Next Article in Journal
Adult Medulloblastoma: Updates on Current Management and Future Perspectives
Next Article in Special Issue
Recent Trends in Nanomedicine-Based Strategies to Overcome Multidrug Resistance in Tumors
Previous Article in Journal
Deep Neural Network Models for Colon Cancer Screening
Previous Article in Special Issue
Repositioning of Old Drugs for Novel Cancer Therapies: Continuous Therapeutic Perfusion of Aspirin and Oseltamivir Phosphate with Gemcitabine Treatment Disables Tumor Progression, Chemoresistance, and Metastases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Drug Repurposing, a Fast-Track Approach to Develop Effective Treatments for Glioblastoma

by
Ioannis Ntafoulis
1,
Stijn L. W. Koolen
2,3,
Sieger Leenstra
1 and
Martine L. M. Lamfers
1,*
1
Brain Tumor Center, Department of Neurosurgery, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
2
Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
3
Department of Hospital Pharmacy, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(15), 3705; https://doi.org/10.3390/cancers14153705
Submission received: 11 June 2022 / Revised: 25 July 2022 / Accepted: 26 July 2022 / Published: 29 July 2022
(This article belongs to the Special Issue Drug Repurposing for Cancer Therapy)

Abstract

:

Simple Summary

Introducing novel and effective treatments against glioblastoma (GBM) remains an arduous journey as reflected in the negative outcome of most clinical trials. The blood–brain barrier and the tremendous heterogeneity of the disease comprise major obstacles in this process. Drug repurposing is a drug discovery approach that can accelerate the drug development timeline and identify promising candidates for GBM treatment. Obtaining insights already at preclinical stage into drug sensitivity and physicochemical properties for central nervous system (CNS) penetration of these candidates could shift research outcomes to more effective drugs for clinical investigation against GBM.

Abstract

Glioblastoma (GBM) remains one of the most difficult tumors to treat. The mean overall survival rate of 15 months and the 5-year survival rate of 5% have not significantly changed for almost 2 decades. Despite progress in understanding the pathophysiology of the disease, no new effective treatments to combine with radiation therapy after surgical tumor debulking have become available since the introduction of temozolomide in 1999. One of the main reasons for this is the scarcity of compounds that cross the blood–brain barrier (BBB) and reach the brain tumor tissue in therapeutically effective concentrations. In this review, we focus on the role of the BBB and its importance in developing brain tumor treatments. Moreover, we discuss drug repurposing, a drug discovery approach to identify potential effective candidates with optimal pharmacokinetic profiles for central nervous system (CNS) penetration and that allows rapid implementation in clinical trials. Additionally, we provide an overview of repurposed candidate drug currently being investigated in GBM at the preclinical and clinical levels. Finally, we highlight the importance of phase 0 trials to confirm tumor drug exposure and we discuss emerging drug delivery technologies as an alternative route to maximize therapeutic efficacy of repurposed candidate drug.

1. Introduction

Glioblastoma (GBM) is the most aggressive form of diffuse gliomas and the most lethal among all types of brain tumors, comprising 12–15% of all adult intracranial tumors and 50–60% of astrocytic neoplasms [1]. According to the 2021 WHO classification of CNS tumors, former (grade 4) GBM is now classified based on the presence or absence of mutations in the isocitrate dehydrogenase (IDH) gene: IDH wild-type (IDHwt) glioblastoma or IDH-mutant (IDHmut) grade 4 astrocytoma [2]. Molecularly, IDHwt glioblastomas are characterized by the presence of TERT promoter mutation, EGRF amplification, +7/−10 chromosome copy number changes or any combination of the above [2]. IDHmut grade 4 astrocytomas are characterized by mutations of IDH1/2, ATRX, TP53, CDKN2A/B homozygous deletion, PDGFRA amplification or any combination of the above [2]. These genomic alterations of IDHwt gliomas are associated with fast growth rates and poor prognosis [2].
The standard of care treatment following diagnosis comprises maximal safe surgical resection of the tumor (debulking), followed by radiation therapy (RT) and concurrent and adjuvant chemotherapy with the alkylating agent temozolomide (TMZ) [3,4]. However, the survival rates of the patients diagnosed with GBM and receiving first-line treatment remain very low. The median overall survival (OS) is 12–15 months, while only 3% of patients have a progression-free survival (PFS) of more than 5 years [5]. The MGMT (O6-methylguanine–DNA methyltransferase) promoter is a well-established predictive marker of response in GBM patients receiving TMZ. The epigenetic silencing of the MGMT gene by promoter methylation compromises DNA repair, improving response to TMZ and leading to longer survival of glioblastoma patients [6]. Inevitably, all GBM patients receiving RT + TMZ and/or adjuvant TMZ therapy relapse; the median PFS upon completing the first line of treatment varies between 1, 5 and 6 months [7]. Lomustine (CCNU), an alkylating agent, is sometimes administered as ultimate treatment option to recurrent GBM patients with minor therapeutic benefit [8,9,10,11].
In 2009, the U.S. food and drug administration (FDA) approved bevacizumab for the treatment of GBM with relapse after prior RT + chemotherapy [12]. Bevacizumab is a recombinant humanized monoclonal antibody, with anti-angiogenic properties by blocking vascular endothelial growth factor A (VEGF-A). However, its moderate clinical benefit and unproven OS advantage to date have withheld approval by the European medicine agency (EMA) [13,14,15,16]. The most recent therapeutic approach for recurrent GBM, which received FDA approval in 2011, is a device known as tumor-treating fields (TTF) [17]. In 2015, the device was also granted FDA approval for newly diagnosed GBM [17]. The device delivers low intensity, alternating electric fields to the tumor, therewith inhibiting glioma cell proliferation [18,19]. Moderate improvements in the survival of newly diagnosed GBM patients have been observed by adding TTF as an adjuvant treatment upon completing the standard of care treatment [20,21]. In Europe, the use of TTF is very limited to date, as the appropriate usage and implementation of the device in daily clinical practice presents many challenges [22,23].
Despite these limited additions to the arsenal of treatments, the prognosis of GBM patients remains dismal [24,25]. Two key players are involved in failure of conventional and targeted therapies: (1) the tremendous intra- and inter-tumoral heterogeneity of GBM and (2) the blood–brain barrier (BBB) [26,27,28]. GBM heterogeneity contributes to drug resistance and treatment escape and comprises a complex and arduous obstacle to overcome [29,30,31]. Extensive genetic and epigenetic profiling led to the classification of GBM tumors into three distinct molecular subgroups (classical, mesenchymal and proneural) as well as to the characterization of distinct DNA methylation profiles and/or expression patterns within these GBM subgroups [32,33,34]. Additionally, single-cell RNA sequencing analysis revealed different molecular subtypes within each tumor that can dynamically adapt to micro-environmental cues [34,35,36,37]. To date, these findings provide a better understanding of the heterogeneous nature of GBM; however, their clinical relevance, in particular in relation to drug treatment, is still limited [38].
The second major obstacle in GBM treatment is the BBB, which prevents effective delivery of drugs to the central nervous system (CNS). Therefore, to achieve any therapeutic response, it is of utmost importance that drugs cross the BBB and reach the tumor region in therapeutically effective concentrations. Drug discovery tools have been developed to identify optimal drug candidates for CNS penetration based on their physicochemical properties [39,40]. Moreover, efforts are being directed towards assessing CNS penetration and actual target delivery of new agents, as noted in the increasing number of phase 0 trials for GBM [41,42,43]. In addition, new delivery techniques, such as focused ultrasound sonication (FUS) and/or the use of nanoparticles to encapsulate therapeutic molecules, are being used to enhance systemic drug delivery into the CNS [44,45,46]. Examples include chemotherapeutic agents widely used in clinical practice, such as paclitaxel, cytarabine, carboplatin, etoposide and daunorubicin [47,48,49,50,51,52,53].
Based on these developments, a renewed interest in the available anticancer agents has arisen. With tools available to predict, enhance and assess drug delivery to CNS tumors as well as approaches to define markers of tumor sensitivity to specific compounds, the available arsenal of approved anticancer agents may be re-evaluated for potential GBM treatment. This approach, known as drug repurposing or drug repositioning, is a recognized strategy in drug discovery aiming to identify secondary indications for already approved drugs [54,55,56]. Given the unmet need for novel therapeutic options for GBM, drug repurposing may be a valuable tool, bypassing the delays and high costs of the novel drug development process and providing new drug candidates against GBM within a relatively short timeframe.
In this review, we aim to: (1) describe the role of the BBB and tumor heterogeneity in the failure of treatments; (2) introduce the significance of drug repurposing in identifying new candidate agents against GBM; (3) highlight the importance of selecting candidates based on the physicochemical properties and/or PK profiles for CNS penetration, as well as the development of novel delivery approaches, to optimize drug delivery to GBM; and (4) provide recent examples of repurposed drugs under clinical investigation against GBM.

2. The Blood–Brain Barrier and Drug Efflux Pumps

The BBB is a neurovascular unit that, in physiological conditions, acts as a ‘gatekeeper’ [57]. The main task of the BBB is to maintain the brain homeostasis by controlling the passage of endogenous and exogenous molecules from the blood stream into the CNS [57,58]. Structurally, the BBB consists of endothelial cells interconnected with a complex network of proteins (tight junctions), while pericytes and astrocytic end-feet provide an additional structural support to the brain microvasculature (Figure 1) [57,59]. Tight junctions (TJs) are the key feature of the BBB and responsible for the impediment of polar solutes through intracellular and paracellular diffusion pathways [57]. TJs consist of claudins, occludins, junction adhesion molecules (JAMS) and various cytoplasmic accessory proteins, such as Zonula occludens-1, -2, -3 (ZO-1, ZO-2, ZO-3) (Figure 2) [60]. The transport of molecules across the BBB can be achieved by different routes, including passive diffusion, solute carriers (SLC), ATP-binding cassette (ABC) transporters, transcytosis and receptor-mediated transport [61,62]. Lipid soluble molecules can passively diffuse the BBB and reach the CNS at a rate that is linked to their physicochemical properties [40].
In pathological conditions, such as brain tumors, the BBB is presented with functional abnormalities affecting normal cellular processes. [63]. Such functional abnormalities also affect processes such as angiogenesis, leading to an abnormal production of proangiogenic factors and malformation of blood vessels [64,65,66]. Specifically, the activation of the endothelial angiopoietin-2 (ANG-2)-TIE growth factor receptor pathway promotes the upregulation of VEGF and the induction of tumor angiogenesis [64,67,68]. Additionally, imbalances in the release of chemical mediators, such as substance P, histamine, bradykinin, thrombin matrix metalloproteinases (MMPs) and/or cytokines, including tumor necrosis factor-alpha (TNF-α), transforming growth factor beta (TGF-β), interleukin (IL)-1 beta and IL-6, can cause the loss of TJs and subsequently BBB breakdown and dysfunction (Figure 2) [57,60,69,70,71,72]. The loss of aquaporin 4 (AQP4) can also lead to BBB disruption by inducing the polarization of astrocytic end-feet [73]. These changes can result in a leaky BBB, also known as the blood–tumor barrier (BTB) [74]. In fact, this leakiness forms the basis of contrast-enhanced MRI imaging of CNS tumors. The extent to which the accumulation of therapeutic agents into brain tumor tissue is affected by the BTB is not well known.
Glioblastoma displays intra-tumoral heterogeneity in drug penetration, resulting from localized areas of vasogenic edema and areas with an intact BBB [74]. The highly infiltrative nature of glioma cells allows them to invade the surrounding brain parenchyma, therewith instigating the growth of malignant foci at a distance of the tumor core around blood vessels with an intact BBB [75,76,77]. After the surgical resection of the tumor core, these cells are left behind and are responsible for the recurrence of the tumor. It, therefore, remains crucial for the development of new treatments that drugs effectively penetrate the BBB in order to reach these foci.
Another key player impeding drug delivery into the CNS is the family of drug efflux pumps and more specifically the ATP-binding cassette (ABC) transporters [78]. The family of ABC transporters consists of 48 identified human ABC transporter genes classified in seven subfamilies [79]. The ABC transporters are actively involved in many intracellular processes by importing or exporting substrates through membranes by utilizing ATP [78,79,80]. Mutations in genes encoding ABC transporters can lead to numerous disorders comprising retinal degeneration, skin diseases, cystic fibrosis and hypercholesterolemia [81,82]. In human malignancies, the role of ABC transporters in the development of multidrug resistance (MDR) has been extensively studied [83,84,85,86,87]. The main efflux pumps linked to MDR are the (1) ABCB1 (P-glycoprotein or P-gp), (2) ABCG2 (breast cancer resistance protein, (BCRP)) and (3) ABCC4 (multidrug resistance-associated protein 4 (MRP4)) [88,89]. Approximately 60% of the available drugs on the market are substrates of ABCB1, making it a key player in the regulation of intracellular drug accumulation and cytotoxicity [90].
Under physiological conditions, ABCB1 and ABCG2 are mainly expressed by brain endothelial cells, allowing the efflux of molecules from the brain parenchyma to the bloodstream [91,92]. In brain tumors such as glioma, efflux pumps are present on the (peri)tumoral vasculature as well as on glioma cells [93]. The upregulation of ABCB1 and ABCG2 hampers the CNS delivery of chemotherapeutic agents, including TMZ [92,94,95,96]. Additionally, de Gooijer et al. have shown that drug delivery restriction is observed even when the BBB is disrupted, highlighting the key role of tumor-cell-associated efflux pumps in the development of drug resistance against GBM [78]. The unique anatomical and biological features of GBM make its treatment extremely challenging. Undoubtedly, the role of drug efflux transporters can be linked to the innumerable failures of clinical trials in GBM and, therefore, needs to be taken into consideration in order to design more effective treatments [91,97]. Hence, in drug development, it is a pre-requisite to identify or design drugs with optimal physicochemical properties and PK profiles to cross the BBB, but also with a low affinity for the ABC transporters in order to achieve and maintain therapeutically effective concentrations in brain tumor tissue [75,98,99,100].

3. Tumor Heterogeneity and Drug Resistance

Tumor heterogeneity is another important factor involved in the development of drug resistance, which subsequently leads to limitations or failures of the majority of therapeutic approaches against GBM [26,28]. It can be classified as intra- and inter-tumoral heterogeneity. Intra-tumoral heterogeneity allows molecularly distinct subpopulations to escape treatment, signifying the need for combination therapies to target the whole tumor population [101,102]. On the other hand, the large inter-tumoral heterogeneity, characterized by intrinsically resistant or sensitive tumors to any given drug, reveals the need to identify biomarkers of response and to focus on personalized treatments for GBM. Indeed, drug screening studies on panels of patient-derived GBM cells have uncovered tremendous intertumoral variability in drug sensitivities for almost any given drug [103,104,105]. The most well-studied biomarker in GBM is the methylation status of the MGMT promoter, which is predictive for response to temozolomide [106,107]. Nowadays, great efforts are being made to identify the biomarkers of response for a multitude of potential GBM treatments, including targeted drugs, chemotherapies and immunotherapies [105,108,109,110]. Recently, Fabro et al. described the different biological mechanisms that glioma cells exploit to escape TMZ treatment, which include enhanced DNA repair, epigenetic changes, stem cell characteristics, the tumor microenvironment, metabolism, autophagy, adaptive molecular pathways and enhanced drug efflux [29]. A better understanding of these mechanisms can aid in identifying the biomarkers of response as well as in the development of combination therapies to counteract drug resistance mechanisms [26,27].
The majority of the clinical trials in GBM patients during the past two decades yielded disappointing outcomes, at best prolonging survival in small sub-populations of patients [111]. These failures have been attributed to, among other factors, the lack of BBB penetrance of tested drugs as well as rapidly developing drug resistance, but also to the interpatient variability in intrinsic tumor sensitivity for the tested agent [112]. Many of these trials were considered a failure when only a small percentage (~10%) of patients responded to the treatment. With more emphasis on the selection of compounds with favorable physicochemical properties, as well as the availability of predictive biomarkers of response, some of those drugs may be of interest to be re-evaluated.

4. Drug Repurposing for Glioblastoma

Drug repurposing is a drug discovery approach investigating new indications for registered medicines against various diseases, including cancer [113]. Given the failures of introducing new and effective treatments for GBM over the past decades [114], drug repurposing might offer a way to identify drug candidates with favorable characteristics for CNS penetration that can be provided to patient-(sub)populations in a relatively short timeframe. A significant advantage of this approach is the available knowledge of pharmacokinetics (PK), pharmacodynamics (PD) and drug safety profiles. This knowledge can lead to the considerable shortening of the preclinical and clinical phases of drug development (Figure 3) [115].
Drug repurposing has already been applied in different types of cancers. Examples of successfully repurposed drugs that were granted approval for a second indication include: aspirin for colorectal cancer [116,117], raloxifene for breast cancer [118,119] and thalidomide for multiple myeloma [55,120]. Additional examples of drug repurposing applications in clinical cancer drug development are the MyPathway and DRUP studies [121,122]. The most recent one, the DRUP study, it is a Dutch multicenter personalized therapy trial that aims to expand the use of available targeted anticancer drugs to patients with other types of cancer but who share the same genetic profile [121]. Unfortunately, only a small percentage of GBM patients are enrolled in such trials as genomic-based approaches rarely identify targetable mutations in this patient group for which effective compounds are available. One targetable mutation that can be identified in GBM, and for which a therapeutically effective compound is available, is the BRAF-V600 mutation [123,124]. Recently, a multicenter, open-label, single-arm, phase 2, basket trial showed that the combination of dabrafenib (BRAF inhibitor) with trametinib (MEK inhibitor) has clinical efficacy against low- and high-grade gliomas [125]. Both drugs have previously been granted FDA approval as a combination for the treatment of metastatic BRAF-V600 mutated melanoma [126,127].
Additionally, the DNA damage repair protein MGMT plays a major role in the development of drug resistance to alkylating agents by removing alkyl groups from the O6 position of guanine [128,129]. Disulfiram (ALDH inhibitor) was found to inhibit MGMT activity and sensitize glioma cells to alkylating agents [130]. Disulfiram is an FDA approved drug used for alcoholism treatment [131]. Recently, Wu et al. have shown that the inhibition of PARylation by talazoparib (a PARP inhibitor) reduces MGMT functioning and therefore increases the sensitivity to TMZ of GBM cells with unmethylated MGMT promoter. Talazoparib is an FDA/EMA approved agent for the treatment of advanced breast cancer with BRCA mutation [132].

