ratio of AMSCs to tumor cells for therapy, and asses the therapeutic effect in real time. Treatment of mice bearing prostate tumors plus therapeutic AMSCs with the prodrug ganciclovir induced bystander killing effect, reducing the number of tumor cells to 1.5 % that of control tumors. Thus, AMSCs could be useful vehicles to deliver localized therapy, with potential for clinical application in inoperable tumors and surgical borders after tumor resection. This approach, useful to evaluate efficiency of therapeutic models, should facilitate the selection of cell types, dosages, therapeutic agents and treatment protocols for cell-based therapies of specific tumors.
Collapse
Affiliation(s)
- M Vilalta
- Cardiovascular Research Center (CSIC-ICCC), CIBER-BBN, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
11
|
|
12
|
McMilin KD, Dasgupta S. Allogeneic transplantation and the risk for transmission of genetic disease: the heritable cancer disorders. Stem Cells Dev 2007; 16:191-212. [PMID: 17521232 DOI: 10.1089/scd.2006.0080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
With the development of new approaches to transplantation therapy, such as those building upon the potential found in stem cells, it is vital to pursue a clear understanding of transplantation risks. Allogeneic transplantation presents risk for the transmission of disease of various types, including genetic disease. Predisposition to develop cancer is a feature of numerous genetic disorders, and it may be transmissible by transplantation. Some genetic disorders predisposing to cancer are remarkably common, either worldwide or in specific populations, and they could pose significant risk. Hence, to reduce risk to recipients, there is reason to exclude from donation those potential donors (including embryos) harboring certain germ-line mutations. However, the frequent absence of readily identifiable features might confound the effort to exclude those who harbor mutation. Thus, it is also important to consider the magnitude of risk that they represent. For some disorders, life-threatening cancer is highly likely to develop in those individuals born with germ-line mutation, but whether recipients would face the same risk from transplanted mutation is not always evident. Given the diversity of pathways that lead to cancer, there may be diverse factors that impact the likelihood for cancer to develop in the recipient, with some factors decreasing and others increasing the risk. One factor of special concern is the possibility that manipulation of donor cells, prior to transplantation, might introduce additional genetic or epigenetic abnormality, thereby increasing the risk.
Collapse
Affiliation(s)
- Kenneth D McMilin
- Alabama and Central Gulf Coast Region, American Red Cross Blood Services, Birmingham, AL 35205, USA.
| | | |
Collapse
|
13
|
Hung SC, Deng WP, Yang WK, Liu RS, Lee CC, Su TC, Lin RJ, Yang DM, Chang CW, Chen WH, Wei HJ, Gelovani JG. Mesenchymal stem cell targeting of microscopic tumors and tumor stroma development monitored by noninvasive in vivo positron emission tomography imaging. Clin Cancer Res 2006; 11:7749-56. [PMID: 16278396 DOI: 10.1158/1078-0432.ccr-05-0876] [Citation(s) in RCA: 209] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The aim of this study was to assess the efficacy human mesenchymal stem cells (hMSC) for targeting microscopic tumors and suicide gene or cytokine gene therapy. Immunodeficient mice were transplanted s.c. with human colon cancer cells of HT-29 Inv2 or CCS line, and 3 to 4 days later, i.v. with "tracer" hMSCs expressing herpes simplex virus type 1 thymidine kinase (HSV1-TK) and enhanced green fluorescent protein (EGFP) reporter genes. Subsequently, these tumors were examined for specificity and magnitude of HSV1-TK(+), EGFP(+) stem cell engraftment and proliferation in tumor stroma by in vivo positron emission tomography (PET) with (18)F-labeled 9-(4-fluoro-3-hydroxymethylbutyl)-guanine ([(18)F]-FHBG). In vivo PET images of tumors growing for 4 weeks showed the presence of HSV1-TK(+) tumor stroma with an average of 0.36 +/- 0.24% ID/g [(18)F]-FHBG accumulation. In vivo imaging results were validated by in situ correlative histochemical, immunofluorescent, and cytometric analyses, which revealed EGFP expression in vWF(+) and CD31(+) endothelial cells of capillaries and larger blood vessels, in germinal layer of dermis and hair follicles proximal to the s.c. tumor site. These differentiated HSV1-TK(+), GFP(+) endothelial cells had limited proliferative capacity and a short life span of <2 weeks in tumor fragments transplanted into secondary hosts. We conclude that hMSCs can target microscopic tumors, subsequently proliferate and differentiate, and contribute to formation of a significant portion of tumor stroma. PET imaging should facilitate clinical translation of stem cell-based anticancer gene therapeutic approaches by providing the means for in vivo noninvasive whole-body monitoring of trafficking, tumor targeting, and proliferation of HSV1-tk-expressing "tracer" hMSCs in tumor stroma.
