Você está na página 1de 13

Neuro-Oncology Advance Access published June 9, 2016

Neuro-Oncology
Neuro-Oncology 2016; 0, 1 – 13, doi:10.1093/neuonc/now096

Stem cell-based therapies for tumors in the brain: are we there yet?
Khalid Shah

Stem Cell Therapeutics and Imaging Program, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (K.S.);
Molecular Neurotherapy and Imaging Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
(K.S.); Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (K.S.); Department
of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (K.S.); Harvard Stem Cell Institute,
Harvard University, Cambridge, Massachusetts (K.S.)

Downloaded from http://neuro-oncology.oxfordjournals.org/ at University of Cambridge on June 11, 2016


Corresponding Author: Khalid Shah, MS, PhD 5403, Bldg 149, 13th street, Charlestown, MA 01810 (kshah@mgh.harvard.edu).

Advances in understanding adult stem cell biology have facilitated the development of novel cell-based therapies for cancer. Re-
cent developments in conventional therapies (eg, tumor resection techniques, chemotherapy strategies, and radiation therapy)
for treating both metastatic and primary tumors in the brain, particularly glioblastoma have not resulted in a marked increase in
patient survival. Preclinical studies have shown that multiple stem cell types exhibit inherent tropism and migrate to the sites of
malignancy. Recent studies have validated the feasibility potential of using engineered stem cells as therapeutic agents to target
and eliminate malignant tumor cells in the brain. This review will discuss the recent progress in the therapeutic potential of stem
cells for tumors in the brain and also provide perspectives for future preclinical studies and clinical translation.

Keywords: brain tumors, glioblastoma (GBM), receptors, stem cells, therapeutics.

Tumors in the brain are categorized as either primary brain suffering from recurrence and ,5% of patients surviving
tumors, defined by their origin from cells autochthonous to 5 years after diagnosis.6,7
the brain, and secondary brain tumors, which originate from The current standard therapy for GBM is a combination of
metastatic cells from peripheral tumor sites.1 Although the cytoreductive surgery followed by concurrent and adjuvant
exact incidence and ratio are controversial, metastatic brain chemotherapy with temozolomide (TMZ) and radiation thera-
tumors are known to be far more common than primary py.8 Current limitations in the treatment of GBM involve hin-
brain tumors and have been estimated to outnumber primary drances in achieving complete tumor resection, tumor
brain tumors by more than 4 to 1.2 Primary brain tumors are resistance, challenges in penetrating the blood brain barrier
commonly further subcategorized according to their cell of (BBB), and insufficient accumulation of therapeutic agents at
origin, which can be within the brain tissue itself (eg, glioma), the tumor site (www.ncbi.nlm.nih.gov). Thus, patients with
the membranes around the brain (eg. meningioma), the GBM are in dire need of alternative treatments that can circum-
nerves (eg, nerve sheath tumors), glands (eg, pituitary tu- vent the limitations posed by the current standard of care.
mors), and vessels (eg, choroid plexus tumors) within the cra- Many adult stem cells (SCs) display intrinsic tumor-tropic prop-
nium.3 The most frequent type of primary malignant brain erties, making them attractive candidates for targeted delivery
tumor is glioma, of which . 50% may be categorized as of anticancer biologics by modifying them to stably express/re-
WHO grade IV glioblastoma (GBM). 4 GBM may either arise lease a variety of anticancer agents. This review aims to high-
de novo (referred to as primary GBM) or much less often light the most recent advances in SC-based treatments for
(about 10%) may progress from lower-grade or anaplastic as- treatment of tumors in the brain, particularly GBM, and discuss
trocytomas (secondary GBM).1 GBM is characterized as malig- the most recent SC-based therapeutic clinical trials for these
nant, mitotically active, and predisposed to necrosis, and its tumor types.
very low median survival (14.6 months) is attributed to
unique treatment limitations such as a high average age of
onset, tumor location, and poor current understanding of
Stem Cell Sources
the tumor’s pathophysiology.5 With a low median survival, Stem cells are the natural source of embriogenetic tissue gen-
both primary and secondary GBMs are both associated with eration and continuous regeneration throughout adult life. Over
an extremely poor prognosis, with virtually all patients the last few decades, 3 major stem cell types (ie, embryonic,

Received 22 January 2016; accepted 8 April 2016


# The Author(s) 2016. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved.
For permissions, please e-mail: journals.permissions@oup.com.

1 of 13
Shah: Stem cell-based therapies for brain tumors

adult [mesenchymal and neural], and induced pluripotent) factor 1 (SDF1; also known as CXCL12) is one of the best-
have been identified and extensively characterized. Embryonic studied mediators of stem cell tropism. SDF1 and CXCR4 are
stem cells (ESCs) are derived from the inner cell mass of expressed at high levels, particularly in the DG and SVZ
blastocyst-stage embryos, and closely related embryonic where they are involved in regulating the tropism of endoge-
germ cells were among the first cell types identified as possible nous NSCs.26 The migration of exogenously modified thera-
sources for stem cell therapy.9 However, owing to numerous peutic stem cells has been shown to proceed in a similar
concerns related to the use of ESCs, adult stem cell types fashion to that of endogenous NSCs toward tumors in the
(namely mesenchymal stem cells [MSCs], neural stem cells brain.27,27–30 The chemokine stem cell factor (SCF) has been
[NSCs], and induced pluripotent stem cells [ipSCs]) have been shown to be upregulated by cells that reside in and around le-
explored extensively for therapeutic purposes.10 MSCs are sioned areas and induce the migration of exogenous NSCs to-
spindle-shaped, fibroblast-like multipotent stem cells that are ward pathology within the brain through interaction with the
responsible for regeneration and cellular homeostasis in almost tyrosine receptor kinase c-Kit, as demonstrated in a GBM
all tissues and have the potential of converting to tissue types model.31 NSCs also express CCR2 and migrate in the direction

Downloaded from http://neuro-oncology.oxfordjournals.org/ at University of Cambridge on June 11, 2016


of other lineages both within and across germ lines.11 MSCs of a MCP-1 gradient toward neoplastic lesions within the
have been isolated from a number of organs including bone brain.32 Hypoxia is known to promote NSC tropism in vitro33
marrow, adipose tissue, fetal tissues, dental pulp, umbilical and in vivo,34 mainly due to the upregulation of vascular en-
cord, Wharton’s jelly, and other tissue types.12 Most of the pre- dothelial growth factor (VEGF) by hypoxic cells, which has
clinical studies to date have been performed with bone been observed to result in increased expression of chemotac-
marrow-derived MSCs; however, adipose tissue and umbilical tic factors Ang2 and GROa.35 These 2 proteins promote the
cord blood are other MSC sources that have received consider- migration of NSCs toward regions of hypoxia within the
able attention in recent years. NSCs, also referred to as neural brain. VEGF can induce NSC migration in a reactive oxygen spe-
precursor cells (NPCs), are a heterogeneous population of self- cies (ROS)- and focal adhesion kinase (FAK)-dependent man-
renewing and multipotent cells of both the developing and the ner. 36 Other influential signaling pathways involved in SC
adult CNS13 and are especially enriched in the subventricular homing have been elucidated and include hepatocyte growth
zone (SVZ) and the subgranular zone in the hippocampal den- factor (HGF)/c-MET receptor, 37 urokinase-type plasminogen
tate gyrus DG. NSCs have been proven to be an invaluable activator (uPA)/uPA receptor (uPAR),38 platelet-derived growth
source for developing cell-based therapies, especially for neuro- factor (PDGF)/uPAR/b1 integrin, 39 and transforming growth
degenerative disorders and also for other conditions, particular- factor b (TGFb)/TGFb receptor (TGFbRII).40 Macrophage mi-
ly certain cancer types.10,14 Induced pluripotent stem cells gration inhibitory factor (MIF)/CXCR4 has been recently identi-
(iPSCs) generated from somatic cells have emerged as popular fied as the dominant chemotactic axis for recruiting human
and effective stem cell types for therapy.15 The iPSCs are gen- MSCs to tumors. 41 Although the tumor-homing ability of
erated by allowing a differentiated somatic cell to revert to em- MSCs and NSCs has been extensively studied and established,
bryonic stage via induced expression of transcription factors understanding the ability of iPSC-derived SCs to home to tu-
OCT4, SOX2, KLF4, and C-MYC associated with pluripotency.16 mors is still in its infancy. Recently, iPSC-derived NSCs have
iPSCs can be derived from a wide variety of starting cells, been shown to have potent glioma tropism.42 The degree of
even though easy accessibility to fibroblasts makes them the SC homing to tumors in vivo is influenced by diverse factors
most common source for iPSC generation.17 Recent studies including the nature of the SC and the tumor microenviron-
have shown the ability of ectopic expression of cell type-specific ment.10 In addition to homing to the primary tumor mass,
transcription factors to directly switch cell fates between both NSCs and MSCs have been shown to efficiently track ma-
somatic cells, thus circumventing the pluripotent state and lignant microinvasive deposits in the brain.24,27 Therefore ,the
eliminating any risk of malignant transformation.18 Similarly, potential for migration of SCs to the main tumor mass and the
MSCs derived from induced pluripotent stem cells (iPSC-MSCs) microinvasive deposits offers a strong rationale for the use of
have also evolved as a promising alternative cell source for SCs in treating brain tumors.
MSCs and regenerative medicine. Several studies have revealed
successful derivation of functional iPSC-MSCs19,20 that were
shown to have similar characteristics as bone marrow-derived
Therapeutic Applications for Brain Tumors
MSCs,21 including expression of typical mesenchymal markers
and the capacity to efficiently differentiate into osteogenic The major limitations of current GBM therapies are (1) margin-
and chondrogenic lineages. al chance of curative resection due to the highly invasive na-
ture of GBM, which allows tumor cells to infiltrate deeply into
normal tissue; (2) the frequent relapses after seemingly cura-
tive resection due to the seeding of microsatellite deposits in
Stem Cell Homing and Migration to Tumors
the surrounding neuropil; (3) the pharmacokinetic problems
The intrinsic homing property of a variety of stem cell types to associated with the blood-brain-barrier that impede the accu-
brain pathologies such as ischemic, neoplastic, and demyelinating mulation of therapeutic drug concentrations in the tumoral
lesions has been unraveled during the past decade.22 – 25 A interstitium; and (4) the caution that has to be incorporated
number of studies have shown tumor tropism of both NSCs with high-dose drug treatment and radiation therapy due to
and MSCs when injected via different routes in mouse tumor the vulnerability of normal brain tissue and the serious conse-
models of brain tumors (reviewed in10). The G-protein coupled quences of brain damage to the patient’s quality of life. Trans-
receptor CXCR4 and its only known ligand, stromal cell-derived planted SCs, engineered to release therapeutic agents, can be

