glioma stem cells as a target for treatment

5

Click here to load reader

Upload: katrin-lamszus

Post on 15-Jul-2016

214 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Glioma stem cells as a target for treatment

PERSPECTIVES

Glioma stem cells as a target for treatment

Katrin Lamszus & Hauke S. Günther

Received: 10 May 2010 /Accepted: 4 August 2010 /Published online: 25 August 2010# Springer-Verlag 2010

Abstract Glioma cells with stem cell-like propertiesrepresent a minor subfraction of the total tumor cellpopulation. These cells are highly chemo- and radio-resistent and are held responsible for the inevitablerecurrence of malignant gliomas. This review summarizescurrent strategies for targeting putative glioma stem cells.Target definition approaches comprise extrapolation andadaptation of therapies from general oncology, targetidentification by correlative molecular genetic analyses,and dedicated target discovery research. Targeting strategiesinclude inhibition of tumor-specific signaling pathways,enhancement of tumor cell differentiation, radiosensitiza-tion, indirect targeting of the tumor stem cell niche, oncolyticvirotherapy, and adhesion molecule blockade.

Keywords Glioblastoma . Stem cells . Tumor

Defining glioma stem cells

Gliomas contain a subpopulation of tumor cells that displaycharacteristics of stem cells. These glioma stem cells(GSCs) or glioma-initiating cells are distinguished by (i)the ability to self-renew, (ii) the ability to initiate braintumors upon orthotopic implantation in immunodeficientmice, (iii) the expression of neural stem cell markers, and(iv) multipotency, that is, the capacity to differentiate into

cells with a neuronal, astrocytic or oligodendroglialphenotype. GSCs constitute only a minor subpopulationof the total tumor cell pool and divide slowly, but areessential for sustaining the continuous growth of the tumor.In contrast, the bulk of tumor cells, which are supposed tobe progeny of GSCs and consist of progenitor-like cells aswell as differentiated cells, divide more rapidly but haveonly limited proliferative potential and are incapable oftumor-initiation. Compared to these bulk cells, GSCs arepreferentially equipped with chemo- and radioresistancemechanisms, so that tumor recurrences after cytotoxictreatment are believed to originate from the survivingtumor stem cells. Consequently, these cells are an importanttarget for treatment.

The cancer stem cell paradigmwas originally established forleukemia in the mid-1990s [1], and was translated to solidtumors about 10 years later [2, 3], where it initially sparkedtremendous enthusiasm. However, more recently severalcornerstones of this concept were called into question; tomention only a few: (i) transplantation in limiting cell dilutioninto immunodeficient NOD/SCID mice, the classical tool totest for tumor-initiating capacity, was found to select for cellsresisting the residual immune response, and the proportion ofcells with cancer initiating capacity turned out to beconsiderably higher when more permissive mice were used[4, 5]; (ii) CD133 was described as a highly specific tumorstem cell marker in glioma [3]; however, CD133− cells canalso be capable of self-renewal and initiate tumors, andCD133− cells can generate CD133+ cells [6–11]; (iii)clonogenicity assays, which are used to assess self-renewal,are strongly affected by the microenvironment and, forexample, the presence of serum greatly promotes clonogenicity[12].

Collectively, these studies indicate that the microenvi-ronment (niche) modulates tumor stem cell characteristics.

K. Lamszus (*) :H. S. GüntherLaboratory for Brain Tumor Biology,Department of Neurosurgery,University Medical Center Hamburg-Eppendorf,Martinistrasse 52,20246 Hamburg, Germanye-mail: [email protected]

Targ Oncol (2010) 5:211–215DOI 10.1007/s11523-010-0155-4

Page 2: Glioma stem cells as a target for treatment

It was recently demonstrated that CD133 is inducible byhypoxia [13–15], so that the expression of this marker isnot a static feature of a defined GSC population but subjectto regulation by external factors. Thus, the use of CD133 asa read-out of stemness in many studies is obviouslyquestionable. It is conceivable that a subpopulation oftumor cells exists that are metabolically flexible and able toswitch between different bioenergetic pathways underalternating microenvironmental conditions. Thus, a moredynamic model is plausible, in which cancer stem cells areconsidered to be highly adaptable cells [16]. While ahierarchy of diverse cancer cells may well exist, these cellsmight all be capable of dedifferentiation and tumorpropagation [17]. According to these qualified models, allcells and not just a temporary population with stem cellcharacteristics need to be targeted and eliminated.

