Skip to main content
Organogenesis logoLink to Organogenesis
. 2013 Jan 1;9(1):34–39. doi: 10.4161/org.24317

Safeguarding clinical translation of pluripotent stem cells with suicide genes

Weiqiang Li 1, 2,, Andy Peng Xiang 1, 2, 3,*
PMCID: PMC3674039  PMID: 23511011

Abstract

The generation of human induced pluripotent stem cells (hiPSCs) opens a new avenue in regenerative medicine. However, transplantation of hiPSC-derived cells carries a risk of tumor formation by residual pluripotent stem cells. Numerous adaptive strategies have been developed to prevent or minimize adverse events and control the in vivo behavior of transplanted stem cells and their progeny. Among them, the application of suicide gene modifications, which is conceptually similar to cancer gene therapy, is considered an ideal means to control wayward stem cell progeny in vivo. In this review, the choices of vectors, promoters, and genes for use in suicide gene approaches for improving the safety of hiPSCs-based cell therapy are introduced and possible new strategies for improvements are discussed. Safety-enhancing strategies that can selectively ablate undifferentiated cells without inducing virus infection or insertional mutations may greatly aid in translating human pluripotent stem cells into cell therapies in the future.

Keywords: induced pluripotent stem cells, suicide gene, stem cell therapy, vector, regenerative medicine

Introduction

Stem cell therapies are one of the most promising areas in medicine and hold great potential for the treatment of degenerative diseases, genetic disorders, and severe injuries that were previously considered refractory to therapeutic intervention.1 Pluripotent stem cells (PSCs), which can undergo extensive proliferation in vitro and give rise to lineages that represent any of the three embryonic germ layers, serve as an unlimited resource for cell-replacement therapy and tissue engineering.2 However, the use of human embryonic stem cells (ESCs), one type of PSCs, for clinical applications has been plagued by highly controversial ethical and legal questions because it requires the destruction of a human embryo.3

It is also possible to reprogram somatic cells to a pluripotent state through somatic cell nuclear transfer (SCNT),4 cell fusion,5 or gene transfer of defined transcription factors.6 Human induced pluripotent stem cells (hiPSCs) derived from adult cells by forced expression of defined transcription factors have attracted considerable attention because their characteristics are indistinguishable from those of inner cell mass-derived hESCs and they offer relatively high reprogramming efficiency without associated ethical dilemmas. These hiPSCs offer an exciting opportunity for elucidating underlying mechanisms of pluripotency and establishin g in vitro models for human disease; they also hold the potential for future clinical applications in regenerative medicine.7,8

Traditionally, hiPSCs have been generated from different kinds of somatic cells, including ðbroblasts, hematopoietic cells, meningiocytes and keratinocytes,9 using a variety of gene delivery methods, including retrovirus (RV) and lentivirus (LV) transduction. hiPSCs generated by these latter methods may cause permanent, and random, transgene insertion into the host genome.6,8 More recently, various non-viral and non-integrating methods, which may enable safe, efficient derivation of hiPSCs suitable for clinical applications, have been developed. These include transient DNA transfection using transposons or minicircle plasmids, protein transduction, and RNA/miRNA (micro RNA) transfection.10 Nevertheless, transcriptional, genetic and epigenetic abnormalities acquired from the corresponding somatic cells of origin or during reprogramming stress and culture adaptation increase the tumorigenicity of hiPSCs.11 In a karyotype analysis of more than 1,700 human iPSC and ESC cultures collected from 97 investigators in 29 laboratories, Taapken et al. reported that trisomy 12 was the predominant abnormality in iPSCs cultures (31.9%), and trisomy 8 occurred more frequently in iPSCs (20%) than in ESCs (10%). More importantly, these authors found that the frequency and types of karyotypic abnormalities were not affected by the reprogramming method.12 Athurva et al.13 reported that 22 hiPSCs lines reprogrammed by different methods (RV, LV, and non-integrating methods including episomal and mRNA delivery) each contained an average of five protein-coding point mutations, and the majority of these mutations were enriched in genes that are cancer promoting or mutated in cancers. Tong et al.14 found that mice generated from tetraploid complementation-competent iPS cells are prone to tumorigenesis. Pancreatic and bone tumors were identified among the iPS-derived mice, whereas ES-derived mice and control mice were all tumor free. Kyoko et al.15 compared the tumorigenicity of neurospheres generated from 36 mouse induced pluripotent stem cell lines. They found that neurospheres from tail tip, fibroblast-derived miPSCs showed the highest propensity for teratoma formation owing to the persistence of undifferentiated cells. Moreover, hiPSCs need to be induced to differentiate before transplantation. To the best of our knowledge, all methods previously used to trigger in vitro differentiation of ES/iPS cells have yielded diverse cell mixtures. These may include undifferentiated or partially differentiated cells that proliferate inappropriately. Cell transplants may also de-differentiate or become transformed to produce tumors, particularly in an in vivo microenvironment.16 Accordingly, it is crucial that these methodological hurdles be overcome before hiPSCs can be translated into the clinic.