4.1. Drug Screening Platforms for the (Re)Evaluation of Registered Compounds

Drug repurposing has also found its way to preclinical drug development research. Patient-derived preclinical model systems (2D and 3D cell cultures and PDX models) have become the gold-standard for GBM drug discovery and development [31,133,134,135,136,137,138,139,140]. These preclinical models have been shown to recapitulate the human GBM biology and to predict clinical response to the standard of care agent temozolomide in GBM patients [141,142,143]. Large-scale drug screening efforts applying such platforms can identify drug candidates for GBM based on dose–response profiles. Importantly, panels of patient-derived GBM models representing the whole spectrum of GBM patients can be employed, allowing for the identification of biomarkers of response to tested agents as well as pathways of resistance. Such approaches can also aid in the identification of effective combination strategies [104,144].
Promising drug candidates identified based on such approaches include actinomycin-D. Taylor et al. identified actinomycin-D as a promising candidate among the approved oncology drug set VIII against recurrent GBM, showing that actinomycin-D is more effective than TMZ [145]. Recently, our group also identified actinomycin-D as a highly active compound in IDHmut high-grade glioma cell cultures [146]. Actinomycin-D is an antineoplastic antibiotic agent, approved in the 1960s and is still widely used for the treatment of various human malignancies [147]. Mechanistically, actinomycin-D exhibits anticancer activity by targeting RNA polymerase and inhibiting transcription [147]. Clinical trials testing actinomycin-D in GBM patients have not yet been reported, which may be related to its poor PK profile for CNS penetration. However, given the emerging new delivery approaches such as FUS and liposomal delivery forms, actinomycin-D may comprise a promising repurposed drug candidate for further investigation.
Interestingly, omacetaxine mepesuccinate, an anticancer drug approved for the treatment of hematological malignancies, has been identified as a potent anti-glioma candidate in various GBM preclinical studies [104,105,146,148]. Omacetaxine mepesuccinate is characterized by a favorable PK profile for CNS penetration [149] and, mechanistically, it targets the ribosomal protein 3 (RPL3), therewith inhibiting protein translation [150]. Other examples of available anticancer agents that have been identified to exhibit potent anti-glioma effect at preclinical level include plicamycin an antineoplastic antibiotic agent [148,151], trametinib a MEK inhibitor [152,153], afatinib an EGFR inhibitor and topotecan a topoisomerase-I inhibitor [105].
A different category of drugs being evaluated concerns non-anticancer agents that may have indirect or off-target effects on glioma cells. In particular, drugs developed for CNS diseases are of interest for glioblastoma due to their optimal PK characteristics for CNS penetration. Their well-established safety profile makes them of interest for combination strategies with anticancer drugs. An example is the recently published study by Bi et al. on the anticancer effects of fluoxetine [154]. Fluoxetine is a second-generation antidepressant drug, which selectively inhibits serotonine reuptake. It possesses a good safety profile and optimal PK characteristics for CNS penetration [155,156]. Preclinical studies in various types of cancer have shown that fluoxetine induces its anticancer effect by inhibiting the NF-κβ signaling and inducing Ca2+ related apoptosis [156,157,158]. Furthermore, fluoxetine has been found to act synergistically with temozolomide against glioma cells [154,159]. A retrospective survival analysis revealed that GBM patients taking fluoxetine during their chemo-radiation treatment had a prolonged overall survival [154]. Other non-oncological drugs being investigated as anti-glioma agents include various statins [160], metformin [161,162] and disulfiram [163] as well as drugs developed for treating various pathogens, such as mebendazole, chloroquine and lumefantrine [164,165,166,167]. CNS compounds under investigation include anti-schizophrenia drugs (fluphenazine and fluspirilene) [168] and anti-epileptics (valproic acid [169] and levetericam [170,171]) as well as drugs developed for Alzheimer’s (memantine) [172] or Parkinson’s disease (pimavanserin) [173] (Table 1).
Finally, a clinical trial has been initiated combining nine different non-oncological agents, the CUSP9 treatment protocol, with a low dose of temozolomide in recurrent GBM patients [174]. Adapted versions of the treatment regiments include the CUSP9* [175] and CUSP9v3 [176], with the latter revealing safety of this combination treatment approach. CUSP9v3 is currently being further investigated in a phase 1/2 clinical trial (NCT02770378).

4.2. Drug Selection for CNS Delivery

Selecting drug candidates for systemic delivery, which can achieve therapeutic concentrations in brain tumor tissue, may significantly improve the outcomes of clinical studies [177]. This approach requires, already from a preclinical stage, a focus on drugs showing a higher probability of crossing the BBB. Various algorithms and drug discovery tools have been developed to predict the CNS permeability of drugs [178,179,180,181,182,183,184]. Among these is the well-recognized CNS Multiparametric Optimization (CNS-MPO) desirability tool, which is characterized by a simple-to-use design algorithm and multiparameter approach in drug discovery [40]. Wager et al. based this drug discovery method on an algorithm by which molecules can be characterized based on their physicochemical properties to predict CNS penetration [40]. These properties include the partition of coefficient (logP), constant of distribution (logD), polar surface area (PSA), number of hydrogen atoms and acid dissociation constant (pKa) as well as molecular weight (MW) [40]. The algorithm combines these parameters into a single value that provides an estimation of the probability that the drug will reach the CNS. The availability of such scores can aid in selecting compounds of interest in drug repurposing programs for GBM. Additionally, the selection of compounds with negative substrate properties for the drug efflux pumps can help to maintain and prolong therapeutic drug levels in the CNS and contribute to better treatments. Although highly valuable in the pre-selection process, these algorithms and properties do not provide a guarantee for drug CNS penetration. Factors such as the absorption, distribution, metabolism and excretion (ADME) profiles of the drug candidates can strongly influence the ultimate drug concentrations in the tumor tissue. Therefore, it remains essential to perform additional in vivo studies to confirm adequate target delivery and modulation.

4.3. The Importance of Phase 0 Clinical Trials in Glioblastoma

Findings derived from pre-clinical drug development studies can be a milestone in bringing promising compounds to clinical implementation [141]. In this process, the PK profile of these compounds has a significant role and confirmation of drug penetration into the tumor tissue at therapeutically effective concentrations provides important support for further development [208]. Various pre-clinical models have been developed to recapitulate the biology of the disease and predict BBB penetration, providing valuable findings for the research community [78,137,182,209,210]. However, these models will not have the weight of evidence of a clinical trial and can only serve as an aid in identifying more or less promising compounds. Currently, a paradigm shift is taking place, where PK/PD and drug delivery studies are forming an essential step in drug development for CNS malignancies. These so-called “window-of-opportunity” studies, or phase 0 trials, are exploratory studies that can promote the rapid clinical implementation of promising preclinical findings on registered compounds and therefore circumvent phase 1 trials given the available knowledge on the safety profiles of these agents [185]. In phase 0 trials, patients are exposed shortly to a drug prior to tumor resection. The resected material is analyzed by liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) to confirm drug delivery to the tumor, and by additional assays to verify modulation of the intended molecular targets [41]. Upon confirmation of these two key points, eligible patients are subsequently enrolled into a phase 2 study arm or into a different study in order to avoid exposure to an ineffective treatment.
The AZD1775, a first in-class Wee1 inhibitor, is one of the first anticancer agents where the tumor penetration and clinical biological activity were evaluated in recurrent GBM patients [211]. Since then, multiple phase 0/2 clinical trials with registered anticancer agents have been initiated in recurrent glioma patients. Examples include (1) ixazomib, a proteasome inhibitor registered for multiple myeloma and relapsed or refractory systemic light chain (AL) amyloidosis [212,213]; (2) ribociclib, a cyclin-dependent kinase (CDK) 4/6 inhibitor in combination with everolimus an mTOR inhibitor, both registered for advanced or metastatic breast cancer (NCT03834740) [214,215,216]; (3) infigratinib, a kinase inhibitor registered for unresectable locally advanced or metastatic cholangiocarcinoma (NCT04424966) [217]; and (4) pamiparib, a poly(ADP-ribose) polymerase (PARP) 1/2 inhibitor approved in China for the treatment of recurrent advanced ovarian cancer, fallopian tube or primary peritoneal cancer previously treated with two or more lines of chemotherapy (NCT04614909) [218].
These “window-of-opportunity” phase 0 trials, as recently reviewed by Vogelbaum et al. [219], have successfully demonstrated target delivery and drug activity for some of these agents, subsequently allowing them to move to phase 2 clinical investigations. As a next step, even more personalized approaches can be envisioned, in which confirmed drug delivery is a pre-requisite for follow-up treatment with any drug being investigated, therewith accounting for inter-patient variability in BBB functionality. Together, these approaches are highly valuable as phase 0/2 studies cover the unmet need for a rapid early phase drug development track by preventing patient exposure to ineffective treatments, identifying novel drugs for further investigation and thus increasing the success rate of later-stage clinical studies [185].

4.4. Clinical Applications of Repurposed Drugs Systemically Delivered in Glioblastoma

As a result of these developments, a plethora of approved drugs, initially developed for other malignancies or non-oncological indications, are now being (re)evaluated as potential treatments for GBM patients [166,194,220,221,222]. An overview of previous and ongoing clinical trials assessing repurposed drugs in GBM patients is presented in Table 2. Such examples include the approved anticancer agent regorafenib, an inhibitor of multiple kinases [223] that is currently being investigated in the phase II/III AGILE trial for newly diagnosed or recurrent GBM (NCT03970447). Previously, a randomized, multicenter, open-label phase 2 trial (REGOMA) indicated enhanced efficacy of regorafenib compared to CCNU in recurrent GBM patients [224]. These findings were further supported by a retrospective clinical study analyzing the clinical outcomes of recurrent GBM patients receiving regorafenib outside clinical trials [225].
Another repurposed registered anticancer drug is abemaciclib [226], which targets the CDK4/6-RB1 signaling pathway and it is under development for brain tumors. Preclinically, abemaciclib was found to reach the brain at therapeutically effective concentrations and exhibit a strong anti-glioma effect [185,227]. The PK and PD profile of abemaciclib was assessed in GBM patients where the drug was detected in the cerebrospinal fluid (CSF) and associated with the inhibition of CDK4/6 [228]. According to Li et al., abemaciclib is considered the most optimal candidate of its class and is currently being investigated as monotherapy as well as in combination with LY3214996 in GBM patients (NCT02981940 and NCT04391595) [185].
Niraparib is a highly selective inhibitor of PARP 1/2 and registered for the treatment of advanced ovarian cancer [229]. In glioblastoma, various PARP inhibitors (PARPi) were clinically tested in patients with recurrent glioblastoma [230]. However, the poor CNS penetration, high affinity to efflux pumps transporters as well as the adverse events caused by the combination of PARPi with temozolomide did not lead to significant therapeutic benefit [231,232,233]. Recently, the implementation of TTF in glioblastoma treatment was associated with the downregulation of breast cancer type 1 susceptibility protein (BRCA1) signaling and the reduction in DNA double-strand break repair capacity [234]. The therapeutic effect of niraparib in combination with TFF is currently being investigated in a phase 2 clinical study (NCT04221503).
Ibrutinib, a small-molecule inhibitor of Bruton’s tyrosine kinase (BTK) and bone marrow X-linked (BMX) non-receptor tyrosine kinase, has been approved for the treatment of patients with chronic lymphocytic leukemia (CLL) and mantle cell lymphoma [235]. In glioblastoma, BMX non-receptor tyrosine kinase is overexpressed, promoting the abnormal activation of the signal transducer and activator of transcription 3 (STAT3), which is involved in self-renewal of glioma stem cells (GSCs) and maintaining GSC tumorigenic potential [236]. Preclinical studies suggest that targeting GSCs through BMX inhibition by ibrutinib may effectively improve GBM treatment [196]. The tolerability and safety of Ibrutinib is currently being investigated in phase 1 clinical study (NCT03535350) in combination with the standard-of-care treatment.
Research interest has also been placed in hydroxyl-chloroquine (CHQ), an anti-malaria drug, of which the anti-tumor effects have been investigated in different types of cancer, including GBM [237,238,239]. CHQ expresses its anticancer effect through the inhibition of autophagy or interference with PI3K/Akt or EGFR signaling pathways in glioma cells [240,241]. A phase 1/2 clinical trial investigated the therapeutic benefit of combining CHQ it to the standard-of-care treatment [242]. Additionally, Compter et al. have shown that, although a slight improvement of the overall survival was observed in GBM patients with EGFRvIII positive tumors, significant side effects may arise by combining CHQ to radiotherapy and TMZ [166].
Hydroxyurea is another FDA-approved compound against various diseases including cancer, which has been found to sensitize glioma cells to temozolomide [243]. A phase 1 clinical study is currently investigating the therapeutic benefit of hydroxyurea in combination with TMZ in recurrent GBM patients (NCT03463733).
Disulfiram (DSF), an approved anti-alcoholism agent, in combination with nutritional copper (Cu) supplement was found to enhance the therapeutic effect of temozolomide in glioma cells [244]. These preclinical data suggest that DSF/Cu in combination with alkylating chemotherapy decreases glioma cell invasion and impairs the DNA repair pathways, thereby improving the efficacy of DNA alkylating agents [163].The safety of this approach was demonstrated in a phase 1 trial for recurrent GBM and the efficacy of this combination was investigated in phase 2 trial, which showed limited activity in the unselected IDH-wild type GBM population [245,246]. Currently, a phase 2 clinical study is ongoing to assess the effect of the DSF/Cu combined with TMZ in GBM patients with unmethylated MGMT promoter (NCT03363659).
A highly interesting approach being investigated in a phase 3 trial is the direct screening of patient-derived tumor cells to determine the most promising anticancer drug for a particular patient (ChemoID) (NCT03632135). This panel of approved anti-cancer agents includes carboplatin, irinotecan, etoposide, carmustine, lomustine, temozolomide, procarbazine, vincristine, imatinib or combinations of these drugs. A personalized pre-screen takes the intertumoral heterogeneity into account and therewith avoids treating patients with drugs for which the tumor is intrinsically resistant. Such approaches are expected to yield much higher response rates [141].

5. Novel Approaches for Delivering Repurposed Drugs across the BBB

The limitations in developing novel drugs that overcome the constraints of the BBB brought a new technological era in delivery systems. Novel approaches aim to reinforce the therapeutic potential of drugs lacking favorable PK characteristics for CNS penetration. One such approach is the focused ultrasound sonication (FUS) technology. This non-invasive technology is designed to improve drug delivery to the CNS by opening the BBB with ultrasonic waves [44,247]. Preclinical and clinical studies have shown that FUS technology can enhance the systemic delivery of compounds with poor PK profile, such as paclitaxel, doxorubicin and carboplatin across the BBB into the CNS [52,53,248,249]. The promising results of these preclinical studies has prompted further investigations in multiple clinical studies combining FUS technology to the standard-of-care treatment (NCT04614493) and/or widely used anticancer agents such as carboplatin (NCT04440358) [250] and paclitaxel (NCT04528680) in GBM patients.
Other technologies being developed for improved CNS drug delivery include nanomaterials, such as liposomes and nanoparticles, which can be used as carriers of therapeutic agents [251,252,253]. Encapsulated drugs can be designed to provide higher drug stability together with elevated accumulated concentrations in the tumor tissue and lower drug-related toxicities [254]. The systemic delivery of encapsulated drugs can also be achieved using exosomes [255]. These nano-sized extracellular vesicles can act as nanocarriers releasing therapeutic agents to the tumor cells and improving drug efficacy [256].
Additionally, local delivery methods, such convection-enhanced delivery (CED) as well as intranasal delivery, are being used to circumvent the BBB/BTBB and achieve the direct delivery of repurposed drugs into the (peri)tumoral area. Such examples include topotecan [257], which is delivered via CED against recurrent GBM, as well as perillyl alcohol via intranasal delivery [258]. Similarly, a novel introduced catheter systems for direct delivery of therapeutics to the brain (Neuroinfuse™) aims to improve chronic and acute implantable intra-parenchymal drug delivery [259]. To date, the only FDA-approved local delivery approach against GBM are Gliadel® wafers, showing an enhanced efficacy when combined with standard-of-care treatment [260]. However, this approach has limitations as only patients undergoing a gross total tumor resection are eligible for this treatment and therapeutic benefit is dependent on factors such as age and Karnofsky performance score (KPS) [261].
With the emergence of more and more (repurposed) candidate drugs with potent anti-glioma activity from patient-derived tumor drug screening platforms, and the parallel development of improved CNS delivery techniques, it is expected that the arsenal of compounds becoming available for clinical assessment in GBM patients will greatly expand.

6. Conclusions

Glioblastoma has one of the worst prognoses among all cancers. The lack of progress in developing new treatments is inextricably linked to the hurdle of delivering drugs to the CNS as well as the heterogeneity of GBM. Therefore, there remains a high need for therapeutic agents, either as monotherapy or in combination with standard-of-care treatment, from which GBM patients will benefit. Drug repurposing is an important player in this battle, offering novel treatment options with rapid clinical implementation by circumventing the standard drug development process. Key in drug repurposing is the availability of patient-derived, clinically relevant, preclinical model systems that allow for the screening of available drugs and the identification of potent compounds for GBM (subtypes). Moreover, the development of novel algorithms and the available PK data can significantly aid in selecting drug candidates that can effectively cross the BBB. To date, this approach has already led to the identification of candidate drugs registered for various other types of cancer. The validation of drug delivery in relevant in vivo models can further reduce the risk of bringing ineffective drugs to clinical trials. Finally, the clinical implementation of these findings in phase 0/2 trials, as well as the development of novel technologies to improve CNS delivery, is expected to significantly improve the success rate of anti-glioma treatments evaluated in patients.
Taken together, the vast number of registered drugs available for (re)evaluation in clinically relevant GBM model systems, combined with improved tools to predict, validate and achieve CNS penetration, is expected to rapidly increase the entry of more effective compounds against GBM into the clinical arena.