Collapse
Affiliation(s)
- Shih-Chieh Hung
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Ossevoort M, Zaldumbide A, Cramer SJ, van der Voort EIH, Toes REM, Hoeben RC. Characterization of an immuno ‘stealth’ derivative of the herpes simplex virus thymidine-kinase gene. Cancer Gene Ther 2006; 13:584-91. [PMID: 16456552 DOI: 10.1038/sj.cgt.7700925] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The cellular immune response against transgene-encoded neoantigens is a potential hurdle in gene therapy applications where long-term expression of transgenes is desired. Here a new optimized derivative of the herpes simplex virus 1-thymidine-kinase (HSV1-TK) gene is described. The HSV-TK gene is frequently used in experimental studies on gene-directed enzyme prodrug therapy. In the optimized gene, the HSV-TK coding region is fused with the codons for the Gly-Ala repeat of the Epstein-Barr virus nuclear-antigen 1 to prevent proteasomal degradation of the HSV-TK. To measure the protective effect in vitro, a model cytotoxic T lymphocyte epitope derived from the ovalbumin was inserted in the TK. Cells expressing the GAr-modified TK do not present TK-derived peptides in the major histocompatibility complex. Furthermore, conservative nucleotide substitutions were introduced, which prevent splicing, as well as mutations that render the TK-expressing cells more sensitive to ganciclovir (GCV). The GAr HSV-TK fusion protein is fully functional in vitro. This HSV-TK gene may be especially useful in those gene therapy applications where an immune response against the transgene-encoded product would frustrate the treatment.
Collapse
Affiliation(s)
- M Ossevoort
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
15
|
Lawrenz B, Schiller H, Willbold E, Ruediger M, Muhs A, Esser S. Highly sensitive biosafety model for stem-cell-derived grafts. Cytotherapy 2005; 6:212-22. [PMID: 15203978 DOI: 10.1080/14653240410006031] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The recent success in the derivation of differentiated cell types from stem cells has raised prospects for the application of regenerative cell therapy. In particular, embryonic stem cells are attractive sources for cell transplantation, due to their immortality and rapid growth. These cells, however, also possess tumorigenic properties, which raises serious safety concerns and makes biosafety testing mandatory. Our goal was to establish a highly sensitive animal model for testing the proliferative potential of stem-cell grafts. METHODS BALB/c nude mice received cell grafts of non-neoplastic MRC-5 cells containing defined numbers of mouse embryonic stem cells. We either injected 1 million viable cells into the kidney capsule, or mixed 2 million cells with Matrigel for s.c. transplantation. To analyze the possible impact of an intact immune response on tumor development, we also transplanted the cells into immunocompetent mice. Animals were sacrificed when the tumors became >1 cm and were analyzed in detail. RESULTS The nude mouse model reproducibly allowed detection of 20 tumorigenic cells, and even as few as 2 ES cells were found to form teratoma. Interestingly, the administration of cell grafts at two different application sites resulted in different growth kinetics and tumor phenotypes. The highest level of sensitivity (100% detection of 20 tumorigenic ES cells) was achieved by s.c. injection of cells mixed with Matrigel. The influence of the immune system on tumor-cell development was demonstrated by a higher tumor rate of transplants in immunodeficient nude mice compared with immunocompetent mice. DISCUSSION We have established a reliable animal model for routine assessment of the biosafety profile of stem-cell-derived cell transplants. This model will facilitate the generation of homogenous non-tumorigenic cell populations, and will help to integrate standardized safety systems into the application of stem-cell-derived grafts for clinical purposes.