A number of strategies, including the use of monoclonal antibodies, recombinant proteins and pharmaceuticals, have been developed to eliminate transferred cells that have gone awry and thereby prevent or minimize the aforementioned adverse events. However, the application of such approaches to date has been limited because they have a finite half-life and/or are only active in dividing cells.16 Suicide genes that can be stably expressed in both quiescent and replicating cells can lead to selective ablation of gene-modified cells without the likelihood of causing collateral damage to contiguous cells and/or tissues. Therefore, suicide gene applications are considered among the most attractive approaches for controlling wayward stem cell progeny in vivo.

Here, we focus on the key strategies that will be used for suicide gene applications in hiPSCs-based therapy, including the choice of vectors, promoters selection markers, and suicide genes, then subsequently provide an update on recent advances of suicide gene modifications of PSCs in order to control the behavior of their wayward progenies in vitro and in animal models.

Vector targeting strategies

Vectors or vehicles are required in suicide gene therapy for efficient and selective delivery of function genes to hiPSCs. A wide variety of vectors for use as gene transfer delivery vehicles have been developed to date for cancer gene therapy, including viral vectors17 and non-viral plasmids18 (Table 1). RV, LV, adenovirus (AV), and adeno-associated virus (AAV) are widely used in cancer gene therapy studies, whereas Epstein Barr virus (EBV), herpes simplex virus (HSV), and baculovirus (BV) are occasionally used. Of these, AV19 and RV20 have been used in clinical trials. Each of these viral vectors has certain drawbacks that restrict their applications in hiPSCs-based cell therapies. Although AVs have wide cellular tropism, they do not integrate into the host genome and cannot be expressed in progenies of transplanted cells, resulting in limited duration of gene expression. AAV,21 a small virus that does not cause disease in humans, integrates at a specific site on chromosome 19 (AAVS1) in the human genome; however, it has a lower transduction efficiency in embryonic stem cells and cannot accommodate a large amount of foreign DNA (≤ 4.7 kb). RVs have relatively high transduction efficiency, but are readily silenced. And when randomly incorporated into the host genome, they may cause insertional mutations. Although LVs22 show high transduction efficiency and are seldom inactivated, they too pose a risk of insertional mutagenesis. EBV,23 HSV,24 and BV25 are all non-integrating viruses and mediate transient transgene expression in human cells, although they can transduce both dividing and non-dividing cells. Non-viral plasmids usually inefficiently transfect target cells and may cause insertional mutagenesis when transferred by liposome or electroporation.

Table 1. Vector targeting strategies.

Vector Integrate Transduction efficiency for PSCs Advantages Obstacles
Adenovirus
no
low
Transduce dividing and non-diving cells
Transient gene expression; post-transduction silencing
Retrovirus
yes
high
Stable transduction
of dividing cells
Post-transduction silencing; random chromosome integration/ oncogene activation
Lentivirus
yes
high
Transduce dividing and non-diving cells; stable transgene expression
Random chromosome integration/ oncogene activation
Adeno-associated virus
yes
low
Stable transduction, integrate in specific site
Accommodates a small amount of foreign DNA
Epstein Barr virus
no
high
Large
cloning capacity (≤ 330 kb)
Transient gene expression
Herpes simplex virus
no
high
Large cloning capacity (≤ 20 kb); neuron specificity
Transient gene expression
Baculovirus
no
high
Large
cloning capacity
(≤ 30 kb)
Transient gene expression
Non-viral plasmid yes low Stable transfection Random chromosome integration; membrane damage

Promoter and selection strategies

The promoters for controlling suicide gene expression can mediate control of cell transplants in vivo and a number of promoters has already been investigated with positive results in cancer gene therapy.16 The promoter chosen to control wayward pluripotent stem cells and their progenies in vivo for clinical applications of hiPSCs should be capable of mediating stable, highly active suicide gene expression in human PSCs. Three kinds of promoters are available for driving suicide gene expression: constitutive, cell specific and inducible.16

Constitutive promoters, such as the cytomegalovirus (CMV) promoter, phosphoglycerate kinase (PGK) promoter and ubiquitin C promoter, are commonly used to drive ectopic gene expression in various gene transfer applications in vitro and in vivo. They usually achieve high levels of gene expression in most cell types.26 However, these promoters may be transcriptional silenced due to extensive methylation. Previous studies have also shown that elongation factor 1α (EF1α) and constitutive cytomegalovirus (CMV) enhancer/chicken β-actin promoter (CAGG promoters are consistently strong in all mammalian cell types tested, whereas the CMV promoter is the most variable, being very strong in some cell types and rather weak in others.27 Norrman et al. found that ACTB (human β-actin promoter), EF1α and PGK promoters showed stable activities during long-term culture of undifferentiated hESCs.26 Therefore, a promoter that can mediate stable, highly active suicide gene expression in human PSCs should be chosen for the clinical translation of hiPSCs in order to control wayward pluripotent stem cells and their progenies in vivo.