Author Contributions

Conceptualization, I.N. and M.L.M.L.; writing, I.N.; review and editing, S.L.W.K., S.L. and M.L.M.L.; visualization, I.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the European Union’s Horizon 2020 Research and Innovation program under the Marie Skłodowska-Curie Actions (No. 766069 GLIOTRAIN).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Thakkar, J.P.; Dolecek, T.A.; Horbinski, C.; Ostrom, Q.T.; Lightner, D.D.; Barnholtz-Sloan, J.S.; Villano, J.L. Epidemiologic and molecular prognostic review of glioblastoma. Cancer Epidemiol. Biomark. Prev. 2014, 23, 1985–1996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. WHO Classification of Tumours Editorial Board. World Health Organization Classification of Tumours of the Central Nervous System, 5th ed.; International Agency for Research on Cancer: Lyon, France, 2021; Volume 6.
  3. Stupp, R.; Mason, W.P.; Bent, M.V.D.; Weller, M.; Fisher, B.; Taphoorn, M.J.B.; Belanger, K.; Brandes, A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef] [PubMed]
  4. Ostrom, Q.T.; Bauchet, L.; Davis, F.G.; Deltour, I.; Fisher, J.L.; Langer, C.E.; Pekmezci, M.; Schwartzbaum, J.A.; Turner, M.C.; Walsh, K.M.; et al. The epidemiology of glioma in adults: A “state of the science” review. Neuro Oncol. 2014, 16, 896–913. [Google Scholar] [CrossRef] [Green Version]
  5. Stupp, R.; Hegi, M.; Mason, W.P.; Bent, M.V.D.; Taphoorn, M.J.B.; Janzer, R.C.; Ludwin, S.K.; Allgeier, A.; Fisher, B.; Belanger, K.; et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009, 10, 459–466. [Google Scholar] [CrossRef]
  6. Hegi, M.E.; Diserens, A.-C.; Gorlia, T.; Hamou, M.-F.; de Tribolet, N.; Weller, M.; Kros, J.M.; Hainfellner, J.A.; Mason, W.; Mariani, L.; et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N. Engl. J. Med. 2005, 352, 997–1003. [Google Scholar]
  7. Birzu, C.; French, P.; Caccese, M.; Cerretti, G.; Idbaih, A.; Zagonel, V.; Lombardi, G. Recurrent Glioblastoma: From Molecular Landscape to New Treatment Perspectives. Cancers 2020, 13, 47. [Google Scholar] [CrossRef]
  8. Herrlinger, U.; Rieger, J.; Koch, D.; Loeser, S.; Blaschke, B.; Kortmann, R.-D.; Steinbach, J.P.; Hunds-berger, T.; Wick, W.; Meyermann, R.; et al. Phase II trial of lomustine plus temozolomide chemotherapy in addition to radiotherapy in newly diagnosed glioblastoma: UKT-03. J. Clin. Oncol. 2006, 24, 4412–4417. [Google Scholar] [CrossRef] [PubMed]
  9. Herrlinger, U.; Tzaridis, T.; Mack, F.; Steinbach, J.P.; Schlegel, U.; Sabel, M.; Hau, P.; Kortmann, R.-D.; Krex, D.; Grauer, O.; et al. Lomustine-temozolomide combination therapy versus standard temozolomide therapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter (CeTeG/NOA-09): A randomised, open-label, phase 3 trial. Lancet 2019, 393, 678–688. [Google Scholar] [CrossRef]
  10. Glas, M.; Happold, C.; Rieger, J.; Wiewrodt, D.; Bähr, O.; Steinbach, J.P.; Wick, W.; Kortmann, R.-D.; Reifenberger, G.; Weller, M.; et al. Long-term survival of patients with glioblastoma treated with radiotherapy and lomustine plus temozolomide. J. Clin. Oncol. 2009, 27, 1257–1261. [Google Scholar] [CrossRef] [Green Version]
  11. Weller, M.; Le Rhun, E. How did lomustine become standard of care in recurrent glioblastoma? Cancer Treat. Rev. 2020, 87, 102029. [Google Scholar] [CrossRef]
  12. Cohen, M.H.; Shen, Y.; Li, K.; Patricia, P.R. FDA drug approval summary: Bevacizumab (Avastin) as treatment of recurrent glioblastoma multiforme. Oncologist 2009, 14, 1131–1138. [Google Scholar] [CrossRef] [PubMed]
  13. Friedman, H.S.; Prados, M.D.; Wen, P.Y.; Mikkelsen, T.; Schiff, D.; Abrey, L.E.; Yung, W.K.; Alfred, P.; Nina, N.; Martin, K.; et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J. Clin. Oncol. 2009, 27, 4733–4740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Vredenburgh, J.J.; Desjardins, A.; Herndon, J.E.; Marcello, J.; Reardon, D.A.; Quinn, J.A.; Rich, J.N.; Sathornsumetee, S.; Gururangan, S.; Sampson, J.; et al. Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J. Clin. Oncol. 2007, 25, 4722–4729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Chinot, O.L.; Wick, W.; Mason, W.; Henriksson, R.; Saran, F.; Nishikawa, R.; Carpentier, A.F.; Hoang-Xuan, K.; Kavan, P.; Cernea, D.; et al. Bevacizumab plus Radiotherapy–Temozolomide for Newly Diagnosed Glioblastoma. N. Engl. J. Med. 2014, 370, 709–722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Wick, W.; Gorlia, T.; Bendszus, M.; Taphoorn, M.; Sahm, F.; Harting, I.; Brandes, A.A.; Taal, W.; Domont, J.; Idbaih, A.; et al. Lomustine and Bevacizumab in Progressive Glioblastoma. N. Engl. J. Med. 2017, 377, 1954–1963. [Google Scholar] [CrossRef] [PubMed]
  17. Mehta, M.; Wen, P.; Nishikawa, R.; Reardon, D.; Peters, K. Critical review of the addition of tumor treating fields (TTFields) to the existing standard of care for newly diagnosed glioblastoma patients. Crit. Rev. Oncol. 2017, 111, 60–65. [Google Scholar] [CrossRef]
  18. Mun, E.J.; Babiker, H.M.; Weinberg, U.; Kirson, E.D.; Hoff, D.D.V. Tumor-Treating Fields: A Fourth Modality in Cancer Treatment. Clin. Cancer Res. 2018, 24, 266–275. [Google Scholar] [CrossRef] [Green Version]
  19. Kirson, E.D.; Gurvich, Z.; Schneiderman, R.; Dekel, E.; Itzhaki, A.; Wasserman, Y.; Schatzberger, R.; Palti, Y. Disruption of cancer cell replication by alternating electric fields. Cancer Res. 2004, 64, 3288–3295. [Google Scholar] [CrossRef] [Green Version]
  20. Stupp, R.; Taillibert, S.; Kanner, A.A.; Kesari, S.; Steinberg, D.M.; Toms, S.A.; Taylor, L.P.; Lieberman, F.; Silvani, A.; Fink, K.L.; et al. Maintenance Therapy With Tumor-Treating Fields Plus Temozolomide vs Temozolomide Alone for Glioblastoma: A Randomized Clinical Trial. JAMA 2015, 314, 2535–2543. [Google Scholar] [CrossRef]
  21. Stupp, R.; Stupp, R.; Taillibert, S.; Kanner, A.; Read, W.; Steinberg, D.; Lhermitte, B.; Toms, S.; Idbaih, A.; Ahluwalia, M.S.; et al. Effect of Tumor-Treating Fields Plus Maintenance Temozolomide vs Maintenance Temozolomide Alone on Survival in Patients with Glioblastoma: A Randomized Clinical Trial. JAMA 2017, 318, 2306–2316. [Google Scholar] [CrossRef] [Green Version]
  22. Fabian, D.; Eibl, M.D.P.G.P.; Alnahhas, I.; Sebastian, N.; Giglio, P.; Puduvalli, V.; Gonzalez, J.; Palmer, J.D. Treatment of Glioblastoma (GBM) with the Addition of Tumor-Treating Fields (TTF): A Review. Cancers 2019, 11, 174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Lassman, A.B.; Joanta-Gomez, A.e.; Pan, P.C.; Wick, W. Current usage of tumor treating fields for glioblastoma. Neurooncol. Adv. 2020, 2, vdaa069. [Google Scholar] [CrossRef] [PubMed]
  24. De Witt Hamer, P.C. Small molecule kinase inhibitors in glioblastoma: A systematic review of clinical studies. Neuro Oncol. 2010, 12, 304–316. [Google Scholar] [CrossRef] [PubMed]
  25. Touat, M.; Idbaih, A.; Sanson, M.; Ligon, K.L. Glioblastoma targeted therapy: Updated approaches from recent biological insights. Ann. Oncol. 2017, 28, 1457–1472. [Google Scholar] [CrossRef]
  26. Osuka, S.; Van Meir, E.G. Overcoming therapeutic resistance in glioblastoma: The way forward. J. Clin. Investig. 2017, 127, 415–426. [Google Scholar] [CrossRef] [Green Version]
  27. Dirkse, A.; Golebiewska, A.; Buder, T.; Nazarov, P.V.; Muller, A.; Poovathingal, S.K.; Brons, N.H.C.; Leite, S.; Sauvageot, N.; Sarkisjan, D.; et al. Stem cell-associated heterogeneity in Glioblastoma results from intrinsic tumor plasticity shaped by the microenvironment. Nat. Commun. 2019, 10, 1787. [Google Scholar] [CrossRef] [PubMed]
  28. Qazi, M.A.; Vora, P.; Venugopal, C.; Sidhu, S.S.; Moffat, J.; Swanton, C.; Singh, S.K. Intratumoral heterogeneity: Pathways to treatment resistance and relapse in human glioblastoma. Ann. Oncol. 2017, 28, 1448–1456. [Google Scholar] [CrossRef] [PubMed]
  29. Fabro, F.; Lamfers, M.L.M.; Leenstra, S. Advancements, Challenges, and Future Directions in Tackling Glioblastoma Resistance to Small Kinase Inhibitors. Cancers 2022, 14, 600. [Google Scholar] [CrossRef]
  30. Yabo, Y.A.; Niclou, S.P.; Golebiewska, A. Cancer cell heterogeneity and plasticity: A paradigm shift in glioblastoma. Neuro-Oncology 2022, 24, 669–682. [Google Scholar] [CrossRef] [PubMed]
  31. Jacob, F.; Salinas, R.D.; Zhang, D.Y.; Nguyen, P.T.T.; Schnoll, J.G.; Wong, S.Z.H.; Thokala, R.; Sheikh, S.; Saxena, D.; Prokop, S.; et al. A Patient-Derived Glioblastoma Organoid Model and Biobank Recapitulates Inter- and Intra-tumoral Heterogeneity. Cell 2020, 180, 188–204.e22. [Google Scholar] [CrossRef]
  32. Verhaak, R.G.; Hoadley, K.A.; Purdom, E.; Wang, V.; Qi, Y.; Wilkerson, M.D.; Miller, C.R.; Ding, L.; Golub, T.; Mesirov, J.P.; et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010, 17, 98–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Ceccarelli, M.; Barthel, F.P.; Malta, T.M.; Sabedot, T.S.; Salama, S.R.; Murray, B.A.; Morozova, O.; Newton, Y.; Radenbaugh, A.; Pagnotta, S.M.; et al. Molecular Profiling Reveals Biologically Discrete Subsets and Pathways of Progression in Diffuse Glioma. Cell 2016, 164, 550–563. [Google Scholar] [CrossRef] [Green Version]
  34. Wang, Q.; Hu, B.; Hu, X.; Kim, H.; Squatrito, M.; Scarpace, L.; deCarvalho, A.C.; Lyu, S.; Li, P.; Li, Y.; et al. Tumor Evolution of Glioma-Intrinsic Gene Expression Subtypes Associates with Immunological Changes in the Microenvironment. Cancer Cell 2017, 32, 42–56.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Patel, A.P.; Tirosh, I.; Trombetta, J.J.; Shalek, A.K.; Gillespie, S.M.; Wakimoto, H.; Cahill, D.P.; Nahed, B.V.; Curry, W.T.; Martuza, R.L.; et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 2014, 344, 1396–1401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Neftel, C.; Laffy, J.; Filbin, M.G.; Hara, T.; Shore, M.E.; Rahme, G.J.; Richman, A.R.; Silverbush, D.; Shaw, M.L.; Hebert, C.M.; et al. An Integrative Model of Cellular States, Plasticity, and Genetics for Glioblastoma. Cell 2019, 178, 835–849.e21. [Google Scholar] [CrossRef]
  37. Bhaduri, A.; Lullo, D.E.; Jung, D.; Müller, S.; Crouch, E.E.; Espinosa, C.S.; Ozawa, T.; Al-varado, B.; Spatazza, J.; Cadwell, C.R.; et al. Outer Radial Glia-like Cancer Stem Cells Contribute to Heterogeneity of Glioblastoma. Cell Stem Cell 2020, 26, 48–63.e6. [Google Scholar] [CrossRef]
  38. Weller, M.; Bent, M.V.D.; Preusser, M.; Rhun, L.E.; Tonn, J.C.; Minniti, G.; Bendszus, M.; Balana, C.; Chinot, O.; Dirven, L.; et al. EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood. Nat. Rev. Clin. Oncol. 2020, 18, 170–186. [Google Scholar] [CrossRef]
  39. Wager, T.T.; Hou, X.; Verhoest, P.R.; Villalobos, A. Moving beyond rules: The development of a central nervous system multiparameter optimization (CNS MPO) approach to enable alignment of druglike properties. ACS Chem. Neurosci. 2010, 1, 435–449. [Google Scholar] [CrossRef] [Green Version]
  40. Wager, T.T.; Hou, X.; Verhoest, P.R.; Villalobos, A. Central Nervous System Multiparameter Optimization Desirability: Application in Drug Discovery. ACS Chem. Neurosci. 2016, 7, 767–775. [Google Scholar] [CrossRef] [Green Version]
  41. Sanai, N. Phase 0 Clinical Trial Strategies for the Neurosurgical Oncologist. Neurosurgery 2019, 85, E967–E974. [Google Scholar] [CrossRef] [Green Version]
  42. Sarapa, N. Exploratory IND: A new regulatory strategy for early clinical drug development in the United States. Ernst Scher. Res. Found Workshop 2007, 151–163. [Google Scholar]
  43. Yamashita, S.; Sugiyama, Y. New strategy for drug development with exploratory IND studies: Scientific basis and future directions. Adv. Drug Deliv. Rev. 2011, 63, 493. [Google Scholar] [CrossRef] [PubMed]
  44. Bunevicius, A.; McDannold, N.J.; Golby, A.J. Focused Ultrasound Strategies for Brain Tumor Therapy. Oper. Neurosurg. 2020, 19, 9–18. [Google Scholar] [CrossRef] [PubMed]
  45. Rathi, S.; Griffith, J.I.; Zhang, W.; Zhang, W.; Oh, J.-H.; Talele, S.; Sarkaria, J.N.; Elmquist, W.F. The influence of the blood–brain barrier in the treatment of brain tumours. J. Intern. Med. 2022, 292, 3–30. [Google Scholar] [CrossRef]
  46. Juhairiyah, F.; de Lange, E.C.M. Understanding Drug Delivery to the Brain Using Liposome-Based Strategies: Studies that Provide Mechanistic Insights Are Essential. AAPS J. 2021, 23, 114. [Google Scholar] [CrossRef]
  47. Weaver, B.A. How Taxol/paclitaxel kills cancer cells. Mol. Biol. Cell 2014, 25, 2677–2681. [Google Scholar] [CrossRef]
  48. Markman, M.; Mekhail, T.M. Paclitaxel in cancer therapy. Expert Opin. Pharmacother. 2002, 3, 755–766. [Google Scholar] [CrossRef]
  49. Krauss, A.C.; Gao, X.; Li, L.; Manning, M.L.; Patel, P.; Fu, W.; Janoria, K.G.; Gieser, G.; Bateman, D.A.; Przepiorka, D.; et al. FDA Approval Summary: (Daunorubicin and Cytarabine) Liposome for Injection for the Treatment of Adults with High-Risk Acute Myeloid Leukemia. Clin. Cancer Res. 2019, 25, 2685–2690. [Google Scholar] [CrossRef]
  50. Dicko, A.; Kwak, S.; Frazier, A.A.; Mayer, L.D.; Liboiron, B.D. Biophysical characterization of a liposomal formulation of cytarabine and daunorubicin. Int. J. Pharm. 2010, 391, 248–259. [Google Scholar] [CrossRef]
  51. Wei, H.J.; Upadhyayula, P.S.; Pouliopoulos, A.N.; Englander, Z.K.; Zhang, X.; Jan, C.-I.; Guo, J.; Mela, A.-g.; Zhang, Z.; Wang, T.J.C.; et al. Focused Ultrasound-Mediated Blood-Brain Barrier Opening Increases Delivery and Efficacy of Etoposide for Glioblastoma Treatment. Int. J. Radiat. Oncol. Biol. Phys. 2021, 110, 539–550. [Google Scholar] [CrossRef]
