NIH Public Access Author Manuscript Curr Opin Mol Ther. Author manuscript; available in PMC 2011 October 1.

NIH-PA Author ManuscriptPublished NIH-PA Author Manuscript in final edited NIH-PA Author Manuscript form as: Curr Opin Mol Ther. 2010 October ; 12(5): 546±552.

The use of neural stem cells in therapy: Predicting the path to the clinic

Atique U Ahmed, Nikita G Alexiades, and Maciej S Lesniak* The University of Chicago, The Tumor Center, 5841 South Maryland Avenue, MC 3026, Chicago, IL 60637, USA

Abstract Gene therapy is a novel means of anticancer treatment that has led to preliminary positive results in the preclinical setting, as well as in clinical trials; however, successful clinical application of this approach has been hampered by the inability of gene delivery systems to target tumors and to deliver a therapeutic payload to disseminated tumor foci efficiently. Along with viral vector systems, various mammalian cells with tropism for tumor cells have been considered as vehicles for delivery of anticancer therapeutics. The discovery of the inherent tumor-tropic properties of neural stem cells (NSCs) has provided a unique opportunity to develop targeted therapies that use NSCs as a vehicle to track invasive tumor cells and deliver anticancer agents selectively to diseased areas. Many in vivo and studies have demonstrated that the targeted migration of NSCs to infiltrative brain tumors, including malignant glioma, provides a potential therapeutic approach. In this review, the development of NSCs as targeted carriers for anticancer gene therapy is discussed, and barriers in the path to the clinic, as well as approaches to overcoming such barriers are presented.

Keywords Brain malignancy; carrier; gene therapy; glioma; NSC; neural stem cell; targeted tumor therapy

Introduction Neural stem cells (NSCs) are defined as CNS progenitor cells that have the capacity for self- renewal and the potential to differentiate into three major types of CNS cells: (i) ; (ii) astrocytes; and (iii) oligodendrocytes [1]. NSCs are derived from fetal, neonatal or postnatal tissue, and have received substantial research attention because of their potential for treating neurodegenerative . Researchers have speculated that it may be possible to formulate a novel cell replacement platform for such diseases by harnessing the multipotent nature of NSCs.

Since 2000, a number of in vitro and in vivo studies have demonstrated the unique migratory capacity of NSCs throughout the brain [2,3]. In 2000, data from several research groups demonstrated that NSCs transplanted into animal models of brain neoplasia were detected near metastatic tumor beds, far from the original transplant site [2,3]. This finding galvanized the initiation of research into stem cell-based delivery of anticancer agents targeted toward disseminated tumors in the brain. This review focuses on the characteristics of NSCs that make

© Thomson Reuters (Scientific) Ltd *To whom correspondence should be addressed: [email protected]. Ahmed et al. Page 2

these cells ideal as therapeutic delivery vehicles for CNS malignancies, and discusses possible barriers to the translation of NSC-based gene therapy into the clinic. NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript Neural stem cells as gene therapy carriers in CNS malignancies The ideal cell carrier system for cancer gene therapy must exhibit three important characteristics [4]. First, the carrier cell must possess tumor-selective migratory capacity. Second, the carrier cell must be receptive to in vitro genetic manipulation to allow for the expression of a large quantity of selected therapeutic . Alternatively, if the carrier system is used to carry tumor-selective oncolytic viruses, then the carrier cell must be permissive to the virus and must be able to support viral replication. Finally, the carrier cell must be able to carry the therapeutic agent to the tumor while protecting it from the host immune system. In the following sections, the different properties of NSCs are described, to demonstrate their potential as cell carriers for antiglioma gene therapy.

Inherent tumor tropism The ability of NSCs to travel extensively throughout the brain and to migrate to tumor beds is central to their role as cellular vehicles for targeted anticancer therapies [2,5]. Most of the early preclinical studies investigating the tumor-homing properties of NSCs were conducted in intracranial glioma models [6]. When fluorescence-labeled NSCs were transplanted into rodent in the hemisphere contralateral to established tumors, the cells rapidly crossed the midline and migrated significant distances to locate the tumor mass [2]. The ability of NSCs to ‘seek out’ tumors in the brain is not limited to glial ; human NSCs can also target [7] and brain metastases [8], as well as intracerebral medulloblastomas [9] and disseminated [10].

The precise mechanism governing the tumor-tropic properties of NSCs is not fully understood. It is possible that gradients of agents such as chemokines and pro-angiogenic growth factors produced in the tumor microenvironment may act as chemoattractants for NSCs [11]. For example, stem cell trafficking toward ischemic tissue is mediated by hypoxia through the associated upregulation of the factor hypoxia-inducible factor-1α (HIF-1α) [12]. Similar to ischemia, hypoxia is a critical feature of gliomas that results in HIF-1α-mediated upregulation of numerous pro-angiogenic factors and chemoattractants [13,14]. In 2008, Zhao et al investigated the role of hypoxia in NSC migration and observed that NSCs preferentially distribute to hypoxic areas within intracranial glioma xenografts [15]. SiRNA-mediated knockdown of HIF-1α in glioma cells reduced the expression of stromal cell-derived factor-1 (SDF-1), urokinase-type plasminogen activator (uPA) and VEGF, resulting in blocked tumor- tropic migration of the NSCs [15]. Numerous other , growth factors and receptors have been implicated in the tumor-homing properties of NSCs, including stem cell factor (SCF)/c-Kit [16], monocyte chemotactic -1 (MCP-1)/CCL2 [17], annexin A2 [18], hepatocyte growth factor (HGF)/c-Met [19] and VEGF/VEGFR [20] (summarized in Table 1). The multiple homing mechanisms used by NSCs support their use as delivery vehicles over other targeting strategies that primarily use a single-factor approach, such as conjugation or tumor-selective recombinant viral vectors, as NSCs should enable dynamic targeting of heterogenous malignancies. Although the reason why NSCs are recruited to tumor beds remains unclear, one hypothesis is that the cells migrate to the tumor site to repair damaged tissue [5,21]. By pursuing a better understanding of this process, approaches leading to improved neurological recovery following antiglioma therapy can likely be formulated.