Cell-specific promoters can be used for applications in which the suicide gene is active in certain cell types in order to restrict transgene expression to targeted tissues, thereby reducing side effects while increasing therapeutic efficacy. Wu et al.28 developed a viral vector platform combining glial fibrillary acidic protein (GFAP) promoter-based transcriptional targeting with miRNA regulation to control the expression of a gene for glioma suicide gene therapy in the mouse brain. They found that these vectors enabled effective elimination of human glioma xenografts while producing negligible toxic effects on normal astrocytes. Niess et al.29 used bone marrow mesenchymal stem cells (MSCs) that were modified by a suicide gene under the control of the Chemokine (C-C motif) ligand 5 (CCL5) promoter for gene therapy of hepatocellular carcinoma. They found that transplanted MSCs were recruited to the tumor site, where they differentiated and participated in tumor angiogenesis following tumor-speciðc activation of CCL5 promoters. CCL5/HSV-TK-transfected MSCs in combination with ganciclovir (GCV) supplementation signiðcantly reduced tumor growth by 56.4% compared with the control group. As for the removal of PSCs in vitro and in vivo, control of suicide gene expression using a pluripotent cell-specific promoter (e.g., OCT4, Nanog) is a better choice and helps to specifically eliminate residual PSCs or progeny cells that have reverted to a pluripotent state.

An ideal promoter for hiPSC-based cell replacement therapies should allow a suicide gene to be regulated quickly and effectively in vivo by an exogenous signal that activates/inactivates the control cassette. For example, the expression of a suicide gene might be under the control of a regulatable promoter so as to turn on only when the stem cell or its progeny form tumors or over-proliferate following addition of an exogenous signal. Inducible promoters regulated by tetracycline (Tet), cAMP, rapamycin, or steroid hormones have been constructed and successfully utilized in cell lines and animal models.16 The construction of TetON/TetOFF regulatable systems has been reported for all AV, LV, RV, HSV, and AAV vectors. These vectors can tightly regulate transgene expression downstream of a TRE promoter in the presence/absence of tetracycline in vitro and in vivo.30

It is necessary that the genetic modification of all hiPSCs-derived cells should be confirmed before cell replacement therapy. Thus, an efficient selection method is essential for obtaining a pure population of transduced cells. Insertion of an antibiotic coding sequence into vectors is an effective strategy that allows single colonies to be selected for further expansion after antibiotic selection.

Suicide gene strategies

It is of great importance to choose an appropriate functional gene to eliminate tumor-initiating cells before and/or after cell implantation. Suicide genes, including toxin, apoptotic, tagging and drug-conversion genes, are widely used in cancer gene therapy.16 Genes for toxins (e.g., Shiga toxin A1, Pseudomonas exotoxin A) induce cell death by inhibiting protein synthesis,31,32 whereas apoptotic genes (caspase-3, caspase-8, et al.) eliminate cancer cells by inducing apoptosis.33,34 Both kinds of genes are active in dividing and non-dividing cells. For such genes, control by inducible promoters is much preferred to ensure that their expression is totally shut off and is activated only when appropriate signals are administered. A tagging gene expressed in the plasma membrane can be transferred into cells before implantation.18 An anti-tag monoclonal antibody carrying a therapeutic agent is then able to subsequently destroy the tagged cells. This tagging strategy also risks bystander toxicity because of the speciðcity and afðnity of the anti-tag antibody and potential immunogenicity to our body.

Gene-directed enzyme prodrug therapy (GDEPT) is the most widely used strategy in cancer gene therapy.35 This type of suicide gene converts a nontoxic drug to a toxic drug in gene-modified cells. Examples of genes used in GDEPT include herpes simplex virus thymidine kinase (HSV-TK), cytosine deaminase (CD), varicella-zoster thymidine kinase (VZV-TK), and nitroreductase (Table 2). HSV-TK phosphorylates nucleoside analogs, including acyclovir and ganciclovir (GCV, a prodrug approved by the US Food and Drug Administration). The resulting acyclovir triphosphate or GCV triphosphate incorporate into DNA via the action of DNA polymerase, leading to chain termination and cell death.36 As such, they are highly toxic toward dividing cells. The CD gene encodes cytosine deaminase, which converts 5-fluorocytosine (5FC) into the cytotoxic 5-fluorouracil (5-FU), which eradicates cells.37 The VZV-TK enzyme monophosphorylates 6-methoxypurine arabinonucleoside (ara-M) to ara-MMP. Then ara-MMP is metabolized to the highly toxic form, ara-AMP, which can significantly inhibit cell growth.38 Nitroreductase (NR) converts CB1954 to its 4-hydroxylamino derivative and then to acetylate, becoming a powerful bifunctional alkylating agent that causes the formation of poorly repaired DNA crosslinks.39 One other promising suicide gene is carboxypeptidase G2 (CPG2), which catalyzes the hydrolysis of nitrogen mustard prodrugs, then releases glutamic acid and the cognate drug. It has no mammalian equivalent, and no additional activating steps are required to produce the active DNA crosslinking molecule.19,40 All GDEPT systems described above produce a toxic agent that can remove cancer cells efficiently, except for VZV-TK/ara-M, which generates weakly toxic metabolites; thus, other prodrugs need to be developed to augment the efficacy of the VZV-TK suicide gene system against cancer cells. CD, NR, and CPG2 usually convert prodrugs to diffusible toxic drugs; this offers an advantage for in vivo cancer gene therapy because it affects cells in the local milieu besides the tumor (bystander effect). However, HSV-TK and VZV-TK metabolites are phosphorylated and cannot cross the cell membrane, resulting in a lesser bystander effect. GDEPT is preferable in cancer gene therapy because the toxic drug product produces a bystander effect, acting on both the cell expressing the enzyme and cells in the local milieu. However, application of GDEPT to regenerative medicine can problematic because the bystander effect may cause undesirable damage to adjacent normal tissue.