Collapse
|
16
|
Abstract
Embryonic stem (ES) cells are typically derived from the inner cell mass of the preimplantation blastocyst and can both self-renew and differentiate into all the cells and tissues of the embryo. Because they are pluripotent, ES cells have been used extensively to analyze gene function in development via gene targeting. The embryonic stem cell is also an unsurpassed starting material to begin to understand a critical, largely inaccessible period of development. If their differentiation could be controlled, they would also be an important source of cells for transplantation to replace cells lost through disease or injury or to replace missing hormones or genes. Traditionally, ES cells have been differentiated in suspension culture as embryoid bodies, named because of their similarity to the early postimplantation-staged embryo. Unlike the pristine organization of the early embryo, differentiation in embryoid bodies appears to be largely unpatterned, although multiple cell types form. It has recently been possible to separate the desired cell types from differentiating ES cells in embryoid bodies by using cell-type-restricted promoters driving expression of either antibiotic resistance genes or fluorophores such as EGFP. In combination with growth factor exposure, highly differentiated cell types have successfully been derived from ES cells. Recent technological advances such as RNA interference to knock down gene expression in ES cells are also producing enriched populations of cells and elucidating gene function in early development.
Collapse
Affiliation(s)
- K Sue O'Shea
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109-0616, USA.
| |
Collapse
|
17
|
Zhao LX, Zhang J, Cao F, Meng L, Wang DM, Li YH, Nan X, Jiao WC, Zheng M, Xu XH, Pei XT. Modification of the brain-derived neurotrophic factor gene: a portal to transform mesenchymal stem cells into advantageous engineering cells for neuroregeneration and neuroprotection. Exp Neurol 2004; 190:396-406. [PMID: 15530878 DOI: 10.1016/j.expneurol.2004.06.025] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2004] [Revised: 05/04/2004] [Accepted: 06/03/2004] [Indexed: 02/05/2023]
Abstract
Multipotential mesenchymal stem cells (MSCs) are ideal seed cells for recruiting the loss of neural cells due to their strong proliferative capacity, easy acquisition, and considerable tolerance of genetic modifications. After transduction of brain-derived neurotrophic factor (BDNF) gene via recombinant retroviral vectors into the human MSCs, nearly 100% of cells expressed BDNF (which were therefore transformed into BNDF-MSCs) as detected by immunocytochemistry, and the quantity of BDNF in the culture medium was increased by approximately 20,000-fold. In spite of the genomic integration of an exogenous gene, BDNF-MSCs did not present any structural aberration in the chromosomes. All-trans-retinoic acid (RA) induction caused the BDNF-MSCs to differentiate into neural cells with significantly increased expressions of such neural-specific proteins as nestin, NeuN, O4, and glial fibrillary acidic protein (GFAP). The voltage-dependent K+/Ca2+ currents were recorded from the induced BDNF-MSCs using patch-clamp technique. Compared with the MSCs induced by both RA and BDNF, BDNF-MSCs survived in significantly greater number in the induction medium, and also more cells were induced into neuron-like cells (NeuN, P < 0.01) and oligodendrocyte-like cells (O4, P < 0.05). We suppose that, once engrafted into human central nervous system, the BDNF-MSCs would not only recruit the neuronal losses, but also provide, by way of paracrine, large quantities of BDNF that effectively perform the functions of neuroprotection and neuroregeneration, promoting the activation of endogenous neural stem/progenitor cells and their chemotactic migration. On the other hand, the BDNF-MSCs that can survive in the host environment and differentiate subsequently into functional mature cells may also serve as specifically targeting vectors for ex vivo gene therapy.