  52. Dréan, A.; Lemaire, N.; Bouchoux, G.; Goldwirt, L.; Canney, M.; Goli, L.; Bouzidi, A.; Schmitt, C.; Guehennec, J.; Verreault, M.; et al. Temporary blood-brain barrier disruption by low intensity pulsed ultrasound increases carboplatin delivery and efficacy in preclinical models of glioblastoma. J. Neurooncol. 2019, 144, 33–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Idbaih, A.; Canney, M.; Belin, L.; Desseaux, C.; Vignot, A.; Bouchoux, G.; Asquier, N.; Law-Ye, B.; Leclercq, D.; Bissery, A.; et al. Safety and Feasibility of Repeated and Transient Blood-Brain Barrier Disruption by Pulsed Ultrasound in Patients with Recurrent Glioblastoma. Clin. Cancer Res. 2019, 25, 3793–3801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Xue, H.; Li, J.; Xie, H.; Wang, Y. Review of Drug Repositioning Approaches and Resources. Int. J. Biol. Sci. 2018, 14, 1232–1244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Pushpakom, S.; Pushpakom, S.; Iorio, F.; Eyers, P.A.; Escott, K.J.; Hopper, S.; Wells, A.; Doig, A.; Guilliams, T.; Latimer, J.; et al. Drug repurposing: Progress, challenges and recommendations. Nat. Rev. Drug Discov. 2019, 18, 41–58. [Google Scholar] [CrossRef] [PubMed]
  56. Abbruzzese, C.; Matteoni, S.; Signore, M.; Cardone, L.; Nath, K.; Glickson, J.D.; Paggi, M.G. Drug repurposing for the treatment of glioblastoma multiforme. J. Exp. Clin. Cancer Res. 2017, 36, 169. [Google Scholar] [CrossRef]
  57. Abbott, N.J.; Patabendige, A.; Dolman, D.E.M.; Yusof, S.R.; Begley, D.J. Structure and function of the blood-brain barrier. Neurobiol. Dis. 2010, 37, 13–25. [Google Scholar] [CrossRef] [PubMed]
  58. Cardoso, F.L.; Brites, D.; Brito, M.A. Looking at the blood-brain barrier: Molecular anatomy and possible investigation approaches. Brain Res. Rev. 2010, 64, 328–363. [Google Scholar] [CrossRef]
  59. Abbott, N.J.; Rönnbäck, L.; Hansson, E. Astrocyte-endothelial interactions at the blood-brain barrier. Nat. Rev. Neurosci. 2006, 7, 41–53. [Google Scholar] [CrossRef]
  60. Kadry, H.; Noorani, B.; Cucullo, L. A blood–brain barrier overview on structure, function, impairment, and biomarkers of integrity. Fluids Barriers CNS 2020, 17, 69. [Google Scholar] [CrossRef]
  61. Theodorakis, P.E.; Müller, E.A.; Craster, R.V.; Matar, O.K. Physical insights into the blood-brain barrier translocation mechanisms. Phys. Biol. 2017, 14, 041001. [Google Scholar] [CrossRef] [Green Version]
  62. Wong, A.D.; Ye, M.; Levy, A.F.; Rothstein, J.D.; Bergles, D.E.; Searson, P.C. The blood-brain barrier: An engineering perspective. Front. Neuroeng. 2013, 6, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Kim, M.; Kizilbash, S.H.; Laramy, J.K.; Gampa, G.; Parrish, K.E.; Sarkaria, J.N.; Elmquist, W.F. Barriers to Effective Drug Treatment for Brain Metastases: A Multifactorial Problem in the Delivery of Precision Medicine. Pharm. Res. 2018, 35, 1–20. [Google Scholar] [CrossRef] [PubMed]
  64. Nduom, E.; Yang, C.; Merrill, M.J.; Zhuang, Z.; Lonser, R.R. Characterization of the blood-brain barrier of metastatic and primary malignant neoplasms. J. Neurosurg. 2013, 119, 427–433. [Google Scholar] [CrossRef] [PubMed]
  65. Brat, D.J.; Van Meir, E.G. Vaso-occlusive and prothrombotic mechanisms associated with tumor hypoxia, necrosis, and accelerated growth in glioblastoma. Lab. Investig. 2004, 84, 397–405. [Google Scholar] [CrossRef] [PubMed]
  66. Zhao, C.; Wang, H.; Xiong, C.; Liu, Y. Hypoxic glioblastoma release exosomal VEGF-A induce the permeability of blood-brain barrier. Biochem. Biophys. Res. Commun. 2018, 502, 324–331. [Google Scholar] [CrossRef]
  67. Saharinen, P.; Eklund, L.; Alitalo, K. Therapeutic targeting of the angiopoietin–TIE pathway. Nat. Rev. Drug Discov. 2017, 16, 635–661. [Google Scholar] [CrossRef]
  68. Thurston, G.; Daly, C. The Complex Role of Angiopoietin-2 in the Angiopoietin-Tie Signaling Pathway. Cold Spring Harb. Perspect. Med. 2012, 2, a006650. [Google Scholar] [CrossRef] [Green Version]
  69. de Vries, H.E.; Blom-Roosemalen, M.C.; van Oosten, M.; de Boer, A.G.; van Berkel, T.J.; Breimer, D.D.; Kuiper, J. The influence of cytokines on the integrity of the blood-brain barrier in vitro. J. Neuroimmunol. 1996, 64, 37–43. [Google Scholar] [CrossRef]
  70. Schneider, S.W.; Ludwig, T.; Tatenhorst, L.; Braune, S.; Oberleithner, H.; Senner, V.; Paulus, W. Glioblastoma cells release factors that disrupt blood-brain barrier features. Acta Neuropathol. 2004, 107, 272–276. [Google Scholar] [CrossRef]
  71. Martin, T.A.; Jiang, W.G. Loss of tight junction barrier function and its role in cancer metastasis. Biochim. et Biophys. Acta (BBA)-Biomembr. 2009, 1788, 872–891. [Google Scholar] [CrossRef] [Green Version]
  72. Ishihara, H.; Kubota, H.; Lindberg, R.L.; Leppert, D.; Gloor, S.M.; Errede, M.; Virgintino, D.; Fontana, A.; Yonekawa, Y.; Frei, K. Endothelial Cell Barrier Impairment Induced by Glioblastomas and Transforming Growth Factor β2Involves Matrix Metalloproteinases and Tight Junction Proteins. J. Neuropathol. Exp. Neurol. 2008, 67, 435–448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Mader, S.; Brimberg, L. Aquaporin-4 Water Channel in the Brain and Its Implication for Health and Disease. Cells 2019, 8, 90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Arvanitis, C.D.; Ferraro, G.B.; Jain, R.K. The blood–brain barrier and blood–tumour barrier in brain tumours and metastases. Nat. Cancer 2019, 20, 26–41. [Google Scholar] [CrossRef] [PubMed]
  75. Sarkaria, J.N.; Hu, L.S.; Parney, I.F.; Pafundi, D.H.; Brinkmann, D.H.; Laack, N.N.; Giannini, C.; Burns, T.C.; Kizilbash, S.; Laramy, J.K.; et al. Is the blood–brain barrier really disrupted in all glioblastomas? A critical assessment of existing clinical data. Neuro-Oncology 2017, 20, 184–191. [Google Scholar] [CrossRef] [PubMed]
  76. Pacioni, S.; D’Alessandris, Q.G.; Buccarelli, M.; Boe, A.; Martini, M.; Larocca, L.M.; Bolasco, G.; Ricci-Vitiani, L.; Falchetti, M.L.; Pallini, R. Brain Invasion along Perivascular Spaces by Glioma Cells: Relationship with Blood–Brain Barrier. Cancers 2019, 12, 18. [Google Scholar] [CrossRef] [Green Version]
  77. Watkins, S.; Robel, S.; Kimbrough, I.F.; Robert, S.M.; Ellisdavies, G.C.R.; Sontheimer, H. Disruption of astrocyte–vascular coupling and the blood–brain barrier by invading glioma cells. Nat. Commun. 2014, 5, 1–15. [Google Scholar] [CrossRef] [Green Version]
  78. de Gooijer, M.C.; Kemper, E.M.; Buil, L.C.M.; Citirikkaya, C.H.; Buckle, T.; Beijnen, J.H.; van Tellingen, O. ATP-binding cassette transporters restrict drug delivery and efficacy against brain tumors even when blood-brain barrier integrity is lost. Cell Rep. Med. 2021, 2, 100184. [Google Scholar] [CrossRef]
  79. Robey, R.W.; Pluchino, K.M.; Hall, M.D.; Fojo, A.T.; Bates, S.E.; Gottesman, M.M. Revisiting the role of ABC transporters in multidrug-resistant cancer. Nat. Rev. Cancer 2018, 18, 452–464. [Google Scholar] [CrossRef]
  80. Hollenstein, K.; Dawson, R.; Locher, K.P. Structure and mechanism of ABC transporter proteins. Curr. Opin. Struct. Biol. 2007, 17, 412–418. [Google Scholar] [CrossRef]
  81. Dean, M. The Genetics of ATP-Binding Cassette Transporters. Methods Enzymol. 2005, 400, 409–429. [Google Scholar] [CrossRef]
  82. Uitto, J. The gene family of ABC transporters—Novel mutations, new phenotypes. Trends Mol. Med. 2005, 11, 341–343. [Google Scholar] [CrossRef] [PubMed]
  83. Kadioglu, O.; Saeed, M.E.M.; Munder, M.; Spuller, A.; Greten, H.J.; Efferth, T. Effect of ABC transporter expression and mutational status on survival rates of cancer patients. Biomed. Pharmacother. 2020, 131, 110718. [Google Scholar] [CrossRef] [PubMed]
  84. Fletcher, J.I.; Haber, M.; Henderson, M.J.; Norris, M.D. ABC transporters in cancer: More than just drug efflux pumps. Nat. Cancer 2010, 10, 147–156. [Google Scholar] [CrossRef] [PubMed]
  85. Gottesman, M.M.; Fojo, T.; Bates, S.E. Multidrug resistance in cancer: Role of ATP–dependent transporters. Nat. Cancer 2002, 2, 48–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Choi, C.-H. ABC transporters as multidrug resistance mechanisms and the development of chemosensitizers for their reversal. Cancer Cell Int. 2005, 5, 30. [Google Scholar] [CrossRef] [Green Version]
  87. Choi, Y.H.; Yu, A.-M. ABC transporters in multidrug resistance and pharmacokinetics, and strategies for drug development. Curr. Pharm. Des. 2014, 20, 793–807. [Google Scholar] [CrossRef]
  88. Ambudkar, S.V.; Kim, I.-W.; Sauna, Z.E. The power of the pump: Mechanisms of action of P-glycoprotein (ABCB1). Eur. J. Pharm. Sci. 2006, 27, 392–400. [Google Scholar] [CrossRef]
  89. Seelig, A. P-Glycoprotein: One Mechanism, Many Tasks and the Consequences for Pharmacotherapy of Cancers. Front. Oncol. 2020, 10, 576559. [Google Scholar] [CrossRef]
  90. van Tellingen, O.; Yetkin-Arik, B.; de Gooijer, M.; Wesseling, P.; Wurdinger, T.; de Vries, H. Overcoming the blood–brain tumor barrier for effective glioblastoma treatment. Drug Resist. Updates 2015, 19, 1–12. [Google Scholar] [CrossRef]
  91. Mason, W.P. Blood-brain barrier-associated efflux transporters: A significant but underappreciated obstacle to drug development in glioblastoma. Neuro-Oncology 2015, 17, 1181–1182. [Google Scholar] [CrossRef] [Green Version]
  92. Dréan, A.; Rosenberg, S.; Lejeune, F.-X.; Goli, L.; Nadaradjane, A.A.; Guehennec, J.; Schmitt, C.; Verreault, M.; Bielle, F.; Mokhtari, K.; et al. ATP binding cassette (ABC) transporters: Expression and clinical value in glioblastoma. J. Neuro-Oncol. 2018, 138, 479–486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Bronger, H.; König, J.; Kopplow, K.; Steiner, H.-H.; Ahmadi, R.; Herold-Mende, C.; Keppler, D.; Nies, A.T. ABCC Drug Efflux Pumps and Organic Anion Uptake Transporters in Human Gliomas and the Blood-Tumor Barrier. Cancer Res. 2005, 65, 11419–11428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Durmus, S.; Hendrikx, J.J.; Schinkel, A.H. Apical ABC Transporters and Cancer Chemotherapeutic Drug Disposition. Adv. Cancer Res. 2015, 125, 1–41. [Google Scholar] [CrossRef]
  95. Bao, X.; Wu, J.; Xie, Y.; Kim, S.; Michelhaugh, S.; Jiang, J.; Mittal, S.; Sanai, N.; Li, J. Protein Expression and Functional Relevance of Efflux and Uptake Drug Transporters at the Blood–Brain Barrier of Human Brain and Glioblastoma. Clin. Pharmacol. Ther. 2020, 107, 1116–1127. [Google Scholar] [CrossRef] [PubMed]
  96. de Trizio, I.; Errede, M.; d’Amati, A.; Girolamo, F.; Virgintino, D. Expression of P-gp in Glioblastoma: What we can Learn from Brain Development. Curr. Pharm. Des. 2020, 26, 1428–1437. [Google Scholar] [CrossRef] [PubMed]
  97. Roy, L.-O.; Lemelin, M.; Poirier, M.-B.; Blanchette, M.; Fortin, D. SCDT-18. EXPRESSION OF ABC TRANSPORTERS AS PROGNOSTIC BIOMARKERS FOR GLIOBLASTOMA. Neuro-Oncology 2017, 19, vi268. [Google Scholar] [CrossRef]
  98. Parrish, K.E.; Sarkaria, J.N.; Elmquist, W.F. Improving drug delivery to primary and metastatic brain tumors: Strategies to overcome the blood-brain barrier. Clin. Pharmacol. Ther. 2015, 97, 336–346. [Google Scholar] [CrossRef]
  99. Arvanitis, C.D.; Askoxylakis, V.; Guo, Y.; Datta, M.; Kloepper, J.; Ferraro, G.B.; Bernabeu, M.O.; Fukumura, D.; McDannold, N.; Jain, R.K. Mechanisms of enhanced drug delivery in brain metastases with focused ultrasound-induced blood–tumor barrier disruption. Proc. Natl. Acad. Sci. USA 2018, 115, E8717–E8726. [Google Scholar] [CrossRef] [Green Version]
  100. Oberoi, R.K.; Parrish, K.E.; Sio, T.T.; Mittapalli, R.K.; Elmquist, W.F.; Sarkaria, J.N. Strategies to improve delivery of anticancer drugs across the blood–brain barrier to treat glioblastoma. Neuro-Oncology 2015, 18, 27–36. [Google Scholar] [CrossRef] [Green Version]
  101. Sottoriva, A.; Spiteri, I.; Piccirillo, S.G.M.; Touloumis, A.; Collins, V.P.; Marioni, J.C.; Curtis, C.; Watts, C.; Tavaré, S. Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc. Natl. Acad. Sci. USA 2013, 110, 4009–4014. [Google Scholar] [CrossRef] [Green Version]
  102. Stieber, D.; Golebiewska, A.; Evers, L.; Lenkiewicz, E.; Brons, N.H.C.; Nicot, N.; Oudin, A.; Bougnaud, S.; Hertel, F.; Bjerkvig, R.; et al. Glioblastomas are composed of genetically divergent clones with distinct tumourigenic potential and variable stem cell-associated phenotypes. Acta Neuropathol. 2013, 127, 203–219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Lenin, S.; Ponthier, E.; Scheer, K.; Yeo, E.; Tea, M.; Ebert, L.; Mansilla, M.O.; Poonnoose, S.; Baumgartner, U.; Day, B.; et al. A Drug Screening Pipeline Using 2D and 3D Patient-Derived In Vitro Models for Pre-Clinical Analysis of Therapy Response in Glioblastoma. Int. J. Mol. Sci. 2021, 22, 4322. [Google Scholar] [CrossRef] [PubMed]
  104. Johansson, P.; Krona, C.; Kundu, S.; Doroszko, M.; Baskaran, S.; Schmidt, L.; Vinel, C.; Almstedt, E.; Elgendy, R.; Elfineh, L.; et al. A Patient-Derived Cell Atlas Informs Precision Targeting of Glioblastoma. Cell Rep. 2020, 32, 107897. [Google Scholar] [CrossRef] [PubMed]
  105. Skaga, E.; Kulesskiy, E.; Brynjulvsen, M.; Sandberg, C.J.; Potdar, S.; Langmoen, I.A.; Laakso, A.; Gaál-Paavola, E.; Perola, M.; Wennerberg, K.; et al. Feasibility study of using high-throughput drug sensitivity testing to target recurrent glioblastoma stem cells for individualized treatment. Clin. Transl. Med. 2019, 8, 33. [Google Scholar] [CrossRef] [Green Version]
  106. Jansen, M.; Yip, S.; Louis, D.N. Molecular pathology in adult gliomas: Diagnostic, prognostic, and predictive markers. Lancet Neurol. 2010, 9, 717–726. [Google Scholar] [CrossRef] [Green Version]
  107. Śledzińska, P.; Bebyn, M.G.; Furtak, J.; Kowalewski, J.; Lewandowska, M.A. Prognostic and Predictive Biomarkers in Gliomas. Int. J. Mol. Sci. 2021, 22, 10373. [Google Scholar] [CrossRef]
  108. Lynes, J.P.; Nwankwo, A.K.; Sur, H.P.; Sanchez, V.E.; Sarpong, K.A.; Ariyo, O.I.; Dominah, G.A.; Nduom, E.K. Biomarkers for immunotherapy for treatment of glioblastoma. J. Immunother. Cancer 2020, 8, e000348. [Google Scholar] [CrossRef]
  109. Bark, J.M.; Kulasinghe, A.; Chua, B.; Day, B.W.; Punyadeera, C. Circulating biomarkers in patients with glioblastoma. Br. J. Cancer 2019, 122, 295–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Stavrakaki, E.; Dirven, C.; Lamfers, M. Personalizing Oncolytic Virotherapy for Glioblastoma: In Search of Biomarkers for Response. Cancers 2021, 13, 614. [Google Scholar] [CrossRef]
  111. Da Silva, E.C.; Mercier, M.-C.; Etienne-Selloum, N.; Dontenwill, M.; Choulier, L. A Systematic Review of Glioblastoma-Targeted Therapies in Phases II, III, IV Clinical Trials. Cancers 2021, 13, 1795. [Google Scholar] [CrossRef]