Although there is sufficient evidence from rodent tumor models to support the glioma-tropic properties of NSCs, data to demonstrate whether NSCs can track and target glioma cells in the human brain are lacking. Because of the size difference between the rodent and human brain, the distances that delivery agents are required to travel in order to locate disseminated tumor

Curr Opin Mol Ther. Author manuscript; available in PMC 2011 October 1. Ahmed et al. Page 3

cells in human brains will be considerably further from the site of transplantation than in rodent brains. Moreover, it is not clear how tumor size will influence the migration kinetics of NSCs.

NIH-PA Author Manuscript NIH-PA Author ManuscriptCareful NIH-PA Author Manuscript preclinical characterization of these parameters will be critical for the successful translation of NSC-based cell carriers to the clinical setting.

Immunosuppressive properties As noted, the clinical outcome of an anticancer gene therapy depends on the successful delivery of the therapeutic payload at tumor sites, as well as the three-way interaction among the delivery vehicle, tumor microenvironment and host immune system [4]. The majority of currently available cancer gene therapies fail to sustain antitumor effects in the tumor microenvironment for a sufficient length of time to achieve clinically relevant therapeutic efficacy, partly because of the development of a host immune response against the administered therapeutic agents [22]. For example, in both preclinical [23] and clinical [24] settings, the anticancer activity of an oncolytic adenovirus (ONYX-015) was significantly restricted by the host immune response. Attempts to suppress the host immune system with cyclophosphamide in order to enhance the effects of anticancer gene therapy have been successful in rat models [23].

A significant amount of preclinical data suggests that in vivo transplanted NSCs can act as immunosuppressants [25]. Results from several studies in both rodent and non-human primate models of experimental autoimmune encephalomyelitis (EAE) indicate that NSCs transplanted by either intrathecal or intravenous injection promote bystander immunomodulation within the CNS through the release of various soluble molecules [25-27]. The NSCs migrate to areas of inflammation [28], where their presence is associated with a significant downregulation of the effector functions of EAE-specific reactive T-cells, APCs and microglia [27,29] and, thus, contribute to a reduction in the clinical severity of EAE.

The immunosuppressive properties of NSCs represent an attractive attribute for a therapeutic delivery agent, as they allow oncolytic viruses and other therapeutic agents to be shielded from host immunosurveillance. In theory, NSCs can suppress the immune system locally at delivery sites, allowing the therapeutic gene/oncolytic virus to be expressed, or to replicate for a longer time period, thereby killing tumor cells without immune interference. In 2009, Mader et al demonstrated that a mesenchymal stem cell (MSC) carrier system, which possesses similar immunosuppressive properties to NSCs, protected an oncolytic measles virus from antibody neutralization in an orthotopic model in mice [30]. Further characterization of the molecular nature of NSC-mediated immunosuppression and its potential application to enhance the therapeutic efficacy of cancer gene therapy will likely support the argument for the use of NSCs as a cell carrier system.

Tumor-selective therapeutic gene delivery approaches using neural stem cells In the past 10 years, NSCs have been used primarily in two ways as delivery vehicles for anticancer gene therapy. In one approach, NSCs have been genetically engineered to express various therapeutic genes and to carry these products selectively to tumor foci. A number of therapeutic systems have been evaluated in preclinical studies for this purpose, including prodrug-activating enzymes, immunomodulatory cytokines and with anti-angiogenic activity. In a second approach, NSCs have been used as tumor-specific carriers for oncolytic viruses or other viral vectors, protecting the viruses from host immunosurveillance and selectively delivering the therapeutic agent to the tumor site. The genetically engineered approaches of suicide gene therapy and immunomodulatory gene therapy, and the tumor- specific carrier approach involving oncolytic virus-loaded NSCs, are discussed in more detail in the following sections.

Curr Opin Mol Ther. Author manuscript; available in PMC 2011 October 1. Ahmed et al. Page 4

Suicide gene therapy approaches The majority of studies on NSC-based anticancer therapy have used an enzyme-prodrug suicide

NIH-PA Author Manuscript NIH-PA Author Manuscriptgene NIH-PA Author Manuscript therapy system in which NSCs are modified to express genes coding for an enzyme that can convert a systemically administered inactive prodrug into toxic metabolites at tumor sites [31]. Two systems have been used to modify NSCs for this purpose. The most commonly used system involves the HSV-thymidine kinase (HSV-tk) gene, which converts the inactive prodrug ganciclovir (GCV) into its toxic metabolite GCV-triphosphate [32]. This system has been tested in several orthotopic malignant glioma models using various immortalized NSC lines. In 2010, Zhao and Wang delivered HSV-tk-expressing immortalized human NT2 NSCs to a mouse glioblastoma model, and observed a significant increase in the survival of treated animals relative to controls [33]. One of the earliest suicide gene therapy systems used to modify NSCs involved the cytosine deaminase (CD) gene, which converts inactive 5- fluorocytosine (5-FC) into highly toxic 5-fluorouracil (5-FU) [2]. Aboody and colleagues used this system extensively to modify HB1.F3 NSC lines, demonstrating a decrease in the tumor volume of mice bearing orthotopic gliomas or intracranial of 70 to 80% [34].