Collapse
Affiliation(s)
- Lian-Xu Zhao
- Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, Guangdong, 515031, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Hoggarth JH, Jones E, Ensser A, Meredith DM. Functional expression of thymidine kinase in human leukaemic and colorectal cells, delivered as EGFP fusion protein by herpesvirus saimiri-based vector. Cancer Gene Ther 2004; 11:613-24. [PMID: 15243631 DOI: 10.1038/sj.cgt.7700729] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Herpesvirus saimiri (HVS) has the capacity to incorporate large amounts of heterologous DNA and can infect many different human cell types. To develop its potential as a gene therapy vector, we cloned herpes simplex virus thymidine kinase (TK) gene into the HVS genome in the form of an enhanced green fluorescent protein (EGFP) fusion protein, using a cosmid-based approach. At multiplicity of infection = 100 over 90% of human leukemic K562 and Jurkat cells were transduced with HVS/EGFP-TK. Conditions of no selective pressure expression were maintained at > 92% per cell division. Expression of the EGFP-TK fusion protein rendered transfected leukaemic cells sensitive to cytotoxic treatment with the prodrugs ganciclovir (GCV) and (E)-5-(2-bromovinyl)-2'deoxyuridine (BVDU) at concentrations as low as 10 ng/ml. The viral vector was also screened against a panel of colorectal and pancreatic carcinoma cell lines. All cell lines were transduced but showed a range of sensitivity to infection. Three of the most easily transduced cell lines: Mia PaCa, HCT116 and SW948 transduced with HVS/EGFP-TK were effectively ablated by subsequent treatment with GCV or BVDU. Our results show that in its current form HVS/EGFP-TK could be utilized as an antitumour agent, or it could be developed further by inclusion of a therapeutic gene, with TK presence ensuring a mechanism of controlled removal of modified cells when no longer necessary. These results suggest that HVS/EGFP-TK has a great potential for a number of gene therapy applications.
Collapse
Affiliation(s)
- Jennifer H Hoggarth
- Molecular Medicine Unit, University of Leeds, St James' University Hospital, Leeds, UK.
| | | | | | | |
Collapse
|
19
|
Abstract
Cell therapy refers to the transplantation of healthy, functional and propagating cells to restore the viability or function of deficient tissues. Stem cells are characterized by self-renewal and the potential to form differentiated cells. In early mammalian embryos, at the blastocyst stage, the inner cell mass is pluripotent. Thus, it has been recognized that human embryonic stem cells (hESCs), which are derived from such cells of blastocysts, may serve as a source of numerous types of differentiated cells. The first part of this review summarizes different techniques for the derivation and maintenance of undifferentiated hESCs. In the second part, issues concerning the safety and bulk production, which may enable hESCs use in future clinical applications, are presented. The last part of this review details accumulated data regarding the in vitro differentiation potential of hESCs.
Collapse
Affiliation(s)
- Sharon Gerecht-Nir
- Biotechnology Interdisciplinary Unit, Technion - Israel Institute of Technology, Haifa
| | | |
Collapse
|
20
|
Gerecht-Nir S, Itskovitz-Eldor J. Human embryonic stem cells: a potential source for cellular therapy. Am J Transplant 2004; 4 Suppl 6:51-7. [PMID: 14871274 DOI: 10.1111/j.1600-6135.2004.0345.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Many degenerative human diseases reflect damage to cells that are not normally repaired or replaced, such as diabetes, Parkinson's disease, hepatic failure and congestive heart failure. Preliminary studies in animals and humans have suggested that these diseases may be treatable by transplantation of healthy cells. Such cells may be obtained by in vitro culture of embryonic stem cells, which are capable of differentiating into many cell types. This review discusses applicative approaches for the derivation, maintenance and safety of human embryonic stem (hES) cells as well as ethical concerns surrounding their possible source for cellular therapy. hES cells offer broad application in cellular therapy; however, this review specifically emphasizes on cardiovascular repair, generation and characterization of hES cell-derived cardiomyocytes, vascular progenitors and differentiation of derivatives.