  112. Noch, E.K.; Ramakrishna, R.; Magge, R. Challenges in the Treatment of Glioblastoma: Multisystem Mechanisms of Therapeutic Resistance. World Neurosurg. 2018, 116, 505–517. [Google Scholar] [CrossRef] [PubMed]
  113. Corsello, S.; Bittker, J.A.; Liu, Z.; Gould, J.; McCarren, P.; Hirschman, J.E.; Johnston, S.E.; Vrcic, A.; Wong, B.; Khan, M.; et al. The Drug Repurposing Hub: A next-generation drug library and information resource. Nat. Med. 2017, 23, 405–408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Jain, K.K. A Critical Overview of Targeted Therapies for Glioblastoma. Front. Oncol. 2018, 8, 419. [Google Scholar] [CrossRef]
  115. Chong, C.R.; Sullivan, D.J., Jr. New uses for old drugs. Nature 2007, 448, 645–646. [Google Scholar] [CrossRef] [PubMed]
  116. Sandler, R.S.; Halabi, S.; Baron, J.A.; Budinger, S.; Paskett, E.; Keresztes, R.; Petrelli, N.; Pipas, J.M.; Karp, D.D.; Loprinzi, C.L.; et al. A Randomized Trial of Aspirin to Prevent Colorectal Adenomas in Patients with Previous Colorectal Cancer. N. Engl. J. Med. 2003, 348, 883–890. [Google Scholar] [CrossRef]
  117. Reimers, M.S.; Bastiaannet, E.; van Herk-Sukel, M.; Lemmens, V.E.P.; Broek, C.B.M.V.D.; Van De Velde, C.J.H.; De Craen, A.J.M.; Liefers, G.J. Aspirin Use After Diagnosis Improves Survival in Older Adults with Colon Cancer: A Retrospective Cohort Study. J. Am. Geriatr. Soc. 2012, 60, 2232–2236. [Google Scholar] [CrossRef]
  118. Lippman, M.E.; Krueger, K.A.; Eckert, S.; Sashegyi, A.; Walls, E.L.; Jamal, S.; Cauley, J.A.; Cummings, S.R. Indicators of Lifetime Estrogen Exposure: Effect on Breast Cancer Incidence and Interaction with Raloxifene Therapy in the Multiple Outcomes of Raloxifene Evaluation Study Participants. J. Clin. Oncol. 2001, 19, 3111–3116. [Google Scholar] [CrossRef]
  119. Dunn, B.K.; Ford, L.G. From Adjuvant Therapy to Breast Cancer Prevention: BCPT and STAR. Breast J. 2001, 7, 144–157. [Google Scholar] [CrossRef]
  120. Singhal, S.; Mehta, J.; Desikan, R.; Ayers, D.; Roberson, P.; Eddlemon, P.; Munshi, N.; Anaissie, E.; Wilson, C.; Dhodapkar, M.; et al. Antitumor Activity of Thalidomide in Refractory Multiple Myeloma. N. Engl. J. Med. 1999, 341, 1565–1571. [Google Scholar] [CrossRef] [Green Version]
  121. Van Der Velden, D.L.; Hoes, L.R.; Van Der Wijngaart, H.; van Berge Henegouwen, J.M.; Van Werkhoven, E.; Roepman, P.; Schilsky, R.L.; De Leng, W.W.J.; Huitema, A.D.R.; Nuijen, B.; et al. The Drug Rediscovery protocol facilitates the expanded use of existing anticancer drugs. Nature 2019, 574, 127–131. [Google Scholar] [CrossRef]
  122. Hainsworth, J.D.; Meric-Bernstam, F.; Swanton, C.; Hurwitz, H.; Spigel, D.R.; Sweeney, C.; Burris, H.A.; Bose, R.; Yoo, B.; Stein, A.; et al. Targeted Therapy for Advanced Solid Tumors on the Basis of Molecular Profiles: Results from MyPathway, an Open-Label, Phase IIa Multiple Basket Study. J. Clin. Oncol. 2018, 36, 536–542. [Google Scholar] [CrossRef] [PubMed]
  123. Kaley, T.; Touat, M.; Subbiah, V.; Hollebecque, A.; Rodon, J.; Lockhart , A.C.; Keedy, V.; Bielle, F.; Hofheinz, R.-D.; Joly, F.; et al. BRAF Inhibition in BRAF(V600)-Mutant Gliomas: Results From the VE-BASKET Study. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2018, 36, 3477–3484. [Google Scholar] [CrossRef] [PubMed]
  124. Bouchè, V.; Aldegheri, G.; Antonio Donofrio, C.; Fioravanti , A.; Roberts-Thomson, S.; Fox, S.B.; Schettini, F.; Generali, D. BRAF Signaling Inhibition in Glioblastoma: Which Clinical Perspectives? Front. Oncol. 2021, 11, 772052. [Google Scholar] [CrossRef] [PubMed]
  125. Wen, P.Y.; Stein, A.; van den Bent, M.; De Greve, J.; Wick, A.; de Vos, F.Y.F.L.; von Bubnoff, N.; van Linde, M.E.; Lai, A.; Prager, G.W.; et al. Dabrafenib plus trametinib in patients with BRAF(V600E)-mutant low-grade and high-grade glioma (ROAR): A multicentre, open-label, single-arm, phase 2, basket trial. Lancet Oncol. 2022, 23, 53–64. [Google Scholar] [CrossRef]
  126. Robert, C.; Karaszewska, B.; Schachter, J.; Rutkowski, P.; Mackiewicz, A.; Stroiakovski, D.; Lichinitser, M.; Dummer, R.; Grange, F.; Mortier, L.; et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N. Engl. J. Med. 2015, 372, 30–39. [Google Scholar] [CrossRef] [Green Version]
  127. Long, G.V.; Hauschild, A.; Santinami, M.; Atkinson, V.; Mandalá, G.V.; Chiarion-Sileni, V.; Larkin, J.; Nyakas, M.; Dutriaux, C.; Haydon, A.; et al. Adjuvant Dabrafenib plus Trametinib in Stage III BRAF-Mutated Melanoma. N. Engl. J. Med. 2017, 377, 1813–1823. [Google Scholar] [CrossRef] [Green Version]
  128. Liu, L.; Gerson, S.L. Targeted Modulation of MGMT: Clinical Implications. Clin. Cancer Res. 2006, 12, 328–331. [Google Scholar] [CrossRef] [Green Version]
  129. Sarkaria, J.N.; Kitange, G.J.; James, C.D.; Plummer, R.; Calvert, H.; Weller, M.; Wick, W. Mechanisms of Chemoresistance to Alkylating Agents in Malignant Glioma. Clin. Cancer Res. 2008, 14, 2900–2908. [Google Scholar] [CrossRef] [Green Version]
  130. Paranjpe, A.; Zhang, R.; Ali-Osman, F.; Bobustuc, G.C.; Srivenugopal, K.S. Disulfiram is a direct and potent inhibitor of human O6-methylguanine-DNA methyltransferase (MGMT) in brain tumor cells and mouse brain and markedly increases the alkylating DNA damage. Carcinogenesis 2013, 35, 692–702. [Google Scholar] [CrossRef]
  131. Jørgensen, C.H.; Pedersen, B.; Tønnesen, H. The Efficacy of Disulfiram for the Treatment of Alcohol Use Disorder. Alcohol. Clin. Exp. Res. 2011, 35, 1749–1758. [Google Scholar] [CrossRef]
  132. Wu, S.; Li, X.; Gao, F.; de Groot, J.F.; Koul, D.; Yung, W.K.A. PARP-mediated PARylation of MGMT is critical to promote repair of temozolomide-induced O6-methylguanine DNA damage in glioblastoma. Neuro-Oncology 2021, 23, 920–931. [Google Scholar] [CrossRef] [PubMed]
  133. Balvers, R.K.; Kleijn, A.; Kloezeman, J.J.; French, P.J.; Kremer, A.; van den Bent, M.J.; Dirven, C.M.F.; Leenstra, S.; Lamfers, M.L.M. Serum-free culture success of glial tumors is related to specific molecular profiles and expression of extracellular matrix–associated gene modules. Neuro-Oncology 2013, 15, 1684–1695. [Google Scholar] [CrossRef] [Green Version]
  134. Lee, J.; Kotliarova, S.; Kotliarov, Y.; Li, A.; Su, Q.; Donin, N.M.; Pastorino, S.; Purow, B.W.; Christopher, N.; Zhang, W.; et al. Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell 2006, 9, 391–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Kleijn, A.; Kloezeman, J.J.; Balvers, R.K.; van der Kaaij, M.; Dirven, C.M.F.; Leenstra, S.; Lamfers, M.L.M. A Systematic Comparison Identifies an ATP-Based Viability Assay as Most Suitable Read-Out for Drug Screening in Glioma Stem-Like Cells. Stem Cells Int. 2016, 2016, 5623235. [Google Scholar] [CrossRef] [Green Version]
  136. Joseph, J.V.; Blaavand, M.S.; Daubon, T.; Kruyt, F.A.; Thomsen, M.K. Three-dimensional culture models to study glioblastoma—Current trends and future perspectives. Curr. Opin. Pharmacol. 2021, 61, 91–97. [Google Scholar] [CrossRef]
  137. Golebiewska, A.; Hau, A.-C.; Oudin, A.; Stieber, D.; Yabo, Y.A.; Baus, V.; Barthelemy, V.; Klein, E.; Bougnaud, S.; Keunen, O.; et al. Patient-derived organoids and orthotopic xenografts of primary and recurrent gliomas represent relevant patient avatars for precision oncology. Acta Neuropathol. 2020, 140, 919–949. [Google Scholar] [CrossRef]
  138. Huszthy, P.C.; Daphu, I.; Niclou, S.P.; Stieber, D.; Nigro, J.M.; Sakariassen, P.; Miletic, H.; Thorsen, F.; Bjerkvig, R. In vivo models of primary brain tumors: Pitfalls and perspectives. Neuro-Oncology 2012, 14, 979–993. [Google Scholar] [CrossRef] [Green Version]
  139. Klein, E.; Hau, A.-C.; Oudin, A.; Golebiewska, A.; Niclou, S.P. Glioblastoma Organoids: Pre-Clinical Applications and Challenges in the Context of Immunotherapy. Front. Oncol. 2020, 10. [Google Scholar] [CrossRef] [PubMed]
  140. Kerstetter-Fogle, A.E.; Harris, P.L.R.; Brady-Kalnay, S.M.; Sloan, A.E. Generation of Glioblastoma Patient-Derived Intracranial Xenografts for Preclinical Studies. Int. J. Mol. Sci. 2020, 21, 5113. [Google Scholar] [CrossRef]
  141. Howard, C.; Valluri, J.; Alberico, A.; Julien, T.; Mazagri, R.; Marsh, R.; Alastair, H.; Cortese, A.; Griswold, M.; Wang, W.; et al. Analysis of Chemopredictive Assay for Targeting Cancer Stem Cells in Glioblastoma Patients. Transl. Oncol. 2017, 10, 241–254. [Google Scholar] [CrossRef]
  142. Stockslager, M.A.; Malinowski, S.; Touat, M.; Yoon, J.C.; Geduldig, J.; Mirza, M.; Kim, A.S.; Wen, P.Y.; Chow, K.-H.; Ligon, K.L.; et al. Functional drug susceptibility testing using single-cell mass predicts treatment outcome in patient-derived cancer neurosphere models. Cell Rep. 2021, 37. [Google Scholar] [CrossRef] [PubMed]
  143. Shuford, S.; Lipinski, L.; Abad, A.; Smith, A.M.; Rayner, M.; O’Donnell, L.; Stuart, J.; Mechtler, L.L.; Fabiano, A.J.; Edenfield, J.; et al. Prospective prediction of clinical drug response in high-grade gliomas using an ex vivo 3D cell culture assay. Neuro-Oncol. Adv. 2021, 3, vdab065. [Google Scholar] [CrossRef] [PubMed]
  144. Narayan, R.S.; Molenaar, P.; Teng, J.; Cornelissen, F.M.G.; Roelofs, I.; Menezes, R.; Dik, R.; Lagerweij, T.; Broersma, Y.; Petersen, N.; et al. A cancer drug atlas enables synergistic targeting of independent drug vulnerabilities. Nat. Commun. 2020, 11, 1–14. [Google Scholar] [CrossRef]
  145. Taylor, J.T.; Ellison, S.; Pandele, A.; Wood, S.; Nathan, E.; Forte, G.; Parker, H.; Zindy, E.; Elvin, M.; Dickson, A.; et al. Actinomycin D downregulates Sox2 and improves survival in preclinical models of recurrent glioblastoma. Neuro-Oncology 2020, 22, 1289–1301. [Google Scholar] [CrossRef]
  146. Verheul, C.; Ntafoulis, I.; Kers, T.V.; Hoogstrate, Y.; Mastroberardino, P.G.; Barnhoorn, S.; Payán-Gómez, C.; Yen, R.T.C.; Struys, E.A.; Koolen, S.L.W.; et al. Generation, characterization, and drug sensitivities of 12 patient-derived IDH1-mutant glioma cell cultures. Neuro-Oncol. Adv. 2021, 3. [Google Scholar] [CrossRef]
  147. Finocchiaro, G. Actinomycin D: A new opening for an old drug. Neuro-Oncology 2020, 22. [Google Scholar] [CrossRef] [PubMed]
  148. Dao Trong, P.; Jungwirth, G.; Yu, T.; Pusch, S.; Unterberg, A.; Herold-Mende, C.; Warta, R. Large-Scale Drug Screening in Patient-Derived IDH(mut) Glioma Stem Cells Identifies Several Efficient Drugs among FDA-Approved Antineoplastic Agents. Cells 2020, 9, 1389. [Google Scholar] [CrossRef]
  149. Feun, L.G.; Savaraj, N.; Landy, H.; Levin, H.; Lampidis, T. Phase II study of homoharringtonine in patients with recurrent primary malignant central nervous system tumors. J. Neuro-Oncol. 1990, 9, 159–163. [Google Scholar] [CrossRef]
  150. Tujebajeva, R.; Graifer, D.; Karpova, G.; Ajtkhozhina, N. Alkaloid homoharringtonine inhibits polypeptide chain elongation on human ribosomes on the step of peptide bond formation. FEBS Lett. 1989, 257, 254–256. [Google Scholar] [CrossRef] [Green Version]
  151. Grohar, P.J.; Woldemichael, G.M.; Griffin, L.B.; Mendoza, A.; Chen, Q.-R.; Yeung, C.; Currier, D.; Davis, S.; Khanna, C.; Khan, J.; et al. Identification of an Inhibitor of the EWS-FLI1 Oncogenic Transcription Factor by High-Throughput Screening. JNCI J. Natl. Cancer Inst. 2011, 103, 962–978. [Google Scholar] [CrossRef] [Green Version]
  152. Berberich, A.; Kessler, T.; Thomé, C.M.; Pusch, S.; Hielscher, T.; Sahm, F.; Oezen, I.; Schmitt, L.-M.; Ciprut, S.; Hucke, N.; et al. Targeting Resistance against the MDM2 Inhibitor RG7388 in Glioblastoma Cells by the MEK Inhibitor Trametinib. Clin. Cancer Res. 2019, 25, 253–265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Schreck, K.C.; Allen, A.N.; Wang, J.; Pratilas, C.A. Combination MEK and mTOR inhibitor therapy is active in models of glioblastoma. Neurooncol. Adv. 2020, 2, vdaa138. [Google Scholar] [CrossRef] [PubMed]
  154. Bi, J.; Khan, A.; Tang, J.; Armando, A.M.; Wu, S.; Zhang, W.; Gimple, R.C.; Reed, A.; Jing, H.; Koga, T.; et al. Targeting glioblastoma signaling and metabolism with a re-purposed brain-penetrant drug. Cell Rep. 2021, 37. [Google Scholar] [CrossRef] [PubMed]
  155. Otto-Meyer, S.; DeFaccio, R.; Dussold, C.; Ladomersky, E.; Zhai, L.; Lauing, K.L.; Bollu, L.R.; Amidei, C.; Lukas, R.V.; Scholtens, D.M.; et al. A retrospective survival analysis of Glioblastoma patients treated with selective serotonin reuptake inhibitors. Brain Behav. Immun. Health 2020, 2, 100025. [Google Scholar] [CrossRef]
  156. Liu, K.-H.; Yang, S.-T.; Lin, Y.-K.; Lin, J.-W.; Lee, Y.-H.; Wang, J.-Y.; Hu, C.-J.; Lin, E.-Y.; Chen, S.-M.; Then, C.-K.; et al. Fluoxetine, an antidepressant, suppresses glioblastoma by evoking AMPAR-mediated calcium-dependent apoptosis. Oncotarget 2014, 6, 5088–5101. [Google Scholar] [CrossRef] [Green Version]
  157. Hsu, L.-C.; Tu, H.-F.; Hsu, F.-T.; Yueh, P.-F.; Chiang, I.-T. Beneficial effect of fluoxetine on anti-tumor progression on hepatocellular carcinoma and non-small cell lung cancer bearing animal model. Biomed. Pharmacother. 2020, 126, 110054. [Google Scholar] [CrossRef]
  158. Abadi, B.; Shahsavani, Y.; Faramarzpour, M.; Rezaei, N.; Rahimi, H. Antidepressants with anti-tumor potential in treating glioblastoma: A narrative review. Fundam. Clin. Pharmacol. 2021, 36, 35–48. [Google Scholar] [CrossRef]
  159. Ma, J.; Yang, Y.-R.; Chen, W.; Chen, M.-H.; Wang, H.; Wang, X.-D.; Sun, L.-L.; Wang, F.-Z.; Wang, D.-C. Fluoxetine synergizes with temozolomide to induce the CHOP-dependent endoplasmic reticulum stress-related apoptosis pathway in glioma cells. Oncol. Rep. 2016, 36, 676–684. [Google Scholar] [CrossRef] [Green Version]
  160. Rendon, L.F.; Tewarie, I.A.; Cote, D.J.; Gabriel, A.; Smith, T.R.; Broekman, M.L.D.; Mekary, R.A. Statins and Gliomas: A Systematic Review of the Preclinical Studies and Meta-Analysis of the Clinical Literature. Drugs 2022, 82, 293–310. [Google Scholar] [CrossRef]
  161. Mazurek, M.; Litak, J.; Kamieniak, P.; Kulesza, B.; Jonak, K.; Baj, J.; Grochowski, C. Metformin as Potential Therapy for High-Grade Glioma. Cancers 2020, 12, 210. [Google Scholar] [CrossRef] [Green Version]
