Glioblastoma multiforme targeted therapy: The Chlorotoxin story

Glioblastoma multiforme targeted therapy: The Chlorotoxin story

Journal of Clinical Neuroscience xxx (2016) xxx–xxx Contents lists available at ScienceDirect Journal of Clinical Neuroscience journal homepage: www...

1MB Sizes 23 Downloads 150 Views

Journal of Clinical Neuroscience xxx (2016) xxx–xxx

Contents lists available at ScienceDirect

Journal of Clinical Neuroscience journal homepage: www.elsevier.com/locate/jocn

Review

Glioblastoma multiforme targeted therapy: The Chlorotoxin story Or Cohen-Inbar a,b,c,d,⇑, Menashe Zaaroor a,b,c a

Department of Neurological Surgery, Rambam Health Care Center, Haifa, Israel Molecular Immunology Laboratory, Technion Israel Institute of Technology, Haifa, Israel c Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel d Department of Neurological Surgery and Gamma-Knife Radiosurgical Center, University of Virginia Health Care Centre, 1215 Lee St, Charlottesville, VA 22908, USA b

a r t i c l e

i n f o

Article history: Received 3 March 2016 Accepted 2 April 2016 Available online xxxx Keywords: Annexin-2 Chlorotoxin Cl-Channel Glioblastoma Immunotherapy MMP-2

a b s t r a c t Glioblastoma multiforme (GBM) is the most common malignant primary brain neoplasm having a mean survival of <24 months. Scorpion toxins are considered promising cancer drug candidates, primarily due to the discovery of hlorotoxin, derived from the venom of the Israeli yellow scorpion. This intriguing short peptide of only 36 amino-acids length and tight configuration, possess the ability to bind to GBM cells in a grade-related manner with 100% of GBM cells staining positive and no cross reactivity to normal brain. Chlorotoxin has an anti-angiogenic effect as well. Molecular targets for Chlorotoxin include voltage gated chloride channels (GCC), calcium-dependent phospholipid-binding protein Annexin-2, and the inducible extracellular enzyme Matrix Metalloproteinase-2 (MMP-2). Of all its targets, MMP-2 seems to bear the most anti-neoplastic potential. Chlorotoxin is a promising tumortargeting peptide. Its small size and compact shape are convenient for intracranial delivery. We present a short discussion on Chlorotoxin. The structure, biological activity, molecular targets and possible clinical role of Chlorotoxin are discussed. Chlorotoxin can be utilized as a targeting domain as well, attaching different effector functions to it. Clinical applications in GBM therapy, intraoperative imaging, nano-probes and nano-vectors based technology; targeted chemotherapy and immunotherapy are discussed as well. Chlorotoxin is likely to play a significant role in effective GBM immunotherapy in the future. Ó 2016 Elsevier Ltd. All rights reserved.

1. Introduction Grade-IV astrocytoma, better known as glioblastoma multiforme (GBM) is unfortunately ranked as both the most common and the most malignant glioma in adults. GBM is associated with a median overall survival of 1–2 years and a 5-year survival rate of less than 10% [1–3]. The unique nature of Glioblastoma multiforme (GBM), featuring both local and distant aggressive recurrence, and its inherent challenging features was evident as early as 80 years ago. Patients with GBM exhibit altered and suppressed function spanning different elements of the immune system [4–21]. These reports set forth the basic premise that GBM cells have cloaking abilities in addition to suppressive mechanism which allow them to persevere. Another premise laid by these reports, is that homing of immune elements to the tumor site may prove sufficient for an effective anti-tumor response. Targeted immunotherapy is rapidly becoming one of the pillars of anti-cancer therapy. Its targeted nature and reduced treatment ⇑ Corresponding author at: 137 Yellowstone Drive, Charlottesville, VA 22903, USA. Tel.: +972 54 6660565. E-mail addresses: [email protected], [email protected], or_coheni@ rambam.health.gov.il (O. Cohen-Inbar).

related toxicity, stemming from recruiting and activating own cells, selective cytotoxic mechanisms, and effector responses, makes it intuitively an attractive option [22,23]. Metastatic melanoma treatment is one such example. FDA approval of cytokine based therapy with interleukin-2 in 1998 and checkpoint blockade with Ipilimumab in 2011 were major developments in the treatment of melanoma [24,25]. Ipilimumab was shown to increase survival in patients with unresectable stage III/IV melanoma [25–27]. GBM has not received similar clinical successes as of yet [4–8]. This may be attributed to its relative inaccessibility (its location beyond the confines of the blood–brain barrier), any of the multiple mechanisms of active or passive immunosuppression reported [4,20,21] which result in overall poor immunogenicity, or the lack of characterized cancer antigens. One should note that the immune response against tumors is limited by the fact that these arise from the organism’s own tissue, and, therefore, mainly express selfantigens. The patient’s T cells are rendered tolerant to these selfantigens via either central or peripheral tolerance [4–8,28,29], and thus do not act to eradicate these cells. Preclinical studies suggest that targeted therapies can elicit significant antitumor responses in GBM, overcoming some of the barriers and tumor-related escape mechanisms, while others fail at specific points [4]. We will next briefly describe the structure,

http://dx.doi.org/10.1016/j.jocn.2016.04.012 0967-5868/Ó 2016 Elsevier Ltd. All rights reserved.