Collapse
Affiliation(s)
- Sharon Gerecht-Nir
- Biotechnology Interdisciplinary Unit, Technion - Israel Institute of Technology, Haifa, Israel
| | | |
Collapse
|
21
|
Dawson L, Bateman-House AS, Mueller Agnew D, Bok H, Brock DW, Chakravarti A, Greene M, King PA, O'Brien SJ, Sachs DH, Schill KE, Siegel A, Solter D, Suter SM, Verfaillie CM, Walters LB, Gearhart JD, Faden RR. Safety issues in cell-based intervention trials. Fertil Steril 2003; 80:1077-85. [PMID: 14607552 DOI: 10.1016/s0015-0282(03)02218-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
We report on the deliberations of an interdisciplinary group of experts in science, law, and philosophy who convened to discuss novel ethical and policy challenges in stem cell research. In this report we discuss the ethical and policy implications of safety concerns in the transition from basic laboratory research to clinical applications of cell-based therapies derived from stem cells. Although many features of this transition from lab to clinic are common to other therapies, three aspects of stem cell biology pose unique challenges. First, tension regarding the use of human embryos may complicate the scientific development of safe and effective cell lines. Second, because human stem cells were not developed in the laboratory until 1998, few safety questions relating to human applications have been addressed in animal research. Third, preclinical and clinical testing of biologic agents, particularly those as inherently complex as mammalian cells, present formidable challenges, such as the need to develop suitable standardized assays and the difficulty of selecting appropriate patient populations for early phase trials. We recommend that scientists, policy makers, and the public discuss these issues responsibly, and further, that a national advisory committee to oversee human trials of cell therapies be established.
Collapse
Affiliation(s)
- Liza Dawson
- Phoebe R Berman Bioethics Institute, Johns Hopkins University, Baltimore, Maryland 21205, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Affiliation(s)
- Sharon Gerecht-Nir
- Biotechnology Interdisciplinary Unit, Technion-Israel Institute of Technology, Haifa, Israel
| | | | | |
Collapse
|
23
|
He Q, Li J, Bettiol E, Jaconi ME. Embryonic stem cells: new possible therapy for degenerative diseases that affect elderly people. J Gerontol A Biol Sci Med Sci 2003; 58:279-87. [PMID: 12634295 DOI: 10.1093/gerona/58.3.m279] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The capacity of embryonic stem (ES) cells for virtually unlimited self renewal and differentiation has opened up the prospect of widespread applications in biomedical research and regenerative medicine. The use of these cells would overcome the problems of donor tissue shortage and implant rejection, if the cells are made immunocompatible with the recipient. Since the derivation in 1998 of human ES cell lines from preimplantation embryos, considerable research is centered on their biology, on how differentiation can be encouraged toward particular cell lineages, and also on the means to enrich and purify derivative cell types. In addition, ES cells may be used as an in vitro system not only to study cell differentiation but also to evaluate the effects of new drugs and the identification of genes as potential therapeutic targets. This review will summarize what is known about animal and human ES cells with particular emphasis on their application in four animal models of human diseases. Present studies of mouse ES cell transplantation reveal encouraging results but also technical barriers that have to be overcome before clinical trials can be considered.
Collapse
Affiliation(s)
- Qing He
- Biology of Aging Laboratory, Department of Geriatrics, Geneva University Hospitals, Switzerland
| | | | | | | |
Collapse
|
24
|
Gerecht-Nir S, Fishman B, Itskovitz-Eldor J. Cardiovascular potential of embryonic stem cells. ACTA ACUST UNITED AC 2003; 276:58-65. [PMID: 14699634 DOI: 10.1002/ar.a.10136] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Initial events involved in the process of heart formation consist of myocardial differentiation as well as development of endothelial and endocardial tissues. As only limited means are allocated to the studying of cardiovascular system development, embryonic stem cells (ESCs) isolated from the inner cell mass (ICM) of developing mice or human blastocysts offer the first step toward the understanding of these complex and intriguing events. ESCs are able to differentiate into a wide range of cell types, including various vascular cells and cardiomyocytes, and their self-renewal capability renders them a unique, homogeneous, and unlimited preliminary population of cells for the investigation of early developmental events of cardiovascular system and lineage commitment. This review summarizes the accumulated knowledge of the cellular and molecular mechanisms involved in the development of the cardiovascular system.
Collapse
Affiliation(s)
- Sharon Gerecht-Nir
- Biotechnology Interdisciplinary Unit, Technion, Israel Institute of Technology, Haifa, Israel
| | | | | |
Collapse
|