Table 2. Prodrug-activating system.

Gene Prodrug Function Bystander effect
HSV-TK
Ganciclovir
Metabolizes to a triphosphate nucleotide that competes with dGTP for DNA polymerases
weak
CD
5-fluorocytosine
Inhibits thymidylate synthetase and DNA synthesis
strong
VZV-TK
ara-M
Competes with dATP for incorporation into DNA, leading to termination of DNA synthesis
weak
NR
CB1954
DNA interstrand
crosslinking
strong
CPG2 Nitrogen mustard
L-glutamates
DNA interstrand
crosslinking
strong

Suicide gene applications for pluripotent stem cells

The pluripotency of PSCs and the malignant transformation and/or reversion to pluripotency of differentiated cells pose the risks of tumor formation posttransplantation. Genetic strategies of suicide gene system can provide effective and safe control to prevent adverse events. Suicide gene applications for improving the safety of embryonic stem cells have been reported by several different groups (Table 3). Most have utilized lentivectors containing the HSV-TK gene under the control of a constitutive promoter (CMV, ubiquitin C, PGK).41-43 Both prevention and elimination of teratomas has been demonstrated using these GDEPT strategies. More recently, Tang et al.44 found that an antibody against SSEA-5 glycan on human embryonic stem cells enabled the removal of teratoma-forming cells in vitro. However, this method does not function well if a progeny cell reverts to pluripotency in vivo; moreover, it is not known whether normal somatic cells express SSEA-5. Wang et al.45 demonstrated that addition of mifepristone-inducible caspase-1 to mouse ES cells eliminated tumor formation but spared differentiated dopamine neurons, both in vitro and in vivo. However, whether other lineages formed from differentiated caspase-1 cells can survive mifepristone is not known.

Table 3. Safeguarding strategies in pluripotent stem cells.

Vector Promoter Suicide gene Cell types Function Reference
Non-viral plasmid
PGK
HSV-TK
hESCs
Ablation of teratomas
43
Lentivirus
Ubiquitin C
HSV-TK
mESCs
Prevention of teratoma formation
42
Lentivirus
CMV
HSV-TK
mESCs
Ablation of teratoma formation
41
Knock-in at the endogenous locus of OCT4
OCT4
HSV-TK
mESCs
Prevention of teratoma formation in spheroids culture
46
Non-viral plasmid
OCT4
HSV-TK
mESCs
Prevention of teratoma formation
47
SSEA5
NA
Anti-SSEA5 antibody
hESCs/hiPSCs
Prevention of teratoma formation
44
Lentivirus
SFFV
CD/ iCaspase9
Macaca nemestrina iPS cells
Prevention and ablation of teratoma formation
51
Non-viral plasmid
mifepristone-inducible promoter (GAL4 UAS)
Caspase-1
mESCs
Ablation of teratoma formation
45
Knock-in at the endogenous locus of Nanog
Nanog
Enhanced mutant version
of HSV-TK
hESCs
Prevention of teratoma formation
50
Lentivirus EF1a/Nanog DeltaTK/CD hESCs/hiPSCs
mECSs/miPSCs
Prevention and ablation of teratoma formation 52, 53

Selective ablation of undifferentiated ES cells has been achieved through expression of the HSV-TK gene under the control of the Oct4 promoter, followed by GCV treatment.46,47 Oct4 is essential for the maintenance of the pluripotency and self-renewal capacities of ES/iPS cells. However, it has been reported that constitutive expression of Oct4 from an exogenous promoter is not sufficient to prevent ES cell differentiation,48 and it was found that Oct4 is broadly expressed in different cell types.49 Accordingly, Oct4-controlled suicide genes may cause undesirable damage to differentiated cell populations. Furthermore, Naujok et al. reported that Oct4-TK ES cells treated with 1 μM (255 μg/mL) GCV during in vitro differentiation over a relatively long period (14 d) prior to implantation still developed into tumors, albeit significantly smaller tumors than those formed from untreated cells.47 Rong et al.50 reported a scalable approach for preventing teratoma formation by human embryonic stem cells. They introduced a hyperactive variant of the HSV-TK gene into the 3′ untranslated region of the endogenous Nanog gene of hESCs through homologous recombination. Using this approach, they were able to demonstrate elimination of teratomas generated by hESCs without apparent negative impacts on the differentiated cell types derived from them.

