Abstract
Malignant gliomas are heterogeneous, diffuse and highly infiltrating by nature. Despite wide surgical resection and improvements in radio- and chemotherapies, the prognosis of patients with glioblastoma multiforme remains extremely poor, with a median survival time of only 14.5 months from diagnosis to death. Particular challenges for glioblastoma multiforme therapy are posed by limitations in the extent of feasible surgical resections, distinct tumor heterogeneity, difficulties in drug delivery across the blood-brain barrier and low drug distribution within the tumor. Therefore, new paradigms permitting tumor-specific targeting and extensive intratumoral distribution must be developed to allow an efficient therapeutic delivery. This review highlights the latest advances in the treatment of glioblastoma multiforme and the recent developments that have resulted from the interchange between preclinical and clinical efforts. We also summarize and discuss novel therapies for malignant glioma, focusing on advances in the following main topics of glioblastoma multiforme therapy: immunotherapy, gene therapy, stem cell-based therapies and nanotechnology. We discuss strategies and outcomes of emerging therapeutic approaches in these fields, and the main challenges associated with the integration of discoveries that occur in the laboratory into clinical practice.
Introduction
Glioblastoma multiforme, the most common and aggressive primary malignant brain tumor, carries a dismal prognosis. Patients with glioblastoma multiforme have a survival rate of less than 10% at five years [1]. Particular challenges to treating glioblastoma multiforme are the inability of treatments to reach all tumor cells and its distinct cellular heterogeneity that result in rapid and aggressive relapse [2]. Surgical resection is usually inadequate for local control, and residual tumors often lead to recurrent disease. Although sensitive to high doses of radiation, glioblastoma multiforme treatment with radiotherapy is limited by normal tissue toxicity. Moreover, chemotherapy has had only modest effects on improving outcomes for glioma patients, mostly due to the protective and selective characteristics of the blood-brain barrier. Therefore, new therapeutic strategies are urgently needed [3,4].
Recent developments in tumor immunology, genetics, cell-signaling pathways, and cancer stem cells have significantly contributed to our understanding of gliomagenesis. Even though significant advances in basic research rendered promising results in the preclinical setting, many of these treatments have not yet made their way into the clinic. In this review, we will analyze the latest advances for the treatment of glioblastoma, presenting a critical review of their benefits, drawbacks and their potential use in a clinical setting.
Glioma immunotherapies
Glioblastoma multiformes secrete various immunosuppressive cytokines, such as prostaglandin-E2, transforming growth factor-β (TGF-β) and interleukin (IL)-10, that dampen the tumor-specific immune response [5]. Currently, there is renewed interest in developing therapies that specifically target this problem. An attractive method is to utilize dendritic cells to generate host immune response against the tumor. Dendritic cells can be employed in various manners: 1) priming of dendritic cells with tumor antigens, followed by re-administration of these cells into the patient [6]; 2) mobilization of dendritic cells into brain tumors by using an FMS-like tyrosine kinase 3 ligand (Flt3L) [7]; 3) ex-vivo expansion of tumor-reactive cytotoxic T lymphocytes (CTLs) by peptide-pulsed dendritic cells [8]; and 4) targeting of overexpressed epitopes in glioblastoma multiforme, such as epidermal growth factor receptor variant III (EGFR vIII), by dendritic cell-based vaccines [9]. These strategies may serve as potential immunotherapies that target glioblastoma multiformes (Table 1).
Table 1. Novel glioma immunotherapeutic targeting strategies.