2 of 13 Neuro-Oncology
Shah: Stem cell-based therapies for brain tumors

used as an engraftable, highly mobile, “organic” delivery vehi- broad categories: direct and indirect effectors. The direct effec-
cle for tumor treatment. This is reliant on the biological and tors include proapoptotic and antiproliferative proteins.
physiological differences between normal and malignant
cells and also on the GBM tumor-homing properties of engi-
neered SCs. The latter is a crucial factor in clinical effective- Proapoptotic Proteins
ness as engineered SCs can “home in” on microsatellite TRAIL (tumor necrosis factor-related apoptosis inducing ligand)
deposits of escaping GBM cells that are known to give rise to binds to death receptors (DR)4/5 specifically expressed on
recurrent tumors in the brain. Both unmodified SCs, which tumor cells and induces caspase-mediated apoptosis. 24,45
possess intrinsic antitumor effects attributed to their released Previous studies on intratumoral implantation of murine pri-
factors and physical interactions with tumor cells,43 and mod- mary NSCs engineered with full length TRAIL in established
ified SCs have been utilized to treat tumors in the brain. Some GBM revealed a significant reduction of tumor burden.46 How-
of the most promising are outlined in Fig. 1 and are discussed ever, TRAIL is a type 2 receptor membrane protein, and its re-
below. lease into the SC environment requires accidental cleavage

Downloaded from http://neuro-oncology.oxfordjournals.org/ at University of Cambridge on June 11, 2016


from its membrane docking site. We have engineered recom-
binant TRAIL that consists of a fusion between the extracellu-
lar domain of TRAIL and the extracellular domain of the hFlt3
Modifying Stem Cells Genetically to Release Direct and
ligand, which is a ligand for Flt3 tyrosine kinase receptor and
Indirect Effector Proteins mediates both secretion through the endoplasmic retic-
SCs are typically modified by viral transduction, although non- ulum. 47 Both MSCs and NSCs transduced with a lentiviral
viral methods to express transgenes encoding secretable effec- expression cassette for secretable (S)-TRAIL have shown anti-
tor proteins have been reported.25,44 Depending on whether the tumor effects in mouse tumor models of both nodular and in-
therapeutic proteins act directly on malignant tumor cells in the vasive GBM.24,48 Recent studies have confirmed the cytotoxic
brain or upon tumor-supporting cells in the tumor microenvi- effects of TRAIL-engineered SCs on GBMs, 49 indicating the
ronment, SC secretion of such proteins can be divided into 2 therapeutic potential of this strategy. Recently, we have

Fig. 1. Modifying stem cells for brain tumor therapy. Stem cells can be expanded and manipulated in vitro to express a variety of
tumor-antagonizing transgenes to establish antitumor effect following stem cell implantation in vivo.

Neuro-Oncology 3 of 13
Shah: Stem cell-based therapies for brain tumors

developed in vivo imageable breast-to brain metastasis progression in mice bearing highly malignant human GBM.60
tumor models and shown that intra-arterial injection of These studies suggest that SCs engineered to express antian-
SC-S-TRAIL suppressed metastatic tumor growth and giogenic agents such as PEX and aaTSP-1 could be exploited
prolonged the survival of mice bearing metastatic tumors in for enhanced therapeutic benefit.
the brain.50

Immunostimulatory Molecules
Antiproliferative Proteins
The rationale for utilizing immunostimulatory molecules is
Among the number of antiproliferative molecules, a limited
based on shifting the immunosuppressive tumor milieu to-
number of molecules can be expressed and released in the
wards directing an immune response against the cancer.68 In-
extracellular milieu. The antitumor effects of SCs expressing
terleukins are known to regulate inflammatory and immune
cytokines such as interferon (INF)-b in brain tumors was
responses and have antitumor effects.69 Both murine and
shown in an early study revealing that human SCs expressing
human primary NSCs engineered to express interleukin-12

Downloaded from http://neuro-oncology.oxfordjournals.org/ at University of Cambridge on June 11, 2016


IFN-b were able to track murine GBM tumors when adminis-
have been shown to have treatment efficacy on GBM in mice
tered systemically.51 IFN-b, which can also act as an alterna-
and rats, respectively.70,71 Human MSCs secreting IL-12 or
tive immunogenic candidate, 52,53 and (IFN)-a 54 have both
IL-18 have also shown efficacy in mice-bearing GBM.72–74
been shown to negatively regulate tumor growth in a variety
The sustained presence of SC-delivered ILs resulted in the
of preclinical cancer models. Recent studies have further
activation of natural killer cells and the recruitment of
shown that the release of biologics from SCs can outcompete
tumor-specific T cells.
or sterically block the binding of endogenous ligands to their
Immunotherapy employing peripherally implanted cyto-
cognate receptors such as epidermal growth factor receptor
kine-secreting inactivated tumor cells in syngeneic tumor
(EGFR) or its tumor-specific antigen EGFRvIII variant, 55,56
models has shown promising results by potentially eradicating
resulting in the inhibition of proliferation pathways in
intracranial GBM through the induction of a local proinflam-
tumor cells.
matory response in the tumor microenvironment.75,76 Several
Bone morphogenetic proteins (BMPs) activate their cognate
reports show that MSCs can potentially synergize with this ap-
receptors (BMPRs), and in vivo delivery of BMP4 has been shown
proach when exposed to cytokines such as interferon-gamma
to effectively block the tumor growth post intracerebral grafting
(IFNg). 77,78 As such, peripheral immunotherapy using
of human GBM cells.57 In a recent study, human adipose-
cytokine-secreting tumor cells has been shown to eradicate
derived MSCs engineered to express BMP4 were shown to prolong
experimental GBM by inducing a proinflammatory tumor mi-
survival in mice bearing GBM.58 Other studies on the regulated
croenvironment. 76,79 Recent studies have shown that the
expression of proteins that induce cell cycle arrest, such as
intratumoral implantation of MSCs into rats bearing GBM, in
growth-arrest specific-1 (Gas1) in SCs, have shown antitumor ef-
addition to peripheral immunotherapy with interferon-
fects in mouse tumor models of GBM.59
gamma (IFNg)-secreting inactivated tumor cells, significantly
Indirect effectors of tumor cells include antiangiogenic
improved animal survival.80 Although the complete mecha-
thrombospondin-1 (aaTSP-1),60 PEX (a fragment of metallopro-
nism of action in this context remains unclear, in vitro results
teinase-2),61 and immunostimulatory molecules such as inter-
suggest that MSCs in response to IFNg act as conditional
leukins (ILs).
antigen-presenting cells, upregulate MHC classes I and II mol-
ecules, and secrete low amounts of immunosuppressive mol-
Antiangiogenic Agents ecules, ultimately inducing an intratumoral antigen-specific
cytotoxic CD8+ T cell response.
Tumor angiogenesis represents the ability of a tumor to stimu-
late new blood vessel formation for self-sustained growth.62
Previous clinical studies have shown that intravascular delivery
Modifying Stem Cells Genetically to Release
of antiangiogenic drugs transiently normalizes the abnormal
vasculature, thus reducing tumor-associated vasogenic brain Tumor-targeted Toxins
edema and providing a clinical benefit for GBM patients.63 Pseudomonas exotoxin (PE) potently blocks protein synthesis by
The decrease in mean vessel diameter and permeability64 catalyzing the inactivation of elongation factor-2 (EF-2). Target-
and increased pericyte coating of small vasculature65 have ed PE-cytotoxins have been used as antitumor agents; howev-
been shown to be associated with vessel normalization. Previ- er, their off-target delivery, systemic toxicity, and short
ous studies have shown that MSCs localize to tumor vascula- chemotherapeutic half-life have confounded their effective
ture upon intratumoral implantation,66 and recent findings clinical translation in GBM patients. 81 In a recent study, we
demonstrated that MSCs display pericyte markers and intratu- have created toxin-resistant SCs by modifying endogenous
morally grafted MSCs could possibly function as tumor peri- EF-2 and engineered them to secrete PE-cytotoxins that specfi-
cytes.67 Human NSCs expressing PEX, a naturally occurring cally target expressed interleukin (IL)-13 receptor subunit
fragment of human metalloproteinase-2 when injected directly alpha-2 or overexpressed EGFR in GBM. The release of IL13-PE
into murine GBM, have shown efficacy post-treatment.61 In our from SCs in a clinically relevant GBM resection model resulted
previous studies, we have shown that NSCs engineered to ex- in tumor cell-specific killing and led to increased long-term
press antiangiogenic repeats of anti-angiogeneic three type 1 survival of mice compared with IL13-PE protein infusion.82
repeats (3TSR) of thrombospondin (TSP)-1 markedly reduced A recent study has shown that intratumoral injection of
tumor vessel-density, resulting in the inhibition of tumor MSCs expressing a bispecific immunotoxin, VEGF165-ephrin