Notwithstanding individual preferences as to what themost appropriate definition of cancer stem cells might be,this groundbreaking new concept has revitalized ourrecognition of the importance of intratumoral heterogeneityand tumor-host interactions, and it provides a novelframework to discover new strategies, targeting a relativelyresistant tumor cell subpopulation.

Targeting GSCs

Targeted cancer therapies usually rely on the presence ofspecific molecules expressed on the tumor cells or activatedpathways, offering a selective therapeutic window. Targetidentification can either be achieved by extrapolation andadaptation of therapies from general oncology, or bycorrelative molecular genetic analyses, or by dedicatedtarget discovery research. All of these strategies have beenemployed in the attempt to identify GSC-specific targets,and these are summarized under different categories asshown in Table 1.

Signaling pathways

A number of key signaling pathways that are crucial fornormal neural stem cell development are also activated inGSCs. NOTCH is an important regulator of normalneural stem cell self-renewal and differentiation, andGSCs have higher NOTCH activity than non-stem tumorcells. The NOTCH pathway has successfully beentargeted using gamma-secretase inhibitors (GSIs), depletingCD133-positive putative GSCs and inhibiting tumor initiationand growth in vivo [18]. NOTCH blockade can furthersensitize GSCs to radiation [19]. The phosphoinositide 3-kinase (PI3K) pathway is another candidate for targeting,and a small molecule Akt inhibitor was shown to predom-inantly reduce the number of GSCs, inhibit migration and

invasion of GSCs, and to inhibit tumor growth in vivo [20].The Hedgehog pathway was also found to be activated inGSCs, and the Hedgehog inhibitor cyclopamine depletedclonogenic glioblastoma stem cells in vitro and inhibitedtumor initiation in vivo [21]. A20 (TNFAIP3) is a regulatorof the NF-κB pathway and is overexpressed in glioblastomastem cells relative to non-stem tumor cells. Knockdown ofA20 by lentiviral shRNA transfer reduced glioma stem cellself-renewal, growth and apoptotic resistance [22]. Also thec-myc pathway is required for the maintenance of GSCs andis overexpressed relative to non-stem glioma cells. LentiviralshRNA targeting of c-Myc reduced the proliferation ofGSCs, increased apoptosis and abolished tumorigenicity[23]. The polycomb group protein enhancer of zestehomologue 2 (EZH2) enhances c-Myc expression, andshRNA-mediated downregulation of EZH2 or its pharmaco-logical disruption by DZNep both impaired GSC self-renewal and tumor-initiating capacity [24]. The interleukin6 (IL6) pathway is another candidate target, since GSCspreferentially express the IL6 receptors IL6Rα and gp130.Lentiviral shRNA targeting of IL6Rα or IL-6 blockedgrowth and neurosphere formation in vitro, and bothknockdown as well as anti-IL6 antibody treatment inhibitedgrowth of GSC-derived tumors in vivo [25]. Finally, werecently discovered that CXCR4 is significantly overex-pressed in a panel of 12 glioma stem cell lines (GS cell lines)compared with 32 conventional, serum-grown glioma celllines. GS cell lines contained a minor subpopulation of cellswith high CXCR4 receptor levels, whereas conventionalglioma lines contained only cells with low levels. Treatmentwith a specific CXCR4 antagonist, AMD3100, inhibited cellmigration in vitro and reduced the growth of highly invasiveGS cell line xenografts in vivo [26].

Differentiation

The induction of differentiation is an obvious and attractiveapproach to target GSCs. Bone morphogenic proteins(BMPs) are well known for their influence on differentia-tion during neural development. BMP4 was shown totrigger an overproportional reduction in stem-like glioblas-toma cells in vitro, to abolish tumor initiation in vivo andreduce growth of established glioblastoma xenografts [27].Using an RNA interference screening approach, silencingof the adaptor protein TRRAP was found to inducedifferentiation of GSCs. Knockdown of TRRAP sensitizedcells to apoptotic stimuli, inhibited cell cycle progressionand suppressed tumor formation in vivo, suggesting thatTRRAP maintains GSCs in a stem cell-like, tumorigenicstate [28]. Retinoids have long been known to promotetumor cell differentiation, and recently all-trans retinoicacid was shown to exert an antitumor effect specifically onstem-like glioma cells [29]. However, previous clinical

212 Targ Oncol (2010) 5:211–215

Page 3: Glioma stem cells as a target for treatment

trials have so far reported limited efficacy of both all-transretinoic acid as well as cis-retinoic acid [30–32], and itremains to be seen whether any particular form of retinoicacid can be useful in combination to sensitize GSCs tocytotoxic treatment.