| Therapeutic approach | Mechanism | Target | Preclinical model | Clinical trial | Reference |
|---|---|---|---|---|---|
| To elicit host immune response |
Immunization with autologous DCs |
MHCII and T cell costimulatory molecules |
X | [6,33] | |
| To elicit host immune response |
Cotransfection of Poly (I:C) and siRNA into DCs and glioma cells |
MHCII and Th1 cell induction |
X | [34] | |
| Recruitment of DC to elicit immune repsonse |
Differentiation of precursor cells into DC through STAT3-dependent mechanism |
Flt3L targets precursor cells |
X | [7] | |
|
Ex-vivo generation of tumor-antigen primed T cells |
Peptide-pulsed DCs for ex-vivo expansion of CTLs |
IL-2, IL-7, IL-15 targeting CD62L and CCR7 |
X | [8] | |
| DC-based vaccine and peptide vaccine |
Targeting EGFRvIII | EGFRvIII | X | X | [9,10] |
| Radiolabeled monoclonal antibodies |
Targeting EGFRvIII | EGFRvIII | X | [11] | |
| Plasmid-based vaccine | Induction of T-cell immunity against glioma cells |
EphA2 | X | [35] |
Abbreviations: DC, dendritic cells; MHCII, major histocompatibility complex class II; siRNA, small interference RNA; Poly (I:C), polyriboinosinic polyribocytidylic acid; Th1, type 1 helper T cell; Flt3L, adenovirus expressing human FMS-like tyrosine kinase 3 ligand; STAT3, Signal transducer and activator of transcription 3; CTLs, Cytotoxic T Lymphocytes; IL-2, Interleukin-2; IL-7, Interleukin-7; IL-15, Interleukin-15; CD62L, L-selectin; CCR7, C-C chemokine receptor type 7; EGFRvIII, epidermal growth factor receptor variant III; EphA2, ephrin type-A receptor 2.
EGFRvIII is a cell-surface receptor commonly mutated in malignant gliomas, whose overexpression leads to uncontrolled cell division. Rindopepimut, a 14-mer peptide vaccine against EGFRvIII is currently being tested in Phase III Clinical trial [10]. Moreover, EGFRvIII can also be targeted by radiolabeled monoclonal antibodies that are being tested in a Phase II clinical trial [11]. Due to their high specificity and potential generation of immune response against the tumor, immunotherapies are currently in the spotlight of glioblastoma multiforme therapies.
Novel gene therapeutic approaches in glioblastoma treatment
Gene therapy nowadays offers what appear to be infinite possibilities to target different populations of tumor cells (Table 2). It works primarily through (1) immune stimulatory approaches and induction of immunologic memory against the tumor; (2) conditional cytotoxic approaches; and (3) RNA interference-based therapies, utilizing transcriptional inhibition as a therapeutic tool.
Table 2. Novel gene therapies.
| Therapeutic approach | Mechanism | Target | Preclinical model | Clinical trial | Reference |
|---|---|---|---|---|---|
| Antigen-based therapies |
In situ production of mutated hIL-13 fused to PE via adenovirus vectors |
IL-13 alfa 2R | X | [24] | |
| Direct cytotoxic effect and induction of anti-glioma immune responses |
Dual therapy: HC adenovirus expressing HC-Ad-TK + immunostimulatory HC-Ad-TetON-Flt3L |
Flt3L | X | X | [12] |
| Decrease tumor invasion | Reduction of MMP-2 via siRNA | MMP-2 | X | [36] | |
| Reduce tumor cell proliferation, increase apoptosis |
Blocking miR-21 function | miR-21 | X | [37] | |
| Repression of cancer stem cell population |
Up-regulation of miR-128 | miR-128 | X | [38] | |
| Increasing anti-glioma immune response |
Inhibition of TGFβ2 through antisense oligonucleotides |
TGFβ2 | X | [14] |
Abbreviations: hIL-13, human interleukin 13; PE, Pseudomonas exotoxin; IL-13 alfa 2R, interleukin 13 receptor alfa 2; HC-Ad-TK, high capacity (HC) adenoviruses expressing the herpes simplex tyrosine kinase; HC-Ad-TetON-Flt3L, immunostimulatory cytokine fms-like tyrosine kinase ligand 3; siRNA, small interference RNA; MMP2, matrix metalloproteinase-2; miR-21, micro-RNA 21; miR-128, micro-RNA 128; TGFβ2, transforming growth factor β 2.
Gene therapy allows targeting of pathways that can induce synergy of both conditional cytotoxic and immune stimulatory approaches. A recent study, which is about to be tested in a clinical trial, developed a combination of newly engineered high-capacity adenoviral vectors that encode both HSV1-TK (conditionally cytotoxic herpes simplex virus type 1 thymidine kinase) and Flt3L. Flt3L, whose expression is under Tet-ON system control, recruits bone marrow-derived dendritic cells to the tumor site, triggering an anti-glioblastoma multiforme-specific immune response. HSV1-TK works through a conditional cytotoxic approach in which, upon virus (HSV1) infection, thymidine kinase (TK) is delivered to tumor cells. In the presence of TK and upon administration of a prodrug these tumor cells end up producing cytotoxic metabolites, which culminate in tumor death. The combination of HSV1-TK and Flt3L has been shown to kill proliferating tumor cells in a rat model, leading to an increased animal survival and the development of a long-term anti-glioma immunological memory [12].