  162. Würth, R.; Pattarozzi, A.; Gatti, M.; Bajetto, A.; Corsaro, A.; Parodi, A.; Sirito, R.; Massollo, M.; Marini, C.; Zona, G.; et al. Metformin selectively affects human glioblastoma tumor-initiating cell viability: A role for metformin-induced inhibition of Akt. Cell Cycle 2013, 12, 145–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Azar, K.; Kannappan, V.; Liu, Y.; Butcher, K.; Morris, M.; Armesilla, A.; Darling, J.; Wang, W. Disulfiram targets glioblastoma-stem-like cells in vitro and in vivo. Neuro-Oncology 2018, 20, i20. [Google Scholar] [CrossRef] [Green Version]
  164. Gallia, G.L.; Holdhoff, M.; Brem, H.; Joshi, A.D.; Hann, C.L.; Bai, R.-Y.; Staedtke, V.; Blakeley, J.O.; Sengupta, S.; Jarrell, T.C.; et al. Mebendazole and temozolomide in patients with newly diagnosed high-grade gliomas: Results of a phase 1 clinical trial. Neuro-Oncol. Adv. 2020, 3, vdaa154. [Google Scholar] [CrossRef] [PubMed]
  165. Weyerhäuser, P.; Kantelhardt, S.R.; Kim, E.L. Re-purposing Chloroquine for Glioblastoma: Potential Merits and Confounding Variables. Front. Oncol. 2018, 8, 335. [Google Scholar] [CrossRef] [PubMed]
  166. Compter, I.; Eekers, D.B.P.; Hoeben, A.; Rouschop, K.M.A.; Reymen, B.; Ackermans, L.; Beckervordersantforth, J.; Bauer, N.J.C.; Anten, M.M.; Wesseling, P.; et al. Chloroquine combined with concurrent radiotherapy and temozolomide for newly diagnosed glioblastoma: A phase IB trial. Autophagy 2020, 17, 2604–2612. [Google Scholar] [CrossRef] [PubMed]
  167. Rajesh, Y.; Biswas, A.; Kumar, U.; Banerjee, I.; Das, S.; Maji, S.; Das, S.K.; Emdad, L.; Cavenee, W.K.; Mandal, M.; et al. Lumefantrine, an antimalarial drug, reverses radiation and temozolomide resistance in glioblastoma. Proc. Natl. Acad. Sci. USA 2020, 117, 12324–12331. [Google Scholar] [CrossRef]
  168. Persico, M.; Abbruzzese, C.; Matteoni, S.; Matarrese, P.; Campana, A.M.; Villani, V.; Pace, A.; Paggi, M.G. Tackling the Behavior of Cancer Cells: Molecular Bases for Repurposing Antipsychotic Drugs in the Treatment of Glioblastoma. Cells 2022, 11, 263. [Google Scholar] [CrossRef]
  169. Eckert, M.; Klumpp, L.; Huber, S.M. Cellular Effects of the Antiepileptic Drug Valproic Acid in Glioblastoma. Cell. Physiol. Biochem. 2017, 44, 1591–1605. [Google Scholar] [CrossRef]
  170. Scicchitano, B.M.; Sorrentino, S.; Proietti, G.; Lama, G.; Dobrowolny, G.; Catizone, A.; Binda, E.; LaRocca, L.M.; Sica, G. Levetiracetam enhances the temozolomide effect on glioblastoma stem cell proliferation and apoptosis. Cancer Cell Int. 2018, 18, 136. [Google Scholar] [CrossRef]
  171. Jabbarli, R.; Ahmadipour, Y.; Rauschenbach, L.; Santos, A.; Oppong, M.D.; Pierscianek, D.; Quesada, C.; Kebir, S.; Dammann, P.; Guberina, N.; et al. How about Levetiracetam in Glioblastoma? An Institutional Experience and Meta-Analysis. Cancers 2021, 13, 3770. [Google Scholar] [CrossRef]
  172. Cacciatore, I.; Fornasari, E.; Marinelli, L.; Eusepi, P.; Ciulla, M.; Ozdemir, O.; Tatar, A.; Turkez, H.; DI Stefano, A. Memantine-derived drugs as potential antitumor agents for the treatment of glioblastoma. Eur. J. Pharm. Sci. 2017, 109, 402–411. [Google Scholar] [CrossRef] [PubMed]
  173. Liu, Z.-Z.; Liu, X.-N.; Fan, R.-C.; Jia, Y.-P.; Zhang, Q.-K.; Gao, X.-Q.; Wang, Y.-Q.; Yang, M.-Q.; Ji, L.-Z.; Zhou, Y.-Q.; et al. Identification of pimavanserin tartrate as a potent Ca2+-calcineurin-NFAT pathway inhibitor for glioblastoma therapy. Acta Pharmacol. Sin. 2020, 42, 1860–1874. [Google Scholar] [CrossRef] [PubMed]
  174. Kast, R.E.; Boockvar, J.A.; Brüning, A.; Cappello, F.; Chang, W.-W.; Cvek, B.; Dou, Q.P.; Duenas-Gonzalez, A.; Efferth, T.; Focosi, D.; et al. A conceptually new treatment approach for relapsed glioblastoma: Coordinated undermining of survival paths with nine repurposed drugs (CUSP9) by the International Initiative for Accelerated Improvement of Glioblastoma Care. Oncotarget 2013, 4, 502–530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Kast, R.E.; Karpel-Massler, G.; Halatsch, M.-E. CUSP9* treatment protocol for recurrent glioblastoma: Aprepitant, artesunate, auranofin, captopril, celecoxib, disulfiram, itraconazole, ritonavir, sertraline augmenting continuous low dose temozolomide. Oncotarget 2014, 5, 8052–8082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Halatsch, M.-E.; Dwucet, A.; Schmidt, C.J.; Mühlnickel, J.; Heiland, T.; Zeiler, K.; Siegelin, M.D.; Kast, R.E.; Karpel-Massler, G. In Vitro and Clinical Compassionate Use Experiences with the Drug-Repurposing Approach CUSP9v3 in Glioblastoma. Pharmaceuticals 2021, 14, 1241. [Google Scholar] [CrossRef]
  177. Jacus, M.O.; Daryani, V.M.; Harstead, K.E.; Patel, Y.T.; Throm, S.L.; Stewart, C.F. Pharmacokinetic Properties of Anticancer Agents for the Treatment of Central Nervous System Tumors: Update of the Literature. Clin. Pharmacokinet. 2015, 55, 297–311. [Google Scholar] [CrossRef] [Green Version]
  178. Doniger, S.; Hofmann, T.; Yeh, J. Predicting CNS Permeability of Drug Molecules: Comparison of Neural Network and Support Vector Machine Algorithms. J. Comput. Biol. 2002, 9, 849–864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Castillo-Garit, J.A.; Martin, G.M.C.; Le-Thi-Thu, H.; Pham-The, H.; Barigye, S.J. A Simple Method to Predict Blood-Brain Barrier Permeability of Drug-Like Compounds Using Classification Trees. Med. Chem. 2017, 13, 1. [Google Scholar] [CrossRef]
  180. Carpenter, T.S.; Kirshner, D.A.; Lau, E.Y.; Wong, S.E.; Nilmeier, J.P.; Lightstone, F.C. A Method to Predict Blood-Brain Barrier Permeability of Drug-Like Compounds Using Molecular Dynamics Simulations. Biophys. J. 2014, 107, 630–641. [Google Scholar] [CrossRef] [Green Version]
  181. Alsenan, S.; Al-Turaiki, I.; Hafez, A. A deep learning approach to predict blood-brain barrier permeability. PeerJ Comput. Sci. 2021, 7, e515. [Google Scholar] [CrossRef]
  182. Massey, S.C.; Urcuyo, J.C.; Marin, B.M.; Sarkaria, J.N.; Swanson, K.R. Quantifying Glioblastoma Drug Response Dynamics Incorporating Treatment Sensitivity and Blood Brain Barrier Penetrance From Experimental Data. Front. Physiol. 2020, 11. [Google Scholar] [CrossRef] [PubMed]
  183. Westerhout, J.; Ploeger, B.; Smeets, J.; Danhof, M.; De Lange, E.C.M. Physiologically Based Pharmacokinetic Modeling to Investigate Regional Brain Distribution Kinetics in Rats. AAPS J. 2012, 14, 543–553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. de Lange, E.C.M.; van den Brink, W.; Yamamoto, Y.; de Witte, W.E.A.; Wong, Y.C. Novel CNS drug discovery and development approach: Model-based integration to predict neuro-pharmacokinetics and pharmacodynamics. Expert Opin. Drug Discov. 2017, 12, 1207–1218. [Google Scholar] [CrossRef] [PubMed]
  185. Li, J.; Jiang, J.; Wu, J.; Bao, X.; Sanai, N. Physiologically Based Pharmacokinetic Modeling of Central Nervous System Pharmacokinetics of CDK4/6 Inhibitors to Guide Selection of Drug and Dosing Regimen for Brain Cancer Treatment. Clin. Pharmacol. Ther. 2021, 109, 494–506. [Google Scholar] [CrossRef]
  186. Vengoji, R.; Macha, M.A.; Nimmakayala, R.K.; Rachagani, S.; Siddiqui, J.A.; Mallya, K.; Gorantla, S.; Jain, M.; Ponnusamy, M.P.; Batra, S.K.; et al. Afatinib and Temozolomide combination inhibits tumorigenesis by targeting EGFRvIII-cMet signaling in glioblastoma cells. J. Exp. Clin. Cancer Res. 2019, 38, 266. [Google Scholar] [CrossRef]
  187. Halatsch, M.-E.; Kast, R.E.; Karpel-Massler, G.; Mayer, B.; Zolk, O.; Schmitz, B.; Scheuerle, A.; Maier, L.; Bullinger, L.; Mayer-Steinacker, R.; et al. A phase Ib/IIa trial of 9 repurposed drugs combined with temozolomide for the treatment of recurrent glioblastoma: CUSP9v3. Neuro-Oncol. Adv. 2021, 3, vdab075. [Google Scholar] [CrossRef]
  188. Rabik, C.A.; Dolan, M.E. Molecular mechanisms of resistance and toxicity associated with platinating agents. Cancer Treat. Rev. 2007, 33, 9–23. [Google Scholar] [CrossRef] [Green Version]
  189. Ciak, J.; Hahn, F.E. Chloroquine: Mode of Action. Science 1966, 151, 347–349. [Google Scholar] [CrossRef]
  190. Liu, P.; Brown, S.; Goktug, T.; Channathodiyil, P.; Kannappan, V.; Hugnot, J.-P.; Guichet, P.-O.; Bian, X.; Armesilla, A.L.; Darling, J.L.; et al. Cytotoxic effect of disulfiram/copper on human glioblastoma cell lines and ALDH-positive cancer-stem-like cells. Br. J. Cancer 2012, 107, 1488–1497. [Google Scholar] [CrossRef] [Green Version]
  191. Thorn, C.F.; Oshiro, C.; Marsh, S.; Hernandez-Boussard, T.; McLeod, H.; Klein, T.E.; Altman, R.B. Doxorubicin pathways: Pharmacodynamics and adverse effects. Pharm. Genom. 2011, 21, 440–446. [Google Scholar] [CrossRef]
  192. Montecucco, A.; Zanetta, F.; Biamonti, G. Molecular mechanisms of etoposide. EXCLI J. 2015, 14, 95–108. [Google Scholar] [CrossRef] [PubMed]
  193. Houghton, P.J. Everolimus. Clin. Cancer Res. 2010, 16, 1368–1372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Tan, S.K.; Jermakowicz, A.; Mookhtiar, A.K.; Nemeroff, C.B.; Schürer, S.C.; Ayad, N.G. Drug Repositioning in Glioblastoma: A Pathway Perspective. Front. Pharmacol. 2018, 9, 218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Dong, Y.; Furuta, T.; Sabit, H.; Kitabayashi, T.; Jiapaer, S.; Kobayashi, M.; Ino, Y.; Todo, T.; Teng, L.; Hirao, A.; et al. Identification of antipsychotic drug fluspirilene as a potential anti-glioma stem cell drug. Oncotarget 2017, 8, 111728–111741. [Google Scholar] [CrossRef] [Green Version]
  196. Shi, Y.; Guryanova, O.A.; Zhou, W.; Liu, C.; Huang, Z.; Fang, X.; Wang, X.; Chen, C.; Wu, Q.; He, Z.; et al. Ibrutinib inactivates BMX-STAT3 in glioma stem cells to impair malignant growth and radioresistance. Sci. Transl. Med. 2018, 10. [Google Scholar] [CrossRef] [Green Version]
  197. Frolov, A.; Evans, I.M.; Li, N.; Sidlauskas, K.; Paliashvili, K.; Lockwood, N.; Barrett, A.; Brandner, S.; Zachary, I.C.; Frankel, P. Imatinib and Nilotinib increase glioblastoma cell invasion via Abl-independent stimulation of p130Cas and FAK signalling. Sci. Rep. 2016, 6, 27378. [Google Scholar] [CrossRef]
  198. Ardizzone, A.; Scuderi, S.; Giuffrida, D.; Colarossi, C.; Puglisi, C.; Campolo, M.; Cuzzocrea, S.; Esposito, E.; Paterniti, I. Role of Fibroblast Growth Factors Receptors (FGFRs) in Brain Tumors, Focus on Astrocytoma and Glioblastoma. Cancers 2020, 12, 3825. [Google Scholar] [CrossRef]
  199. Vredenburgh, J.J.; Desjardins, A.; Reardon, D.A.; Friedman, H.S. Experience with irinotecan for the treatment of malignant glioma. Neuro-Oncology 2009, 11, 80–91. [Google Scholar] [CrossRef] [Green Version]
  200. Muz, B.; Ghazarian, R.N.; Ou, M.; Luderer, M.J.; Kusdono, H.D.; Azab, A.K. Spotlight on ixazomib: Potential in the treatment of multiple myeloma. Drug Des. Dev. Ther. 2016, 10, 217–226. [Google Scholar] [CrossRef] [Green Version]
  201. Bai, R.-Y.; Staedtke, V.; Aprhys, C.M.; Gallia, G.L.; Riggins, G.J. Antiparasitic mebendazole shows survival benefit in 2 preclinical models of glioblastoma multiforme. Neuro-Oncology 2011, 13, 974–982. [Google Scholar] [CrossRef]
  202. Gritti, M.; Würth, R.; Angelini, M.; Barbieri, F.; Peretti, M.; Pizzi, E.; Pattarozzi, A.; Carra, E.; Sirito, R.; Daga, A.; et al. Metformin repositioning as antitumoral agent: Selective antiproliferative effects in human glioblastoma stem cells, via inhibition of CLIC1-mediated ion current. Oncotarget 2014, 5, 11252–11268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Calinescu, A.-A.; Castro, M.G. Microtubule targeting agents in glioma. Transl. Cancer Res. 2016, 5, S54–S60. [Google Scholar] [CrossRef] [PubMed]
  204. Stahl, S.M. Mechanism of action of pimavanserin in Parkinson’s disease psychosis: Targeting serotonin 5HT2A and 5HT2C receptors. CNS Spectr. 2016, 21, 271–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Bernstock, J.D.; Ye, D.; Gessler, F.A.; Lee, Y.-J.; Peruzzotti-Jametti, L.; Baumgarten, P.; Johnson, K.R.; Maric, D.; Yang, W.; Kögel, D.; et al. Topotecan is a potent inhibitor of SUMOylation in glioblastoma multiforme and alters both cellular replication and metabolic programming. Sci. Rep. 2017, 7, 1–14. [Google Scholar] [CrossRef]
  206. El-Habr, E.A.; Dubois, L.G.; Burel-Vandenbos, F.; Bogeas, A.; Lipecka, J.; Turchi, L.; Lejeune, F.-X.; Coehlo, P.L.C.; Yamaki, T.; Wittmann, B.M.; et al. A driver role for GABA metabolism in controlling stem and proliferative cell state through GHB production in glioma. Acta Neuropathol. 2017, 133, 645–660. [Google Scholar] [CrossRef]
  207. Bent, M.J.V.D.; Brandes, A.; Taphoorn, M.J.B.; Kros, J.M.; Kouwenhoven, M.; Delattre, J.-Y.; Bernsen, H.J.J.A.; Frenay, M.; Tijssen, C.C.; Grisold, W.; et al. Adjuvant Procarbazine, Lomustine, and Vincristine Chemotherapy in Newly Diagnosed Anaplastic Oligodendroglioma: Long-Term Follow-Up of EORTC Brain Tumor Group Study 26951. J. Clin. Oncol. 2013, 31, 344–350. [Google Scholar] [CrossRef]
  208. Straehla, J.P.; Warren, K.E. Pharmacokinetic Principles and Their Application to Central Nervous System Tumors. Pharmaceutics 2020, 12, 948. [Google Scholar] [CrossRef]
  209. Lightbody, G.; Haberland, V.; Browne, F.; Taggart, L.; Zheng, H.; Parkes, E.; Blayney, J.K. Review of applications of high-throughput sequencing in personalized medicine: Barriers and facilitators of future progress in research and clinical application. Brief. Bioinform. 2019, 20, 1795–1811. [Google Scholar] [CrossRef]
  210. Cho, C.-F.; Wolfe, J.M.; Fadzen, C.M.; Calligaris, D.; Hornburg, K.; Chiocca, E.A.; Agar, N.Y.R.; Pentelute, B.L.; Lawler, S.E. Blood-brain-barrier spheroids as an in vitro screening platform for brain-penetrating agents. Nat. Commun. 2017, 8, 15623. [Google Scholar] [CrossRef]
  211. Sanai, N.; Li, J.; Boerner, J.; Stark, K.; Wu, J.; Kim, S.; Derogatis, A.; Mehta, S.; Dhruv, H.D.; Heilbrun, L.K.; et al. Phase 0 Trial of AZD1775 in First-Recurrence Glioblastoma Patients. Clin. Cancer Res. 2018, 24, 3820–3828. [Google Scholar] [CrossRef] [Green Version]
  212. Quillin, J.; Patel, R.; Herzberg, E.; Alton, D.; Bikzhanova, G.; Geisler, L.; Olson, J. A phase 0 analysis of ixazomib in patients with glioblastoma. Mol. Clin. Oncol. 2020, 13, 1. [Google Scholar] [CrossRef] [PubMed]