Only a few groups have reported the use of suicide genes to improve the safety of induced pluripotent stem cells. CD/5FC and iCaspase/AP20187 (caspase 9) systems have been used to safeguard nonhuman primate iPS cells.51 However, in vitro killing of iCaspase cells by AP20187 was relatively slow compared with that observed in CodA/5-FC cells, and the in vivo function of AP20187 has not yet been determined. In our research,52,53 we modified mouse/human ES/iPS cells to contain the suicide gene deltaTK or CodA under the transcriptional control of the EF1α or Nanog promoter. The suicide gene was introduced via lentivirus transduction without interfering with the self-renewal or pluripotency characteristics of ES/iPS cells. We found that EF1α promoter-controlled deltaTK/CodA expression efficiently eliminated pluripotent stem cells and their derivatives, both in vitro and in vivo. When the suicide gene was under the control of the Nanog promoter, tumor-initiating, undifferentiated pluripotent stem cells were selectively ablated in vitro after prodrug treatment. deltaTK was chosen because this truncated form of HSV-TK enhances the specificity of cell ablation and because deltaTK transgenic male mice are fertile, in contrast to animals expressing intact TK.54 Chambers et al. found that transgenic expression of Nanog was sufficient for clonal expansion of ES cells, independent of Stat3 (signal transducer and activator of transcription 3) activation status, indicating that Nanog is central in the transcription factor hierarchy that defines the ES cell state.55 Our results showed that a much lower concentration of GCV (10 μg/mL vs. 255 μg/mL) and much less time (5 d vs. 14 d) was required to eradicate pluripotent stem cells using the Nanog-deltaTK system than was observed with the Oct4-TK system.

Future directions

An optimal suicide gene strategy for the safe application of hiPSCs is one that has high transfection efficiency and does not cause insertional mutagenesis. Transgene expression should be stable and maintained in all stem cells and their progeny in vitro and in vivo. Unfortunately, the vectors described above do not yet meet all these requirements. One of the best approaches for improvement is to recombine suicide genes in “safe harbor” sites or in the intron of a specific gene (e.g., Nanog, β-actin) of the hiPSCs genome through homologous recombination using new genetic manipulation technologies with high efficiency, such as zinc finger nucleases (ZFNs)56 and transcription activator-like effector nucleases (TALENs).57 ZFNs combine the nonspecific cleavage domain of the FokI endonuclease with DNA-binding domains of zinc finger proteins.59 Genomic alterations, including point mutations, deletions, insertions, inversions, duplications and translocations, can be introduced by ZFNs with high efficiency.60 TALENs that can be very easily and rapidly designed have similar structure and function to ZFNs and have been an alternative to ZFNs for genome editing by introducing targeted double-strand breaks into specific sites of the genome with similar efficiency.60 However, the gene-targeting technology used for controlling wayward stem cells may result in the destruction of all transplanted cells, including properly functioning cells, through constitutive expression of the suicide gene or bystander effects. And when excess proliferation, inappropriate differentiation, and/or improper localization of transplanted cells occur, pluripotent gene promoter-controlled suicide genes may not function well. Therefore, a dual system comprising a suicide gene under the control of constitutive and a cell-specific promoter, both transferred by knock-in technology, would greatly aid in controlling transplanted cells in a “failsafe” manner.

Acknowledgments

This work was supported by National Basic Research Program of China (2012CBA01302, 2010CB945401), National Natural Science Foundation of China (30971675, 81270646, 81271265), the National High Technology Research and Development Program of China (2011AA020108).