One of the attractive characteristics of suicide gene therapy is the in vivo bystander effect, defined as the ability of surrounding tumor cells that do not carry the therapeutic transgene to be killed [32,35]. This approach enables the amplification of the antitumor activity of NSCs carrying a suicide gene. Moreover, the inherent tumor-homing properties of NSCs allow these cells to seek tumor sites that are at a far distance from the original mass, and to kill non- transduced tumor cells via this potent bystander effect. In addition to these benefits, the majority of the inactive prodrug, delivered systemically, is able to cross the blood-brain barrier when carried by NSCs; thus, this approach is particularly well suited for the treatment of CNS malignancy [2]. In the future, it will be critical to improve the understanding of the molecular mechanisms underlying the tumor-tropic properties of NSCs, and to develop ways to improve their migratory capacity so that a greater number of engineered NSCs can track multiple disparate tumors for more effective therapy.

Immunomodulatory gene therapy approaches Immortalized NSCs have been engineered to express immunomodulatory genes that have been established to act as effective anticancer agents. IL-4 [3], IL-12 [36], IL-23 [37] and IFNβ [36] produced by NSCs have been demonstrated to be effective in controlling tumor progression. For example, in rats with glioma, IFNβ-expressing NSCs in combination with CD resulted in a 60% reduction in tumor volume with 100% survival, compared with a rate of 70% survival with CD alone [38]. However, inflammatory -induced cellular signals alter the NSC properties of self-renewal and progenitor cell differentiation [39], with potential implications for the intrinsic tumor-homing properties of the cells. The immunomodulatory gene to be expressed must be selected carefully to ensure that the tumor-tropism of NSCs is not negatively affected, thus avoiding suppression of the sustained migration to the tumor and to distant sites.

Oncolytic virus-loaded neural stem cells for antiglioma therapy Oncolytic virus (OV) therapy for cancer is a novel approach in which viruses are modified to replicate preferentially in tumor cells. These viral vectors have the ability to amplify a therapeutic gene in a tumor-selective manner, and can also kill neoplastic cells through tumor cell-specific replication. In the past 20 years, several promising OVs with antiglioma activity have been developed [40]. The human immune system, however, poses a significant challenge in the development of effective oncolytic viral therapy against gliomas. Early in vivo experiments with OVs demonstrated that infected virus-producing cells could enhanced atitumor activity when administered instead of naked virus [41,42]. This finding led to the hypothesis that carrier cells can be used to conceal the therapeutic virus from the host immune

Curr Opin Mol Ther. Author manuscript; available in PMC 2011 October 1. Ahmed et al. Page 5

system and amplify its volume while being transported toward the target site. In 2009, Tyler et al demonstrated the enhanced delivery of an oncolytic adenovirus when loaded into NSCs

NIH-PA Author Manuscript NIH-PA Author Manuscriptin a NIH-PA Author Manuscript mouse xenograft model [43]. Tumors receiving virus-loaded NSCs exhibited an overall reduction in the mean tumor volume compared with tumors treated with the virus alone [43].

Choosing a neural stem cell line for cancer gene therapy There is substantial diversity among the NSC lines currently available, with some being potentially unsuitable as delivery vehicles. The cell lines differ in their source, required culture conditions, tumor-tropic migratory kinetics, ability to express therapeutic genes, and even in their methods of immortalization. A comprehensive approach is necessary to compare the various cell lines to assess these parameters in order to select the most suitable NSC line for clinical application. By 2008, only NSCs from fetal tissue-derived cells had been scaled up under GMP grade conditions, and these were the only NSCs available for clinical trials [44]. The use of these GMP-grade cell lines may be favorable for generating consistent experimental outcomes, and for potentially accelerating the translation of NSC-based anticancer therapies into the clinic. In addition, once each cell line has been genetically manipulated, its phenotype and karyotype, as well as its microbiological status, should be carefully assessed. According to existing EMEA and FDA guidelines, information detailing the source of the stem cell line and its method of manipulation should also be freely available to the public and the scientific community [45,46]. In the future, an important challenge for the rapid clinical translation of NSC-based anticancer therapy will be the consensus among researchers worldwide on a common set of parameters for different NSC lines (eg, culture conditions and characterization criteria).

The use of immortalized neural stem cells An ideal cell delivery system should be stable in tissue culture and be capable of the sustained expression of therapeutic molecules. A primary cell must be immortalized by the introduction of an in order to become stable in culture; such stability allows the cell to continue to expand beyond the time at which senescence would usually be reached. Several genes have been used to immortalize NSCs for their use as cell carriers, including the human telomerase gene (hTERT) [47], SV40 (simian vacuolating virus 40) large T- [48] and the proto- oncogene v-myc [49]. Although most of these immortalized cells are well characterized, and have been demonstrated not to result in the formation of tumors in SCID mice for up to 12 months [6], there are concerns regarding the safety of such cells in the clinical setting. Results from earlier cell-based gene therapy clinical trials have suggested that the most serious toxicity related to this approach is the potential of developing secondary malignancies [48]. In 2009, it was reported that a boy with ataxia telangiectasia (AT) had been diagnosed with a donor- derived multifocal brain tumor 4 years following treatment with intracerebellar injections of allogeneic human fetal NSCs [50]. Although the stem cell line used in that study was not well characterized, and patients with AT have a higher incidence of cancer as a result of their severely compromised immune systems [51], the findings described in this report provided the first clinical evidence that such concerns may be valid. In addition, the most common method of introducing therapeutic genes into NSCs is through retro- and lentiviral vectors. This method can induce insertional mutagenesis, which may also lead to secondary malignancies [52]. To circumvent these problems, clonal populations of modified NSCs must be isolated and expanded, and the site of DNA integration must be mapped prior to the clinical use of NSCs.

The use of allogeneic neural stem cell lines As a result of HLA mismatch, the transplantation of allogeneic NSCs into patients may require immunosuppression to prevent host rejection. Despite the low expression of MHC class II and costimulatory molecules on NSCs, in vitro allorecognition of these cells by peripheral blood

Curr Opin Mol Ther. Author manuscript; available in PMC 2011 October 1. Ahmed et al. Page 6

lymphocytes has been reported in cell models [53]. In a phase II clinical trial in patients with Huntington’s , alloimmunization was observed in 4 out of 13 patients transplanted with

NIH-PA Author Manuscript NIH-PA Author Manuscriptfetal NIH-PA Author Manuscript NSCs [54]. Furthermore, one patient exhibited a significant rejection reaction, although a full recovery was made following immunosuppressive treatment. These observations support the use of autologous NSC sources. However, currently available technologies for isolating and expanding autologous NSCs in culture exhibit serious limitations in terms of their ability to produce a sufficient number of viable cells for successful transplantation. The acquisition of autologous NSCs from an adult patient currently requires invasive procedures, which generally obtain only small numbers of cells that can be difficult to expand in culture [55,56]. In 2009, Casalbore and colleagues were able to isolate and expand NSCs from patients who had undergone unilateral olfactory bulb removal for benign conditions [57]; however, this process requires further optimization before it can be used routinely. Until technology allowing unlimited expansion of autologous primary cells to express a therapeutic gene becomes available, well-characterized NSC lines are the most viable option.

Conclusion NSC derivatives have demonstrated potent inherent tumor-tropic properties in a variety of preclinical brain tumor models. The available evidence supports the theory that an NSC-based delivery system can be clinically useful in the targeted delivery of a therapeutic agent to widely disseminated tumors. Genetic manipulation of NSCs to express a variety of anticancer molecules has resulted in clinically relevant therapeutic efficacy in various xenograft animal models. However, the main issues that remain to be addressed in preclinical studies are the identification of an optimal NSC type and determination of the most effective therapeutic molecules. The tumor-homing efficiency of NSCs will likely be the most important aspect to consider for the successful translation of NSC-based cell carriers in the treatment of brain malignancies in humans. The number of NSCs that have the capacity to reach tumor satellites located a significant distance from the implantation site will likely be limited. Therefore, NSCs must not only maintain their natural tropism following modification, but may even require enhancement. The therapeutic molecule expressed by an engineered NSC must have strong antitumor activity to be able to suppress the spread of malignancies throughout the CNS. The amplification of therapeutic effects, through mechanisms such as oncolytic viruses or the bystander effect, would therefore significantly facilitate the successful translation of this strategy into the clinic. As a result of their intrinsic characteristics, NSCs are more suitable as therapeutic delivery vehicles for CNS malignancies compared with other cell types such as MSCs. Unlike MSCs, which might be the preferred cell carrier for other organs, such as the breast and ovaries, [30], NSCs have the ability to integrate into the host brain without disrupting usual brain function. However, this innovative therapeutic approach carries a substantial risk of secondary malignancy following transformation of the implanted NSCs. Thus, engineered NSCs must be extensively characterized before they can be considered appropriate for clinical use. Safety measures should be established for any potential NSC-based therapy, such as the introduction of a suicide gene (eg, encoding CD) that could facilitate the rapid killing of abnormally growing transplanted NSCs by the administration of a prodrug (eg, 5- fluorocytosine). Once these issues have been addressed in the preclinical setting, the implantation of genetically modified NSCs into the resection cavity wall during removal of the primary tumor might be sufficient to allow the NSCs to migrate to residual disseminated tumor sites and to deliver their associated therapeutic agent.

Acknowledgments

Research by the authors was supported by the NCI (R01CA122930, R01CA138587), the National Institute of Neurological Disorders and Stroke (U01NS069997), and the American Cancer Society (RSG-07-276-01-MGO).

Curr Opin Mol Ther. Author manuscript; available in PMC 2011 October 1. Ahmed et al. Page 7

References •• of outstanding interest NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

• of special interest

1. Brustle O, Spiro AC, Karram K, Choudhary K, Okabe S, McKay RD. In vitro-generated neural precursors participate in mammalian brain development. Proc Natl Acad Sci USA 1997;94(26):14809– 14814. [PubMed: 9405695] • An important paper establishing the feasibility of the in vitro manipulation of NSCs and their transplantation into the mammalian brain. 2. Aboody KS, Brown A, Rainov NG, Bower KA, Liu S, Yang W, Small JE, Herrlinger U, Ourednik V, Black PM, Breakefield XO, et al. Neural stem cells display extensive tropism for pathology in adult brain: Evidence from intracranial gliomas. Proc Natl Acad Sci USA 2000;97(23):12846–12851. [PubMed: 11070094] •• Presents the first suicide gene therapy approach to NSC-mediated targeted therapy for glioblastoma. Identified robust tumor-targeted migration from distant sites, via different methods of administration and to disseminated tumor foci. 3. Benedetti S, Pirola B, Pollo B, Magrassi L, Bruzzone MG, Rigamonti D, Galli R, Selleri S, Di Meco F, De Fraja C, Vescovi A, et al. Gene therapy of experimental brain tumors using neural progenitor cells. Nat Med 2000;6(4):447–450. [PubMed: 10742153] •• The first study to use NSCs as vectors for the targeted delivery of therapeutic agents. Highlights both the tumor tropism of NSCs and the feasibility of their use for localized drug delivery. 4. Willmon C, Harrington K, Kottke T, Prestwich R, Melcher A, Vile R. Cell carriers for oncolytic viruses: Fed Ex for cancer therapy. Mol Ther 2009;17(10):1667–1676. [PubMed: 19690519] 5. Muller FJ, Snyder EY, Loring JF. Gene therapy: Can neural stem cells deliver? Nat Rev Neurosci 2006;7(1):75–84. [PubMed: 16371952] 6. Aboody KS, Najbauer J, Danks MK. Stem and progenitor cell-mediated tumor selective gene therapy. Gene Ther 2008;15(10):739–752. [PubMed: 18369324] •• An excellent review, highlighting in detail the particular approaches that different research groups have taken to target malignancies with carrier- based gene therapy. 7. Joo KM, Park IH, Shin JY, Jin J, Kang BG, Kim MH, Lee SJ, Jo MY, Kim SU, Nam DH. Human neural stem cells can target and deliver therapeutic genes to breast cancer brain metastases. Mol Ther 2009;17(3):570–575. [PubMed: 19127251] •• Provides important evidence, along with references [8] and [9], supporting the use of NSC-based therapies for a variety of CNS malignancies, both primary and secondary. Demonstrates that NSCs migrate to a variety of CNS pathologies effectively, thus encouraging further research into their therapeutic potential in other disease paradigms. 8. Aboody KS, Bush RA, Garcia E, Metz MZ, Najbauer J, Justus KA, Phelps DA, Remack JS, Yoon KJ, Gillespie S, Kim SU, et al. Development of a tumor-selective approach to treat metastatic cancer. PLoS One 2006;1:e23. [PubMed: 17183650] •• Provides important evidence, along with references [7] and [9], supporting the use of NSC-based therapies for a variety of CNS malignancies, both primary and secondary. 9. Kim SK, Kim SU, Park IH, Bang JH, Aboody KS, Wang KC, Cho BK, Kim M, Menon LG, Black PM, Carroll RS. Human neural stem cells target experimental intracranial medulloblastoma and deliver a therapeutic gene leading to tumor regression. Clin Cancer Res 2006;12(18):5550–5556. [PubMed: 17000692] •• Provides important evidence, along with references [7] and [8], supporting the use of NSC-based therapies for a variety of CNS malignancies, both primary and secondary. Demonstrates that NSCs migrate to a variety of CNS pathologies effectively, thus encouraging further research into their therapeutic potential in other disease paradigms. 10. Sims TL Jr, Hamner JB, Bush RA, Fischer PE, Kim SU, Aboody KS, McCarville B, Danks MK, Davidoff AM. Neural progenitor cell-mediated delivery of osteoprotegerin limits disease progression in a preclinical model of bone . J Pediatr Surg 2009;44(1):204–210. discussion 210-201. [PubMed: 19159744] 11. Imitola J, Raddassi K, Park KI, Mueller FJ, Nieto M, Teng YD, Frenkel D, Li J, Sidman RL, Walsh CA, Snyder EY, et al. Directed migration of neural stem cells to sites of CNS injury by the stromal cell-derived factor 1α/CXC chemokine receptor 4 pathway. Proc Natl Acad Sci USA 2004;101(52): 18117–18122. [PubMed: 15608062] •• Demonstrated directed migration of NSCs to sites of CNS injury by the SDF1 α/CXCR4 pathway.

Curr Opin Mol Ther. Author manuscript; available in PMC 2011 October 1. Ahmed et al. Page 8