4 of 13 Neuro-Oncology
Shah: Stem cell-based therapies for brain tumors

A1-PE38KDEL, was effective at inhibiting tumor growth in a ma- Stem Cells Loaded With Oncolytic Virus
lignant GBM tumor model.83
Oncolytic viruses (OVs) are natural or genetically modified vi-
ruses that, upon infection, selectively replicate in and kill neo-
Modifying Stem Cells Genetically to Induce Suicide plastic cells while sparing healthy cells.101 However, clearance
Therapy of the virus by host defense mechanisms post systemic ad-
ministration and insufficient viral spread post intratumoral
SC-mediated suicide therapy is a strategy whereby SCs are en- administration confound their therapeutic efficacy. In an ef-
gineered to express an enzyme that converts a nontoxic pro- fort to improve delivery of OV-based therapeutics and circum-
drug into a cytotoxic drug, resulting in efficiently killing vent antiviral immunity, a number of studies have explored
surrounding cells by the bystander effect. Three major prodrug the possibility of using SCs as delivery vehicles.11,102 Different
systems are currently being used: cytosine deaminase (CD)/ SC types have been utilized for local release of intact replicat-
5-fluorocytosine (5-FC); herpes simplex virus thymidine kinase ing oncolytic adenoviruses administered systemically or intra-
(HSV-TK)/ganciclovir (GCV); and carboxylesterase (CE)/ tumorally into mouse models of GBM.103 – 105 Previous studies

Downloaded from http://neuro-oncology.oxfordjournals.org/ at University of Cambridge on June 11, 2016


camptothecin-11 (CPT-11). Both MSCs and NSCs have been have shown effective homing of NSCs loaded with the onco-
modified with the CD/5-FC system, tested in mouse models lytic adenovirus, CRAd-Survivin-pk7, to intracranial GBM, re-
of GBM84,85 and medulloblastoma,86 and shown to result in sulting in increased animal survival. 106 Recently, we have
tumor regression and prolonged survival. The HSV-TK system shown that hyaluronan or hyaluronic acid (HA) is highly ex-
has been used in modified MSCs and relies on the formation pressed in a majority of tumor xenografts established from
of gap junctions between the SCs and surrounding target patient-derived GBM lines that present both invasive and nod-
cells for an efficient bystander effect. MSCs expressing HSV-TK ular phenotypes. 107 Intratumoral injection of MSCs loaded
have shown efficacy in animal models of GBM.87 – 89 Human with a conditionally replicating adenovirus expressing soluble
NSCs bearing the CE/CPT-11 system have proven effective in hyaluronidase (ICOVIR17) into GBM enhanced viral spread
preclinical models of medulloblastoma.90 The feasibility of and resulted in a significant antitumor effect and mice sur-
treating GBM with SCs expressing double prodrug enzymes vival.107 Oncolytic herpes simplex virus (oHSV) is inherently
has been explored. Human NSCs expressing CD and TK were neurotropic and has been modified for tumor-selective repli-
shown to have efficient antitumor efficacy and eradication of cation, making it a promising candidate for brain tumor ther-
the SCs.91 In an effort to develop efficient SC-based therapeutic apy. 108 Previous clinical studies have revealed suboptimal
strategy that simultaneously allows killing of GBM tumor cells response rates in patients after delivery of oHSV in GBM post
and eradication of SCs post tumor treatment, we have engi- tumor resection,109 which could be attributed to “wash out”
neered MSCs to co-express HSV-TK and S-TRAIL and shown of the oHSV in the tumor resection cavity. We have utilized
that they induce caspase-mediated GBM cell death and can MSCs for local release of oHSV and its proapoptotic variant
be selectively eradicated post tumor treatment in mouse GBM oHSV-TRAIL into mouse models of GBM and shown prolonged
models.88 Similar studies utilizing SCs expressing CD/IFNb57,92 median survival in mice as compared with direct injection of
and CE/TRAIL93 have also shown efficacy in malignant GBM purified oHSV.110
mouse models.

Stem Cells Carrying Nanoparticles Stem Cell-released Extracellular Vesicles


Nanoparticles are emerging as novel therapeutic and diagnos- Extracellular vesicles (EVs) (exosomes and shedding micro-
tic tools with great potential in cancer biology. The surface of vesicles) are released by various cell types and contain
nanoparticles can be modified, thus making them ideally suited mRNA, miRNA, proteins, and lipids. 111 Accumulating evi-
to contain high concentrations of often insoluble therapeutic dence supports the notion that MSCs act in a paracrine man-
reagents.94 Although the use of nanoparticles offers consid- ner112 by transferring the biological material to adjacent or
erable delivery benefits, their efficient clearance, inefficient distant cells to facilitate communication between different
dissemination in solid tumors, and inability to target microme- cell types.113 Previous studies have shown that MSCs secrete
tastatic lesions94 have presented a number of challenges. SCs and transfer microvesicles containing specific miRNAs that
have the potential to overcome these barriers and can be load- can serve in intercellular communication. 114 The ability of
ed with nanoparticles and administered in vivo, where they can adult MSCs to transfer miRNA (miR-124 and miR-145) mim-
deposit the loaded nanoparticles in close proximity to the ics to GBM cells, which express very low levels of these miR-
tumor.95 Previous studies have shown the potential for loading NAs, has been explored and shown to have antitumor effects
cell membranes of MSCs with porous silica “nanorattles” con- in a mouse model of GBM.115 Similarly, intratumoral injection
taining doxorubicin (DOX)96 and their efficacy in intracranial tu- of exosomes derived from miR-146-expressing MSCs signifi-
mors.96 Recent studies have shown that NSCs can improve the cantly reduced tumor growth in a rat model of primary
tumor-selective distribution and retention of nanoparticles GBM. 116 Recent studies have reported that MSCs are able
within invasive brain tumors.97 The release of nanoparticles to package and deliver active drugs such as paclitaxel
from SCs is either due to membrane rupture or by facilitating (PTX) through their exosomes and increase survival of mice
death via photoinduction98,99 or inducing hyperthermia.100 bearing GBM. 117,118 Additional, recent studies have shown
The combination of nanoparticle-based therapeutics with SC that exosomes derived from MSCs engineered to express dif-
delivery has exciting prospects for the treatment of tumors in ferent miRs have antitumor effects in mouse tumor
the brain. models.119

Neuro-Oncology 5 of 13
Shah: Stem cell-based therapies for brain tumors

Synergistic Approaches Enhancing Therapeutic Stem Cell model by promoting cell apoptosis, antitumor cytokine produc-
Efficacy in Combination With Other Antitumor Agents tion, and CD4+ and CD8+ T-cell infiltration.122
An emerging branch of cancer therapies is the use of smaller
Brain tumors comprise a heterogeneous population of cells
antibody fragments such as single-chain variable fragments
that are genetically and epigenetically unstable.120 Given the
(scFv) and nanobodies (consisting of just the VHH domain)
GBM heterogeneity, it is unlikely that any one effective strategy
that bind to epitopes overexpressed on tumor cells123 and cre-
will provide a satisfactory treatment regimen for such tumors.
ate bifunctional proteins that are able to perturb multiple sig-
Therefore, using a combination approach would be more effec-
naling pathways specifically in tumor cells (Fig. 2). In one
tive for treating GBM. Because brain tumor cells and the cells in
the tumor microenvironment either specifically express or over- study, we fused S-TRAIL to an EGFR-specific nanobody to
express cell surface receptors, one strategy is to create SCs that make a proapoptotic immunoconjugate (ENb-TRAIL) that con-
simultaneously express/secrete different therapeutic agents currently targets cell proliferation and death pathways.56 SCs
targeting these receptor types. This will allow targeting multiple engineered to express ENb-TRAIL were shown to efficiently in-
pathways in tumor cells and associated cells in the microenvi- hibit EGFR signaling and induce caspase-mediated cell death in