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Footnotes

References

  • 1.Weissman IL. Stem cells: units of development, units of regeneration, and units in evolution. Cell. 2000;100:157–68. doi: 10.1016/S0092-8674(00)81692-X. [DOI] [PubMed] [Google Scholar]
  • 2.Keller G. Embryonic stem cell differentiation: emergence of a new era in biology and medicine. Genes Dev. 2005;19:1129–55. doi: 10.1101/gad.1303605. [DOI] [PubMed] [Google Scholar]
  • 3.Hyun I. The bioethics of stem cell research and therapy. J Clin Invest. 2010;120:71–5. doi: 10.1172/JCI40435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Byrne JA, Pedersen DA, Clepper LL, Nelson M, Sanger WG, Gokhale S, et al. Producing primate embryonic stem cells by somatic cell nuclear transfer. Nature. 2007;450:497–502. doi: 10.1038/nature06357. [DOI] [PubMed] [Google Scholar]
  • 5.Cowan CA, Atienza J, Melton DA, Eggan K. Nuclear reprogramming of somatic cells after fusion with human embryonic stem cells. Science. 2005;309:1369–73. doi: 10.1126/science.1116447. [DOI] [PubMed] [Google Scholar]
  • 6.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–76. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
  • 7.Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–72. doi: 10.1016/j.cell.2007.11.019. [DOI] [PubMed] [Google Scholar]
  • 8.Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318:1917–20. doi: 10.1126/science.1151526. [DOI] [PubMed] [Google Scholar]
  • 9.Robinton DA, Daley GQ. The promise of induced pluripotent stem cells in research and therapy. Nature. 2012;481:295–305. doi: 10.1038/nature10761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Narsinh K, Narsinh KH, Wu JC. Derivation of human induced pluripotent stem cells for cardiovascular disease modeling. Circ Res. 2011;108:1146–56. doi: 10.1161/CIRCRESAHA.111.240374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ben-David U, Benvenisty N. The tumorigenicity of human embryonic and induced pluripotent stem cells. Nat Rev Cancer. 2011;11:268–77. doi: 10.1038/nrc3034. [DOI] [PubMed] [Google Scholar]
  • 12.Taapken SM, Nisler BS, Newton MA, Sampsell-Barron TL, Leonhard KA, McIntire EM, et al. Karotypic abnormalities in human induced pluripotent stem cells and embryonic stem cells. Nat Biotechnol. 2011;29:313–4. doi: 10.1038/nbt.1835. [DOI] [PubMed] [Google Scholar]
  • 13.Gore A, Li Z, Fung HL, Young JE, Agarwal S, Antosiewicz-Bourget J, et al. Somatic coding mutations in human induced pluripotent stem cells. Nature. 2011;471:63–7. doi: 10.1038/nature09805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Tong M, Lv Z, Liu L, Zhu H, Zheng QY, Zhao XY, et al. Mice generated from tetraploid complementation competent iPS cells show similar developmental features as those from ES cells but are prone to tumorigenesis. Cell Res. 2011;21:1634–7. doi: 10.1038/cr.2011.143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Miura K, Okada Y, Aoi T, Okada A, Takahashi K, Okita K, et al. Variation in the safety of induced pluripotent stem cell lines. Nat Biotechnol. 2009;27:743–5. doi: 10.1038/nbt.1554. [DOI] [PubMed] [Google Scholar]
  • 16.Kiuru M, Boyer JL, O’Connor TP, Crystal RG. Genetic control of wayward pluripotent stem cells and their progeny after transplantation. Cell Stem Cell. 2009;4:289–300. doi: 10.1016/j.stem.2009.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Yang NS, Sun WH. Gene gun and other non-viral approaches for cancer gene therapy. Nat Med. 1995;1:481–3. doi: 10.1038/nm0595-481. [DOI] [PubMed] [Google Scholar]
  • 18.Hewitt Z, Priddle H, Thomson AJ, Wojtacha D, McWhir J. Ablation of undifferentiated human embryonic stem cells: exploiting innate immunity against the Gal alpha1-3Galbeta1-4GlcNAc-R (alpha-Gal) epitope. Stem Cells. 2007;25:10–8. doi: 10.1634/stemcells.2005-0481. [DOI] [PubMed] [Google Scholar]
  • 19.Schepelmann S, Hallenbeck P, Ogilvie LM, Hedley D, Friedlos F, Martin J, et al. Systemic gene-directed enzyme prodrug therapy of hepatocellular carcinoma using a targeted adenovirus armed with carboxypeptidase G2. Cancer Res. 2005;65:5003–8. doi: 10.1158/0008-5472.CAN-05-0393. [DOI] [PubMed] [Google Scholar]
  • 20.Hiraoka K, Kimura T, Logg CR, Kasahara N. Tumor-selective gene expression in a hepatic metastasis model after locoregional delivery of a replication-competent retrovirus vector. Clin Cancer Res. 2006;12:7108–16. doi: 10.1158/1078-0432.CCR-06-1452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Schoensiegel F, Paschen A, Sieger S, Eskerski H, Mier W, Rothfels H, et al. MIA (melanoma inhibitory activity) promoter mediated tissue-specific suicide gene therapy of malignant melanoma. Cancer Gene Ther. 2004;11:408–18. doi: 10.1038/sj.cgt.7700721. [DOI] [PubMed] [Google Scholar]