Radiosensitization

Gliomas show only a moderate response to radio- andchemotherapy. Resistance mechanisms are preferentiallyactivated in cancer stem cells, and in GSCs in particular,display activation of the DNA damage checkpoint responseas well as an increase in DNA repair capacity. CD133expressing putative GSCs survive radiation in vitro and invivo at increased proportions relative to cells lackingCD133 expression [33]. They repair radiation-inducedDNA damage more effectively than CD133-negative tumorcells, and their radioresistance could be reversed usingsmall molecule inhibitors of the Chk1 and Chk2 checkpointkinases. Thus, targeting the DNA damage checkpointresponse in GSCs might represent a promising therapeuticstrategy. In addition, as mentioned above, inhibition of theNOTCH pathway using GSIs may also sensitize GSCs toradiotherapy [19]. Interestingly an analysis that correlatedclinical outcome with gene expression profiles showed thata stem cell-related “self-renewal” signature dominatedby HOX genes was a predictor of poor survival inglioblastoma patients treated with chemoradiotherapy [34].

GSCs have further been demonstrated to be relativelyresistant to chemotherapy with temozolomide [35], al-though this issue is controversial [36]. Chemoresistancecan in part be explained by elevated expression of drugtransporter genes such as ABCG2 and ABCA3 [37]. Itremains to be demonstrated which other resistance mech-anisms contribute to GSC chemoresistance and whethertheir blockade can sensitize cells to treatment.

Indirect targeting of the GSC niche

Several studies suggest that GSCs preferentially reside inperivascular areas, termed the “vascular niche”, wherethey receive paracrine cues from endothelial cells thatmimic the normal neural stem cell niche and where theymaintain a stem-like state [38]. Disruption of nichehomeostasis therefore is another theoretically attractivetargeting approach. Compared with non stem-like gliomacells, GSCs secrete elevated levels of vascular endothelialgrowth factor (VEGF) and have a strong angiogenic effect[39], suggesting mutual interdependence of the GSCcompartment and the vascular compartment. Bevacizumab,an anti-VEGF antibody with potent antiangiogenic activity,was found to suppress angiogenesis and growth of GSC-derived xenografts, whereas it had limited efficacy againstxenografts derived from non-GSC populations [39]. Inanother study, treatment with bevacizumab ablated self-renewing cells from glioblastoma xenografts and arrested

Table 1 Successful targeting of glioma stem cells

Targeting strategy Target Tool Reference

Signaling pathway inhibition NOTCH γ-secretase inhibitors Fan et al., 2010 [18]

PI3K pathway AKT inhibitors Eyler et al., 2008 [20]

SHH pathway Cyclopamine Bar et al., 2007 [21]

NF-kappaB pathway shRNA against A20 (TNFAIP3) Hjelmeland et al, 2010 [22]

c-Myc pathway shRNA against c-Myc Wang et al., 2008 [23]

c-Myc pathway shRNA against EZH2 or DZNep Suva et al., 2009 [24]

IL6 shRNA/antibody Wang et al., 2009 [25]

CXCR4 AMD3100 Lamszus et al., 2010 [26]

Differentiation BMP receptors BMP4 Piccirillo et al., 2006 [27]

TRRAP Wurdak et al., 2010 [28]

Retinoic acid receptors All-trans retinoic acid Campos et al., 2010 [29]

Radiosensitization Chk1, Chk2 Small molecule inhibitors Bao et al., 2006 [33]

NOTCH pathway γ-secretase inhibitors Wang et al., 2010 [19]

Indirect targeting of the GSC niche VEGF Bevacizumab Bao et al., 2006 [39]

VEGF Bevacizumab Calabrese et al., 2007 [38]

VEGFR-2 DC101(+cyclophosphamide) Folkins et al., 2007 [40]

Oncolytic virotherapy Abnormal p16INK4/Rb cells Oncolytic Adenovirus Delta-24-RGD Jiang et al., 2007 [41]

Neoplastic cells Oncolytic herpes simplex virus Wakimoto et al., 2009 [42]

Adhesion molecule inhibition L1CAM shRNA Bao et al., 2008 [43]

Only studies are included that provide in vitro as well as in vivo preclinical evidence of successful GSC targeting

Targ Oncol (2010) 5:211–215 213

Page 4: Glioma stem cells as a target for treatment

tumor growth [38]. In a third study, the combination ofantiangiogenic therapy using an anti-VEGF receptor anti-body with cytotoxic treatment resulted in efficient reductionof the GSC population in vivo, however, antiangiogenictreatment alone had no such effect, suggesting that disruptionof the niche sensitized GSCs to chemotherapy [40].Bevacizumab is currently in phase III clinical trials for treatingnewly diagnosed glioblastoma, and it will be interesting todetermine whether this treatment leads to depletion of tumorcells expressing stem cell markers in patients.