Please cite this article in press as: Cohen-Inbar O, Zaaroor M. Glioblastoma multiforme targeted therapy: The Chlorotoxin story. J Clin Neurosci (2016), http://dx.doi.org/10.1016/j.jocn.2016.04.012

2

O. Cohen-Inbar, M. Zaaroor / Journal of Clinical Neuroscience xxx (2016) xxx–xxx

biological activity. Molecular target and possible clinical role of Chlorotoxin (ClTx), an intriguing and short peptide derived from the venom of the Israeli Yellow Scorpion. We will present both pre-clinical laboratory data as well as clinical data from our own laboratory as well as from other colleagues, supporting the premise that ClTx will play a significant role in effective GBM targeted therapy in the future. ClTx is a promising tumortargeting peptide. Its small size and compact shape are convenient for intracranial delivery. 2. Classification, structure and synthesis of Chlorotoxin Scorpion toxins are considered promising cancer drug candidates [30,31], primarily due to the discovery of ClTx, a peptide from the venom of the giant Israeli yellow scorpion Leiurus quinquestriatus [32], which can preferentially bind to cancer cells [30]. Its venom is known to inhibit reconstituted smallconductance chloride channels, when applied to the cytoplasmic surface [33] (discussed later). Of note, this scorpion’s venom is rich in many potent physiologically active substances in addition to ClTx. An array of bio-polysaccharides, hyaluronic acid derivate, serotonin, histamine, histamine-releasing factors and protease inhibitors are known to comprise the crude venom [34,35]. These bioactive polypeptides selectively bind to and modulate specific ion channels of excitable cell membranes. Scorpion-derived peptide toxins can be classified according to their specificity in inhibiting various ion channel receptors. So far scorpion-derived toxins identified specifically target either Na+ [36,37], K+ [38], Cl [39] and Ca2+ channels [40]. Since its original description in 1992, a number of Chlorotoxin related peptides have been isolated and identified [32]. ClTx is 4070 Da and 36 amino acids basic peptide with considerable sequence homology to the class of small insectotoxins [30,32,41,42]. Its amino acids sequence includes eight cysteines and four disulfide bonds, and it is therefore classified as a shortchain, disulfide-containing peptide. Other scorpion peptides with a similar primary structure to Chlorotoxin have been collectively referred to as Chlorotoxin-like peptides (i.e. ClTx-a, -b, -c, -d, BmKCL1, Lqh-8:6, Be I5A, BeI1, AmmP2 and GaTx1) [43–45]. The secondary structure of ClTx was first determined using nuclear magnetic resonance (NMR) spectroscopy in 1995 [46]. The NMR data showed that ClTx comprises an a-helix, formed by amino acids 11–21, and three b-sheets, formed by amino acids 1–4, 26–29, and 32 –36 (Fig. 1) [46,47]. The amount of ClTx present in venom is limited. Thus, an alternative or synthetic source of the peptide is necessary for

biochemical and biophysical studies, as well as for potential therapeutic applications. Chlorotoxin has been successfully synthesized and folded in vitro, using classical molecular biology laboratory techniques. Ojeda et al. [30] described synthesis using a solid-phase peptide synthesis (SPPS). An alternative and simple method utilized in our laboratory entailed cloning the ClTx gene [7]. The widely published and available amino-acid sequence of the 36-amino-acids ClTx, was converted into a nucleic acids sequence empirically. Next, this sequence was commercially optimized for expression in Escherichia coli (GENEART Inc.). In the optimization process, the codon usage is adapted to the codon bias of E. coli genes. In addition, regions of very high (>80%) or very low (<30%) GC (Guanine-thymine) has been avoided when possible. The expressed protein can then be purified and refolded in vitro. This allows for a cheap, easy and highly reliable method of ClTx production. It also allows for unique chimeric molecular designs to be planned and produced from the DNA level and then expressed in E. coli (discussed later) [7]. 3. Chlorotoxin activity ClTx is assigned two clinical effects; a tumor binding activity and an antiangiogenic activity. Soroceanu et al. [48] first described in 1998 the tumor binding activity of ClTx using a 125I-labeled peptide (125I-ClTx) [48]. The authors showed that upon 125I-Cltx administration to GBM tumor-bearing mice, the peptides accumulated within the tumor. Soroceanu et al. [48] also showed the specific binding of 125I-Cltx to glioma cells, sparing normal rat astrocytes or neurons [48]. ClTx was initially thought to bind only to the so-called glioma-specific chloride ion channel (GCC) [49–53] which was found to be abundantly expressed in glioma cells but absent in normal brain tissue. Ullrich et al. [49] reported a ClTx-sensitive Cl current, present in human astrocytoma (glioma) cell lines [49]. From then, ClTx has been widely used as a Cl -channel blocker to analyze Cl channels [50–52] and has been proposed as a glioma-specific marker with diagnostic and therapeutic potential [53,54]. GCC expression in situ using labeled ClTx was found to correlate with the tumor’s grade, with only 40–45% of low-grade astrocytoma’s (World Health Organization [WHO] grade I–II) binding it, as opposed to over 90% of high-grade tumors (WHO grade III) and essentially all of the GBM samples, binding labeled ClTx. Comparison tissues including normal human brain, kidney, and colon were consistently negative for ClTx immunostaining [51,53,54]. Sontheimer et al. [55] also showed ClTx binding to tumors of neuro-ectodermal descent (sharing a common embryonic origin

Fig. 1. Chlorotoxin (Cltx). (A) The three-dimensional structure of Cltx. a-Helix is in red, b-sheets are in yellow arrows. The disulfide bonds are not shown. Toxicon 38 (2000). (B) Amino-acids sequence of Cltx. The Disulfide bones are shown as black lines, the cysteines are labeled with roman letters. The cyclic configuration is allowed using an extra amino-acids linker sequence, marked in purple. Adapted from Ojeda et al. [14].

Please cite this article in press as: Cohen-Inbar O, Zaaroor M. Glioblastoma multiforme targeted therapy: The Chlorotoxin story. J Clin Neurosci (2016), http://dx.doi.org/10.1016/j.jocn.2016.04.012

O. Cohen-Inbar, M. Zaaroor / Journal of Clinical Neuroscience xxx (2016) xxx–xxx

with glial cells), in addition to GBM’s, such as melanoma, neuroblastomas, meduloblastomas, and small cell lung carcinomas in over 200 human surgical biopsy samples [55]. Biotinylated ClTx did not bind to normal tissues from brain, skin, kidney and lung, showing impressive selectivity. In vivo studies, using ClTx conjugated to a fluorescent dye (Cy5.5, BLZ-100, 800CW), have further confirmed the specific binding of ClTx to GBM’s [56–58]. Xu et al. [59] reported on two new ClTx derivatives (termed CA4 and Cltx-23) that show highly selective binding to malignant glioma cells [59]. A much less pronounced, yet proven ClTx feature is an antiangiogenic property. ClTx has been shown in vitro to inhibit the migration and invasion of glioma cells and human umbilical vein endothelial cell (HUVEC) [53,60]. ClTx was shown to inhibit the migration of endothelial cells when these were stimulated with VEGF and bFGF (vascular related growth factors implicated in the proliferation, migration and differentiation of endothelial cells) [60]. This antiangiogenic effect was noted in the absence of any other ClTx-related toxic effects, suggesting that inhibition of migration of HUVECs was due to a mechanism other than cell death [60]. 4. Molecular targets of Chlorotoxin 4.1. Chloride channels ClTx was first described, as the name suggests, as a chloride channel inhibitor [32]. In this report, Strichartz et al. [32] showed an inhibitory activity only when ClTx was applied to the intracellular face of reconstituted small-conductance chloride ion channels on rat epithelia using patch-clamping technique [32]. The inhibitory effect of ClTx on human GBM associated chloride channels was described [50,61]. Cheng et al. [62] recently described the scorpion venom blocking activity on single-channel as a firstorder binding reaction and revealed the identification of ClTx amongst others as a single Cl specific peptide blocker [62]. The cell cycle-dependent expression of a GCC current was suggested to be linked to cellular cytoskeletal changes [51], shape glioma cell morphology, foster proliferation and migration, and regulates apoptosis [51,63–65].

3

extracellular matrix during cancer invasion of normal tissue [79,80]. MMPs are active at neutral pH and can therefore catalyze the normal turnover of extracellular matrix (ECM) macromolecules such as the interstitial and basement membrane collagens, proteoglycans such as aggrecan, decorin, biglycan, fibromodulin and versican as well as accessory ECM proteins (i.e. fibronectin). Members of the MMP family include the ‘‘classical” MMPs, the membrane-bound MMPs (MT-MMPs) the ADAMs (a disintegrin and metalloproteinase; adamlysins) and the ADAMTS (a disintegrin and metalloproteinase with thrombospondin motif) [81]. Most members are secreted as a latent zymogen requiring activation. Once activated, the latent 72-kDa MMP-2 is converted via a 64-kDa activation intermediate to a 62-kDa mature protease [82,83]. Efficient activation of the pro-enzyme to the mature enzyme has shown to contribute to the invasive potential of tumor cells. The mechanism of activation and regulation of MMP-2 is tightly regulated by several other proteins that form a macromolecular complex [84]. Specifically, this involves interactions with membrane-associated MT1-MMP and avb3 integrin, matrix proteins, and its endogenous inhibitor TIMP-2. These proteins are stoichiometrically balanced in equimolar ratios to facilitate tumor cell migration, invasion, as well as control and maintenance of ECM proteolysis [84]. Glioma cells require the activation of MMP-2, which degrades the ECM during invasion and migration [85,86]. In the central nervous system, membrane type MMP-1 (MT1-MMP) has a more important role than MMP-2 during ECM remodeling, migration, infiltration, and invasion of gliomas [87]. MT1-MMP on cell surfaces is replenished by auto-degradation or clathrin-dependent internalization, and its concentration is stabilized by the tissue inhibitor of MMP (TIMP)-2 [88,89]. Malignant human gliomas express membrane-anchored MMPs and their endogenous TIMPs [90–92]. MMP-2 is specifically up-regulated in gliomas as well as in other tumors of neuroectodermal origin, but is not normally expressed in the central nervous system (Fig. 2, 3) [55,93]. Additionally, MMP-2 expression was shown to be in direct correlation to the tumor’s aggressiveness and grade [20,52,55]. Sontheimer et al. [93] demonstrated in 2003, using a recombinant His-Cltx, that principal ClTx-receptor on the surface of glioma cells was cell surface Matrix Metalloproteinase-2 (MMP-2) [93].

4.2. Annexin A2 The Annexin protein family is a group of calcium-dependent phospholipid-binding proteins that are found implicated in both sensing and repairing plasma membrane lesions triggered by various stimuli [66–68]. Besides their role in membrane repair, Annexin family members bind anionic phospholipids and regulate various cellular functions including vesicle trafficking, vesicle fusion, and membrane segregation, in a Ca2+-dependent manner [69,70]. Furthermore, Annexins and their binding partners (the S100 proteins) are known regulators of the cellular actin cytoskeleton [71–73]. Annexin-A2 was proposed as a molecular target for ClTx in tumor and vascular endothelial cells [74]. Using AnnexinA2 knockdown cells, the binding of ClTx to the surface of the cells was abolished [74]. The A2-complex, comprised of annexin-A2 and the protein p11 [75,76], was shown to be over-expressed at the cell surface in GBM as well as in many other human cancers and is correlated with poor prognosis [77,78]. 4.3. Matrix Metalloproteinase-2 Matrix Metalloproteinases (MMPs) are members of an enzyme family that require a zinc ion in their active site for catalytic activity (zinc-dependent endopeptidases). MMPs are critical for maintaining tissue allostasis, able to degrade structural proteins of the

Fig. 2. Glioblastoma multiforme (GBM) cell lines express Matrix Metalloproteinase2 (MMP-2). A Reverse-Transcriptase Polymerase-Chain-Reaction (RT-PCR) for the presence of MMP-2 intracellularly. GBM cell lines tested are U118, D392. First, messenger-RNA (mRNA) was extracted from the cells, followed by the production of cloned-DNA (cDNA) from the mRNA template. Both cell lines show intracellular presence of MMP-2 [4]. GAP-DH is a positive control. 100 base pairs (100 bp) marker is shown on the left. NTC = no template control.

Please cite this article in press as: Cohen-Inbar O, Zaaroor M. Glioblastoma multiforme targeted therapy: The Chlorotoxin story. J Clin Neurosci (2016), http://dx.doi.org/10.1016/j.jocn.2016.04.012

4

O. Cohen-Inbar, M. Zaaroor / Journal of Clinical Neuroscience xxx (2016) xxx–xxx

ClTx was shown to inhibit the enzymatic activity of MMP-2 in a dose-dependent fashion and reduced MMP-2 surface expression. The ClTx MMP-2 binding was shown specific, with MMP-1, MMP-3, or MMP-9 (also expressed by glioma cells) unaffected or bound by ClTx [93]. Veiseh et al. [58] showed that ClTx binding to GBM cells was reduced in the presence of a MMP-2 inhibitor. 5. Can Chlorotoxin penetrate through the blood–brain barrier? The short answer to this critical question is yes. ClTx analogues (i.e., conjugated to Cy5.5, 800CW and BLZ-100) were shown to bind to GBM’s tumors in mice when delivered via tail injection [56–58]. Xiang et al. [94] described the used of ClTx as a carrier for delivering levodopa for the treatment of Parkinson’s disease, and showed an increased distribution of dopamine in the brain of Parkinson’s disease mice model [94]. Still, in studies quoted, the integrity of the blood–brain barrier (BBB) has not been proven conclusively. This is an extension of a general discussion on the local integrity of the BBB in brain tumors, and its evolution during the course of the disease [95]. Adding to the complexity of the question, even if we assume that ClTx in its basic form can cross through the BBB, any chimeric molecular structure, in which ClTx serves as a targeting domain, will not necessarily preserve such as ability. 6. Therapeutic applications of Chlorotoxin: present and future The clinical applications of ClTx in GBM therapy are only limited by our imagination. ClTx serves as an optimal ‘‘magic bullet”, specifically targeting the GBM cells, while posing no danger to

normal cells of the brain or the body. One could argue that administering ClTx by itself may have some tumorocidic effect, binding to MMP-2 and inhibiting infiltration as well binding to GCC and influencing membrane potential. Still, multiple applications using a chimeric molecule that utilizes the ClTx as a targeting domain and bringing an effector domain to the tumor microenvironment were reported [96] and are summarized briefly next. These molecules differ in function based on the action of its effector domain only. In addition, it seems that potential side effects, toxicity and effectiveness are also governed by the effector domain molecule rather than by ClTx. One such application is in optical imaging [97], or an intraoperative ‘‘tumor paint” [58]. ClTx conjugated to a fluorescent molecular probe (Cy5.5) [58], which absorbs light in the near infrared spectrum (poorly absorbed by water or haemoglobin), makes it compatible with intra-operative imaging. This probe was proven to be the most sensitive probe for intraoperative visualization of GBM foci, having approximately 500-times sensitivity than normal MRI [58]. ClTx has been used to deliver magnetic resonancece imaging (MRI) contrast agents or nano-probes to tumorigenic tissue [98]. ClTx nanoparticles, i.e. iron oxide nanoparticles conjugated to both a therapeutic molecule or an imaging agent and ClTx were first published in 2005, using ClTx and Cy5.5 [99]. Radionuclide iodine–131–labeled, chemically synthesized ClTx (commercial name 131I–TM601) has been designated as an orphan drug for the treatment of malignant gliomas [100] and melanomas by the U.S. Food and Drug Administration (FDA) [100–102]. In a phase I clinical trial, a single dose of 131I–TM601, delivered directly to the brain (via a catheter located in the tumor bed), was shown to be well tolerated and was eliminated from the body within

Fig. 3. Human glioblastoma multiforme samples, Immunohistochemistry. (A) Chlorotoxin (ClTx) staining. Cltx was detected using a primary (BB7.2) antibody and a secondary fluorescent-labeled anti-mouse antibody. A granular membranal staining is detected in high power (100). (B) Positive control. Commercially available anti-MMP-2 antibodies and fluorescent-labeled anti-rabbit secondary antibody. A similar granular staining pattern is shown. (100) (C–E) Negative controls to samples presented in A and B (40). (C) Primary and secondary antibody without Cltx, (D) Secondary anti-mouse antibody and (E) anti-rabbit antibody, all showing negative staining [4].

Please cite this article in press as: Cohen-Inbar O, Zaaroor M. Glioblastoma multiforme targeted therapy: The Chlorotoxin story. J Clin Neurosci (2016), http://dx.doi.org/10.1016/j.jocn.2016.04.012

O. Cohen-Inbar, M. Zaaroor / Journal of Clinical Neuroscience xxx (2016) xxx–xxx

24–48 h [100]. Testing of 131I–TM601 has now moved into phase III trials. ClTx nano-probes have also been used to deliver potent chemotherapeutic drugs or biologics to GBM cells. Sun et al. [103] reported that the conjugation of ClTx to Methotrexate (a potent chemotherapeutic agent used for intracerebral administration), was shown to have increased cytotoxicity towards cancer cells when compared to the effect of the drug alone [103]. Similarly, Graf et al. [104] reported the conjugation of ClTx to platinum chemotherapy (cisplatin) [104] and for alisertib, which is currently in Phase II clinical trials for relapsed and refractory aggressive B/T-cell non-Hodgkin lymphomas [105,106]. Cytotoxicity of the nano-probe on these cancer cells was shown to increase as compared to the drug alone. Mu et al. [107] reported the development of a ClTx-gemcitabine conjugated iron oxide nanoparticle for GBM therapy, having a small size (32 nm) and uniform size distribution, stable in biological medium. This nanoparticle was shown to effectively enter GBM cells without losing potency compared to free drug. Significantly, these conjugate nanoparticles showed a prolonged blood half-life and the ability to cross the BBB in wild type mice [107]. ClTx homing can has also been used to deliver siRNA or DNA to cancer cells as a targeting gene delivery system [45,96,108–114]. Veiseh et al. [108] reported the development of a nano-vector construct comprised of a super-paramagnetic iron oxide nanoparticle core coated with polyethylene glycol (PEG)-grafted chitosan, and polyethylenimine (PEI). The construct was then functionalized with siRNA and a ClTx to improve its tumor specificity and potency. The authors concluded that ClTx enabled nanoparticle carrier may be well suited for delivery of RNAi therapeutics to brain cancer cells. Yue et al. [114] reported the use of OX26/CTX-conjugated PEGylated liposome as a dual-targeting gene delivery system for GBM. The authors concluded that with the addition of ClTx, these liposomes were ‘‘able to significantly promote cell transfection, increase the transport of plasmid DNA across the BBB and afterwards target the brain glioma cells in vitro and in vivo, exhibiting the most significant therapeutic efficacy” [114]. Tamborini et al. [115] reported a combined approach employing ClTx nano-vectors and low dose ionizing radiation to reach infiltrating tumor niches in a GBM model. Poly (lactic–co-glycolic acid) (PLGA) nanoparticles (PNP) were conjugated to ClTx. Silver nanoparticles were entrapped inside the functionalized nanoparticles (Ag-PNP-CTX), to allow detection and quantification of the cellular uptake by confocal microscopy, both in vitro and in vivo. In vitro experiments showed higher cytoplasmic uptake of Ag-PNP-CTX, with respect to non-functionalized nanoparticles. Utilizing a single whole brain X-ray irradiation, performed 20 h before nanoparticle injection, served to enhance the expression of the ClTx targets (MMP-2 and ClC-3), and through BBB permeabilization, potently increases the amount of internalized Ag-PNP-CTX. Notably, the application of Ag-PNP-CTX to irradiated tumor cells decreases the extracellular activity of MMP-2 [115]. A novel immunotherapeutic concept involves the use of targeting domains, such as the ClTx for the delivery of highly potent immunesystem components [cytotoxic T-cells (CTL’s) or NK-cells] to the tumor milieu. This assisted targeting of immune-competent cells helps circumvent some of the GBM’s immune-evasion mechanisms [4–8]. These immune cells, serving the effector function of the construct, carry the potential benefit of minimal toxicity, able to discern self from non-self in their effector functions [4].

7. Summary and conclusions GBM patients exhibit a mean survival of less than 2 years. GBM tumors are highly aggressive and infiltrative to healthy tissue,

5

rendering current multimodality treatment less effective. We present herein a new immunotherapeutic approach for GBM patients. This approach utilizes the targeting of different effector molecules and vectors to the tumor site using the specific binding of chlorotoxin. Chlorotoxin, a short 36-amino-acid compact folded peptide, has been shown to specifically bind neuroectodermal originated neoplasms including malignant gliomas via specific recognition of MMP-2, GCC Chloride Channels and Annexin-2. This GBM targeting moiety can be safely used to deliver specifically to GBM cells, and not healthy tissues, different effector mechanism, circumventing some of the GBM immune-evasion mechanisms. It seems that in current day of targeted selective immune-mediated therapy, chlorotoxin will play a key role in the battle against GBM.

Conflicts of Interest/Disclosures The authors declare that they have no financial or other conflicts of interest in relation to this research and its publication.

References [1] Buckner JC. Factors influencing survival in high-grade gliomas. Semin Oncol 2003;30:10–4. [2] Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005;352:987–96. [3] DeAngelis LM. Brain tumors. N Engl J Med 2001;344:114–23. [4] Cohen-Inbar O, Zaaroor M. Immunological aspects of malignant gliomas. Can J Neurol Sci 2016 [in press]. [5] Cohen-Inbar O. Focused neurosurgery book. Jaypee Brothers Publishers; 2016. [6] Cohen-Inbar O, Xu Z, Sheehan JP. Focused ultrasound-aided immunomodulation in glioblastoma multiforme: a therapeutic concept. J Ther Ultrasound 2016;4:2. [7] Cohen-Inbar O. Recruitment of immune effector cells against astrocytoma by MHC-chlorotoxin chimeric proteins PhD Theses. Department of Medicine, Technion Israel Institute of Technology; 2014. [8] Cohen-Inbar O. Can high intensity focused ultrasound facilitate immunomodulation in glioblastoma multiforme? Research Highlights. Inflammation and Cell Signaling; 2016 [in press]. [9] Aghi MK, Batchelor TT, Louis DN, et al. Decreased rate of infection in glioblastoma patients with allelic loss of chromosome 10q. J Neurooncol 2009;93:115–20. [10] Parsa AT, Waldron JS, Panner A, et al. Loss of tumor suppressor PTEN function increases B7–H1 expression and immunoresistance in glioma. Nat Med 2007;13:84–8. [11] Fecci PE, Mitchell DA, Whitesides JF, et al. Increased regulatory T-cell fraction amidst a diminished CD4 compartment explains cellular immune defects in patients with malignant glioma. Cancer Res 2006;66:3294–302. [12] El Andaloussi A, Lesniak MS. An increase in CD4+CD25+FOXP3+ regulatory T cells in tumor-infiltrating lymphocytes of human glioblastoma multiforme. Neuro Oncol 2006;8:234–43. [13] Choe G, Horvath S, Cloughesy TF, et al. Analysis of the phosphatidylinositol 3’kinase signaling pathway in glioblastoma patients in vivo. Cancer Res 2003;63:2742–6. [14] Ermoian RP, Furniss CS, Lamborn KR, et al. Dysregulation of PTEN and protein kinase B is associated with glioma histology and patient survival. Clin Cancer Res 2002;8:1100–6. [15] Louis DN, Holland EC, Cairncross JG. Glioma classification: a molecular reappraisal. Am J Pathol 2001;159:779–86. [16] Yang L, Ng KY, Lillehei KO. Cell-mediated immunotherapy: a new approach to the treatment of malignant glioma. Cancer Control 2003;10:138–47. [17] Pistoia V, Morandi F, Wang X, et al. Soluble HLA-G: are they clinically relevant? Semin Cancer Biol 2007;17:469–79. [18] Wiendl H, Mitsdoerffer M, Weller M. Hide-and-seek in the brain: a role for HLA-G mediating immune privilege for glioma cells. Semin Cancer Biol 2003;13:343–51. [19] Das A, Tan WL, Teo J, et al. Expression of survivin in primary glioblastomas. J Cancer Res Clin Oncol 2002;128:302–6. [20] Hao C, Parney IF, Roa WH, et al. Cytokine and cytokine receptor mRNA expression in human glioblastomas: evidence of Th1, Th2 and Th3 cytokine dysregulation. Acta Neuropathol 2002;103:171–8. [21] See AP, Parker JJ, Waziri A. The role of regulatory T cells and microglia in Glioblastoma associated immunosuppression. J Neurooncol 2015;123:405–12. [22] Wainwright DA, Chang AL, Dey M, et al. Durable therapeutic efficacy utilizing combinatorial blockade against IDO, CTLA-4, and PD-L1 in mice with brain tumors. Clin Cancer Res 2014;20:5290–301.

Please cite this article in press as: Cohen-Inbar O, Zaaroor M. Glioblastoma multiforme targeted therapy: The Chlorotoxin story. J Clin Neurosci (2016), http://dx.doi.org/10.1016/j.jocn.2016.04.012

6

O. Cohen-Inbar, M. Zaaroor / Journal of Clinical Neuroscience xxx (2016) xxx–xxx

[23] Castro MG, Baker GJ, Lowenstein PR. Blocking immunosuppressive checkpoints for glioma therapy: the more the Merrier! Clin Cancer Res 2014;20:5147–9. [24] Shah DJ, Dronca RS. Latest advances in chemotherapeutic, targeted, and immune approaches in the treatment of metastatic melanoma. Mayo Clin Proc 2014;89:504–19. [25] O’Day SJ, Maio M, Chiarion-Sileni V, et al. Efficacy and safety of ipilimumab monotherapy in patients with pretreated advanced melanoma: a multicenter single-arm phase II study. Ann Oncol 2010;21:1712–7. [26] Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melaoma. N Engl J Med 2010;363:711–23. [27] Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 2011;364:2517–26. [28] Mapara MY, Sykes M. Tolerance and cancer: mechanisms of tumor evasion and strategies for breaking tolerance. J Clin Oncol 2004;22:1136–51. [29] Khong HT, Restifo NP. Natural selection of tumor variants in the generation of ‘‘tumor escape” phenotypes. Nat Immunol 2002;3:999–1005. [30] Ojeda PG, Wang CK, Craik DJ. Chlorotoxin: structure, activity, and potential uses in cancer therapy. Biopolymers 2016;106:25–36. [31] Ding J, Chua PJ, Bay BH, et al. Scorpion venoms as a potential source of novel cancer therapeutic compounds. Exp Biol Med (Maywood) 2014;239:387–93. [32] DeBin JA, Maggio JE, Strichartz GR. Purification and characterization of chlorotoxin, a chloride channel ligand from the venom of the scorpion. Am J Physiol 1993;264:C361–9. [33] DeBin JA, Strichartz GR. Chloride channel inhibition by the venom of the scorpion Leiurus quinquestriatus. Toxicon 1991;29:1403–8. [34] MF ME FC. Scorpion neurotoxins: effects and mechanisms. In: Chang LW, Dyer RS, editors. Handbook on neurotoxicology. New York: Marcel Dekker; 1995. p. 683–716. [35] Seyedian R, Pipelzadeh MH, Jalali A, et al. Enzymatic analysis of Hemiscorpius lepturus scorpion venom using zymography and venom-specific antivenin. Toxicon 2010;56:521–5. [36] Quintero-Hernández V, Ortiz E, Rendón-Anaya M, et al. Scorpion and spider venom peptides: gene cloning and peptide expression. Toxicon 2011;58:644–63. [37] Possani LD, Becerril B, Delepierre M, et al. Scorpion toxins specific for Na+-channels. Eur J Biochem 1999;264:287–300. [38] Valdivia HH, Possani LD. Peptide toxins as probes of ryanodine receptor structure and function. Trends Cardiovasc Med 1998;8:111–8. [39] Tytgat J, Chandy KG, Garcia ML, et al. A unified nomenclature for short-chain peptides isolated from scorpion venoms: alpha-KTx molecular subfamilies. Trends Pharmacol Sci 1999;20:444–7. [40] Batista CV, Gómez-Lagunas F, Rodríguez de la Vega RC, et al. Two novel toxins from the Amazonian scorpion Tityus cambridgei that block Kv1.3 and Shaker B K(+)-channels with distinctly different affinities. Biochim Biophys Acta 2002;1601:123–31. [41] Tytgat J, Debont T, Rostoll K, et al. Purification and partial characterization of a ‘short’ insectotoxin-like peptide from the venom of the scorpion Parabuthus schlechteri. FEBS Lett 1998;441. [42] Ali SA, Alam M, Abbasi A, et al. Structure-activity relationship of chlorotoxinlike peptides. Toxins (Basel) 2016;8. [43] Rosso JP, Rochat H. Characterization of ten proteins from the venom of the Moroccan scorpion Androctonus mauretanicus mauretanicus, six of which are toxic to the mouse. Toxicon 1985;23:113–25. [44] Ali SA, Stoeva S, Schütz J, et al. Purification and primary structure of low molecular mass peptides from scorpion (Buthus sindicus) venom. Comp Biochem Physiol 1998;121:323–32. [45] Dardevet L, Rani D, Aziz TA, et al. Chlorotoxin: a helpful natural scorpion peptide to diagnose glioma and fight tumor invasion. Toxins 2015;7:1079–101. [46] Lippens G, Najib J, Wodak SJ, et al. NMR sequential assignments and solution structure of chlorotoxin, a small scorpion toxin that blocks chloride channels. Biochemistry 1995;34:13–21. [47] Ojeda PG, Chan LY, Poth AG, et al. The role of disulfide bonds in structure and activity of chlorotoxin. Future Med Chem 2014;6:1617–28. [48] Soroceanu L, Gillespie Y, Khazaeli MB, et al. Use of chlorotoxin for targeting of primary brain tumors. Cancer Res 1998;58:4871–9. [49] Ullrich N, Gillespie GY, Sontheimer H. Human astrocytoma cells express a unique chloride current. NeuroReport 1995;7:343–7. [50] Ullrich N, Sontheimer H. Biophysical and pharmacological characterization of chloride currents in human astrocytoma cells. Am J Physiol 1996;270: C1511–21. [51] Ullrich N, Sontheimer H. Cell cycle-dependent expression of a glioma-specific chloride current: proposed link to cytoskeletal changes. Am J Physiol 1997;273:C1290–7. [52] Ullrich N, Bordey A, Gillespie GY, et al. Expression of voltage-activated chloride currents in acute slices of human gliomas. Neuroscience 1998;83:1161–73. [53] Soroceanu L, Manning Jr TJ, Sontheimer H. Modulation of glioma cell migration and invasion using Cl( ) and K(+) ion channel blockers. J Neurosci 1999;19:5942–54. [54] Huys I, Waelkens E, Tytgat J. Structure-function study of a chlorotoxin-chimer and its activity on Kv1.3 channels. J Chromatogr B Analyt Technol Biomed Life Sci 2004;803:67–73.

[55] Lyons SA, O’Neal J, Sontheimer H. Chlorotoxin, a scorpion-derived peptide, specifically binds to gliomas and tumors of neuroectodermal origin. Glia 2002;39:162–73. [56] Kovar JL, Curtis E, Othman SF, et al. Characterization of IRDye 800CW chlorotoxin as a targeting agent for brain tumors. Anal Biochem 2013;440:212–9. [57] Butte PV, Mamelak A, Parrish-Novak J, et al. Near-infrared imaging of brain tumors using the Tumor Paint BLZ-100 to achieve near-complete resection of brain tumors. Neurosurg Focus 2014;36:E1. [58] Veiseh M, Gabikian P, Bahrami SB, et al. Tumor paint: a chlorotoxin:Cy5.5 bioconjugate for intraoperative visualization of cancer foci. Cancer Res 2007;67:6882–8. [59] Xu T, Fan Z, Li W, et al. Identification of two novel chlorotoxin derivatives CA4 and CTX-23 with chemotherapeutic and anti-angiogenic potential. Sci Rep 2016;6:19799. [60] Jacoby DB, Dyskin E, Yalcin M, et al. Potent pleiotropic anti-angiogenic effects of TM601, a synthetic chlorotoxin peptide. Anticancer Res 2010;30:39–46. [61] Ullrich N, Gillespie GY, Sontheimer H. Human astrocytoma cells express a unique chloride current. NeuroReport 1996;7:1020–4. [62] Cheng Y, Zhao J, Qiao W, et al. Recent advances in diagnosis and treatment of gliomas using chlorotoxin-based bioconjugates. Am J Nucl Med Mol Imag 2014;4:385–405. [63] Haas BR, Sontheimer H. Inhibition of the Sodium-Potassium-Chloride Cotransporter Isoform-1 reduces glioma invasion. Cancer Res 2010;70:5597–606. [64] Habela CW, Olsen ML, Sontheimer H. ClC3 is a critical regulator of the cell cycle in normal and malignant glial cells. J Neurosci 2008;28:9205–17. [65] Habela CW, Ernest NJ, Swindall AF, et al. Chloride accumulation drives volume dynamics underlying cell proliferation and migration. J Neurophysiol 2009;101:750–7. [66] Draeger A, Monastyrskaya K, Babiychuk EB. Plasma membrane repair and cellular damage control: the annexin survival kit. Biochem Pharmacol 2011;81:703–12. [67] Mamelak AN, Jacoby DB. Targeted delivery of antitumoral therapy to glioma and other malignancies with synthetic chlorotoxin (TM-601). Expert Opin Drug Deliv 2007;4:175–86. [68] Moss SE, Morgan RO. The annexins. Genome Biol 2004;5:219. [69] Gerke V, Moss SE. Annexins: from structure to function. Physiol Rev 2002;82:331–71. [70] Miwa N, Uebi T, Kawamura S. S100-annexin complexes–biology of conditional association. FEBS J 2008;275:4945–55. [71] Zhao XQ, Naka M, Muneyuki M, et al. Ca(2+)-dependent inhibition of actinactivated myosin ATPase activity by S100C (S100A11), a novel member of the S100 protein family. Biochem Biophys Res Commun 2000;267:77–9. [72] Hayes MJ, Rescher U, Gerke V, et al. Annexin-actin interactions. Traffic 2004;5:571–6. [73] Lauritzen SP, Boye TL, Nylandsted J. Annexins are instrumental for efficient plasma membrane repair in cancer cells. Semin Cell Dev Biol 2015;45:32–8. [74] Kesavan K, Ratliff J, Johnson EW, et al. Annexin A2 is a molecular target for TM601, a peptide with tumor targeting and anti-angiogenic effects. J Biol Chem 2010;285:4366–74. [75] Waisman DM. Annexin II tetramer: structure and function. Mol Cell Biochem 1995;149–150:301–22. [76] Deora AB, Kreitzer G, Jacovina AT, et al. An annexin 2 phosphorylation switch mediates p11-dependent translocation of annexin 2 to the cell surface. J Biol Chem 2004;279:43411–8. [77] Jia JW, Li KL, Wu JX, et al. Clinical significance of annexin II expression in human non-small cell lung cancer. Tumour Biol 2013;34:1767–71. [78] Seckinger A, Meissner T, Moreaux J, et al. Clinical and prognostic role of annexin A2 in multiple myeloma. Blood 2012;120:1087–94. [79] Sternlicht MD, Werb Z. How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol 2001;17:463–516. [80] Cox G, O’Byrne KJ. Matrix metalloproteinases and cancer. Anticancer Res 2001;21:4207–19. [81] Malemud CJ. Matrix metalloproteinases (MMPs) in health and disease: an overview. Front Biosci 2006;11:1696–701. [82] Sato H, Seiki M. Membrane-type matrix metalloproteinases (MT-MMPs) in tumor metastasis. J Biochem 1996;119:209–15. [83] Strongin AY, Collier I, Bannikov G, et al. Mechanism of cell surface activation of 72-kDa type IV collagenase. Isolation of the activated form of the membrane metalloprotease. J Biol Chem 1995;270:5331–8. [84] Toth M, Bernardo MM, Gervasi DC, et al. Tissue inhibitor of metalloproteinase (TIMP)-2 acts synergistically with synthetic matrix metalloproteinase (MMP) inhibitors but not with TIMP-4 to enhance the (Membrane type 1)-MMP-dependent activation of pro-MMP-2. J Biol Chem 2000;275:41415–23. [85] Roomi MW, Ivanov V, Kalinovsky T, et al. Inhibition of glioma cell line A-172 MMP activity and cell invasion in vitro by a nutrient mixture. Med Oncol 2007;24:231–8. [86] Wild-Bode C, Weller M, Wick W. Molecular determinants of glioma cell migration and invasion. J Neurosurg 2001;94:978–84. [87] Beliën AT, Paganetti PA, Schwab ME. Membrane-type 1 matrix metalloprotease (MT1-MMP) enables invasive migration of glioma cells in central nervous system white matter. J Cell Biol 1999;144:373–84.