  • 22.Yu D, Scott C, Jia WW, De Benedetti A, Williams BJ, Fazli L, et al. Targeting and killing of prostate cancer cells using lentiviral constructs containing a sequence recognized by translation factor eIF4E and a prostate-specific promoter. Cancer Gene Ther. 2006;13:32–43. doi: 10.1038/sj.cgt.7700885. [DOI] [PubMed] [Google Scholar]
  • 23.Paillard F. Epstein-Barr virus vectors for the treatment of Epstein-Barr virus-associated cancers. Hum Gene Ther. 1998;9:1119–20. doi: 10.1089/hum.1998.9.8-1119. [DOI] [PubMed] [Google Scholar]
  • 24.Detta A, Harland J, Hanif I, Brown SM, Cruickshank G. Proliferative activity and in vitro replication of HSV1716 in human metastatic brain tumours. J Gene Med. 2003;5:681–9. doi: 10.1002/jgm.396. [DOI] [PubMed] [Google Scholar]
  • 25.Zhao Y, Lam DH, Yang J, Lin J, Tham CK, Ng WH, et al. Targeted suicide gene therapy for glioma using human embryonic stem cell-derived neural stem cells genetically modified by baculoviral vectors. Gene Ther. 2012;19:189–200. doi: 10.1038/gt.2011.82. [DOI] [PubMed] [Google Scholar]
  • 26.Norrman K, Fischer Y, Bonnamy B, Wolfhagen Sand F, Ravassard P, Semb H. Quantitative comparison of constitutive promoters in human ES cells. PLoS One. 2010;5:e12413. doi: 10.1371/journal.pone.0012413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Qin JY, Zhang L, Clift KL, Hulur I, Xiang AP, Ren BZ, et al. Systematic comparison of constitutive promoters and the doxycycline-inducible promoter. PLoS One. 2010;5:e10611. doi: 10.1371/journal.pone.0010611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Wu C, Lin J, Hong M, Choudhury Y, Balani P, Leung D, et al. Combinatorial control of suicide gene expression by tissue-specific promoter and microRNA regulation for cancer therapy. Mol Ther. 2009;17:2058–66. doi: 10.1038/mt.2009.225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Niess H, Bao Q, Conrad C, Zischek C, Notohamiprodjo M, Schwab F, et al. Selective targeting of genetically engineered mesenchymal stem cells to tumor stroma microenvironments using tissue-specific suicide gene expression suppresses growth of hepatocellular carcinoma. Ann Surg. 2011;254:767–74, discussion 774-5. doi: 10.1097/SLA.0b013e3182368c4f. [DOI] [PubMed] [Google Scholar]
  • 30.Goverdhana S, Puntel M, Xiong W, Zirger JM, Barcia C, Curtin JF, et al. Regulatable gene expression systems for gene therapy applications: progress and future challenges. Mol Ther. 2005;12:189–211. doi: 10.1016/j.ymthe.2005.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Nakayama K, Pergolizzi RG, Crystal RG. Gene transfer-mediated pre-mRNA segmental trans-splicing as a strategy to deliver intracellular toxins for cancer therapy. Cancer Res. 2005;65:254–63. [PubMed] [Google Scholar]
  • 32.Mazor R, Vassall AN, Eberle JA, Beers R, Weldon JE, Venzon DJ, et al. Identification and elimination of an immunodominant T-cell epitope in recombinant immunotoxins based on Pseudomonas exotoxin A. Proc Natl Acad Sci U S A. 2012;109:E3597–603. doi: 10.1073/pnas.1218138109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Yamabe K, Shimizu S, Ito T, Yoshioka Y, Nomura M, Narita M, et al. Cancer gene therapy using a pro-apoptotic gene, caspase-3. Gene Ther. 1999;6:1952–9. doi: 10.1038/sj.gt.3301041. [DOI] [PubMed] [Google Scholar]
  • 34.Carlotti F, Zaldumbide A, Martin P, Boulukos KE, Hoeben RC, Pognonec P. Development of an inducible suicide gene system based on human caspase 8. Cancer Gene Ther. 2005;12:627–39. doi: 10.1038/sj.cgt.7700825. [DOI] [PubMed] [Google Scholar]
  • 35.Springer CJ, Niculescu-Duvaz I. Prodrug-activating systems in suicide gene therapy. J Clin Invest. 2000;105:1161–7. doi: 10.1172/JCI10001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Moolten FL. Tumor chemosensitivity conferred by inserted herpes thymidine kinase genes: paradigm for a prospective cancer control strategy. Cancer Res. 1986;46:5276–81. [PubMed] [Google Scholar]
  • 37.Tiraby M, Cazaux C, Baron M, Drocourt D, Reynes JP, Tiraby G. Concomitant expression of E. coli cytosine deaminase and uracil phosphoribosyltransferase improves the cytotoxicity of 5-fluorocytosine. FEMS Microbiol Lett. 1998;167:41–9. doi: 10.1111/j.1574-6968.1998.tb13205.x. [DOI] [PubMed] [Google Scholar]
  • 38.Huber BE, Richards CA, Krenitsky TA. Retroviral-mediated gene therapy for the treatment of hepatocellular carcinoma: an innovative approach for cancer therapy. Proc Natl Acad Sci U S A. 1991;88:8039–43. doi: 10.1073/pnas.88.18.8039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Clark AJ, Iwobi M, Cui W, Crompton M, Harold G, Hobbs S, et al. Selective cell ablation in transgenic mice expression E. coli nitroreductase. Gene Ther. 1997;4:101–10. doi: 10.1038/sj.gt.3300367. [DOI] [PubMed] [Google Scholar]
  • 40.Hedley D, Ogilvie L, Springer C. Carboxypeptidase-G2-based gene-directed enzyme-prodrug therapy: a new weapon in the GDEPT armoury. Nat Rev Cancer. 2007;7:870–9. doi: 10.1038/nrc2247. [DOI] [PubMed] [Google Scholar]