Another recent successful approach to block important glioma pro-survival pathways has been transcriptional inhibition through RNA interference. One of the most promising RNA interference tools has been the use of antisense oligonucleotides. They inhibit gene expression at the translational level by binding to a specific RNA sequence. A phase III clinical trial to evaluate the efficacy of antisense transforming growth factor β II (TGFβ2) (in patients is also underway. It is believed that, by silencing the TGFβ2 isoform, it will be possible to reduce glioma growth and proliferation by indirectly activating the immune response [13, 14]
Stem cells and stem cell modification
A number of in vitro and in vivo studies have demonstrated the unique migratory capacity of neural and mesenchymal stem cells to target glioma. In the setting of glioblastoma multiforme therapy, mesenchymal stem cells are attractive because it is relatively easy to isolate them from patients [15], while neural stem cells have shown more specific migratory potential towards glioblastoma multiforme.
Stem cell-based therapies rely mostly on expression of therapeutic genes and delivery of therapeutic agents (Table 3), and offer certain advantages over vector-based approaches. Modified stem cells can not only disperse into the tumor and reach more malignant cells but also allow for longer therapeutic gene expression. Such cells can be modified to express either direct cytotoxic molecules (such as tumor necrosis factor-related apoptosis inducing ligand, TRAIL) [16] or suicide genes that can convert pro-drugs into their active agents (cytosine deaminase or HSV-thymidine kinase). Following promising pre-clinical data, recently, a clinical trial was launched using human immortalized neural stem cells expressing a suicide CD gene (cytosine deaminase – HB1.F3.CD), followed by oral 5-fluocytosine. The introduction of the CD gene in a tumor induces the activation of the pro-drug 5-fluocytosine into 5-fluorouracil, resulting in intratumoral chemotherapy and, consequently, tumor shrinkage. (ClinicalTrails.gov Identifier: NCT 01172964) [17].
Table 3. Stem cell-based carriers’ therapies.
| Therapeutic approach | Mechanism | Target | Preclinical model |
Clinical trial |
Reference |
|---|---|---|---|---|---|
| Induction of apoptosis in glioma cells |
NSC/MSC engineered to express therapeutic genes, such as TRAIL |
Tumor cells | X | [16] | |
| Selective killing glioma cells | Oncolytic virus delivery | Tumor cells | X | [39] | |
| Selective killing glioma cells | Expression of suicide genes (CD) in NSCs – HB1.F3-CD |
Tumor cells | X | [17]; ClinicalTrails.gov Identifier: NCT 01172964 |
|
| Selective killing glioma cells | Delivery of therapeutic agents (scFv) | EGFRvIII | X | X | [18] |
Abbreviations: NSC, neural stem cell; MSC, mesenchymal stem cell; TRAIL, TNF-related apoptosis-inducing ligand; CD, cytosine deaminase; scFv, single-chain variable fragment; EGFRvIII, epidermal growth factor receptor variant III.
Alternatively, stem cells can function as in situ factories of therapeutic agents: such as immunomodulatory cytokines, antibodies (single-chain scFv–EGFRvIII) [18] or even oncolytic viruses [19]. Our lab has focused on the inherent tumor-tropic and adenovirus-replicating capabilities of such cells to target glioblastoma multiforme. We have shown that loading stem cells with conditionally replicative adenovirus (CRAds-pk7) does not significantly compromise their homing abilities, protects the payload from host immunosurveillance and, most importantly, prolongs animal survival better than direct adenovirus injection. Lastly, we noticed that neural stem cell-delivery approaches display superior therapeutic efficacy than mesenchymal stem cells in glioblastoma multiforme [20].
Overall, regardless of some pitfalls, carrier stem cells have proven to be a promising and an exciting new therapeutic approach for malignant gliomas.