Downloaded from http://neuro-oncology.oxfordjournals.org/ at University of Cambridge on June 11, 2016


ronment and thus increase the efficacy of targeted therapies. a panel of GBM expressing varying levels of EGFR and DR4/5.56
As such, there are a growing number of studies using bimodal In vivo, when mice bearing intracranial GBM were treated with
SCs or SCs that secrete bifunctional molecules. We have shown stem cell-delivered ENb-TRAIL, tumor burden was significantly
that 3TSR of TSP-1 upregulates death receptor (DR) 4/5 expres- decreased and mice survival was prolonged.56 Taken together,
sion in a CD36-dependent manner and primes resistant GBMs bifunctional antibody fragments possess many attributes,
to TRAIL-induced apoptosis. A single administration of MSCs ex- making this a novel and potentially effective means for treating
pressing 3TSR and S-TRAIL targets both tumor cells and vascu- malignancies.
lar component of GBMs and extends survival of mice bearing A number of different studies have combined an additional
intracranial TRAIL-resistant and highly vascularized GBM.121 Re- agent that synergizes with SC therapy or sensitizes an other-
cent studies have shown that SCs coexpressing immunostimu- wise resistant population to SCs delivering a single therapy,
latory cytokines IL-18 and (IFN)-b significantly prolonged the thus increasing the effectiveness of locally delivered biological
survival and inhibited tumor growth in a rat intracranial GBM agent to the tumor site. As such, different drugs that can be

Fig. 2. Enhancing stem-cell (SC) efficacy by engineering bifunctional molecules. The efficacy of SC-delivered therapeutic payload can be enhanced
by engineering bispecific molecules that target multiple cell surface receptor mediated signaling pathways in glioblastoma cells.

6 of 13 Neuro-Oncology
Shah: Stem cell-based therapies for brain tumors

delivered systemically have been shown to synergize with


SC-delivered TRAIL to potentiate its p53-independent proapop-
totic mode of action in brain tumors. These include proteasome
inhibitors such as bortezomib, HDAC inhibitors, genotoxic drugs
such as cisplatin, the PI3-kinase/mTOR inhibitor PI-103, shRNA,
and micro-RNA inhibitors.48,124 We have also designed addi-
tional supplementary treatments such as microRNA-21
(miR-21) inhibitors52 and novel PI3-kinase/mTOR inhibitor
PI-103125 to augment the antitumor effect of different
SC-mediated S-TRAIL therapy in mouse models of GBM in
vivo. The combination of valproic acid (VPA) with MSCs express-
ing HSV-TK was shown to enhance the bystander effect, allow-
ing enhanced cellular transmission of GCV-monophosphate in

Downloaded from http://neuro-oncology.oxfordjournals.org/ at University of Cambridge on June 11, 2016


culture and increased survival in preclinical models of GBM.89
GBM patients are often treated by maximal surgical resec-
tion followed by ionizing radiation and TMZ.6 Therefore, it is im-
perative to initially test SC therapies with preclinical models and
study designs that incorporate current standard of care in an
effort to be translated into the clinic. Previous studies have
shown that tumor irradiation enhances the tumor tropism of
human umbilical cord blood-derived MSCs mediated by in-
creased IL-8 expression on glioma cells.126 Similarly, MSCs ex-
pressing TRAIL127 or interferon-b128 were shown to be more
effective at killing malignant GBM in the presence of TMZ. In an-
other study, NSCs carrying OV used in combination with radia- Fig. 3. Encapsulating engineered therapeutic stem cells in clinically
tion and TMZ129 were shown to prolong the survival of mice relevant mouse tumor models of GBM. Stem cells expressing
bearing patient-derived GBM xenografts. bifunctional molecules or loaded with oncolytic viruses can be
encapsulated in biodegradable synthetic extracellular matrix and
placed in the tumor resection cavity created after GBM tumor
Promising Preclinical Studies and Clinical Trials debulking in mouse tumor models of GBM.
The CNS lesion site(s) (focal vs multifocal) very much define(s)
the route of SC transplantation.14 Direct local transplantation is
suggested as a preferred route of NSC transplantation for focal soluble hyaluronidase (ICOVIR17) compared with direct injec-
CNS disorders, whereas the multifocality of certain CNS dis- tion in a clinically relevant mouse model of GBM resection.107
orders represents a major limitation for intralesional cell- Similarly, we have shown that a low dose of cisplatin in combi-
transplantation approaches. In mouse models of GBM, our nation with sECM-encapsulated SC-TRAIL transplanted in the
earlier studies have shown that the intraventricular route of GBM resection cavity significantly decreases tumor regrowth
delivery is much more efficacious than the intravenous or intra- and increases survival in mice bearing GBM.135 Together,
peritoneal route.33 Recent studies suggest that intranasal deliv- these studies provide a platform for clinical translation of engi-
ery of SCs is an emerging noninvasive means for the treatment neered SCs to target residual brain tumor cells post surgery.
of brain tumors.130,131 Given that about 75% of GBM patients Currently, a few trials are testing if modified SCs have active
undergo tumor debulking,31,132 another innovative strategy is antitumor activity in brain tumor patients (Table 1). Genexine,
the encapsulation and implantation of SCs in the resected Inc is sponsoring NCT02079324, which is intended to establish
GBM tumors in intracranial mouse GBM models. Biodegradable the safety and efficacy of MSCs expressing IL-12 (GX-051) ad-
hydrogels and synthetic extracellular matrix (sECM) materials ministered intratumorally in patients with advanced head and
composed of hyaluronic acid, alginate, agarose, and other poly- neck cancer. In another trial, allogeneic NSCs modified to ex-
mers133 have been utilized to encapsulate SCs in a variety of press CD are being tested in recurrent GBM (NCT01172964).
rodent cancer models133,134 and thus offer an important pros- Two more complementary trials have been approved that
pect for harnessing on-site delivery of tumor-specific agents. combine NSC-CD therapy with either leucovorin calcium
We have investigated a new approach to GBM treatment by en- (NCT02015819) or irinotecan hydrochloride (NCT02055196)
capsulating therapeutic SCs in GBM resection xenograft mouse to further sensitize GBM cells.
models that recapitulate the clinical scenario of surgical
debulking of GBM31 (Fig. 3). Human MSCs and NSCs encapsulat-
ed in sECM were retained in the resection cavity and permitted Prospects and Caveats on the way to the Clinics
tumor-selective migration. sECM-encapsulated SCs engineered The adult SC represents a potentially powerful tool and offers
to express proapoptotic TRAIL protein or loaded with oHSV- great potential for use in the treatment of tumors in the
TRAIL were placed into the mouse GBM tumor resection cavity brain. Transplantation experiments have revealed that both
and shown to increase survival significantly.31,110 Recent stud- NSCs and MSCs migrate extensively towards both primary and
ies have further shown significant therapeutic efficacy of MSCs metastatic tumors in the brain. These intrinsic abilities have
loaded with conditionally replicating adenovirus expressing been harnessed to deliver apoptosis-inducing proteins,

Neuro-Oncology 7 of 13
Shah: Stem cell-based therapies for brain tumors

Table 1. Stem cell therapies for cancer in clinical trials

Stem-cell Type Therapeutics Loaded Phase (study start) National Clinical Trial Number Targeted Cancer Types

NSC Cytosine deaminase I (12/2013) NCT02015819 Recurrent high-grade glioma


NSC Carboxylesterase I (01/2016) NCT02192359 Recurrent high-grade glioma
MSC IL-12 I (03.2014) NCT02079324 Head and neck cancer
MSC MV-NIS I/II (03/2014) NCT02068794 Recurrent ovarian cancer
MSC IFNb I (03/2016) NCT02530047 Ovarian cancer
MSC ICOVIR5 I/II (01/2013) NCT01844661 Metastatic and refractory solid tumor
HSPC rHIV7-shl-TAR-CCR5RZ N/L (08/2015) NCT02337985 AIDS-related non-Hodgkin’s lymphoma
HSPC LVsh5/C46 (CAL-1) I (11/2015) NCT02378922 Relapsed or refractory acute myeloid leukemia
HSPC LV-C46/CCR5/P140K I (03/2016) NCT02343666 Lymphoma with HIV infection

Downloaded from http://neuro-oncology.oxfordjournals.org/ at University of Cambridge on June 11, 2016


Abbreviations: HSPC, hematopoietic stem/progenitor cell; ICOVIR5, oncolytic adenovirus; IFNb, Interferon-b; L-12, interleukin-12; LV-C46/CCR5/
P140K, lentiviral vector encoding multiple inhibitors for HIV replication; LVsh5/C46 (CAL-1), dual anti-HIV lentiviral vector; MSC, mesenchymal
stem cell; MV-NIS, oncolytic measles virus encoding thyroidal sodium iodide symporter; N/L, not listed in clinical trial database; NSC, neuronal
stem cell; rHIV7-shl-TAR-CCR5RZ, lentiviral vector encoding multiple anti-HIV RNAs.

immunostimulatory signals, antiangiogenic factors, cell cycle The use of genetically engineered mouse brain tumor models
modulators, inducers of differentiation, and OVs. However, in which tumors arise in situ or incorporation of surgical resec-
the integration of nascent therapeutic technologies such as ge- tion into GBM tumor studies might hold more validity and need
netically modified SCs would require considerable understand- exploration.
ing of fundamental mechanisms before they can be efficiently In clinical settings, the safety of the grafted SC is a major
translated into clinical settings. The use of a particular SC type concern as there are still scientifically contentious issues re-
in clinical trials will depend on the ability SCs to preferentially garding basic SC biology. Of particular concern is whether SCs
home to tumors in the brain, their easy availability, and their promote the growth of certain tumors141 – 143 or indeed form
relative ease of manipulation in vitro without requiring immor- tumors themselves.144,145 Importantly, non-immortalized
talization. A major advantage of using autologous stem cells is adult stem cells provide fewer safety concerns than immortal-
their immunological compatibility, which has a profound effect ized adult stem cells and may be used without posing a risk to
on cell survival post transplantation. MSCs with multilineage the patient. The incorporation of suicide genes such as HSV-TK
potential show a broad migratory capacity for different brain into therapeutic SCs would enable their controlled eradication
tumor types including glioblastoma, medulloblastoma, epen- post treatment and alleviate this concern. Because patient
dymoma, and astrocytoma.136,137 Hence, they have been stud- safety is vital, these issues should be resolved before starting
ied as a better alternative to NSCs that, despite their strong the next generation of clinical trials.
tumor-tropic properties, have limited availability and ethical is-
sues.138 Due to their abundant availability, easy isolation and
expansions, fewer ethical concerns and, most importantly,
eligibility for autologous transplantation, human adipose Funding
tissue-derived MSCs are one of the most attractive vehicles This work was supported by R01CA138922 and R01CA173077.
for the delivery of therapeutic agents.138 Although iPS-derived
NSCs have been considered a feasible, effective, and autolo-
gous source for clinical applications, their therapeutic ability
Acknowledgments
has not yet been fully addressed.19 The selection of the SC
We thank Dr. Sung Hugh Choi and Dr. Clemens Reinshagen with their
type, its purity, thorough understanding of its biology, and
help with literature search for the manuscript and Anusha
the effect of any modifications on its function should be estab- Venogopalan for their help with the references for this review. We
lished in preclinical studies before its consideration for clinical also thank Dr. Daniel Stuckey for his help with literature search and
translation. Fig. 2 of the review.
In recent years, patient-derived primary GBM lines have
been created from isolated human brain tumor tissue and uti-
lized for preclinical studies. A number of studies have shown
Conflict of interest statement. There is no conflict of interest.
that xenografts of these primary patient-derived cell lines
often recapitulate clinical GBM more faithfully, thus providing
additional insights and more stringent testing of promising
new anti-GBM therapies.139 A thorough biological understand- References
ing of such GBM tumor types will improve the chances of 1. Ohgaki H, Kleihues P. The definition of primary and secondary
therapeutic success by yielding new therapeutic targets glioblastoma. Clin Cancer Res. 2013;19(4):764–772.
and enabling mechanism-based SC therapies to be developed, 2. Alexandru D, Bota DA, Linskey ME. Epidemiology of central
tested, and refined in response to a specific cancer profile.140 nervous system metastases. Prog Neurol Surg. 2012;25:13– 29.

8 of 13 Neuro-Oncology
Shah: Stem cell-based therapies for brain tumors

3. Filippini G. Epidemiology of primary central nervous system through CXCR4 in a viral model of multiple sclerosis. Proc Natl
tumors. Handb Clin Neurol. 2012;104:3 –22. Acad Sci USA. 2011;107(24):11068– 11073.
4. Ostrom QT, Gittleman H, Farah P, et al. CBTRUS statistical report: 23. Pluchino S, Zanotti L, Rossi B, et al. Neurosphere-derived
Primary brain and central nervous system tumors diagnosed in multipotent precursors promote neuroprotection by an
the United States in 2006 – 2010. Neuro Oncol. 2013;15(Suppl immunomodulatory mechanism. Nature. 2005;436(7048):
2):ii1–i56. 266–271.
5. Louis DN. Molecular pathology of malignant gliomas. Annu Rev 24. Sasportas LS, Kasmieh R, Wakimoto H, et al. Assessment of
Pathol. 2006;1:97–117. therapeutic efficacy and fate of engineered human mesenchymal
6. Darefsky AS, King JT Jr., Dubrow R. Adult glioblastoma multiforme stem cells for cancer therapy. Proc Natl Acad Sci USA. 2009;106(12):
survival in the temozolomide era: a population-based analysis of 4822–4827.
Surveillance, Epidemiology, and End Results registries. Cancer. 25. Shah K, Hingtgen S, Kasmieh R, et al. Bimodal viral vectors
2012;118(8):2163– 2172. and in vivo imaging reveal the fate of human neural stem
7. Ostrom QT, Bauchet L, Davis FG, et al. The epidemiology of glioma cells in experimental glioma model. J Neurosci. 2008;28(17):

Downloaded from http://neuro-oncology.oxfordjournals.org/ at University of Cambridge on June 11, 2016


in adults: a “state of the science” review. Neuro Oncol. 2014; 4406– 4413.
16(7):896 – 913. 26. Tran PB, Banisadr G, Ren D, Chenn A, Miller RJ. Chemokine
8. Stupp R, Hegi ME, Mason WP, et al. Effects of radiotherapy with receptor expression by neural progenitor cells in neurogenic
concomitant and adjuvant temozolomide versus radiotherapy regions of mouse brain. J Comp Neurol. 2007;500(6):1007 – 1033.
alone on survival in glioblastoma in a randomised phase III 27. Aboody KS, Brown A, Rainov NG, et al. Neural stem cells display
study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. extensive tropism for pathology in adult brain: evidence from
2009;10(5):459– 466. intracranial gliomas. Proc Natl Acad Sci USA. 2000;97(23):
9. Reubinoff BE, Pera MF, Fong CY, Trounson A, Bongso A. Embryonic 12846– 12851.
stem cell lines from human blastocysts: somatic differentiation in 28. Kauer TM, Figueiredo JL, Hingtgen S, Shah K. Encapsulated
vitro. Nat Biotechnol. 2000;18(4):399 –404. therapeutic stem cells implanted in the tumor resection cavity
10. Stuckey DW, Shah K. Stem cell-based therapies for cancer induce cell death in gliomas. Nat Neurosci. 2011;15(2):197–204.
treatment: separating hope from hype. Nat Rev Cancer. 2014; 29. Shah K, Tung CH, Breakefield XO, Weissleder R. In vivo imaging of
14(10):683 –691. S-TRAIL-mediated tumor regression and apoptosis. Mol Ther.
11. Anderson DJ, Gage FH, Weissman IL. Can stem cells cross lineage 2005;11(6):926– 931.
boundaries? Nat Med. 2001;7(4):393 –395. 30. Tang Y, Shah K, Messerli SM, Snyder E, Breakefield X, Weissleder R.
12. Sun Y, Williams A, Waisbourd M, Iacovitti L, Katz LJ. Stem cell In vivo tracking of neural progenitor cell migration to
therapy for glaucoma: science or snake oil? Surv Ophthalmol. glioblastomas. Hum Gene Ther. 2003;14(13):1247– 1254.
2015;60(2):93– 105. 31. Koizumi S, Gu C, Amano S, et al. Migration of mouse-induced
13. Martino G, Pluchino S. The therapeutic potential of neural stem pluripotent stem cells to glioma-conditioned medium is
cells. Nat Rev Neurosci. 2006;7(5):395– 406. mediated by tumor-associated specific growth factors. Oncol
Lett. 2011;2(2):283 –288.
14. Yip S, Shah K. Stem-cell based therapies for brain tumors. Curr
Opin Mol Ther. 2008;10(4):334 –342. 32. Magge SN, Malik SZ, Royo NC, et al. Role of monocyte
chemoattractant protein-1 (MCP-1/CCL2) in migration of neural
15. Abbott A. Cell rewind wins medicine Nobel. Nature. 2012; progenitor cells toward glial tumors. J Neurosci Res. 2009;87(7):
490(7419):151– 152. 1547– 1555.
16. Zomer HD, Vidane AS, Goncalves NN, Ambrosio CE. Mesenchymal 33. Xu Q, Wang S, Jiang X, et al. Hypoxia-induced astrocytes
and induced pluripotent stem cells: general insights and clinical promote the migration of neural progenitor cells via
perspectives. Stem Cells Cloning Adv Appl. 2015;8:125– 134. vascular endothelial factor, stem cell factor, stromal-derived
17. de Lazaro I, Yilmazer A, Kostarelos K. Induced pluripotent stem factor-1alpha and monocyte chemoattractant protein-1
(iPS) cells: a new source for cell-based therapeutics? J Control upregulation in vitro. Clin Exp Pharmacol Physiol. 2007;34(7):
Release. 2014;185:37 –44. 624 – 631.
18. Hong JY, Lee SH, Lee SC, et al. Therapeutic potential of induced 34. Zhao D, Najbauer J, Garcia E, et al. Neural stem cell tropism to
neural stem cells for spinal cord injury. J Biol Chem. 2014; glioma: critical role of tumor hypoxia. Mol Cancer Res. 2008;
289(47):32512– 32525. 6(12):1819 –1829.
19. Bilousova G, Jun du H, King KB, et al. Osteoblasts derived from 35. Schmidt NO, Koeder D, Messing M, et al. Vascular endothelial
induced pluripotent stem cells form calcified structures in growth factor-stimulated cerebral microvascular endothelial
scaffolds both in vitro and in vivo. Stem Cells. 2011;29(2): cells mediate the recruitment of neural stem cells to the
206–216. neurovascular niche. Brain Res. 2009;1268:24– 37.
20. Villa-Diaz LG, Brown SE, Liu Y, et al. Derivation of mesenchymal 36. Hayakawa K, Pham LD, Som AT, et al. Vascular endothelial growth
stem cells from human induced pluripotent stem cells cultured factor regulates the migration of oligodendrocyte precursor cells.
on synthetic substrates. Stem Cells. 2012;30(6):1174– 1181. J Neurosci. 2011;31(29):10666– 10670.
21. Kang R, Zhou Y, Tan S, et al. Mesenchymal stem cells derived from 37. Esencay M, Newcomb EW, Zagzag D. HGF upregulates CXCR4
human induced pluripotent stem cells retain adequate expression in gliomas via NF-kappaB: implications for glioma
osteogenicity and chondrogenicity but less adipogenicity. Stem cell migration. J Neurooncol. 2010;99(1):33 –40.
Cell Res Ther. 2015;6:144. 38. Vallabhaneni KC, Tkachuk S, Kiyan Y, et al. Urokinase receptor
22. Carbajal KS, Schaumburg C, Strieter R, Kane J, Lane TE. Migration mediates mobilization, migration, and differentiation of
of engrafted neural stem cells is mediated by CXCL12 signaling mesenchymal stem cells. Cardiovasc Res. 2011;90(1):113–121.

Neuro-Oncology 9 of 13
Shah: Stem cell-based therapies for brain tumors

39. Chabot V, Dromard C, Rico A, et al. Urokinase-type plasminogen 56. van de Water JA, Bagci-Onder T, Agarwal AS, et al. Therapeutic
activator receptor interaction with beta1 integrin is required for stem cells expressing variants of EGFR-specific nanobodies
platelet-derived growth factor-AB-induced human mesenchymal have antitumor effects. Proc Natl Acad Sci USA. 2012;109(41):
stem/stromal cell migration. Stem Cell Res Ther. 2015;6:188. 16642–16647.
40. Shinojima N, Hossain A, Takezaki T, et al. TGF-beta mediates 57. Piccirillo SG, Binda E, Fiocco R, Vescovi AL, Shah K. Brain cancer
homing of bone marrow-derived human mesenchymal stem stem cells. J Mol Med. 2009;87(11):1087 – 1095.
cells to glioma stem cells. Cancer Res. 2013;73(7):2333– 2344. 58. Li Q, Wijesekera O, Salas SJ, et al. Mesenchymal stem cells from
41. Lourenco S, Teixeira VH, Kalber T, Jose RJ, Floto RA, Janes SM. human fat engineered to secrete BMP4 are nononcogenic,
Macrophage migration inhibitory factor-CXCR4 is the dominant suppress brain cancer, and prolong survival. Clin Cancer Res.
chemotactic axis in human mesenchymal stem cell recruitment 2014;20(9):2375–2387.
to tumors. J Immunol. 2015;194(7):3463–3474. 59. Lopez-Ornelas A, Vergara P, Segovia J. Neural stem cells
42. Yamazoe T, Koizumi S, Yamasaki T, Amano S, Tokuyama T, producing an inducible and soluble form of Gas1 target and
Namba H. Potent tumor tropism of induced pluripotent stem inhibit intracranial glioma growth. Cytotherapy. 2014;16(7):

Downloaded from http://neuro-oncology.oxfordjournals.org/ at University of Cambridge on June 11, 2016


cells and induced pluripotent stem cell-derived neural stem 1011– 1023.
cells in the mouse intracerebral glioma model. Int J Oncol.
60. van Eekelen M, Sasportas LS, Kasmieh R, et al. Human stem cells
2015;46(1):147–152.
expressing novel TSP-1 variant have anti-angiogenic effect on
43. Schichor C, Albrecht V, Korte B, et al. Mesenchymal stem cells and brain tumors. Oncogene. 2010;29(22):3185 –3195.
glioma cells form a structural as well as a functional syncytium in
61. Kim SK, Cargioli TG, Machluf M, et al. PEX-producing human
vitro. Exp Neurol. 2012;234(1):208– 219.
neural stem cells inhibit tumor growth in a mouse glioma
44. Jo J, Hong S, Choi WY, Lee DR. Cell-penetrating peptide model. Clin Cancer Res. 2005;11(16):5965– 5970.
(CPP)-conjugated proteins is an efficient tool for manipulation
62. Jain RK, di Tomaso E, Duda DG, Loeffler JS, Sorensen AG,
of human mesenchymal stromal cells. Sci Rep. 2014;4:4378.
Batchelor TT. Angiogenesis in brain tumours. Nat Rev Neurosci.
45. Stuckey DW, Shah K. TRAIL on trial: preclinical advances in cancer 2007;8(8):610– 622.
therapy. Trends Mol Med. 2013;19(11):685 –694.
63. Batchelor TT, Sorensen AG, di Tomaso E, et al. AZD2171, a
46. Ehtesham M, Kabos P, Gutierrez MA, et al. Induction of pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor
glioblastoma apoptosis using neural stem cell-mediated vasculature and alleviates edema in glioblastoma patients.
delivery of tumor necrosis factor-related apoptosis-inducing Cancer Cell. 2007;11(1):83 –95.
ligand. Cancer Res. 2002;62(24):7170–7174.
64. Tong RT, Boucher Y, Kozin SV, Winkler F, Hicklin DJ, Jain RK.
47. Shah K, Hsich G, Breakefield XO. Neural precursor cells and their Vascular normalization by vascular endothelial growth factor
role in neuro-oncology. Dev Neurosci. 2004;26(2 –4):118–130. receptor 2 blockade induces a pressure gradient across the
48. Shah K, Bureau E, Kim DE, et al. Glioma therapy and real-time vasculature and improves drug penetration in tumors. Cancer
imaging of neural precursor cell migration and tumor Res. 2004;64(11):3731– 3736.
regression. Ann Neurol. 2005;57(1):34 –41. 65. Hormigo A, Gutin PH, Rafii S. Tracking normalization of brain
49. Tang XJ, Lu JT, Tu HJ, et al. TRAIL-engineered bone tumor vasculature by magnetic imaging and proangiogenic
marrow-derived mesenchymal stem cells: TRAIL expression biomarkers. Cancer Cell. 2007;11(1):6– 8.
and cytotoxic effects on C6 glioma cells. Anticancer Res. 2014; 66. Bexell D, Gunnarsson S, Tormin A, et al. Bone marrow multipotent
34(2):729 –734.
mesenchymal stroma cells act as pericyte-like migratory vehicles
50. Bagci-Onder T, Du W, Figueiredo JL, et al. Targeting breast to in experimental gliomas. Mol Ther. 2009;17(1):183–190.
brain metastatic tumours with death receptor ligand
67. Bexell D, Scheding S, Bengzon J. Toward brain tumor gene
expressing therapeutic stem cells. Brain. 2015;138(Pt 6):
therapy using multipotent mesenchymal stromal cell vectors.
1710– 1721.
Mol Ther. 2010;18(6):1067–1075.
51. Nakamizo A, Marini F, Amano T, et al. Human bone
68. Gajewski TF, Woo SR, Zha Y, et al. Cancer immunotherapy
marrow-derived mesenchymal stem cells in the treatment of
strategies based on overcoming barriers within the tumor
gliomas. Cancer Res. 2005;65(8):3307–3318.
microenvironment. Curr Opin Immunol. 2013;25(2):268– 276.
52. Dembinski JL, Wilson SM, Spaeth EL, et al. Tumor stroma
69. Okada H, Pollack IF. Cytokine gene therapy for malignant glioma.
engraftment of gene-modified mesenchymal stem cells as
Expert Opin Biol Ther. 2004;4(10):1609– 1620.
anti-tumor therapy against ovarian cancer. Cytotherapy. 2013;
15(1):20– 32. 70. Ehtesham M, Kabos P, Kabosova A, Neuman T, Black KL, Yu JS.
The use of interleukin 12-secreting neural stem cells for the
53. Ito S, Natsume A, Shimato S, et al. Human neural stem cells
treatment of intracranial glioma. Cancer Res. 2002;62(20):
transduced with IFN-beta and cytosine deaminase genes
5657– 5663.
intensify bystander effect in experimental glioma. Cancer Gene
Ther. 2010;17(5):299 –306. 71. Yang SY, Liu H, Zhang JN. Gene therapy of rat malignant gliomas
using neural stem cells expressing IL-12. DNA Cell Biol. 2004;
54. Ren C, Kumar S, Chanda D, Chen J, Mountz JD, Ponnazhagan S.
23(6):381– 389.
Therapeutic potential of mesenchymal stem cells producing
interferon-alpha in a mouse melanoma lung metastasis 72. Hong X, Miller C, Savant-Bhonsale S, Kalkanis SN. Antitumor
model. Stem Cells. 2008;26(9):2332 –2338. treatment using interleukin- 12-secreting marrow stromal cells
55. Balyasnikova IV, Ferguson SD, Sengupta S, Han Y, Lesniak MS. in an invasive glioma model. Neurosurgery. 2009;64(6):
Mesenchymal stem cells modified with a single-chain antibody 1139– 1146. discussion 1146 –1137.
against EGFRvIII successfully inhibit the growth of human 73. Ryu CH, Park SH, Park SA, et al. Gene therapy of intracranial
xenograft malignant glioma. PloS One. 2010;5(3):e9750. glioma using interleukin 12-secreting human umbilical cord

10 of 13 Neuro-Oncology
Shah: Stem cell-based therapies for brain tumors

blood-derived mesenchymal stem cells. Hum Gene Ther. 2011; 89. Ryu CH, Park KY, Kim SM, et al. Valproic acid enhances anti-tumor
22(6):733 –743. effect of mesenchymal stem cell mediated HSV-TK gene therapy
74. Xu G, Jiang XD, Xu Y, et al. Adenoviral-mediated interleukin-18 in intracranial glioma. Biochem Biophys Res Commun. 2012;
expression in mesenchymal stem cells effectively suppresses 421(3):585 –590.
the growth of glioma in rats. Cell Biol Int. 2009;33(4):466 –474. 90. Hong SH, Lee HJ, An J, et al. Human neural stem cells expressing
75. Agarwalla P, Barnard Z, Fecci P, Dranoff G, Curry WT Jr. Sequential carboxyl esterase target and inhibit tumor growth of lung cancer
immunotherapy by vaccination with GM-CSF-expressing glioma brain metastases. Cancer Gene Ther. 2013;20(12):678–682.
cells and CTLA-4 blockade effectively treats established murine 91. Lee JY, Lee DH, Kim HA, et al. Double suicide gene therapy using
intracranial tumors. J Immunother. 2012;35(5):385– 389. human neural stem cells against glioblastoma: double safety
76. Fritzell S, Eberstal S, Sanden E, Visse E, Darabi A, Siesjo P. measures. J Neurooncol. 2014;116(1):49–57.
IFNgamma in combination with IL-7 enhances immunotherapy 92. Kim KY, Yi BR, Lee HR, et al. Stem cells with fused gene expression
in two rat glioma models. J Neuroimmunol. 2013;258(1 – 2): of cytosine deaminase and interferon-beta migrate to human
91–95. gastric cancer cells and result in synergistic growth inhibition

Downloaded from http://neuro-oncology.oxfordjournals.org/ at University of Cambridge on June 11, 2016


77. Stagg J, Galipeau J. Mechanisms of immune modulation by for potential therapeutic use. Int J Oncol. 2012;40(4):
mesenchymal stromal cells and clinical translation. Curr Mol 1097 –1104.
Med. 2013;13(5):856– 867. 93. Choi SA, Lee YE, Kwak PA, et al. Clinically applicable human
78. Waterman RS, Morgenweck J, Nossaman BD, Scandurro AE, adipose tissue-derived mesenchymal stem cells delivering
Scandurro SA, Betancourt AM. Anti-inflammatory mesenchymal therapeutic genes to brainstem gliomas. Cancer Gene Ther.
stem cells (MSC2) attenuate symptoms of painful diabetic 2015;22(6):302–311.
peripheral neuropathy. Stem Cells Transl Med. 2012;1(7):557–565. 94. Bertrand N, Wu J, Xu X, Kamaly N, Farokhzad OC. Cancer
79. Visse E, Siesjo P, Widegren B, Sjogren HO. Regression of nanotechnology: the impact of passive and active targeting in
intracerebral rat glioma isografts by therapeutic subcutaneous the era of modern cancer biology. Adv Drug Deliv Rev. 2014;66:
immunization with interferon-gamma, interleukin-7, or 2– 25.
B7-1-transfected tumor cells. Cancer Gene Ther. 1999;6(1): 95. Auffinger B, Morshed R, Tobias A, Cheng Y, Ahmed AU, Lesniak MS.
37–44. Drug-loaded nanoparticle systems and adult stem cells: a
80. Strojby S, Eberstal S, Svensson A, et al. Intratumorally implanted potential marriage for the treatment of malignant glioma?
mesenchymal stromal cells potentiate peripheral immunotherapy Oncotarget. 2013;4(3):378– 396.
against malignant rat gliomas. J Neuroimmunol. 2014;274(1–2): 96. Li L, Guan Y, Liu H, et al. Silica nanorattle-doxorubicin-anchored
240–243. mesenchymal stem cells for tumor-tropic therapy. ACS Nano.
81. Wang Y, Jiang T. Understanding high grade glioma: molecular 2011;5(9):7462– 7470.
mechanism, therapy and comprehensive management. Cancer 97. Mooney R, Weng Y, Tirughana-Sambandan R, et al. Neural stem
Lett. 2013;331(2):139–146. cells improve intracranial nanoparticle retention and
tumor-selective distribution. Future Oncol. 2014;10(3):401–415.
82. Stuckey DW, Hingtgen SD, Karakas N, Rich BE, Shah K. Engineering
toxin-resistant therapeutic stem cells to treat brain tumors. Stem 98. Duchi S, Sotgiu G, Lucarelli E, et al. Mesenchymal stem cells as
Cells. 2015;33(2):589– 600. delivery vehicle of porphyrin loaded nanoparticles: effective
photoinduced in vitro killing of osteosarcoma. J Control Release.
83. Zhang Y, Sun X, Huang M, Ke Y, Wang J, Liu X. A novel bispecific
2013;168(2):225–237.
immunotoxin delivered by human bone marrow-derived
mesenchymal stem cells to target blood vessels and 99. Schnarr K, Mooney R, Weng Y, et al. Gold nanoparticle-loaded
vasculogenic mimicry of malignant gliomas. Drug Des Devel neural stem cells for photothermal ablation of cancer. Adv
Ther. 2015;9:2947 –2959. Healthc Mater. 2013;2(7):976 –982.
84. Altaner C, Altanerova V, Cihova M, et al. Complete regression of 100. Rachakatla RS, Balivada S, Seo GM, et al. Attenuation of mouse
glioblastoma by mesenchymal stem cells mediated prodrug melanoma by A/C magnetic field after delivery of bi-magnetic
gene therapy simulating clinical therapeutic scenario. Int J nanoparticles by neural progenitor cells. ACS Nano. 2010;4(12):
Cancer. 2014;134(6):1458– 1465. 7093 –7104.
85. Zhao Y, Lam DH, Yang J, et al. Targeted suicide gene therapy for 101. Chiocca EA, Aghi M, Fulci G. Viral therapy for glioblastoma. Cancer
glioma using human embryonic stem cell-derived neural stem J. 2003;9(3):167 – 179.
cells genetically modified by baculoviral vectors. Gene Ther. 102. Kim J, Hall RR, Lesniak MS, Ahmed AU. Stem cell-Based
2012;19(2):189– 200. cell Carrier for Targeted Oncolytic Virotherapy: Translational
86. Kim SK, Kim SU, Park IH, et al. Human neural stem cells target Opportunity and Open Questions. Viruses. 2015;7(12):
experimental intracranial medulloblastoma and deliver a 6200 – 6217.
therapeutic gene leading to tumor regression. Clin Cancer Res. 103. Ahmed AU, Tyler MA, Thaci B, et al. A comparative study of neural
2006;12(18):5550 – 5556. and mesenchymal stem cell-based carriers for oncolytic
87. de Melo SM, Bittencourt S, Ferrazoli EG, et al. The Anti-Tumor adenovirus in a model of malignant glioma. Mol Pharm. 2011;
Effects of Adipose Tissue Mesenchymal Stem Cell Transduced 8(5):1559–1572.
With HSV-Tk Gene on U-87-Driven Brain Tumor. PLoS One. 104. Sonabend AM, Ulasov IV, Tyler MA, Rivera AA, Mathis JM, Lesniak
2015;10(6):e0128922. MS. Mesenchymal stem cells effectively deliver an oncolytic
88. Martinez-Quintanilla J, Bhere D, Heidari P, He D, Mahmood U, adenovirus to intracranial glioma. Stem Cells. 2008;26(3):
Shah K. Therapeutic efficacy and fate of bimodal engineered 831– 841.
stem cells in malignant brain tumors. Stem Cells. 2013;31(8): 105. Yong RL, Shinojima N, Fueyo J, et al. Human bone
1706– 1714. marrow-derived mesenchymal stem cells for intravascular

Neuro-Oncology 11 of 13
Shah: Stem cell-based therapies for brain tumors

delivery of oncolytic adenovirus Delta24-RGD to human gliomas. 122. Xu G, Guo Y, Seng Z, Cui G, Qu J. Bone marrow-derived
Cancer Res. 2009;69(23):8932 – 8940. mesenchymal stem cells co-expressing interleukin-18 and
106. Ahmed AU, Thaci B, Tobias AL, et al. A preclinical evaluation of interferon-beta exhibit potent antitumor effect against
neural stem cell-based cell carrier for targeted antiglioma intracranial glioma in rats. Oncol Rep. 2015;34(4):1915–1922.
oncolytic virotherapy. J Natl Cancer Inst. 2013;105(13): 123. Muyldermans S. Single domain camel antibodies: current status.
968 – 977. J Biotechnol. 2001;74(4):277–302.
107. Martinez-Quintanilla J, He D, Wakimoto H, Alemany R, Shah K. 124. Du W, Uslar L, Sevala S, Shah K. Targeting c-Met receptor
Encapsulated stem cells loaded with hyaluronidase-expressing overcomes TRAIL-resistance in brain tumors. PLoS One. 2014;
oncolytic virus for brain tumor therapy. Mol Ther. 2015;23(1): 9(4):e95490.
108–118.
125. Bagci-Onder T, Wakimoto H, Anderegg M, Cameron C, Shah K. A
108. Hoffmann D, Wildner O. Comparison of herpes simplex virus- and dual PI3K/mTOR inhibitor, PI-103, cooperates with stem
conditionally replicative adenovirus-based vectors for cell-delivered TRAIL in experimental glioma models. Cancer
glioblastoma treatment. Cancer Gene Ther. 2007;14(7): Res. 2011;71(1):154 –163.