  • 41.Jung J, Hackett NR, Pergolizzi RG, Pierre-Destine L, Krause A, Crystal RG. Ablation of tumor-derived stem cells transplanted to the central nervous system by genetic modification of embryonic stem cells with a suicide gene. Hum Gene Ther. 2007;18:1182–92. doi: 10.1089/hum.2007.078. [DOI] [PubMed] [Google Scholar]
  • 42.Cao F, Lin S, Xie X, Ray P, Patel M, Zhang X, et al. In vivo visualization of embryonic stem cell survival, proliferation, and migration after cardiac delivery. Circulation. 2006;113:1005–14. doi: 10.1161/CIRCULATIONAHA.105.588954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Schuldiner M, Itskovitz-Eldor J, Benvenisty N. Selective ablation of human embryonic stem cells expressing a “suicide” gene. Stem Cells. 2003;21:257–65. doi: 10.1634/stemcells.21-3-257. [DOI] [PubMed] [Google Scholar]
  • 44.Tang C, Lee AS, Volkmer JP, Sahoo D, Nag D, Mosley AR, et al. An antibody against SSEA-5 glycan on human pluripotent stem cells enables removal of teratoma-forming cells. Nat Biotechnol. 2011;29:829–34. doi: 10.1038/nbt.1947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Wang Y, Yang D, Song L, Li T, Yang J, Zhang X, et al. Mifepristone-inducible caspase-1 expression in mouse embryonic stem cells eliminates tumor formation but spares differentiated cells in vitro and in vivo. Stem Cells. 2012;30:169–79. doi: 10.1002/stem.1000. [DOI] [PubMed] [Google Scholar]
  • 46.Hara A, Aoki H, Taguchi A, Niwa M, Yamada Y, Kunisada T, et al. Neuron-like differentiation and selective ablation of undifferentiated embryonic stem cells containing suicide gene with Oct-4 promoter. Stem Cells Dev. 2008;17:619–27. doi: 10.1089/scd.2007.0235. [DOI] [PubMed] [Google Scholar]
  • 47.Naujok O, Kaldrack J, Taivankhuu T, Jörns A, Lenzen S. Selective removal of undifferentiated embryonic stem cells from differentiation cultures through HSV1 thymidine kinase and ganciclovir treatment. Stem Cell Rev. 2010;6:450–61. doi: 10.1007/s12015-010-9148-z. [DOI] [PubMed] [Google Scholar]
  • 48.Niwa H, Miyazaki J, Smith AG. Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat Genet. 2000;24:372–6. doi: 10.1038/74199. [DOI] [PubMed] [Google Scholar]
  • 49.Liedtke S, Stephan M, Kögler G. Oct4 expression revisited: potential pitfalls for data misinterpretation in stem cell research. Biol Chem. 2008;389:845–50. doi: 10.1515/BC.2008.098. [DOI] [PubMed] [Google Scholar]
  • 50.Rong Z, Fu X, Wang M, Xu Y. A scalable approach to prevent teratoma formation of human embryonic stem cells. J Biol Chem. 2012;287:32338–45. doi: 10.1074/jbc.M112.383810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Zhong B, Watts KL, Gori JL, Wohlfahrt ME, Enssle J, Adair JE, et al. Safeguarding nonhuman primate iPS cells with suicide genes. Mol Ther. 2011;19:1667–75. doi: 10.1038/mt.2011.51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Cheng F, Ke Q, Chen F, Cai B, Gao Y, Ye C, et al. Protecting against wayward human induced pluripotent stem cells with a suicide gene. Biomaterials. 2012;33:3195–204. doi: 10.1016/j.biomaterials.2012.01.023. [DOI] [PubMed] [Google Scholar]
  • 53.Chen F, Cai B, Gao Y, Yuan X, Cheng F, Wang T, et al. Suicide gene-mediated ablation of tumor-initiating mouse pluripotent stem cells. Biomaterials. 2013;34:1701–11. doi: 10.1016/j.biomaterials.2012.11.018. [DOI] [PubMed] [Google Scholar]
  • 54.Salomon B, Maury S, Loubière L, Caruso M, Onclercq R, Klatzmann D. A truncated herpes simplex virus thymidine kinase phosphorylates thymidine and nucleoside analogs and does not cause sterility in transgenic mice. Mol Cell Biol. 1995;15:5322–8. doi: 10.1128/mcb.15.10.5322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Chambers I, Colby D, Robertson M, Nichols J, Lee S, Tweedie S, et al. Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell. 2003;113:643–55. doi: 10.1016/S0092-8674(03)00392-1. [DOI] [PubMed] [Google Scholar]
  • 56.Hockemeyer D, Soldner F, Beard C, Gao Q, Mitalipova M, DeKelver RC, et al. Efficient targeting of expressed and silent genes in human ESCs and iPSCs using zinc-finger nucleases. Nat Biotechnol. 2009;27:851–7. doi: 10.1038/nbt.1562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Ding Q, Lee YK, Schaefer EA, Peters DT, Veres A, Kim K, et al. A TALEN Genome-Editing System for Generating Human Stem Cell-Based Disease Models. Cell Stem Cell. 2013;12:238–51. doi: 10.1016/j.stem.2012.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Le Provost F, Lillico S, Passet B, Young R, Whitelaw B, Vilotte JL. Zinc finger nuclease technology heralds a new era in mammalian transgenesis. Trends Biotechnol. 2010;28:134–41. doi: 10.1016/j.tibtech.2009.11.007. [DOI] [PubMed] [Google Scholar]
  • 60.Joung JK, Sander JD. TALENs: a widely applicable technology for targeted genome editing. Nat Rev Mol Cell Biol. 2013;14:49–55. doi: 10.1038/nrm3486. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Organogenesis are provided here courtesy of Taylor & Francis

RESOURCES