Applications of nanotechnology in brain tumors
Nanomaterials offer several therapeutic strategies for the treatment of brain tumors [21, 22]. They can either act as drug carrier systems or induce glioma cytotoxicity directly. Drug carriers consist of polymer or lipid-based systems that allow for intratumoral injection of chemotherapeutic drugs or help such agents to cross the blood-brain barrier [23]. Recently, biodegradable polymer-based nanoparticles and gold nanoparticles have been explored as vehicles to target brain tumors [24-27]. They are mainly delivered by direct intratumoral injection, in situ retention through receptor-mediated endocytosis, transcytosis or membrane permeabilization, and blood-brain barrier disruption.
Once in situ, nanoparticles need an external inducer to generate an oscillatory or rotating momentum to induce glioma cell toxicity. The source can be heat, light or even magnetic field. In fact, induction of nanoparticles via local hyperthermia is already under Phase II study for treatment of brain tumors. Our group has been focused on magnetic field induction of oscillatory microdiscs bound to IL-13Rα2 to kill glioma cells [28] (Table 4). Our in vitro results show that even a low-frequency magnetic field can achieve approximately 90% of cancer cell destruction.
Table 4. Nanoparticle-based therapies.
| Therapeutic approach | Mechanism | Target | Preclinical model |
Clinical trial |
Reference |
|---|---|---|---|---|---|
| Tumor killing through local hyperthermia |
SPIO generates heat under alternating magnetic field |
Tumor cells through direct injection into the tumor |
X | [40] | |
| Tumor killing through spin-vortex magnetic discs under low-frequency alternating magnetic field |
Mechanical oscillation of micro discs trigger apoptosis |
Tumor cells | X | [41] | |
| Use of semiconductor nanomaterials (photo catalyst) |
Generation of cytotoxic reactive oxygen species under light exposure |
Tumor cells | X | [28] | |
| Deliver therapeutic drugs across BBB |
Intravenous injection of PMLA platform conjugated w/AON therapeutic payload |
Tumor cells | X | [24] | |
| Deliver therapeutic drugs | Transferrin conjugated polimersome loaded w/doxorubicin |
Tumor cells | X | [26] | |
| Deliver therapeutic drugs | Gold nanoparticle drug conjugate (5nm core size) |
EGFR | X | [25] | |
| MRI Contrast reagent and drug delivery |
Iron oxide nanoparticle | Tumor cells | X | [42] | |
| Deliver therapeutic drugs | MSC loaded with drug-loaded nanoparticles |
Tumor cells | X | [32] |
Abbreviations: SPIO, superparamagnetic iron oxide nanoparticles; PMLA, polymeric nanobioconjugate drug based on the poly (β-L-malic acid); AON, antisense oligonucleotide; BBB, blood brain barrier; EGFR, epidermal growth factor receptor; MRI, magnetic resonance imaging; MSC, mesenchymal stem cells.
In addition, nanotechnology can be combined with other therapies, such as stem cell-based carriers, offering new concepts for treatment of brain tumors [29,30]. Drug-loaded nanoparticles can be efficiently taken up by mesenchymal stem cells without affecting the cell viability [31,32]. Since they can be combined with a variety of drugs that are not able to cross the blood-brain barrier, drug-loaded nanoparticles greatly improved drug biodistribution and their therapeutic effect in brain tumor tissues. Today, they represent a new form of targeted tissue-specific delivery.
Conclusion
With the recent progresses made in understanding the molecular and genetic composition of malignant gliomas, promising new targets for therapy have emerged.
What does the future hold?
The integration of novel agents into existing treatment algorithms remains challenging. In recent years, basic and preclinical studies have revealed multiple new mechanisms of gliomagenesis and corresponding targets for treatment. Nevertheless, much from the translational research still needs to be explored to allow a better connection between basic science discoveries and clinical trials.
What questions remain?
Despite recent advances in glioblastoma multiforme treatment, there are still many open questions: (1) Determining the unique molecular and genetic profiles of tumors from individual patients; (2) Understanding the role of glioblastoma multiforme heterogeneity in therapeutic resistance; (3) Targeting glioma stem cells specifically without interfering with normal cell function or biologic stressors; (4) Improving drug delivery methods across the blood-brain barrier and into the tumor.
An effective treatment of brain tumors requires certain obstacles to be overcome. Anti-cancer agents that have been shown to work well against glioma cells in vitro are not optimally effective in vivo because they do not reach the desired location in sufficient doses. Therefore, modifications need to be implemented to allow better penetrance throughout the CNS and blood-brain barrier. Although a complete oncobiological understanding can enhance the therapeutic efficacy of traditional anti-glioblastoma strategies, the neurobiological complexity of such a diffuse, limited-access and heterogeneous target structure requires profound studies and new drug delivery concepts. Nanoparticle-based drug delivery systems, in situ production of immunomodulatory and anti-angiogenic agents through gene therapy vectors, or cell-based delivery vehicles are presented as promising options.
Only a better understanding of glioma neurobiology and heterogeneity will allow the design of more efficient approaches that are capable of selectively targeting glioma tumor cells. This strategy will spare normal brain tissue, extending patient survival and improving quality of life.
Acknowledgments
We would like to thank Atique Ahmed for his advice and comments on the paper. This research was supported by the NCI (R01CA122930, R01CA138587), the NINDS (R01NS077388), the National Institute of Neurological Disorders and Stroke (U01NS069997) and the American Cancer Society (RSG-07-276-01-MGO).
Abbreviations
- CD
cytosine deaminase
- CTLs
cytotoxic T lymphocytes
- EGFR
epidermal growth factor receptor
- Flt3L
FMS-like tyrosine kinase 3 ligand
- HSV
herpes simplex virus
- IL
interleukin
- TGF
transporting growth factor
Competing interests
The authors declare that they have no competing interests.
The electronic version of this article is the complete one and can be found at: http://f1000.com/reports/m/4/18
References
- 1.CBTRUS Statistical Report: Primary Brain and Central Nervous System Tumors Diagnosed in the United States in 2004-2007. 2011. [DOI] [PMC free article] [PubMed]
- 2.Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJB, Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, Hau P, Brandes AA, Gijtenbeek J, Marosi C, Vecht CJ, Mokhtari K, Wesseling P, Villa S, Eisenhauer E, Gorlia T, Weller M, Lacombe D, Cairncross JG, Mirimanoff R. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009;10:459–66. doi: 10.1016/S1470-2045(09)70025-7. [DOI] [PubMed] [Google Scholar]; F1000 Factor 6Minesh Mehta and Deepak Khuntia 14 Jul 2009
- 3.Bonavia R, Inda M, Cavenee WK, Furnari FB. Heterogeneity maintenance in glioblastoma: a social network. Cancer Res. 2011;71:4055–60. doi: 10.1158/0008-5472.CAN-11-0153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kesari S. Understanding glioblastoma tumor biology: the potential to improve current diagnosis and treatments. Semin Oncol. 2011;38(Suppl 4):S2–10. doi: 10.1053/j.seminoncol.2011.09.005. [DOI] [PubMed] [Google Scholar]
- 5.Platten M, Wick W, Weller M. Malignant glioma biology: role for TGF-β in growth, motility, angiogenesis, and immune escape. Microsc Res Tech. 2001;52:401–10. doi: 10.1002/1097-0029(20010215)52:4<401::AID-JEMT1025>3.0.CO;2-C. [DOI] [PubMed] [Google Scholar]; F1000 Factor 6Mat Lesniak 15 Aug 2012
- 6.Chang C, Huang Y, Yang D, Kikuta K, Wei K, Kubota T, Yang W. A phase I/II clinical trial investigating the adverse and therapeutic effects of a postoperative autologous dendritic cell tumor vaccine in patients with malignant glioma. J Clin Neurosci. 2011;18:1048–54. doi: 10.1016/j.jocn.2010.11.034. [DOI] [PubMed] [Google Scholar]; F1000 Factor 6Mat Lesniak 15 Aug 2012