12. Ceradini DJ, Kulkarni AR, Callaghan MJ, Tepper OM, Bastidas N, Kleinman ME, Capla JM, Galiano RD, Levine JP, Gurtner GC. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med 2004;10(8):858–864. [PubMed: 15235597] •• Investigated the NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript role of the CXCR4 signaling pathway in NSC tumor tropism, and elucidated, along with reference [11], the important interplay between hypoxia and NSC migration. 13. Lee JW, Bae SH, Jeong JW, Kim SH, Kim KW. Hypoxia-inducible factor (HIF-1)α: Its protein stability and biological functions. Exp Mol Med 2004;36(1):1–12. [PubMed: 15031665] 14. Sun L, Hui AM, Su Q, Vortmeyer A, Kotliarov Y, Pastorino S, Passaniti A, Menon J, Walling J, Bailey R, Rosenblum M, et al. Neuronal and glioma-derived stem cell factor induces within the brain. Cancer Cell 2006;9(4):287–300. [PubMed: 16616334] 15. Zhao D, Najbauer J, Garcia E, Metz MZ, Gutova M, Glackin CA, Kim SU, Aboody KS. Neural stem cell tropism to glioma: Critical role of tumor hypoxia. Mol Cancer Res 2008;6(12):1819–1829. [PubMed: 19074827] 16. Sun L, Lee J, Fine HA. Neuronally expressed stem cell factor induces neural stem cell migration to areas of brain injury. J Clin Invest 2004;113(9):1364–1374. [PubMed: 15124028] 17. Magge SN, Malik SZ, Royo NC, Chen HI, Yu L, Snyder EY, O’Rourke DM, Watson DJ. Role of monocyte chemoattractant protein-1 (MCP-1/CCL2) in migration of neural progenitor cells toward glial tumors. J Neurosci Res 2009;87(7):1547–1555. [PubMed: 19125409] 18. An JH, Lee SY, Jeon JY, Cho KG, Kim SU, Lee MA. Identification of gliotropic factors that induce human stem cell migration to malignant tumor. J Proteome Res 2009;8(6):2873–2881. [PubMed: 19351187] 19. Heese O, Disko A, Zirkel D, Westphal M, Lamszus K. Neural stem cell migration toward gliomas in vitro. Neuro Oncol 2005;7(4):476–484. [PubMed: 16212812] 20. Schmidt NO, Przylecki W, Yang W, Ziu M, Teng Y, Kim SU, Black PM, Aboody KS, Carroll RS. Brain tumor tropism of transplanted human neural stem cells is induced by vascular endothelial growth factor. Neoplasia 2005;7(6):623–629. [PubMed: 16036113] 21. Kim SU, de Vellis J. Stem cell-based cell therapy in neurological diseases: A review. J Neurosci Res 2009;87(10):2183–2200. [PubMed: 19301431] 22. Cattaneo R, Miest T, Shashkova EV, Barry MA. Reprogrammed viruses as cancer therapeutics: Targeted, armed and shielded. Nat Rev Microbiol 2008;6(7):529–540. [PubMed: 18552863] 23. Fulci G, Breymann L, Gianni D, Kurozomi K, Rhee SS, Yu J, Kaur B, Louis DN, Weissleder R, Caligiuri MA, Chiocca EA. Cyclophosphamide enhances glioma virotherapy by inhibiting innate immune responses. Proc Natl Acad Sci USA 2006;103(34):12873–12878. [PubMed: 16908838] 24. Chiocca EA, Abbed KM, Tatter S, Louis DN, Hochberg FH, Barker F, Kracher J, Grossman SA, Fisher JD, Carson K, Rosenblum M, et al. A phase I open-label, dose-escalation, multi-institutional trial of injection with an E1B-Attenuated adenovirus, ONYX-015, into the peritumoral region of recurrent malignant gliomas, in the adjuvant setting. Mol Ther 2004;10(5):958–966. [PubMed: 15509513] • Presents results from a phase I clinical trial highlighting the value of a carrier system to shield therapeutic agents from the host immune system, thus extending the effectiveness of the therapeutics. 25. Einstein O, Ben-Hur T. The changing face of neural stem cell therapy in neurologic diseases. Arch Neurol 2008;65(4):452–456. [PubMed: 18413466] 26. Pluchino S, Quattrini A, Brambilla E, Gritti A, Salani G, Dina G, Galli R, Del Carro U, Amadio S, Bergami A, Furlan R, et al. Injection of adult neurospheres induces recovery in a chronic model of multiple sclerosis. Nature 2003;422(6933):688–694. [PubMed: 12700753] 27. Pluchino S, Gritti A, Blezer E, Amadio S, Brambilla E, Borsellino G, Cossetti C, Del Carro U, Comi G, t Hart B, Vescovi A, et al. Human neural stem cells ameliorate autoimmune encephalomyelitis in non-human primates. Ann Neurol 2009;66(3):343–354. [PubMed: 19798728] 28. Politi LS, Bacigaluppi M, Brambilla E, Cadioli M, Falini A, Comi G, Scotti G, Martino G, Pluchino S. Magnetic-resonance-based tracking and quantification of intravenously injected neural stem cell accumulation in the brains of mice with experimental multiple sclerosis. Stem Cells 2007;25(10): 2583–2592. [PubMed: 17600110]

Curr Opin Mol Ther. Author manuscript; available in PMC 2011 October 1. Ahmed et al. Page 9

29. Pluchino S, Zanotti L, Rossi B, Brambilla E, Ottoboni L, Salani G, Martinello M, Cattalini A, Bergami A, Furlan R, Comi G, et al. Neurosphere-derived multipotent precursors promote neuroprotection by an immunomodulatory mechanism. Nature 2005;436(7048):266–271. [PubMed: 16015332] NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript 30. Mader EK, Maeyama Y, Lin Y, Butler GW, Russell HM, Galanis E, Russell SJ, Dietz AB, Peng KW. Mesenchymal stem cell carriers protect oncolytic measles viruses from antibody neutralization in an orthotopic ovarian cancer therapy model. Clin Cancer Res 2009;15(23):7246–7255. [PubMed: 19934299] 31. Bonini C, Bondanza A, Perna SK, Kaneko S, Traversari C, Ciceri F, Bordignon C. The suicide gene therapy challenge: How to improve a successful gene therapy approach. Mol Ther 2007;15(7):1248– 1252. [PubMed: 17505474] 32. Li S, Tokuyama T, Yamamoto J, Koide M, Yokota N, Namba H. Bystander effect-mediated gene therapy of gliomas using genetically engineered neural stem cells. Cancer Gene Ther 2005;12(7): 600–607. [PubMed: 15775995] 33. Zhao Y, Wang S. Human NT2 neural precursor-derived tumor-infiltrating cells as delivery vehicles for treatment of glioblastoma. Hum Gene Ther 2010;21(6):683–694. [PubMed: 20113165] 34. Aboody KS, Najbauer J, Schmidt NO, Yang W, Wu JK, Zhuge Y, Przylecki W, Carroll R, Black PM, Perides G. Targeting of melanoma brain metastases using engineered neural stem/progenitor cells. Neuro Oncol 2006;8(2):119–126. [PubMed: 16524944] 35. Denny WA. Tumor-activated prodrugs – A new approach to cancer therapy. Cancer Invest 2004;22 (4):604–619. [PubMed: 15565818] 36. Ehtesham M, Kabos P, Kabosova A, Neuman T, Black KL, Yu JS. The use of interleukin 12-secreting neural stem cells for the treatment of intracranial glioma. Cancer Res 2002;62(20):5657–5663. [PubMed: 12384520] 37. Yuan X, Hu J, Belladonna ML, Black KL, Yu JS. Interleukin-23-expressing bone marrow-derived neural stem-like cells exhibit antitumor activity against intracranial glioma. Cancer Res 2006;66(5): 2630–2638. [PubMed: 16510582] 38. Lee DH, Ahn Y, Kim SU, Wang KC, Cho BK, Phi JH, Park IH, Black PM, Carroll RS, Lee J, Kim SK. Targeting rat brainstem glioma using human neural stem cells and human mesenchymal stem cells. Clin Cancer Res 2009;15(15):4925–4934. [PubMed: 19638465] 39. Johansson S, Price J, Modo M. Effect of inflammatory cytokines on major histocompatibility complex expression and differentiation of human neural stem/progenitor cells. Stem Cells 2008;26(9):2444– 2454. [PubMed: 18635871] 40. Sonabend AM, Ulasov IV, Han Y, Lesniak MS. Oncolytic adenoviral therapy for glioblastoma multiforme. Neurosurg Focus 2006;20(4):E19. [PubMed: 16709024] 41. Coukos G, Makrigiannakis A, Kang EH, Caparelli D, Benjamin I, Kaiser LR, Rubin SC, Albelda SM, Molnar-Kimber KL. Use of carrier cells to deliver a replication-selective herpes simplex virus-1 mutant for the intraperitoneal therapy of epithelial ovarian cancer. Clin Cancer Res 1999;5(6):1523– 1537. [PubMed: 10389942] 42. Sonabend AM, Ulasov IV, Tyler MA, Rivera AA, Mathis JM, Lesniak MS. Mesenchymal stem cells effectively deliver an oncolytic adenovirus to intracranial glioma. Stem Cells 2008;26(3):831–841. [PubMed: 18192232] 43. Tyler MA, Ulasov IV, Sonabend AM, Nandi S, Han Y, Marler S, Roth J, Lesniak MS. Neural stem cells target intracranial glioma to deliver an oncolytic adenovirus in vivo. Gene Ther 2009;16(2): 262–278. [PubMed: 19078993] 44. Hayden EC. California institute to help stem-cell biotechs. Nature 2008;455(7212):436–437. [PubMed: 18818612] 45. Guidelines for the clinical translation of stem cells. International Society for Stem Cell Research; Deerfield, IL, USA: 2010. www.isscr.org/clinical_trans/ 46. Halme DG, Kessler DA. FDA regulation of stem-cell-based therapies. N Engl J Med 2006;355(16): 1730–1735. [PubMed: 17050899] 47. Xu G, Li X, Bai Y, Bai J, Li L, Shen L. Improving recovery of spinal cord-injured rats by telomerase- driven human neural progenitor cells. Restor Neurol Neurosci 2004;22(6):469–476. [PubMed: 15798365]

Curr Opin Mol Ther. Author manuscript; available in PMC 2011 October 1. Ahmed et al. Page 10

48. Truckenmiller ME, Vawter MP, Zhang P, Conejero-Goldberg C, Dillon-Carter O, Morales N, Cheadle C, Becker KG, Freed WJ. AF5, a CNS cell line immortalized with an N-terminal fragment of SV40 large T: Growth, differentiation, genetic stability, and . Exp Neurol 2002;175(2): NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript 318–337. [PubMed: 12061863] 49. Cacci E, Villa A, Parmar M, Cavallaro M, Mandahl N, Lindvall O, Martinez-Serrano A, Kokaia Z. Generation of human cortical neurons from a new immortal fetal neural stem cell line. Exp Cell Res 2007;313(3):588–601. [PubMed: 17156776] 50. Amariglio N, Hirshberg A, Scheithauer BW, Cohen Y, Loewenthal R, Trakhtenbrot L, Paz N, Koren- Michowitz M, Waldman D, Leider-Trejo L, Toren A, et al. Donor-derived brain tumor following neural stem cell transplantation in an ataxia telangiectasia patient. PLoS Med 2009;6(2):e1000029. [PubMed: 19226183] 51. Frappart PO, McKinnon PJ. Ataxia-telangiectasia and related diseases. Neuromolecular Med 2006;8 (4):495–511. [PubMed: 17028372] 52. Hacein-Bey-Abina S, Garrigue A, Wang GP, Soulier J, Lim A, Morillon E, Clappier E, Caccavelli L, Delabesse E, Beldjord K, Asnafi V, et al. Insertional oncogenesis in 4 patients after retrovirus- mediated gene therapy of SCID-X1. J Clin Invest 2008;118(9):3132–3142. [PubMed: 18688285] 53. Ubiali F, Nava S, Nessi V, Frigerio S, Parati E, Bernasconi P, Mantegazza R, Baggi F. Allorecognition of human neural stem cells by peripheral blood lymphocytes despite low expression of MHC molecules: Role of TGFβ in modulating proliferation. Int Immunol 2007;19(9):1063–1074. [PubMed: 17660500] 54. Krystkowiak P, Gaura V, Labalette M, Rialland A, Remy P, Peschanski M, Bachoud-Levi AC. Alloimmunisation to donor and immune rejection following foetal neural grafts to the brain in patients with Huntington’s disease. PLoS One 2007;2(1):e166. [PubMed: 17245442] • Presents a different therapeutic paradigm in the treatment of Huntington’s disease through the use of NSC grafts, while highlighting the need to investigate the host graft rejection observed in clinical trials using this approach. 55. Ernst N, Tiede S, Tronnier V, Kruse C, Zechel C, Paus R. An improved, standardised protocol for the isolation, enrichment and targeted neural differentiation of Nestin+ progenitors from adult human dermis. Exp Dermatol 19(6):549–555. [PubMed: 20100195] 56. Mitrecic D, Gajovic S, Pochet R. Toward the treatments with neural stem cells: Experiences from amyotrophic lateral sclerosis. Anat Rec (Hoboken) 2009;292(12):1962–1967. [PubMed: 19943351] 57. Casalbore P, Budoni M, Ricci-Vitiani L, Cenciarelli C, Petrucci G, Milazzo L, Montano N, Tabolacci E, Maira G, Larocca LM, Pallini R. Tumorigenic potential of olfactory bulb-derived human adult neural stem cells associates with activation of TERT and NOTCH1. PLoS One 2009;4(2):e4434. [PubMed: 19209236] 58. Kim JH, Lee JE, Kim SU, Cho KG. Stereological analysis on migration of human neural stem cells in the brain of rats bearing glioma. Neurosurgery 66(2):333–342. [PubMed: 20087133] 59. Mercapide J, Rappa G, Anzanello F, King J, Fodstad O, Lorico A. Primary gene-engineered neural stem/progenitor cells demonstrate tumor-selective migration and antitumor effects in glioma. Int J Cancer 126(5):1206–1215. [PubMed: 19653275] 60. Xu Q, Yuan X, Xu M, McLafferty F, Hu J, Lee BS, Liu G, Zeng Z, Black KL, Yu JS. Chemokine CXC receptor 4-mediated glioma tumor tracking by bone marrow-derived neural progenitor/stem cells. Mol Cancer Ther 2009;8(9):2746–2753. [PubMed: 19723878] 61. Jeon JY, An JH, Kim SU, Park HG, Lee MA. Migration of human neural stem cells toward an intracranial glioma. Exp Mol Med 2008;40(1):84–91. [PubMed: 18305401] 62. Jurvansuu J, Zhao Y, Leung DS, Boulaire J, Yu YH, Ahmed S, Wang S. Transmembrane protein 18 enhances the tropism of neural stem cells for glioma cells. Cancer Res 2008;68(12):4614–4622. [PubMed: 18559506] 63. Brown AB, Yang W, Schmidt NO, Carroll R, Leishear KK, Rainov NG, Black PM, Breakefield XO, Aboody KS. Intravascular delivery of neural stem cell lines to target intracranial and extracranial tumors of neural and non-neural origin. Hum Gene Ther 2003;14(18):1777–1785. [PubMed: 14670128] • Highlights the potential of modes of delivery other than intratumoral injections for the treatment of CNS pathologies with NSCs, providing evidence for the ability of NSCs to cross the blood-brain-barrier.

Curr Opin Mol Ther. Author manuscript; available in PMC 2011 October 1. Ahmed et al. Page 11 NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript [9] [33] [58] [59] [38] [60] [17] [61] [62] [15] [34] [63] [63] Reference in vivo Table 1 Kinetics observed – Robust migration to contralateral hemisphere – Cells crossed the blood brain barrier to reach the intracranial tumor Migratory fraction of 10% at 50 min after injection; signifcant migration continued to 15 days post injection Prominent migration along vascular tracts to interhemipsheric fssure; 80% migration knockdown following anti-VEGR1 treatment Migration by 29.7 ± 5.4% of NSCs at 2 weeks after implantation Signifcant decrease in tumor tropism following anti-CXCR4 treatment Contralateral migration by a subpopulation of cells to the site MCP-1 infusion Migration initiated by day 3, tumor mass surrounded by NSCs day 5 and infltrated throughout by day 7 Signifcant increase in migration following induction of TM-18 overexpression NSCs clustered at the tumor-brain interface with preferential migration toward hypoxic tumor regions Cells colocalized to brain metastases via both injection methods Qualitative observation of cells migrating contralaterally and into the tumor bed Qualitative observation of cells migrating across the blood brain barrier to reach intracranial tumor Robust migration throughout the tumor mass and tracking of in vivo Signals examined n/a n/a VEGFR1 n/a CXCR4 MCP-1 n/a TM-18 Hypoxia n/a n/a n/a n/a Route of administration - Contralateral injection - Tail vein injection Contralateral injection Contralateral injection Ipsilateral forebrain injection Contralateral injection Injection contralateral to infusion site Ipsilateral injection away from tumor site Contralateral injection Contralateral injection Intracarotid or intracranial injections Contralateral injection Tail vein injection Distant ipsilateral site, contralateral, Model – Intracranial glioma (U87) – Lateral ventricle glioma (U87) Intracranial glioma (U373) Intracranial glioma (GL261, U87) Brainstem glioma (F98) Rat intracranial glioma (RG2) Intracranial infusion of MCP-1 in glial tumor cells Rat intracranial glioma (C6) Rat intracranial glioma (C6) Intracranial glioma (U251) Melanoma brain metastases (B16/ F10) Intracranial medulloblastoma (Daoy) Intracranial glioma (U251) Rat intracranial glioma (CNS-1) Cell line hNT2RA2 (human) HB1.F3 (human) Primary murine NSC HB1.F3 (human) Bone marrow- derived neural progenitor/ stem cells C17.2 (murine) HB1.F5, A4, F3 (human) C17.2 (murine) HB1.F3 (human) C17.2 (murine) HB1.F3 (human) C17.2 (murine) C17.2 (murine) Selected studies evaluating the tumor-tropism of neural stem cells

Curr Opin Mol Ther. Author manuscript; available in PMC 2011 October 1. Ahmed et al. Page 12 Reference NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript transmembrane protein 18 TM-18 Kinetics observed microsatellites observed; contralateral migration with no impact of tumor size observed neural stem cell, NSC Signals examined monocyte chemotactic protein-1, Route of administration intraventricular, or intravascular injection MCP-1 Model CXC chemokine receptor 4, Cell line CXCR4

Curr Opin Mol Ther. Author manuscript; available in PMC 2011 October 1.