Downloaded from http://neuro-oncology.oxfordjournals.org/ at University of Cambridge on June 11, 2016


627– 639.
126. Kim SM, Oh JH, Park SA, et al. Irradiation enhances the
109. Markert JM, Liechty PG, Wang W, et al. Phase Ib trial of tumor tropism and therapeutic potential of tumor
mutant herpes simplex virus G207 inoculated pre-and necrosis factor-related apoptosis-inducing ligand-secreting
post-tumor resection for recurrent GBM. Mol Ther. 2009;17(1): human umbilical cord blood-derived mesenchymal stem
199– 207. cells in glioma therapy. Stem Cells. 2010;28(12):2217 –
110. Duebgen M, Martinez-Quintanilla J, Tamura K, et al. Stem cells 2228.
loaded with multimechanistic oncolytic herpes simplex virus
127. Kim SM, Woo JS, Jeong CH, Ryu CH, Jang JD, Jeun SS. Potential
variants for brain tumor therapy. J Natl Cancer Inst. 2014;
application of temozolomide in mesenchymal stem cell-based
106(6):dju090.
TRAIL gene therapy against malignant glioma. Stem Cells
111. Nawaz M, Fatima F, Vallabhaneni KC, et al. Extracellular vesicles: Transl Med. 2014;3(2):172 –182.
evolving factors in stem cell biology. Stem Cells Int. 2016;2016:
128. Park JH, Ryu CH, Kim MJ, Jeun SS. Combination therapy for
1073140.
gliomas using temozolomide and interferon-Beta secreting
112. Bruno S, Collino F, Iavello A, Camussi G. Effects of mesenchymal human bone marrow derived mesenchymal stem cells. J
stromal cell-derived extracellular vesicles on tumor growth. Front Korean Neurosurg Soc. 2015;57(5):323–328.
Immunol. 2014;5:382.
129. Tobias AL, Thaci B, Auffinger B, et al. The timing of neural stem
113. Favaro E, Carpanetto A, Lamorte S, et al. Human mesenchymal cell-based virotherapy is critical for optimal therapeutic efficacy
stem cell-derived microvesicles modulate T cell response to islet when applied with radiation and chemotherapy for the
antigen glutamic acid decarboxylase in patients with type 1 treatment of glioblastoma. Stem Cells Transl Med. 2013;2(9):
diabetes. Diabetologia. 2014;57(8):1664 –1673.
655– 666.
114. Chen TS, Lai RC, Lee MM, Choo AB, Lee CN, Lim SK. Mesenchymal
130. Balyasnikova IV, Prasol MS, Ferguson SD, et al. Intranasal delivery
stem cell secretes microparticles enriched in pre-microRNAs.
of mesenchymal stem cells significantly extends survival of
Nucleic Acids Res. 2010;38(1):215–224.
irradiated mice with experimental brain tumors. Mol Ther.
115. Lee HK, Finniss S, Cazacu S, et al. Mesenchymal stem cells deliver 2014;22(1):140 –148.
synthetic microRNA mimics to glioma cells and glioma stem cells
131. Reitz M, Demestre M, Sedlacik J, et al. Intranasal delivery of
and inhibit their cell migration and self-renewal. Oncotarget.
neural stem/progenitor cells: a noninvasive passage to target
2013;4(2):346 – 361.
intracerebral glioma. Stem Cells Transl Med. 2012;1(12):
116. Katakowski M, Buller B, Zheng X, et al. Exosomes from marrow 866– 873.
stromal cells expressing miR-146b inhibit glioma growth.
Cancer Lett. 2013;335(1):201–204. 132. Chang SM, Parney IF, Huang W, et al. Patterns of care for adults
with newly diagnosed malignant glioma. JAMA. 2005;293(5):
117. Pacioni S, D’Alessandris QG, Giannetti S, et al. Mesenchymal 557– 564.
stromal cells loaded with paclitaxel induce cytotoxic
damage in glioblastoma brain xenografts. Stem Cell Res Ther. 133. Shah K. Encapsulated stem cells for cancer therapy. Biomatter.
2015;6:194. 2013;3(1):1– 7.
118. Pascucci L, Cocce V, Bonomi A, et al. Paclitaxel is incorporated by 134. Goren A, Dahan N, Goren E, Baruch L, Machluf M. Encapsulated
mesenchymal stromal cells and released in exosomes that human mesenchymal stem cells: a unique hypoimmunogenic
inhibit in vitro tumor growth: a new approach for drug delivery. platform for long-term cellular therapy. FASEB J. 2010;24(1):
J Control Release. 2014;192:262 –270. 22– 31.
119. Lou G, Song X, Yang F, et al. Exosomes derived from 135. Redjal N, Zhu Y, Shah K. Combination of systemic chemotherapy
miR-122-modified adipose tissue-derived MSCs increase with local stem cell delivered S-TRAIL in resected brain tumors.
chemosensitivity of hepatocellular carcinoma. J Hematol Stem Cells. 2015;33(1):101– 110.
Oncol. 2015;8(1):122. 136. Choi SA, Lee JY, Wang KC, et al. Human adipose tissue-derived
120. Yong RL, Tsankova NM. Emerging interplay of genetics and mesenchymal stem cells: characteristics and therapeutic
epigenetics in gliomas: a new hope for targeted therapy. potential as cellular vehicles for prodrug gene therapy against
Semin Pediatr Neurol. 2015;22(1):14– 22. brainstem gliomas. Eur J Cancer. 2012;48(1):129– 137.
121. Choi SH, Tamura K, Khajuria RK, et al. Anti-angiogenic variant of 137. Kan I, Melamed E, Offen D. Integral therapeutic potential of bone
TSP-1 targets TRAIL-resistant brain tumor cells. Mol Ther. 2015; marrow mesenchymal stem cells. Curr Drug Targets. 2005;6(1):
23(2):235–243. 31 –41.

12 of 13 Neuro-Oncology
Shah: Stem cell-based therapies for brain tumors

138. Choi SA, Lee JY, Kwon SE, et al. Human Adipose Tissue-Derived Metastasis of Head and Neck Cancer Xenografts. Aesthet Surg J.
Mesenchymal Stem Cells Target Brain Tumor-Initiating Cells. 2016;36(1):93 – 104.
PLoS One. 2015;10(6):e0129292. 143. Yang T, Zhang X, Wang M, et al. Activation of mesenchymal
139. Wakimoto H, Kesari S, Farrell CJ, et al. Human glioblastoma- stem cells by macrophages prompts human gastric
derived cancer stem cells: establishment of invasive glioma cancer growth through NF-kB pathway. PLoS One. 2014;9(5):
models and treatment with oncolytic herpes simplex virus e97569.
vectors. Cancer Res. 2009;69(8):3472– 3481. 144. Amariglio N, Hirshberg A, Scheithauer BW, et al. Donor-
140. Garraway LA, Lander ES. Lessons from the cancer genome. Cell. derived brain tumor following neural stem cell transplantation
2013;153(1):17 –37. in an ataxia telangiectasia patient. PLoS Med. 2009;6(2):
141. Karnoub AE, Dash AB, Vo AP, et al. Mesenchymal stem cells e1000029.
within tumour stroma promote breast cancer metastasis. 145. Rosland GV, Svendsen A, Torsvik A, et al. Long-term cultures of
Nature. 2007;449(7162):557– 563. bone marrow-derived human mesenchymal stem cells frequently
142. Rowan BG, Lacayo EA, Sheng M, et al. Human Adipose undergo spontaneous malignant transformation. Cancer Res.

Downloaded from http://neuro-oncology.oxfordjournals.org/ at University of Cambridge on June 11, 2016


Tissue-Derived Stromal/Stem Cells Promote Migration and Early 2009;69(13):5331 –5339.

Neuro-Oncology 13 of 13

Você também pode gostar