- 7.Xiong W, Candolfi M, Liu C, Muhammad AKMG, Yagiz K, Puntel M, Moore PF, Avalos J, Young JD, Khan D, Donelson R, Pluhar GE, Ohlfest JR, Wawrowsky K, Lowenstein PR, Castro MG. Human Flt3L generates dendritic cells from canine peripheral blood precursors: implications for a dog glioma clinical trial. PLoS ONE. 2010;5:e11074. doi: 10.1371/journal.pone.0011074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Wölfl M, Merker K, Morbach H, van Gool SW, Eyrich M, Greenberg PD, Schlegel PG. Primed tumor-reactive multifunctional CD62L+ human CD8+ T cells for immunotherapy. Cancer Immunol Immunother. 2011;60:173–86. doi: 10.1007/s00262-010-0928-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sampson JH, Heimberger AB, Archer GE, Aldape KD, Friedman AH, Friedman HS, Gilbert MR, Herndon JE, McLendon RE, Mitchell DA, Reardon DA, Sawaya R, Schmittling RJ, Shi W, Vredenburgh JJ, Bigner DD. Immunologic escape after prolonged progression-free survival with epidermal growth factor receptor variant III peptide vaccination in patients with newly diagnosed glioblastoma. J Clin Oncol. 2010;28:4722–9. doi: 10.1200/JCO.2010.28.6963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Del Vecchio CA, Li G, Wong AJ. Targeting EGF receptor variant III: tumor-specific peptide vaccination for malignant gliomas. Expert Rev Vaccines. 2012;11:133–44. doi: 10.1586/erv.11.177. [DOI] [PubMed] [Google Scholar]; F1000 Factor 6Mat Lesniak 15 Aug 2012
- 11.Li L, Quang TS, Gracely EJ, Kim JH, Emrich JG, Yaeger TE, Jenrette JM, Cohen SC, Black P, Brady LW. A Phase II study of anti-epidermal growth factor receptor radioimmunotherapy in the treatment of glioblastoma multiforme. J Neurosurg. 2010;113:192–8. doi: 10.3171/2010.2.JNS091211. [DOI] [PubMed] [Google Scholar]; F1000 Factor 6Mat Lesniak 15 Aug 2012
- 12.Muhammad AKMG, Puntel M, Candolfi M, Salem A, Yagiz K, Farrokhi C, Kroeger KM, Xiong W, Curtin JF, Liu C, Lawrence K, Bondale NS, Lerner J, Baker GJ, Foulad D, Pechnick RN, Palmer D, Ng P, Lowenstein PR, Castro MG. Study of the efficacy, biodistribution, and safety profile of therapeutic gutless adenovirus vectors as a prelude to a phase I clinical trial for glioblastoma. Clin Pharmacol Ther. 2010;88:204–13. doi: 10.1038/clpt.2009.260. [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Factor 6Mat Lesniak 15 Aug 2012
- 13.Schlingensiepen KH, Fischer-Blass B, Schmaus S, Ludwig S. Antisense therapeutics for tumor treatment: the TGF-β2 inhibitor AP 12009 in clinical development against malignant tumors. Recent Results Cancer Res. 2008;177:137–50. doi: 10.1007/978-3-540-71279-4_16. [DOI] [PubMed] [Google Scholar]
- 14.Vallières L. Trabedersen, a TGFβ2-specific antisense oligonucleotide for the treatment of malignant gliomas and other tumors overexpressing TGFβ2. IDrugs. 2009;12:445–53. [PubMed] [Google Scholar]; F1000 Factor 6Mat Lesniak 15 Aug 2012
- 15.Hayes M, Curley G, Laffey JG. Mesenchymal stem cells - a promising therapy for Acute Respiratory Distress Syndrome. F1000 Med Rep. 2012;4:2. doi: 10.3410/M4-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Balyasnikova IV, Ferguson SD, Han Y, Liu F, Lesniak MS. Therapeutic effect of neural stem cells expressing TRAIL and bortezomib in mice with glioma xenografts. Cancer Lett. 2011;310:148–59. doi: 10.1016/j.canlet.2011.06.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Aboody K, Capela A, Niazi N, Stern JH, Temple S. Translating stem cell studies to the clinic for CNS repair: current state of the art and the need for a Rosetta Stone. Neuron. 2011;70:597–613. doi: 10.1016/j.neuron.2011.05.007. [DOI] [PubMed] [Google Scholar]
- 18.Balyasnikova IV, Ferguson SD, Sengupta S, Han Y, Lesniak MS. Mesenchymal stem cells modified with a single-chain antibody against EGFRvIII successfully inhibit the growth of human xenograft malignant glioma. PLoS ONE. 2010;5:e9750. doi: 10.1371/journal.pone.0009750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Sonabend AM, Rolle CE, Lesniak MS. The role of regulatory T cells in malignant glioma. Anticancer Res. 2008;28:1143–50. [PubMed] [Google Scholar]
- 20.Ahmed AU, Alexiades NG, Lesniak MS. The use of neural stem cells in cancer gene therapy: predicting the path to the clinic. Curr Opin Mol Ther. 2010;12:546–52. [PMC free article] [PubMed] [Google Scholar]
- 21.Caruso G, Raudino G, Caffo M, Alafaci C, Granata F, Lucerna S, Salpietro FM, Tomasello F. Nanotechnology platforms in diagnosis and treatment of primary brain tumors. Recent Pat Nanotechnol. 2010;4:119–24. doi: 10.2174/187221010791208786. [DOI] [PubMed] [Google Scholar]
- 22.Mohs AM, Provenzale JM. Applications of nanotechnology to imaging and therapy of brain tumors. Neuroimaging Clin N Am. 2010;20:283–92. doi: 10.1016/j.nic.2010.04.002. [DOI] [PubMed] [Google Scholar]
- 23.Schroeder A, Heller DA, Winslow MM, Dahlman JE, Pratt GW, Langer R, Jacks T, Anderson DG. Treating metastatic cancer with nanotechnology. Nat Rev Cancer. 2012;12:39–50. doi: 10.1038/nrc3180. [DOI] [PubMed] [Google Scholar]; F1000 Factor 6Mat Lesniak 15 Aug 2012
- 24.Candolfi M, Xiong W, Yagiz K, Liu C, Muhammad AKMG, Puntel M, Foulad D, Zadmehr A, Ahlzadeh GE, Kroeger KM, Tesarfreund M, Lee S, Debinski W, Sareen D, Svendsen CN, Rodriguez R, Lowenstein PR, Castro MG. Gene therapy-mediated delivery of targeted cytotoxins for glioma therapeutics. Proc Natl Acad Sci USA. 2010;107:20021–6. doi: 10.1073/pnas.1008261107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Cheng Y, Meyers JD, Agnes RS, Doane TL, Kenney ME, Broome A, Burda C, Basilion JP. Addressing Brain Tumors with Targeted Gold Nanoparticles: A New Gold Standard for Hydrophobic Drug Delivery? Small. 2011 doi: 10.1002/smll.201100628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Pang Z, Gao H, Yu Y, Guo L, Chen J, Pan S, Ren J, Wen Z, Jiang X. Enhanced intracellular delivery and chemotherapy for glioma rats by transferrin-conjugated biodegradable polymersomes loaded with doxorubicin. Bioconjug Chem. 2011;22:1171–80. doi: 10.1021/bc200062q. [DOI] [PubMed] [Google Scholar]; F1000 Factor 6Mat Lesniak 15 Aug 2012
- 27.Wohlfart S, Khalansky AS, Gelperina S, Maksimenko O, Bernreuther C, Glatzel M, Kreuter J. Efficient chemotherapy of rat glioblastoma using doxorubicin-loaded PLGA nanoparticles with different stabilizers. PLoS ONE. 2011;6:e19121. doi: 10.1371/journal.pone.0019121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Rozhkova EA, Ulasov I, Lai B, Dimitrijevic NM, Lesniak MS, Rajh T. A high-performance nanobio photocatalyst for targeted brain cancer therapy. Nano Lett. 2009;9:3337–42. doi: 10.1021/nl901610f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Cheng H, Kastrup CJ, Ramanathan R, Siegwart DJ, Ma M, Bogatyrev SR, Xu Q, Whitehead KA, Langer R, Anderson DG. Nanoparticulate cellular patches for cell-mediated tumoritropic delivery. ACS Nano. 2010;4:625–31. doi: 10.1021/nn901319y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Roger M, Clavreul A, Venier-Julienne M, Passirani C, Montero-Menei C, Menei P. The potential of combinations of drug-loaded nanoparticle systems and adult stem cells for glioma therapy. Biomaterials. 2011;32:2106–16. doi: 10.1016/j.biomaterials.2010.11.056. [DOI] [PubMed] [Google Scholar]; F1000 Factor 6Mat Lesniak 15 Aug 2012
- 31.Li L, Guan Y, Liu H, Hao N, Liu T, Meng X, Fu C, Li Y, Qu Q, Zhang Y, Ji S, Chen L, Chen D, Tang F. Silica nanorattle-doxorubicin-anchored mesenchymal stem cells for tumor-tropic therapy. ACS Nano. 2011;5:7462–70. doi: 10.1021/nn202399w. [DOI] [PubMed] [Google Scholar]; F1000 Factor 6Mat Lesniak 15 Aug 2012
- 32.Roger M, Clavreul A, Venier-Julienne M, Passirani C, Sindji L, Schiller P, Montero-Menei C, Menei P. Mesenchymal stem cells as cellular vehicles for delivery of nanoparticles to brain tumors. Biomaterials. 2010;31:8393–401. doi: 10.1016/j.biomaterials.2010.07.048. [DOI] [PubMed] [Google Scholar]
- 33.Study of a drug [DCVax-Brain] to treat newly diagnosed GBM brain cancer - NCT00045968. Clinical Trials 18/4/2012] Available from: http://clinicaltrials.gov/ct2/show/NCT00045968?term=DCVax&rank=2.
- 34.Akasaki Y, Kikuchi T, Irie M, Yamamoto Y, Arai T, Tanaka T, Joki T, Abe T. Cotransfection of Poly(I: C) and siRNA of IL-10 into fusions of dendritic and glioma cells enhances antitumor T helper type 1 induction in patients with glioma. J Immunother. 2011;34:121–8. doi: 10.1097/CJI.0b013e3181e5c278. [DOI] [PubMed] [Google Scholar]
- 35.Lee J, Han HD, Shahzad MMK, Kim SW, Mangala LS, Nick AM, Lu C, Langley RR, Schmandt R, Kim H, Mao S, Gooya J, Fazenbaker C, Jackson D, Tice DA, Landen CN, Coleman RL, Sood AK. EphA2 immunoconjugate as molecularly targeted chemotherapy for ovarian carcinoma. J Natl Cancer Inst. 2009;101:1193–205. doi: 10.1093/jnci/djp231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kargiotis O, Chetty C, Gondi CS, Tsung AJ, Dinh DH, Gujrati M, Lakka SS, Kyritsis AP, Rao JS. Adenovirus-mediated transfer of siRNA against MMP-2 mRNA results in impaired invasion and tumor-induced angiogenesis, induces apoptosis in vitro and inhibits tumor growth in vivo in glioblastoma. Oncogene. 2008;27:4830–40. doi: 10.1038/onc.2008.122. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 37.Zhou X, Ren Y, Moore L, Mei M, You Y, Xu P, Wang B, Wang G, Jia Z, Pu P, Zhang W, Kang C. Downregulation of miR-21 inhibits EGFR pathway and suppresses the growth of human glioblastoma cells independent of PTEN status. Lab Invest. 2010;90:144–55. doi: 10.1038/labinvest.2009.126. [DOI] [PubMed] [Google Scholar]
- 38.Godlewski J, Nowicki MO, Bronisz A, Williams S, Otsuki A, Nuovo G, Raychaudhury A, Newton HB, Chiocca EA, Lawler S. Targeting of the Bmi-1 oncogene/stem cell renewal factor by microRNA-128 inhibits glioma proliferation and self-renewal. Cancer Res. 2008;68:9125–30. doi: 10.1158/0008-5472.CAN-08-2629. [DOI] [PubMed] [Google Scholar]
- 39.Ahmed AU, Tyler MA, Thaci B, Alexiades NG, Han Y, Ulasov IV, Lesniak MS. A comparative study of neural and mesenchymal stem cell-based carriers for oncolytic adenovirus in a model of malignant glioma. Mol Pharm. 2011;8:1559–72. doi: 10.1021/mp200161f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Müller S. Magnetic fluid hyperthermia therapy for malignant brain tumors--an ethical discussion. Nanomedicine. 2009;5:387–93. doi: 10.1016/j.nano.2009.01.011. [DOI] [PubMed] [Google Scholar]
- 41.Kim D, Rozhkova EA, Ulasov IV, Bader SD, Rajh T, Lesniak MS, Novosad V. Biofunctionalized magnetic-vortex microdiscs for targeted cancer-cell destruction. Nat Mater. 2010;9:165–71. doi: 10.1038/nmat2591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Tang M, Russell PJ, Khatri A. Magnetic nanoparticles: prospects in cancer imaging and therapy. Discov Med. 2007;7:68–74. [PubMed] [Google Scholar]