  213. Shirley, M. Ixazomib: First Global Approval. Drugs 2016, 76, 405–411. [Google Scholar] [CrossRef] [PubMed]
  214. Tien, A.-C.; Li, J.; Bao, X.; Derogatis, A.; Kim, S.; Mehta, S.; Sanai, N. A Phase 0 Trial of Ribociclib in Recurrent Glioblastoma Patients Incorporating a Tumor Pharmacodynamic- and Pharmacokinetic-Guided Expansion Cohort. Clin. Cancer Res. 2019, 25, 5777–5786. [Google Scholar] [CrossRef] [Green Version]
  215. Shah, A.; Bloomquist, E.; Tang, S.; Fu, W.; Bi, Y.; Liu, Q.; Yu, J.; Zhao, P.; Palmby, T.R.; Goldberg, K.B.; et al. FDA Approval: Ribociclib for the Treatment of Postmenopausal Women with Hormone Receptor-Positive, HER2-Negative Advanced or Metastatic Breast Cancer. Clin. Cancer Res. 2018, 24, 2999–3004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Hasskarl, J. Everolimus. Recent Results Cancer Res. 2018, 211, 101–123. [Google Scholar] [PubMed]
  217. Kang, C. Infigratinib: First Approval. Drugs 2021, 81, 1355–1360. [Google Scholar] [CrossRef] [PubMed]
  218. Markham, A. Pamiparib: First Approval. Drugs 2021, 81, 1343–1348. [Google Scholar] [CrossRef] [PubMed]
  219. Vogelbaum, M.A.; Li, G.; Heimberger, A.B.; Lang, F.F.; Fueyo, J.; Gomez-Manzano, C.; Sanai, N. A Window of Opportunity to Overcome Therapeutic Failure in Neuro-Oncology. Am. Soc. Clin. Oncol. Educ. Book 2022, 139–146. [Google Scholar] [CrossRef]
  220. Lyne, S.; Yamini, B. An Alternative Pipeline for Glioblastoma Therapeutics: A Systematic Review of Drug Repurposing in Glioblastoma. Cancers 2021, 13, 1953. [Google Scholar] [CrossRef]
  221. Yadavalli, S.; Yenugonda, V.M.; Kesari, S. Repurposed Drugs in Treating Glioblastoma Multiforme: Clinical Trials Update. Cancer J. 2019, 25, 139–146. [Google Scholar] [CrossRef]
  222. Alomari, S.; Zhang, I.; Hernandez, A.; Kraft, C.Y.; Raj, D.; Kedda, J.; Tyler, B. Drug Repurposing for Glioblastoma and Current Advances in Drug Delivery—A Comprehensive Review of the Literature. Biomolecules 2021, 11, 1870. [Google Scholar] [CrossRef] [PubMed]
  223. Ettrich, T.J.; Seufferlein, T. Regorafenib. Recent Results Cancer Res. 2018, 211, 45–56. [Google Scholar] [PubMed]
  224. Lombardi, G.; De Salvo, G.L.; Brandes, A.A.; Eoli, M.; Rudà, R.; Faedi, M.; Lolli, I.; Pace, A.; Daniele, B.; Pasqualetti, F.; et al. Regorafenib compared with lomustine in patients with relapsed glioblastoma (REGOMA): A multicentre, open-label, randomised, controlled, phase 2 trial. Lancet Oncol. 2018, 20, 110–119. [Google Scholar] [CrossRef]
  225. Lombardi, G.; Caccese, M.; Padovan, M.; Cerretti, G.; Pintacuda, G.; Manara, R.; Di Sarra, F.; Zagonel, V. Regorafenib in Recurrent Glioblastoma Patients: A Large and Monocentric Real-Life Study. Cancers 2021, 13, 4731. [Google Scholar] [CrossRef] [PubMed]
  226. Johnston, S.R.D.; Harbeck, N.; Hegg, R.; Toi, M.; Martin, M.; Shao, Z.M.; Zhang, Q.Y.; Rodriguez, J.L.M.; Campone, M.; Hamilton, E.; et al. Abemaciclib Combined with Endocrine Therapy for the Adjuvant Treatment of HR+, HER2−, Node-Positive, High-Risk, Early Breast Cancer (monarchE). J. Clin. Oncol. 2020, 38, 3987–3998. [Google Scholar] [CrossRef]
  227. Riess, C.; Koczan, D.; Schneider, B.; Linke, C.; del Moral, K.; Classen, C.F.; Maletzki, C. Cyclin-dependent kinase inhibitors exert distinct effects on patient-derived 2D and 3D glioblastoma cell culture models. Cell Death Discov. 2021, 7, 1–15. [Google Scholar] [CrossRef]
  228. Patnaik, A.; Rosen, L.S.; Tolaney, S.M.; Tolcher, A.W.; Goldman, J.W.; Gandhi, L.; Papadopoulos, K.P.; Beeram, M.; Rasco, D.W.; Hilton, J.F.; et al. Efficacy and Safety of Abemaciclib, an Inhibitor of CDK4 and CDK6, for Patients with Breast Cancer, Non-Small Cell Lung Cancer, and Other Solid Tumors. Cancer Discov. 2016, 6, 740–753. [Google Scholar] [CrossRef] [Green Version]
  229. Scott, L.J. Niraparib: First Global Approval. Drugs 2017, 77, 1029–1034. [Google Scholar] [CrossRef]
  230. Lesueur, P.; Chevalier, F.; Austry, J.-B.; Waissi, W.; Burckel, H.; Noël, G.; Habrand, J.-L.; Saintigny, Y.; Joly, F. Poly-(ADP-ribose)-polymerase inhibitors as radiosensitizers: A systematic review of pre-clinical and clinical human studies. Oncotarget 2017, 8, 69105–69124. [Google Scholar] [CrossRef] [Green Version]
  231. Lesueur, P.; LeQuesne, J.; Grellard, J.-M.; Dugué, A.; Coquan, E.; Brachet, P.-E.; Geffrelot, J.; Kao, W.; Emery, E.; Berro, D.H.; et al. Phase I/IIa study of concomitant radiotherapy with olaparib and temozolomide in unresectable or partially resectable glioblastoma: OLA-TMZ-RTE-01 trial protocol. BMC Cancer 2019, 19, 198. [Google Scholar] [CrossRef]
  232. Plummer, R.; Jones, C.; Middleton, M.; Wilson, R.; Evans, J.; Olsen, A.; Curtin, N.; Boddy, A.; McHugh, P.; Newell, D.; et al. Phase I Study of the Poly(ADP-Ribose) Polymerase Inhibitor, AG014699, in Combination with Temozolomide in Patients with Advanced Solid Tumors. Clin. Cancer Res. 2008, 14, 7917–7923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Halford, S.E.R.; Cruickshank, G.; Dunn, L.; Erridge, S.; Godfrey, L.; Herbert, C.; Jefferies, S.; Lopez, J.S.; McBain, C.; Pittman, M.; et al. Results of the OPARATIC trial: A phase I dose escalation study of olaparib in combination with temozolomide (TMZ) in patients with relapsed glioblastoma (GBM). J. Clin. Oncol. 2017, 35, 2022. [Google Scholar] [CrossRef]
  234. Karanam, N.K.; Ding, L.; Aroumougame, A.; Story, M.D. Tumor treating fields cause replication stress and interfere with DNA replication fork maintenance: Implications for cancer therapy. Transl. Res. 2020, 217, 33–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Burger, J.A.; Tedeschi, A.; Barr, P.M.; Robak, T.; Owen, C.; Ghia, P.; Bairey, O.; Hillmen, P.; Bartlett, N.L.; Li, J.; et al. Ibrutinib as Initial Therapy for Patients with Chronic Lymphocytic Leukemia. N. Engl. J. Med. 2015, 373, 2425–2437. [Google Scholar] [CrossRef]
  236. Guryanova, O.A.; Wu, Q.; Cheng, L.; Lathia, J.D.; Huang, Z.; Yang, J.; MacSwords, J.; Eyler, C.E.; McLendon, R.E.; Heddleston, J.M.; et al. Nonreceptor Tyrosine Kinase BMX Maintains Self-Renewal and Tumorigenic Potential of Glioblastoma Stem Cells by Activating STAT3. Cancer Cell 2011, 19, 498–511. [Google Scholar] [CrossRef] [Green Version]
  237. Cook, K.L.; Wärri, A.; Soto-Pantoja, D.R.; Clarke, P.A.G.; Cruz, M.I.; Zwart, A.; Clarke, R. Hydroxychloroquine inhibits autophagy to potentiate antiestrogen responsiveness in ER+ breast cancer. Clin. Cancer Res. 2014, 20, 3222–3232. [Google Scholar] [CrossRef] [Green Version]
  238. Liu, L.-Q.; Wang, S.-B.; Shao, Y.-F.; Shi, J.-N.; Wang, W.; Chen, W.-Y.; Ye, Z.-Q.; Jiang, J.-Y.; Fang, Q.-X.; Zhang, G.-B.; et al. Hydroxychloroquine potentiates the anti-cancer effect of bevacizumab on glioblastoma via the inhibition of autophagy. Biomed. Pharmacother. 2019, 118, 109339. [Google Scholar] [CrossRef]
  239. Al-Bari, M.A. Chloroquine analogues in drug discovery: New directions of uses, mechanisms of actions and toxic manifestations from malaria to multifarious diseases. J. Antimicrob. Chemother. 2015, 70, 1608–1621. [Google Scholar] [CrossRef] [Green Version]
  240. Jutten, B.; Keulers, T.G.; Schaaf, M.B.; Savelkouls, K.; Theys, J.; Span, P.N.; Vooijs, M.A.; Bussink, J.; Rouschop, K.M. EGFR overexpressing cells and tumors are dependent on autophagy for growth and survival. Radiother. Oncol. 2013, 108, 479–483. [Google Scholar] [CrossRef]
  241. Lee, S.W.; Kim, H.-K.; Lee, N.-H.; Yi, H.-Y.; Kim, H.-S.; Hong, S.H.; Hong, Y.-K.; Joe, Y.A. The synergistic effect of combination temozolomide and chloroquine treatment is dependent on autophagy formation and p53 status in glioma cells. Cancer Lett. 2015, 360, 195–204. [Google Scholar] [CrossRef]
  242. Rosenfeld, M.R.; Ye, X.; Supko, J.G.; Desideri, S.; Grossman, S.A.; Brem, S.; Mikkelson, T.; Wang, D.; Chang, Y.C.; Hu, J.; et al. A phase I/II trial of hydroxychloroquine in conjunction with radiation therapy and concurrent and adjuvant temozolomide in patients with newly diagnosed glioblastoma multiforme. Autophagy 2014, 10, 1359–1368. [Google Scholar] [CrossRef] [PubMed]
  243. Teng, J.; Hejazi, S.; Hiddingh, L.; Carvalho, L.; De Gooijer, M.C.; Wakimoto, H.; Barazas, M.; Tannous, M.; Chi, A.S.; Noske, D.P.; et al. Recycling drug screen repurposes hydroxyurea as a sensitizer of glioblastomas to temozolomide targeting de novo DNA synthesis, irrespective of molecular subtype. Neuro-Oncology 2017, 20, 642–654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Lun, X.; Wells, J.C.; Grinshtein, N.; King, J.C.; Hao, X.; Dang, N.-H.; Wang, X.; Aman, A.; Uehling, D.; Datti, A.; et al. Disulfiram when Combined with Copper Enhances the Therapeutic Effects of Temozolomide for the Treatment of Glioblastoma. Clin. Cancer Res. 2016, 22, 3860–3875. [Google Scholar] [CrossRef] [Green Version]
  245. Jakola, A.S.; Werlenius, K.; Mudaisi, M.; Hylin, S.; Kinhult, S.; Bartek, J., Jr.; Salvesen, Ø.; Carlsen, S.M.; Strandéus, M.; Lindskog, M.; et al. Disulfiram repurposing combined with nutritional copper supplement as add-on to chemotherapy in recurrent glioblastoma (DIRECT): Study protocol for a randomized controlled trial. F1000Research 2018, 7, 1797. [Google Scholar] [CrossRef] [PubMed]
  246. Huang, J.; Campian, J.L.; Gujar, A.D.; Tsien, C.; Ansstas, G.; Tran, D.D.; DeWees, T.A.; Lockhart, A.C.; Kim, A.H. Final results of a phase I dose-escalation, dose-expansion study of adding disulfiram with or without copper to adjuvant temozolomide for newly diagnosed glioblastoma. J. Neuro-Oncol. 2018, 138, 105–111. [Google Scholar] [CrossRef] [PubMed]
  247. Paun, L.; Moiraghi, A.; Jannelli, G.; Nouri, A.; DiMeco, F.; Pallud, J.; Meling, T.R.; Momjian, S.; Schaller, K.; Prada, F.; et al. From Focused Ultrasound Tumor Ablation to Brain Blood Barrier Opening for High Grade Glioma: A Systematic Review. Cancers 2021, 13, 5614. [Google Scholar] [CrossRef] [PubMed]
  248. Zhang, D.Y.; Dmello, C.; Chen, L.; Arrieta, V.A.; Gonzalez-Buendia, E.; Kane, J.R.; Magnusson, L.P.; Baran, A.; James, C.D.; Horbinski, C.; et al. Ultrasound-mediated Delivery of Paclitaxel for Glioma: A Comparative Study of Distribution, Toxicity, and Efficacy of Albumin-bound Versus Cremophor Formulations. Clin. Cancer Res. 2020, 26, 477–486. [Google Scholar] [CrossRef] [PubMed]
  249. Carpentier, A.; Canney, M.; Vignot, A.; Reina, V.; Beccaria, K.; Horodyckid, C.; Karachi, C.; Leclercq, D.; Lafon, C.; Chapelon, J.-Y.; et al. Clinical trial of blood-brain barrier disruption by pulsed ultrasound. Sci. Transl. Med. 2016, 8, 343re2. [Google Scholar] [CrossRef]
  250. Idbaih, A.; Ducray, F.; Stupp, R.; Baize, N.; Chinot, O.L.; De Groot, J.F.; Guyotat, J.; Sonabend, A.M.; Menei, P.; Dufour, H.; et al. A phase I/IIa study to evaluate the safety and efficacy of blood-brain barrier (BBB) opening with the SonoCloud-9 implantable ultrasound device in recurrent glioblastoma patients receiving IV carboplatin. J. Clin. Oncol. 2021, 39, 2049. [Google Scholar] [CrossRef]
  251. Jain, K.K. Use of nanoparticles for drug delivery in glioblastoma multiforme. Expert Rev. Neurother. 2007, 7, 363–372. [Google Scholar] [CrossRef]
  252. Zhao, M.; Van Straten, D.; Broekman, M.L.; Préat, V.; Schiffelers, R.M. Nanocarrier-based drug combination therapy for glioblastoma. Theranostics 2020, 10, 1355–1372. [Google Scholar] [CrossRef] [PubMed]
  253. Zhou, J.; Atsina, K.-B.; Himes, B.T.; Strohbehn, G.W.; Saltzman, W.M. Novel Delivery Strategies for Glioblastoma. Cancer J. 2012, 18, 89–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Li, J.; Zhao, J.; Tan, T.; Liu, M.; Zeng, Z.; Zeng, Y.; Zhang, L.; Fu, C.; Chen, D.; Xie, T. Nanoparticle Drug Delivery System for Glioma and Its Efficacy Improvement Strategies: A Comprehensive Review. Int. J. Nanomed. 2020, 15, 2563–2582. [Google Scholar] [CrossRef] [Green Version]
  255. Wu, X.; Wang, X.; Wang, J.; Hao, Y.; Liu, F.; Wang, X.; Yang, L.; Lu, Z. The Roles of Exosomes as Future Therapeutic Agents and Diagnostic Tools for Glioma. Front. Oncol. 2021, 11, 733529. [Google Scholar] [CrossRef] [PubMed]
  256. Dai, J.; Su, Y.; Zhong, S.; Cong, L.; Liu, B.; Yang, J.; Tao, Y.; He, Z.; Chen, C.; Jiang, Y. Exosomes: Key players in cancer and potential therapeutic strategy. Signal Transduct. Target. Ther. 2020, 5, 145. [Google Scholar] [CrossRef] [PubMed]
  257. Spinazzi, E.F.; Argenziano, M.G.; Upadhyayula, P.S.; Banu, M.A.; Neira, J.A.; Higgins, D.M.O.; Wu, P.B.; Pereira, B.; Mahajan, A.; Humala, N.; et al. Treatment of Recurrent Glioblastoma by Chronic Convection-Enhanced Delivery of Topotecan. medRxiv 2021. [Google Scholar] [CrossRef]
  258. Chen, T.C.; Da Fonseca, C.O.; Schönthal, A.H. Intranasal Perillyl Alcohol for Glioma Therapy: Molecular Mechanisms and Clinical Development. Int. J. Mol. Sci. 2018, 19, 3905. [Google Scholar] [CrossRef] [Green Version]
  259. Killick-Cole, C.; Woolley, M.; Johnson, D.; Lewis, O.; Skinner, P.; Bienemann, A.; Gill, S. SCIDOT-36. PREDICTIVE CED INFUSION VOLUMES FOR SURGICAL PLANNING AND INFUSION REGIME STRATEGIES. Neuro-Oncology 2019, 21, vi279. [Google Scholar] [CrossRef]
  260. Ashby, L.S.; Smith, K.A.; Stea, B. Gliadel wafer implantation combined with standard radiotherapy and concurrent followed by adjuvant temozolomide for treatment of newly diagnosed high-grade glioma: A systematic literature review. World J. Surg. Oncol. 2016, 14, 225. [Google Scholar] [CrossRef] [Green Version]
  261. Perry, J.; Chambers, A.; Spithoff, K.; Laperriere, N.; on behalf of the Neuro-Oncology Disease Site Group§ of Cancer Care Ontario’s Program in Evidence-Based Care. Gliadel Wafers in the Treatment of Malignant Glioma: A Systematic Review. Curr. Oncol. 2007, 14, 189–194. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Anatomical features of the blood–brain barrier (BBB). The structure of the BBB in normal physiology consists of endothelial cells interconnected with a complex network of proteins (tight junctions), while mechanical support is provided by pericytes and astrocytic end-feet. The parenchymal and endothelial basal membranes (BM) provide additional strengthening to the cell attachments. Figure was created in BioRender.com (accessed on 5 July 2022).