Please cite this article in press as: Cohen-Inbar O, Zaaroor M. Glioblastoma multiforme targeted therapy: The Chlorotoxin story. J Clin Neurosci (2016), http://dx.doi.org/10.1016/j.jocn.2016.04.012

O. Cohen-Inbar, M. Zaaroor / Journal of Clinical Neuroscience xxx (2016) xxx–xxx [88] Sato H, Takino T. Coordinate action of membrane-type matrix metalloproteinase-1 (MT1-MMP) and MMP-2 enhances pericellular proteolysis and invasion. Cancer Sci 2010;101:843–7. [89] Hernandez-Barrantes S, Toth M, Bernardo MM, et al. Binding of active (57 kDa) membrane type 1-matrix metalloproteinase (MT1-MMP) to tissue inhibitor of metalloproteinase (TIMP)-2 regulates MT1-MMP processing and pro-MMP-2 activation. J Biol Chem 2000;275:12080–9. [90] Emdad L, Dent P, Sarkar D, et al. Future approaches for the therapy of malignant glioma: targeting genes mediating invasion. Future Oncol 2012;8:343–6. [91] Alves TR, Lima FR, Kahn SA, et al. Glioblastoma cells: a heterogeneous and fatal tumor interacting with the parenchyma. Life Sci 2011;89:532–9. [92] Yue J, Zhang K, Chen J. Role of integrins in regulating proteases to mediate extracellular matrix remodeling. Cancer Microenviron 2012;5:275–83. [93] Deshane J, Garner CC, Sontheimer H. Chlorotoxin inhibits glioma cell invasion via matrix metalloproteinase-2. J Biol Chem 2003;278:4135–44. [94] Xiang Y, Wu Q, Liang L, et al. Chlorotoxin-modified stealth liposomes encapsulating levodopa for the targeting delivery against Parkinson’s disease in the MPTP-induced mice model. J Drug Target 2012;20:67–75. [95] On NH, Mitchell R, Savant SD, et al. Examination of blood-brain barrier (BBB) integrity in a mouse brain tumor model. J Neurooncol 2013;111:133–43. [96] Zhao L, Shi X, Zhao J. Chlorotoxin-conjugated nanoparticles for targeted imaging and therapy of glioma. Curr Top Med Chem 2015;15:1196–208. [97] Stroud MR, Hansen SJ, Olson JM. In vivo bio-imaging using chlorotoxin-based conjugates. Curr Pharm Des 2011;17:4362–71. [98] Fu Y, An N, Li K, et al. Chlorotoxin-conjugated nanoparticles as potential glioma-targeted drugs. J Neurooncol 2012;107:457–62. [99] Veiseh O, Sun C, Gunn J, et al. Optical and MRI multifunctional nanoprobe for targeting gliomas. Nano Lett 2005;5:1003–8. [100] Mamelak AN, Rosenfeld S, Bucholz R, et al. Phase I single-dose study of intracavitary-administered iodine-131-TM-601 in adults with recurrent high-grade glioma. J Clin Oncol 2006;24:3644–50. [101] Hockaday DC, Shen S, Fiveash J, et al. Imaging glioma extent with 131I-TM601. J Nucl Med 2005;46:580–6. [102] Wang XM, Luo X, Guo ZY. Recombinant expression and downstream processing of the disulfide-rich tumor-targeting peptide chlorotoxin. Exp Ther Med 2013;6:1049–53.

7

[103] Sun C, Fang C, Stephen Z, et al. Tumor-targeted drug delivery and MRI contrast enhancement by chlorotoxin-conjugated iron oxide nanoparticles. Nanomedicine (Lond) 2008;3:495–505. [104] Graf N, Mokhtari TE, Papayannopoulos IA, et al. Platinum(IV)-chlorotoxin (CTX) conjugates for targeting cancer cells. J Inorg Biochem 2012;110:58–63. [105] Locatelli E, Naddaka M, Uboldi C, et al. Targeted delivery of silver nanoparticles and alisertib: in vitro and in vivo synergistic effect against glioblastoma. Nanomedicine (Lond) 2014;9:839–49. [106] Friedberg JW, Mahadevan D, Cebula E, et al. Phase II study of alisertib, a selective Aurora A kinase inhibitor, in relapsed and refractory aggressive Band T-cell non-Hodgkin lymphomas. J Clin Oncol 2014;32:44–50. [107] Mu Q, Lin G, Patton VK, et al. Gemcitabine and chlorotoxin conjugated iron oxide nanoparticles for glioblastoma therapy. J Mater Chem B Mater Biol Med 2016;4:32–6. [108] Veiseh O, Kievit FM, Fang C, et al. Chlorotoxin bound magnetic nanovector tailored for cancer cell targeting, imaging, and siRNA delivery. Biomaterials 2010;31:8032–42. [109] Veiseh O, Kievit FM, Gunn JW, et al. A ligand-mediated nanovector for targeted gene delivery and transfection in cancer cells. Biomaterials 2009;30:649–57. [110] Mok H, Veiseh O, Fang C, et al. PH-Sensitive siRNA nanovector for targeted gene silencing and cytotoxic effect in cancer cells. Mol Pharm 2010;7:1930–9. [111] Kievit FM, Veiseh O, Fang C, et al. Chlorotoxin labeled magnetic nanovectors for targeted gene delivery to glioma. ACS Nano 2010;4:4587–94. [112] Huang R, Ke W, Han L, et al. Targeted delivery of chlorotoxin-modified DNAloaded nanoparticles to glioma via intravenous administration. Biomaterials 2011;32:2399–406. [113] Costa PM, Cardoso AL, Mendonça LS, et al. Tumor-targeted chlorotoxincoupled nanoparticles for nucleic acid delivery to glioblastoma cells: a promising system for glioblastoma treatment. Mol Ther Nucleic Acid 2013;18:e100. [114] Yue PJ, He L, Qiu SW, et al. OX26/CTX-conjugated PEGylated liposome as a dual-targeting gene delivery system for brain glioma. Mol Cancer 2014;13:191. [115] Tamborini M, Locatelli E, Rasile M, et al. A combined approach employing chlorotoxin-nanovectors and low dose radiation to reach infiltrating tumor niches in glioblastoma. ACS Nano 2016.

Please cite this article in press as: Cohen-Inbar O, Zaaroor M. Glioblastoma multiforme targeted therapy: The Chlorotoxin story. J Clin Neurosci (2016), http://dx.doi.org/10.1016/j.jocn.2016.04.012