Oncolytic virotherapy

Oncolytic viruses are natural or genetically modifiedviruses that preferentially infect and lyse cancer cells whilesparing nonneoplastic cells. Delta-24-RGD is an oncolyticadenovirus that selectively replicates in cancer cells with anabnormal Rb pathway and displays enhanced tropism toglioma cells [41]. Mice bearing intracranial xenografts ofglioblastoma stem cell lines survived significantly longerupon treatment with Delta-24-RGD, and autophagic tumorcell death appeared to be the most likely mechanism. Anoncolytic herpes simplex virus (oHSV) was also describedto inhibit self-renewal of GSCs, to cause cell death, and toprolong survival of mice bearing invasive GSC culture-derived xenografts [42].

Adhesion molecule inhibition

L1CAM is a neuronal cell adhesion molecule that regulatesneural cell growth, survival, and migration, as well asaxonal outgrowth and neurite extension during develop-ment. It is overexpressed in gliomas as well as othercancers and thus represents a potential cell surface target.L1CAM was found to be present on most CD133+ gliomacells, whereas CD113− cells were typically L1CAM-negative. Lentiviral shRNA targeting of L1CAM disruptedneurosphere formation, induced apoptosis and inhibitedgrowth of CD133-positve GSCs; furthermore, L1CAMknockdown inhibited GSC-derived tumor formation in vivoand increased the survival of tumor-bearing animals [43].

Future considerations

Further GSC targeting strategies are also currently beingexplored, an important one of which is immunotherapy, andthis complex area of research, which includes vaccinationapproaches as well as attempts to overcome tumor-inducedimmunosuppressive mechanisms, is covered in a separatearticle in this issue (Hatiboglu et al. [44]). In addition,interesting results are to be expected from the emergingfield of MicroRNA (miRNA) research. These are small

non-coding RNAs, which downregulate gene expressionpost-transcriptionally during various crucial cell processessuch as apoptosis, differentiation and development. Severalrecent studies identified aberrant miRNA expression ingliomas, and linked some of them to GSC maintainanceand growth [45, 46], although specific targeting agentsother than transfection/transduction-based techniques arenot yet available.

A major concern in the design of any GSC-targetingtherapy is to avoid damaging normal adult neural stem/progenitor cells, which share similar gene and antigenexpression profiles with GSCs. Adult neural stem cellsserve many important functions, especially tissue repairafter injury, including traumatic, ischemic, tumor-associated or treatment-induced tissue destruction. Inaddition, endogenous neural precursor cells were foundto possess significant intrinsic anti-tumor activity: theyare attracted by gliomas in vivo, inhibit glioma cellgrowth and induce apoptosis [47]. Thus, one of the mostimportant challenges lies in the definition of a specific-enough therapeutic window within which the GSCs can besuccessfully targeted and eliminated, while normal neuralstem/progenitor cells are saved.

Acknowledgement The authors´ work is supported by the DeutscheForschungsgemeinschaft (LA 1300/3-1 and LA 1300/4-1), theDeutsche Krebshilfe e.V., and the Johannes Bauer Stiftung fürHirntumorforschung.

Conflict of interest statement The authors have no conflict ofinterest to declare.

References

1. Lapidot T, Sirard C, Vormoor J et al (1994) A cell initiatinghuman acute myeloid leukaemia after transplantation into SCIDmice. Nature 367:645–648

2. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, ClarkeMF (2003) Prospective identification of tumorigenic breast cancercells. Proc Natl Acad Sci USA 100:3983–3988

3. Singh SK, Hawkins C, Clarke ID et al (2004) Identification ofhuman brain tumour initiating cells. Nature 432:396–401

4. Kelly PN, Dakic A, Adams JM, Nutt SL, Strasser A (2007) Tumorgrowth need not be driven by rare cancer stem cells. Science 317:337

5. Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM,Morrison SJ (2008) Efficient tumour formation by single humanmelanoma cells. Nature 456:593–598

6. Beier D, Hau P, Proescholdt M et al (2007) CD133(+) and CD133(-)glioblastoma-derived cancer stem cells show differential growthcharacteristics and molecular profiles. Cancer Res 67:4010–4015

7. Gunther HS, Schmidt NO, Phillips HS et al (2008) Glioblastoma-derived stem cell-enriched cultures form distinct subgroupsaccording to molecular and phenotypic criteria. Oncogene27:2897–2909

8. Joo KM, Kim SY, Jin X et al (2008) Clinical and biologicalimplications of CD133-positive and CD133-negative cells inglioblastomas. Lab Invest 88:808–815

214 Targ Oncol (2010) 5:211–215

Page 5: Glioma stem cells as a target for treatment

9. Ogden AT, Waziri AE, Lochhead RA et al (2008) Identification ofA2B5+CD133- tumor-initiating cells in adult human gliomas.Neurosurgery 62:505–514, discussion 14–15

10. Wang J, Sakariassen PO, Tsinkalovsky O et al (2008) CD133negative glioma cells form tumors in nude rats and give rise toCD133 positive cells. Int J Cancer 122:761–768

11. Chen R, Nishimura MC, Bumbaca SM et al (2010) A hierarchy ofself-renewing tumor-initiating cell types in glioblastoma. CancerCell 17:362–375

12. Zheng X, Shen G, Yang X, Liu W (2007) Most C6 cells arecancer stem cells: evidence from clonal and population analyses.Cancer Res 67:3691–3697

13. Blazek ER, Foutch JL, Maki G (2007) Daoy medulloblastomacells that express CD133 are radioresistant relative to CD133-cells, and the CD133+ sector is enlarged by hypoxia. Int J RadiatOncol Biol Phys 67:1–5

14. Platet N, Liu SY, Atifi ME et al (2007) Influence of oxygentension on CD133 phenotype in human glioma cell cultures.Cancer Lett 258:286–290

15. Griguer CE, Oliva CR, Gobin E et al (2008) CD133 is a marker ofbioenergetic stress in human glioma. PLoS ONE 3:e3655

16. Sakariassen PO, Immervoll H, Chekenya M (2007) Cancer stemcells as mediators of treatment resistance in brain tumors: statusand controversies. Neoplasia 9:882–892

17. Hadjipanayis CG, Van Meir EG (2009) Brain cancer propagatingcells: biology, genetics and targeted therapies. Trends Mol Med15:519–530

18. Fan X, Khaki L, Zhu TS et al (2010) NOTCH pathwayblockade depletes CD133-positive glioblastoma cells andinhibits growth of tumor neurospheres and xenografts. StemCells 28:5–16

19. Wang J, Wakeman TP, Lathia JD et al (2010) Notch promotesradioresistance of glioma stem cells. Stem Cells 28:17–28

20. Eyler CE, Foo WC, LaFiura KM, McLendon RE, Hjelmeland AB,Rich JN (2008) Brain cancer stem cells display preferentialsensitivity to Akt inhibition. Stem Cells 26:3027–3036

21. Bar EE, Chaudhry A, Lin A et al (2007) Cyclopamine-mediatedhedgehog pathway inhibition depletes stem-like cancer cells inglioblastoma. Stem Cells 25:2524–2533

22. Hjelmeland AB, Wu Q, Wickman S et al (2010) Targeting A20decreases glioma stem cell survival and tumor growth. PLoS Biol8:e1000319

23. Wang J, Wang H, Li Z et al (2008) c-Myc is required formaintenance of glioma cancer stem cells. PLoS ONE 3:e3769

24. Suva ML, Riggi N, Janiszewska M et al (2009) EZH2 is essentialfor glioblastoma cancer stem cell maintenance. Cancer Res69:9211–9218

25. Wang H, Lathia JD, Wu Q et al (2009) Targeting interleukin 6signaling suppresses glioma stem cell survival and tumor growth.Stem Cells 27:2393–2404

26. Lamszus K, Schulte A, Günther H, Phillips HS, Kemming D,Westphal M (2010) A distinct subset of glioma cell lines withstem cell-like properties reflects the transcriptional phenotype ofhuman glioblastomas and overexpresses CXCR4 as therapeutictarget. Abstract, The 18th Conference on Brain Tumor Researchand Therapy, May 18–20, 2010, Travemünde, Germany

27. Piccirillo SG, Reynolds BA, Zanetti N et al (2006) Bonemorphogenetic proteins inhibit the tumorigenic potential ofhuman brain tumour-initiating cells. Nature 444:761–765

28. Wurdak H, Zhu S, Romero A et al (2010) An RNAi screenidentifies TRRAP as a regulator of brain tumor-initiating celldifferentiation. Cell Stem Cell 6:37–47

29. Campos B, Wan F, Farhadi M et al (2010) Differentiation therapyexerts antitumor effects on stem-like glioma cells. Clin CancerRes 16:2715–2728

30. Butowski N, Prados MD, Lamborn KR et al (2005) A phaseII study of concurrent temozolomide and cis-retinoic acidwith radiation for adult patients with newly diagnosedsupratentorial glioblastoma. Int J Radiat Oncol Biol Phys61:1454–1459

31. Kaba SE, Kyritsis AP, Conrad C et al (1997) The treatment ofrecurrent cerebral gliomas with all-trans-retinoic acid (tretinoin). JNeurooncol 34:145–151

32. Phuphanich S, Scott C, Fischbach AJ, Langer C, Yung WK (1997)All-trans-retinoic acid: a phase II Radiation Therapy OncologyGroup study (RTOG 91-13) in patients with recurrent malignantastrocytoma. J Neurooncol 34:193–200

33. Bao S, Wu Q, McLendon RE et al (2006) Glioma stem cellspromote radioresistance by preferential activation of the DNAdamage response. Nature 444:756–760

34. Murat A, Migliavacca E, Gorlia T et al (2008) Stem cell-related“self-renewal” signature and high epidermal growth factorreceptor expression associated with resistance to concomitantchemoradiotherapy in glioblastoma. J Clin Oncol 26:3015–3024

35. Liu G, Yuan X, Zeng Z et al (2006) Analysis of gene expressionand chemoresistance of CD133+ cancer stem cells in glioblasto-ma. Mol Cancer 5:67

36. Beier D, Rohrl S, Pillai DR et al (2008) Temozolomidepreferentially depletes cancer stem cells in glioblastoma. CancerRes 68:5706–5715

37. Hirschmann-Jax C, Foster AE, Wulf GG et al (2004) A distinct“side population” of cells with high drug efflux capacity in humantumor cells. Proc Natl Acad Sci USA 101:14228–14233

38. Calabrese C, Poppleton H, Kocak M et al (2007) A perivascularniche for brain tumor stem cells. Cancer Cell 11:69–82

39. Bao S, Wu Q, Sathornsumetee S et al (2006) Stem cell-like gliomacells promote tumor angiogenesis through vascular endothelialgrowth factor. Cancer Res 66:7843–7848

40. Folkins C, Man S, Xu P, Shaked Y, Hicklin DJ, Kerbel RS (2007)Anticancer therapies combining antiangiogenic and tumor cellcytotoxic effects reduce the tumor stem-like cell fraction in gliomaxenograft tumors. Cancer Res 67:3560–3564

41. Jiang H, Gomez-Manzano C, Aoki H et al (2007) Examination of thetherapeutic potential of Delta-24-RGD in brain tumor stem cells: roleof autophagic cell death. J Natl Cancer Inst 99:1410–1414

42. Wakimoto H, Kesari S, Farrell CJ et al (2009) Humanglioblastoma-derived cancer stem cells: establishment of invasiveglioma models and treatment with oncolytic herpes simplex virusvectors. Cancer Res 69:3472–3481

43. Bao S, Wu Q, Li Z et al (2008) Targeting cancer stem cells throughL1CAM suppresses glioma growth. Cancer Res 68:6043–6048

44. Hatiboglu MA, Wei J, Wu ASG, Heimberger A (2010) Immunetherapeutic targeting of glioma cancer stem cells. Target Oncol.doi:10.1007/s11523-010-0151-8 (in this issue)

45. Kefas B, Comeau L, Floyd DH et al (2009) The neuronalmicroRNA miR-326 acts in a feedback loop with notch and hastherapeutic potential against brain tumors. J Neurosci 29:15161–15168

46. Guessous F, Zhang Y, Kofman A et al (2010) microRNA-34a istumor suppressive in brain tumors and glioma stem cells. CellCycle 9(6):1031–1036

47. Glass R, Synowitz M, Kronenberg G et al (2005) Glioblastoma-induced attraction of endogenous neural precursor cells isassociated with improved survival. J Neurosci 25:2637–2646

Targ Oncol (2010) 5:211–215 215