Figure 1. Anatomical features of the blood–brain barrier (BBB). The structure of the BBB in normal physiology consists of endothelial cells interconnected with a complex network of proteins (tight junctions), while mechanical support is provided by pericytes and astrocytic end-feet. The parenchymal and endothelial basal membranes (BM) provide additional strengthening to the cell attachments. Figure was created in BioRender.com (accessed on 5 July 2022).
Cancers 14 03705 g001
Figure 2. The integrity of the blood–brain barrier in physiological and malignant conditions. (A) In physiological conditions, tight junctions (claudin 3, occludin, junction adhesion molecules (JAMS) as well as cytoplasmic accessory proteins, such as Zonula occludens-1 (ZO-1)) of the endothelial cells remain intact maintaining the integrity of the BBB. (B) In CNS tumors, the release of chemical mediators by the tumor cells, such as substance P, matrix metalloproteinases (MMPs) and transforming growth factor beta (TGF-β), can cause the loss of tight junctions, which leads to the dysfunction and disruption of the BBB [60,69,70,72]. Additionally, the overexpression of angiopoietin-2 (ANG-2) is linked to vascular malformations and pericyte detachment through the hypoxic upregulation of VEGF, which subsequently promotes angiogenesis at the tumor margin [68]. Figure was created in BioRender.com (accessed on 20 May 2022).
Figure 2. The integrity of the blood–brain barrier in physiological and malignant conditions. (A) In physiological conditions, tight junctions (claudin 3, occludin, junction adhesion molecules (JAMS) as well as cytoplasmic accessory proteins, such as Zonula occludens-1 (ZO-1)) of the endothelial cells remain intact maintaining the integrity of the BBB. (B) In CNS tumors, the release of chemical mediators by the tumor cells, such as substance P, matrix metalloproteinases (MMPs) and transforming growth factor beta (TGF-β), can cause the loss of tight junctions, which leads to the dysfunction and disruption of the BBB [60,69,70,72]. Additionally, the overexpression of angiopoietin-2 (ANG-2) is linked to vascular malformations and pericyte detachment through the hypoxic upregulation of VEGF, which subsequently promotes angiogenesis at the tumor margin [68]. Figure was created in BioRender.com (accessed on 20 May 2022).
Cancers 14 03705 g002
Figure 3. Development stages for repurposing a drug in glioblastoma.
Figure 3. Development stages for repurposing a drug in glioblastoma.
Cancers 14 03705 g003
Table 1. Examples of oncological and non-oncological repurposed drugs under preclinical investigation against GBM.
Table 1. Examples of oncological and non-oncological repurposed drugs under preclinical investigation against GBM.
Drug NameDrug CategoryDrug ClassIndicationsMoaMoa In GliomaReference
AbemaciclibOncologicalKinase inhibitorsBreast cancerCDK4/6 inhibitorCDK4/6 inhibitor[185]
Actinomycin-DOncologicalAntineoplastic antibioticOvarian, testicular, Ewing sarcoma, rhabdomyosarcoma, trophoblastic neoplasms, Wilms tumorRNA polymerase 1 inhibitorSOX-2 downregulation[145]
AfatinibOncologicalKinase inhibitorsNon-small-cell lung cancerEGFR inhibitorInhibition EGFRvIII–cMet signaling pathway[186]
AprepitantNon-OncologicalAntiemeticsNausea/VomitingSubstance P/NK1 receptor antagonistNK1 inhibitor[187]
AuranofinNon-OncologicalDMARDRheumatoid arthritisThioredoxin reductase inhibitorNA[187]
CaptorilNon-OncologicalACE inhibitorsHypertensionACE inhibitorACE, MMPs, AT1 receptors[187]
CarboplatinOncologicalAntineoplastic/Platinum analogOvarian, lung, head and neck cancerDNA cross-linking/alkylationDNA cross-linking/alkylation[188]
CelecoxibNon-OncologicalNSAIDsOsteoarthritis and rheumatoid arthritisCOX-2 inhibitorsCOX-1 and -2, carbonic anhydrase-2 and -9[187]
ChloroquineNon-OncologicalAntimalarial/
amebicide
MalariaDNA replicationinhibitorInhibition PI3K/Akt or EGFR signaling pathways[189]
DabrafenibOncologicalKinase inhibitorsMelanomaBRAF inhibitorBRAF-inhibitor[125]
DisulfiramNon-OncologicalAnti-alcoholismAlcoholismALDH inhibitorIn combination with copper, induces ROS, activates p38 and inhibits NF-κB[190]
DoxorubicinOncologicalAnthracyclinesOvary, prostate, stomach, thyroid, lung, liver; head and neck cancer, multiple myeloma, Hodgkin’s disease, lymphomas, acute lymphocytic leukemia and acute myeloid leukemiaDNA topoisomerase-2 inhibitorDNA topoisomerase-2 inhibitor[191]
EtoposideOncologicalPodophyllotoxin derivativesTesticular cancerDNA topoisomerase-2 inhibitorDNA topoisomerase-2 inhibitor[192]
EverolimusOncologicalKinase inhibitorsBreast cancermTOR inhibitormTOR inhibitor[193]
FluoxetineNon-OncologicalAntidepressantDepressionSerotine uptake inhibitorSMPD1inhibitor[154]
FluphenazineNon-OncologicalAntipsychoticSchizophreniaDopamine D2 receptors inhibitorInhibition of mitochondrial CcO and GPCR σ-receptors, increase AMPK activity[194]
FluspirileneNon-OncologicalAntipsychoticSchizophreniaDopamine D2 receptors inhibitorInactivation of STAT3[195]
IbrutinibOncologicalKinase inhibitorsChronic lymphocytic leukemia and small lymphocytic lymphomaBTK and BMX inhibitorBMX inhibitor[196]
ImatinibOncologicalKinase inhibitorsChronic myeloid leukemiaBcr-Abl inhibitorBcr-Abl and FAK inhibitor[197]
InfigratinibOncologicalKinase inhibitorsMetastatic cholangiocarcinomaFGFR-1, -2, -3FGFR-1[198]
IrinotecanOncologicalAntineoplasticColorectal and pancreatic cancerTopoisomerase-1 inhibitorTopoisomerase-1 inhibitor[199]
ItraconazoleNon-OncologicalAntifungalsSystematic fungal infections14-α demethylase inhibitorP-gp efflux transporters, BCRP, hedgehog, 5-lipoxygenase[187]
IxazomibOncologicalKinase inhibitorsMultiple myelomaProteasome subunit beta type-5 inhibitorProteasome subunit beta type-5 inhibitor[200]
LevetericamNon-OncologicalAnticonvulsantsEpilepsyProlong Na+ channel inactivation and GABA transaminase inhibitorPromoting HDAC4 nuclear translocation and apoptosis[194,170]
LumefantrineNon-OncologicalAntimalarialMalariaβ-hematin inhibitorFli-1 inhibitor[167]
MebendazoleNon-OncologicalAnthelminticsRoundworm and whipworm infectionsMicrotubules inhibitorMicrotubules inhibitor[194,201]
MemantineNon-OncologicalNMDA receptor antagonistAlzheimerblocks current flow through channels of NMDA receptorsNA[172]
MetforminNon-OncologicalAntidiabeticHyperglycemiaComplex 1 of the mitochondrial respiratory chain inhibitorCLIC-1 mediated ion currentinhibitor[202]
MinocyclineNon-OncologicalTetracycline antibioticsBacterial infectionsProtein synthesis inhibitorMonocyte, macrophage and microglial inhibition[187]
Omacetaxine mepessucinateOncologicalAntineoplasticChronic myeloid leukemiaProtein synthesis (RPL3) inhibitorNA[104]
PaclitaxelOncologicalAnti-microtubule agentsOvarian, breast, and non-small cell lung cancerTubulin beta-1 chain inhibitorTubulin beta-1 chain inhibitor[203]
PimavanserinNon-OncologicalAtypical antipsychoticParkinsonInverse agonist/antagonist of serotonin 5HT2A and 5HT2C receptorsCa 2+-calcineurin-NFAT pathway inhibitor[204,173]
TopotecanOncologicalAntineoplasticOvarian and lung cancerTopoisomerase 1inhibitorSUMOylationinhibitor[205]
TrametinibOncologicalKinase inhibitorsMelanomaMEK inhibitorMEK inhibitor[153]
Valproic acidNon-OncologicalAnticonvulsantsEpilepsyHistone deacetylase 9 inhibitorSSADH downregulation[206]
VincristineOncologicalVinca alkaloidsAcute lymphocytic leukemia, lymphoid blast crisis of chronic myeloid leukemia, and Hodgkin and Non-Hodgkin lymphomaTubulin beta chain inhibitorTubulin beta chain inhibitor[207]
CDK4/6: cyclin-dependent kinase 4/6, NK1: neurokinin 1, DMARD: disease modifying anti-rheumatic drug, ACE: angiotensin-converting enzyme, MMPs: matrix metalloproteinases, NSAIDs: non-steroidal anti-inflammatory drugs, COX-2: cyclo-oxygenase-2, mTOR: mammalian target of rapamycin, BTK: Bruton’s tyrosine kinase, BMX: bone marrow tyrosine kinase on chromosome X, FAK: focal adhesion kinase, FGFR: fibroblast growth factor receptors, P-gp: P-glycoprotein, BCRP: breast cancer-resistant protein, RPL3: ribosomal protein 3, EGFR: epidermal growth factor receptor, EGFRvIII: epidermal growth factor receptor variant-III, SUMO: small ubiquitin-like modifier, SMPD1: sphingomyelin phosphodiesterase 1, CLIC-1: chloride intracellular channel 1, ALDH: aldehyde dehydrogenase, ROS: reactive oxygen species, NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells, ERK1/2: extracellular signal-regulated protein kinases 1 and 2, Bcl-2: B-cell lymphoma 2, Akt: protein kinase B, FOXO3a: forkhead box O3, Fli-1: friendleukemia integration 1, CcO: cytochrome c oxidase, GPCR: G protein-coupled receptors, STAT3: signal transducer and activator of transcription 3, GABA: γ aminobutyric acid, NMDA:N-methyl-D-aspartate, NFAT: nuclear factor of activated T cells, SSRIs: selective serotonin reuptake inhibitors, TCTP: translationally controlled tumor protein, SSADH: succinic semialdehyde dehydrogenase.
Table 2. Repurposed drugs under clinical investigation in glioblastoma.
Table 2. Repurposed drugs under clinical investigation in glioblastoma.
NCT CodeStudy TitleInterventionsDiseaseStatusClinical Phase
NCT03834740A phase 0/2 study of ribociclib (LEE011) in combination with everolimus in preoperative recurrent high-grade glioma patients scheduled for resectionDrug: Ribociclib
Drug: Everolimus
GlioblastomaRecruitingPhase 0/2
NCT02981940A phase 0/2 study of abemaciclib in recurrent glioblastomaDrug: AbemaciclibGlioblastomaRecruitingPhase 0/2
NCT04391595A phase 0/2 study of LY3214996 (ERK inhibitor) in combination with abemaciclib (CDK4 and 6 inhibitor) in recurrent glioblastoma participants scheduled for resection to evaluate central nervous system (CNS) penetrationDrug: Abemaciclib
Drug: LY3214996
GlioblastomaRecruitingEarly phase 1
NCT04424966A phase 0 study of infigratinib in recurrent high-grade glioma participants scheduled for resection to evaluate central nervous system (CNS) penetration with PK triggered expansion cohortDrug: InfigratinibGlioblastomaRecruitingEarly Phase 1
NCT04614909A phase 0/2 clinical trial of pamiparib in newly diagnosed and recurrent glioblastoma patientsDrug: Pamiparib
Drug: Olaparib
Radiation therapy
Drug: TMZ
GlioblastomaRecruitingEarly Phase 1
NCT01294735A phase 1 study of MK-4827 in combination with temozolomide in patients with advanced cancerDrug: Niriparib (MK-4827)
Drug: TMZ
Glioblastoma
Melanoma
CompletedPhase 1
NCT03535350Ibrutinib with radiation and temozolomide in patients with newly diagnosed glioblastomaDrug: Ibrutinib
Radiation
Drug: TMZ
GlioblastomaRecruitingPhase 1
NCT03463733Hydroxy-urea and temozolomide in patients with a recurrent malignant brain tumor (glioblastoma) (HUTMZ)Drug: Hydroxyurea
Drug: TMZ
Glioma GlioblastomaRecruitingPhase 1
NCT02770378A proof-of-concept clinical trial assessing the safety of the coordinated undermining of survival paths by 9 repurposed drugs combined with metronomic temozolomide (CUSP9v3 treatment protocol) for recurrent glioblastomaDrug: TMZ
Drug: Aprepitant
Drug: Minocycline
Drug: Disulfiram
Drug: Celecoxib
Drug: Sertraline
Drug: Captopril
Drug: Itraconazole
Drug: Ritonavir
Drug: Auranofin
GlioblastomaCompletedPhase 1/2
NCT04440358Assessment of safety and feasibility of Exablate blood–brain barrier disruption (BBBD) with microbubbles for the treatment of recurrent glioblastoma (rGBM) in subjects undergoing carboplatin monotherapyDevice: Exablate BBBD
Drug: Carboplatin
GlioblastomaRecruitingPhase 1/2
NCT04528680Phase 1/2 trial of blood–brain barrier opening with an implantable ultrasound device SonoCloud-9 and treatment with albumin-bound paclitaxel in patients with recurrent glioblastomaDevice: Sonication for the opening of blood–brain barrier
Drug: albumin-bound paclitaxel
GlioblastomaRecruitingPhase 1/2
NCT04051606Regorafenib in bevacizumab refractory recurrent glioblastomaDrug: RegorafenibRecurrent GlioblastomaRecruitingPhase 2
NCT03970447A trial to evaluate multiple regimens in newly diagnosed and recurrent glioblastoma (GBM AGILE)Drug: TMZ
Drug: Lomustine
Drug: Regorafenib
Radiation
GlioblastomaRecruitingPhase 2/3
NCT02926222Regorafenib in relapsed glioblastoma (REGOMA)Drug: Regorafenib
Drug: Lomustine
GlioblastomaActive, non-recruitingPhase 2
NCT04221503A phase 2 study evaluating the efficacy and safety of niraparib and tumor-treating fields in recurrent glioblastomaDrug: Niraparib
Device: Optune
Glioblastoma Recurrent GlioblastomaRecruitingPhase 2
NCT03243851Study on low-dose temozolomide plus metformin or placebo in patient with recurrent or refractory glioblastoma (METT)Drug: TMZ +Metformin
Drug: TMZ +Placebo
GlioblastomaRecruitingPhase 2
NCT03363659Disulfiram and copper gluconate with temozolomide in unmethylated glioblastoma multiformeDrug: Disulfiram
Dietary Supplement: Copper gluconate
Drug: TMZ
GlioblastomaRecruitingPhase 2
NCT02432417The addition of chloroquine to chemoradiation for glioblastomaDrug: ChloroquineAstrocytoma, Grade IVNot yet recruitingPhase 2
NCT03632135Standard chemotherapy vs. chemotherapy guided by cancer stem cell test in recurrent glioblastoma (CSCRGBM)Diagnostic Test: ChemoID assay
Drug: Chemotherapy
Recurrent GlioblastomaActive, non-recruitingPhase 3
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ntafoulis, I.; Koolen, S.L.W.; Leenstra, S.; Lamfers, M.L.M. Drug Repurposing, a Fast-Track Approach to Develop Effective Treatments for Glioblastoma. Cancers 2022, 14, 3705. https://doi.org/10.3390/cancers14153705

AMA Style

Ntafoulis I, Koolen SLW, Leenstra S, Lamfers MLM. Drug Repurposing, a Fast-Track Approach to Develop Effective Treatments for Glioblastoma. Cancers. 2022; 14(15):3705. https://doi.org/10.3390/cancers14153705

Chicago/Turabian Style

Ntafoulis, Ioannis, Stijn L. W. Koolen, Sieger Leenstra, and Martine L. M. Lamfers. 2022. "Drug Repurposing, a Fast-Track Approach to Develop Effective Treatments for Glioblastoma" Cancers 14, no. 15: 3705. https://doi.org/10.3390/cancers14153705

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop