Next Article in Journal
Multi-View Data Integration Methods for Radiotherapy Structure Name Standardization
Previous Article in Journal
Circulating Biomarkers of Response and Toxicity of Immunotherapy in Advanced Non-Small Cell Lung Cancer (NSCLC): A Comprehensive Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Systematic Review of Glioblastoma-Targeted Therapies in Phases II, III, IV Clinical Trials

by
Elisabete Cruz Da Silva
1,
Marie-Cécile Mercier
1,
Nelly Etienne-Selloum
1,2,
Monique Dontenwill
1 and
Laurence Choulier
1,*
1
CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France
2
Service de Pharmacie, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
*
Author to whom correspondence should be addressed.
Submission received: 12 February 2021 / Revised: 19 March 2021 / Accepted: 26 March 2021 / Published: 9 April 2021

Abstract

:

Simple Summary

This review describes in a very detailed and exhaustive approach the literature of these last 20 years on glioblastoma targeted therapies in Phases II-IV of 257 clinical trials on adults with newly diagnosed or recurrent GBMs (excluding targeted immunotherapies and therapies targeting tumor cell metabolism, well documented in recent reviews). Divided in four Sections, are provided descriptions and lists (in 12 different tables) of, not only main but all drugs, targets, clinical trials and the results of targeted therapies when they are known.

Abstract

Glioblastoma (GBM), the most frequent and aggressive glial tumor, is currently treated as first line by the Stupp protocol, which combines, after surgery, radiotherapy and chemotherapy. For recurrent GBM, in absence of standard treatment or available clinical trials, various protocols including cytotoxic drugs and/or bevacizumab are currently applied. Despite these heavy treatments, the mean overall survival of patients is under 18 months. Many clinical studies are underway. Based on clinicaltrials.org and conducted up to 1 April 2020, this review lists, not only main, but all targeted therapies in phases II-IV of 257 clinical trials on adults with newly diagnosed or recurrent GBMs for the last twenty years. It does not involve targeted immunotherapies and therapies targeting tumor cell metabolism, that are well documented in other reviews. Without surprise, the most frequently reported drugs are those targeting (i) EGFR (40 clinical trials), and more generally tyrosine kinase receptors (85 clinical trials) and (ii) VEGF/VEGFR (75 clinical trials of which 53 involving bevacizumab). But many other targets and drugs are of interest. They are all listed and thoroughly described, on an one-on-one basis, in four sections related to targeting (i) GBM stem cells and stem cell pathways, (ii) the growth autonomy and migration, (iii) the cell cycle and the escape to cell death, (iv) and angiogenesis.

1. Introduction

Since 1926, different classifications of brain tumors have been proposed, based mainly on histological and malignancy criteria [1]. Increasing knowledge on glioma molecular characteristics enabled the proposition of a new classification in 2016. Figure 1 recapitulates the main steps of the modern classification of gliomas. Glioblastoma (GBM) is a high-grade glioma (grade IV), the most aggressive and the most frequent glioma. In the 2016 classification, GBMs are divided into three groups according to the status of the isocitrate dehydrogenase (IDH) gene: (i) GBMs IDHwt [this group represents 90% of GBMs and corresponds to primary GBMs], (ii) mutated IDH GBMs [this group represents 10% of GBMs, corresponds to secondary GBMs, occurs in young patients and has a better prognostic], (iii) Not otherwise specified (NOS) GBMs [status could not be evaluated]. When histological data suggest GBM and immunohistochemical analysis of IDHmut is negative, sequencing is recommended. Sequencing is no longer recommended after the age of 55 [2]. Inhibitors of the mutated IDH proteins are currently evaluated in GBM in Phase I clinical trials (NCT02073994, NCT02273739). They will thus not be further described in this review.
The standard treatment of GBMs is based on surgical resection followed by radiotherapy (RT) and concomitant chemotherapy for 6 weeks. The area around the tumor is irradiated with 2 Gy per day, five days per week for a total dose of 60 Gy. The chemotherapy used is Temozolomide (TMZ) at 75 mg/m2 per day. After this radiochemotherapy, TMZ treatment is pursued alone every four weeks at 150–200 mg/m2 per day for 5 consecutive days [3]. TMZ is an alkylating agent that causes DNA damage, cell cycle arrest and cell apoptosis. After oral administration, it is spontaneously hydrolyzed into an highly instable metabolite: 3-methyl-(triazen-1-yl)imidazole-4-carboxamide (MTIC) which reacts with water and releases highly reactive 5-aminoimidazole-4-carboxamide and methyldiazonium. The latter induces methylation at the O6 and N7 positions of a guanine and N3 position of an adenine [4]. These mutations cause aberrant repairs.
Despite these aggressive treatments, recurrence generally appears within 6–9 months of diagnosis [5]. In 90% of cases, recurrence is at the edge of the surgical resection. At the appearance of recurrence, patients’ survival is low: 3–6 months [6,7]. No protocol has yet been validated in the management of recurrent GBM. An increase in RT doses does not lead to gain in survival but induces more toxicity, including necrosis of healthy tissue [8]. Long-term side effects of radiation exposure (among which neurocognitive, psychosocial, endocrine …) are present months or years after treatment and cause problems in rare people who survive as the effect of side-effects increases with time [9]. Increasing the doses of TMZ is also not more efficient [10]. In most cases, patients with recurrent GBM are included in clinical trials [11]. If not, several therapeutic molecules are proposed in the second line, mainly alkylating agents (lomustine, carmustine, fotemustine, carboplatin or procarbazine), microtubule destabilizing agent (vincristine) or antiangiogenic drug (bevacizumab). In absence of standard protocols, the therapeutic strategy is discussed for individual patients. In addition, corticosteroids, anticonvulsants (lacosamide, levetiracetam) and anticoagulants are used in the progression of tumors in the event of intracranial pressure, stroke and deep venous thrombosis epilepsy which occurs in 30% of patients with primary brain tumors [12].
Different improvements of the current protocol (surgery, radio and chemo therapies) or new strategies based on the particular microenvironment of GBM are increasingly proposed for the effective care of GBM [13,14,15,16,17]. They are briefly mentioned below, but are not the focus of this review. But regardless of strategies, if new treatments allowed for significantly longer survival, they would require more than improving patients’ survival and would minimize long-term side effects to preserve or even improve patients’ quality of life. Late adverse events induced by administered treatments should be addressed [18].
New strategies are proposed to improve the drug passage through the blood brain barrier (BBB) to achieve a higher therapeutic concentration at the tumor site. Delivering chemotherapy directly into the surgical resection cavity has been proposed. Convection-enhanced delivery (CED) allows chemotherapy to be delivered directly via a catheter in the tissue surrounding the GBM resection cavity. This method increases the volume of distribution but results in unpredictable brain diffusion [19]. It requires the use of several surgical procedures, leading to a high risk of infection or bleeding. Another strategy consists of administrating the therapy directly at the tumor resection bed [20,21,22,23,24,25]. The use of small lipophilic molecules, able to passively cross the endothelial cells of the BBB, has been tested in combination with standard therapies [26]. Encapsulating therapies in nanoparticles (10–200 nm) not only increases their solubility but also their release time and stability, while reducing side effects [27,28].
GBM has long been considered as a non-immunogenic tumor due to immunosuppressive adaptation mechanisms, low levels of T cells, dendritic cells and monocytes, decreased IgG and IgA and increased regulatory T cells [29]. Many different recent reviews focus on novel therapies that harness the immune system, including vaccination, T-cell therapies, immune check-point modulators or adaptive immunotherapy [30,31,32,33].
Targeting tumor cell metabolism is another option. GBM is a hypoxic tumor. Hypoxia plays a role via different hypoxia inducing factors, HIF-1α and HIF-2α [34]. HIF1-α or factors implicated in the HIFs pathways have been proposed as potential therapeutic targets (as for examples profilin-1 or FIH1) [35,36,37]. To date, one Phase II clinical trial has been performed via the inhibitor of HIF2α, PT2385 [38] (NCT03216499).
Approaches aiming to exploit the metabolic deregulation of tumor cells compared to healthy cells are also increasing and characterization of specific metabolic pathways and metabolites are under intense investigations. Tumor cells have an increased need for glucose compared to healthy cells [39]. Thus, unlike healthy cells that use mitochondrial oxidative phosphorylation to generate ATP, tumor cells use aerobic glycolysis (the “Warburg effect”) [40]. Based on this concept, reduction of glucose delivery to tumor cells, for example, might influence their growth without influencing normal cells [41].
Delivery of low-intensity, intermediate-frequency (100–300 kHz) alternating electric fields through the TTFields, Optune®, Novocure Inc., Portsmouth, NH USA (tumor treatment fields) device has given an alternative strategy to treat GBM. It was approved by the FDA since 2011 for recurrent GBM. Beside antiproliferative and anti-mitotic effects, this device efficacy might also be related to inhibition of migration, invasion, angiogenesis and DNA repair as well as induction of apoptosis and immune effects [42].
GBMs are characterized by a high molecular and transcriptional inter- and intra-tumoral heterogeneity [43,44,45,46]. Developments in multi-omic analysis have led to identification of specific molecular signatures [47,48,49] discriminating at least 3 different subclasses (mesenchymal, proneural and classical) but also emphasized a high degree of plasticity between cellular states [50]. Nevertheless, proposition of targeted therapies has increased these last years based on promising preclinical data which supported the initiation of clinical trials. The aim of this paper is to make an exhaustive review of the different clinical trials (completed or under way) focusing on drugs considered as targeted therapeutics. We have divided the topic in 4 different sections considering drugs inhibiting (1) stem cells and stem cell pathways (Section 3.1), (2) the growth autonomy and migration (Section 3.2), (3) the cell cycle and escape to cell death (Section 3.3) and (4) angiogenesis (Section 3.4). Clinical trials of phases I/II, II, III or IV have been considered but not those of Phase I.

2. Methods

1 April 2020 has been set as the end date for data collection for this study. The flowchart (Figure 2) lists the clinical trials included and excluded from this manuscript. Briefly, 1519 clinical trials were listed on www.clinicaltrials.com (accessed on 1 April 2021) for GBM. Restrictions were applied to keep only clinical trials on adults and phases I/II to IV. 788 clinical trials remained (212 Phase I/II, 488 Phase II, 14 Phase II/III, 70 Phase III & 4 Phase IV). They have then been sub-classified: 257 clinical trials concerning targeted therapies are described in this review, and 531 clinical trials were excluded from this analysis as they are related to (i) RT, irradiation, imaging, classic cytotoxic chemotherapy, surgery, (ii) immunotherapy and vaccine therapy, (iii) other tumors than adult brain tumors, and (iv) other studies, such as withdrawal trials, trials which did not retain enough patients or did not pass phase II, studies on hypoxia, metabolism, anti-depressants, vitamins, hormones, molecules for sleep disorders, or cognitive decline, or drugs for which molecular targets are not clearly identified.
To recapitulate, the 257 clinical trials described in this review cover 20-years of targeted therapies in clinical phases I/II and over, for adult GBM. In addition to GBM, clinical trials including gliomas, high grade gliomas, gliosarcomas, anaplastic astrocytomas, or other brain tumors were retained. Children and young patient brain tumors were excluded.
Twelve tables detail the different clinical trials underway or completed in phases I/II, II, III or IV. The dates mentioned correspond to the start of the clinical trial and the last date of data update on Clinicaltrials.com. In tables, comparative trials with a significant difference between two treatments are highlighted in green and those with a non-significant difference are highlighted in red.

3. Results-Glioblastoma Targeted Therapies

The different GBM biomarkers targeted in phases I/II, II, III and IV and described in the following paragraphs are presented in Figure 3.

3.1. Targeting Stem Cells and Stem Cell Pathways

The discovery of tumor stem-like cells in solid tumors including glioma [59,60] has changed the landscape of the origin of tumors and their recurrence. These cells also named “GBM initiating cells” (GICs) or “GBM stem cells” (GSC) [61,62] exhibit self-renewal capacity and differentiating ability to form the tumoral mass [63]. The presence of GICs can be explained by the malignant transformation of neural (non-tumor) stem cells [64] and/or by the de-differentiation of tumor cells into tumor stem cells following radiotherapy or chemotherapy [65].
GICs are reported to be more resistant to current treatments than differentiated tumor cells explaining their role in GBM recurrence. This increased resistance can be explained by (1) a quiescent condition, resulting in the ineffectiveness of currently used chemotherapies targeting the cell cycle [66], (2) High expression of efflux transporters, including MRP1 (Multidrug resistance-associated protein 1) and P-gP (Permeability-GlycoProtein), evicting therapeutic molecules and (3) a defective regulation of apoptosis, with higher expression of survival factors and an ability to adapt to a stressful environment [67].
The discovery of GICs has generated hope for new therapeutic targets. Eradicating GICs would prevent the initiation of GBM on the periphery of surgical resection and reduce drug resistance and recurrence [68]. Three strategies are currently being studied to induce apoptosis of GICs: (i) directly targeting the signaling pathways involved in the self-renewal of GICs (Table 1), (ii) inducing their differentiation to sensitize them to therapies, and (iii) inhibiting the pathways that control their resistance.

3.1.1. Targeting the Self-Renewal of GICs

(i)
Wnt pathway
The Wnt signaling pathway is involved in the development of neural stem cells [72]. Aberrant activation of this pathway is involved in their malignant transformation and the development of brain tumors [73]. The Wnt pathway is also involved in the invasion of GBMs and in the epithelial-mesenchymal transition. Inhibiting the Wnt pathway in GICs leads to the sensitization to TMZ by decreasing the transcription of the transport proteins ABCC2 (MRP2) and ABCC4 (MRP4) [74]. Two proteins are being investigated in the inhibition of the Wnt pathway: β-catenin and GSK3-β. Diclofenac and Celecoxib, non-steroidal anti-inflammatory drugs, respectively, have been shown to inhibit β-catenin and to induce a decrease in the proliferation and migration of GBMs cells [75]. Tested in Phase II in newly diagnosed GBMs, combined with TMZ, Celecoxib had no survival benefit (NCT00112502) [69]. Two GSK3-β inhibitors were assayed in preclinical assays on GBMs cells: AR-A01441 and LiCl. These two agents increase the apoptosis of GBMs cells, decrease neurosphere formation and clonogenicity [76]. Two new selective inhibitors of the Wnt pathway have been synthesized: SEN461 and XAV939 [77]. In vitro, SEN461 is known to be responsible for the inhibition of GBM cell growth. However no clinical trials have analyzed the efficacy of GSK3-β inhibition [78] in vivo.
(ii)
Notch pathway
The Notch pathway is involved in invasion, resistance to anti-VEGF (Vascular endothelial growth factor) therapies and recurrences of GBMs [79,80,81]. Activation of this pathway induced by one of its ligands (Delta and Jagged) results in the cleavage of the Notch receptor, allowing the release of the receptor’s intracellular domain and its translocation to the nucleus. Notch’s cleavage is mediated by α and γ-secretase [82]. It has been suggested that targeting the Notch pathway via inhibition of γ-secretase [83,84] may be useful. Several inhibitors have been tested in vitro, such as MRK003 [85], GSI (RO4929097) [86], and dnMAML [87]. Only the GSI compound (R04929097) is currently being tested in clinics (Table 1). A Phase I study, investigating the toxicity of GSI combined with Bevacizumab, showed encouraging results (NCT01189240). The study is being pursued in a Phase II study [88].
(iii)
Hedgehog (SHH) pathway
The SHH signaling pathway is associated with resistance to radiotherapy and chemotherapy. Two main effectors of this pathway exist: SMO (smoothened) and Gli1 (glioma-associated oncogene homolog 1) [89,90,91]. SMO inhibition is achievable via two inhibitors, LDE225/Sonidegib and GDC-0449/Vismodegib [92]. The latter is currently in clinical trials (Phase II) in recurrent GBMs (NCT00980343) and (Phase I/II) in patients with newly diagnosed GBM without O6 methylguanine methyl transferase (MGMT) promoter methylation (NCT03158389, referred below as N2M2 (NOA-20), NCT Neuro Master Match the umbrella protocol for Phase I/IIa trials of molecularly matched targeted therapies combined with RT) [71].
Glasdegib (PF-04449913), another SMO inhibitor that has demonstrated potent and selective inhibition of Hedgehog signaling in vitro, and significant antitumor efficacy in vivo in various solid and hematologic malignancies [93], is a rational therapeutic agent currently in phase I/II for patients with newly diagnosed GBM, since it inhibits SHH pathway interfering with cancer stem cells and endothelial migration.
Gli1 can be inhibited by the cyclopamine. This steroidal alkaloid induces a decrease in the number of GICs and leads to RT sensitization [94]. The optimization of cyclopamine, by addition of a glucuronide group, showed a decrease in the tumor mass without having the toxic effects of Gli1 inhibition in astrocytes. This formulation specifically targets tumor cells expressing the beta-glucuronidase enzyme [95]. Similarly, the formulation of cyclopamine in micelles leads to inhibition of the proliferation and invasion of GBMs cells. This formulation also enhances the cytotoxic effect of TMZ in vivo [96]. No clinical studies have tested Gli1 inhibition.
(iv)
STAT3 pathway
The transcription factor STAT3 has an established function in neural stem cell and astrocyte development. It has been found to play dual tumor suppressive and oncogenic roles in glial malignancy depending on the mutational profile of the tumor [97]. Napabucasin (BBI608), a small molecule that blocks stem cell activity in cancer cells by targeting the STAT3 pathway, is currently in clinical Phase I/II in combination with TMZ in adult patients with recurrent or progressed GBM (NCT02315534, Table 1).

3.1.2. Inducing the Differentiation of GICs or Inhibiting Pathways That Control Resistance

Very few clinical trials addressing these points are currently developed although new targets are suggested through preclinical explorations.
As previously mentioned, inducing differentiation of GICs would sensitize them to current therapies. Simulating the BMP (Bone Morphogenetic Proteins) pathway is possible by different mechanisms:
-
Activation of an effector of the BMP pathway, such as BMP-7, blocks the tumor progression in vitro [98].
-
Using mimic effectors of the BMP pathway: the BMP-2 protein mimicking peptide, GBMP1, has been developed to activate this pathway and is currently being studied [99]. Activation of the BMP pathway is currently tested in clinical trials. A Phase I study is testing the recombinant protein hrBMP4 in recurrent GBMs (NCT02869243).
A new strategy aims to target adenosine, which is involved in GIC chemoresistance [100,101]. Physiologically, adenosine is produced by the degradation of AMP by the factors CD39 and CD73. In GBMs cells, CD73 expression is increased and leads to an increase in adenosine levels [102]. An increase in the A3AR adenosine receptor has also been observed in GBMs cells. Inhibition of A3AR receptor expression induces a decrease in MRP1 activity and increased sensitivity to chemotherapy [102,103]. CD73/A3AR/MRP1 is a potential therapeutic target, not yet tested in a clinical setting.
Two other adenosine receptors, A1B and A2B, are involved in apoptosis and GIC differentiation. The stimulation of these receptors by agonists helps to sensitize GICs to chemotherapy [104].

3.2. Targeting Growth Autonomy and Migration

Mutations in RAS/MAPK and PI3K/AKT pathways are reported in 88% of GBMs [105]. Their hyperactivation plays a central role in cell survival, growth, angiogenesis and cellular metabolism. It is mainly caused by ligand-induced stimulation of tyrosine kinase receptors (RTKs), such as epidermal growth factor receptor (EGFR) and platelet-derived growth factor receptors (PDGFR). The different RTKs are activated by the autophosphorylation of their tyrosine kinase domain, which results in the binding and activation of PI3K. The activated PI3K transforms PIP2 into PIP3. The latter binds to AKT and transports it to the plasma membrane where residues are phosphorylated by PDk-1 (on Thr308) and mTORC2 (on Ser473). The activation of AKT leads to a phosphorylation cascade and to the activation of several proteins involved in cell growth, angiogenesis and apoptosis, including mTOR and its partner mTORC1. One of the main inhibitors of this pathway is PTEN, which prevents the transformation of PIP2 into PIP3 [106].
The RAS/MAPK pathway activation results in the transformation of GDP to GTP, recruitment of RAF to the membrane and its activation, and ERK phosphorylation.
Targeting the different effectors of these pathways would reduce growth autonomy and migration of the GBM.

3.2.1. Inhibition of EGFR and HER2

The ErbB family of proteins contains four receptor tyrosine kinases, structurally related to the epidermal growth factor receptor (EGFR or HER1). EGFR and HER2 are promising anti-tumor targets for the therapy of GBM (Table 2).

i. Inhibition of EGFR

The EGFR signaling drives cancer development. EGFR aberrant expression and signaling promotes cell growth, survival, invasion and angiogenesis, and regulates tumor metabolism and cell stemness [107]. EGFR is a clinical target in solid tumors. In GBM, EGFR is amplified and/or mutated in more than 50% of cases [108]. EGFR and its mutant EGFRvIII are the subjects of extensive research. Several strategies are proposed to inhibit these receptors, including monoclonal antibodies, tyrosine kinase inhibitors (TKI) and anti-tumor vaccines. The first two classes are described in this review (Table 2).

Monoclonal Antibodies

Cetuximab was the first chimeric antibody proposed to target EGFR. Two Phase II studies did not show any therapeutic benefit in patients with recurrent GBM, either as monotherapy [128] or in combination with Bevacizumab and Irinotecan [110].
Panitumumab, the first fully human monoclonal anti-EGFR antibody to enter clinical trials for the treatment of solid tumors, did not prove to be beneficial for GBM patients in a phase II with irinotecan (NCT01017653).
Nimotuzumab, a humanized anti-EGFR antibody, also did not show a gain in overall survival (OS) or progression-free survival (PFS) in patients newly diagnosed and treated with the Stupp protocol (phase III) (NCT00753246) [111]. These results were disappointing compared to an earlier study that showed that the combination of nimotuzumab with RT resulted in prolonged survival [129]. Nimotuzumab remains a potential interesting therapy. Indeed, an enhancement of the cytotoxic activity of TMZ in vivo has recently been observed [130].
GC1118, an anti-EGFR antibody which seems more potent to inhibit EGF binding to EGFR than cetuximab or panitumumab [131] is currently being tested as monotherapy (NCT03618667).
Sym004 is a synergistic antibody combination containing two recombinant mAbs (futuximab and modotuximab) which binds to different non-overlapping epitopes of EGFR and promotes a rapid EGFR internalization and degradation. Sym004 overcame cetuximab resistance in pre-clinical lung cancer cells [132]. However, it did not improve OS in patients with metastatic colorectal cancer [133]. In GBM, it is evaluated as monotherapy (NCT02540161).
Depatuxizumab-mafodotin (ABT-414) is an antibody-drug conjugate (ADC) composed by an anti-EGFR IgG conjugated to the tubulin inhibitor monomethyl auristatin F [134]. Depatuximab-mafodotin failed to show survival benefit in newly diagnosed GBM but used in combination with TMZ in EGFR amplified recurrent GBM presented a possible efficiency [135].

Tyrosine Kinase Activity Inhibitors

Erlotinib is a reversible inhibitor of EGFR tyrosine kinase activity. Two Phase II studies did not show any improvement in OS when combining erlotinib and bevacizumab with TMZ as adjuvant therapy to the Stupp protocol in newly diagnosed patients [114,136]. Similar results were observed in a Phase II study analyzing the efficacy of Erlotinib in combination with sorafenib [113].
Gefitinib is a reversible and specific inhibitor of EGFR tyrosine kinase activity. Combined with RT in newly diagnosed patients, OS is not improved compared to RT alone [124], nor is it improved as adjuvant after RT [137].
Afatinib, an irreversible pan-inhibitor of the ErbB family (including EGFR and EGFRvIII) did not show better results than TMZ in a Phase II study (NCT00727506). Nevertheless, an increase in PFS has been observed in patients with tumors expressing EGFRvIII or with EGFR amplification [125].
Dacomitinib is a pan-HER family inhibitor (EGFR, HER2, and HER4), approved as first-line treatment of EGFR mutant NSCLC. In GBM, dacomitinib was tested as monotherapy in tumors with EGFR amplification or with the presence of the most common EGFR mutation in GBM EGFRvIII, but it provided minimal benefits [126].

ii. Inhibition of HER2

HER2 tends to be activated by forming heterodimers with other members of the family or other receptors, since no activating-ligand is known [138]. HER2 overexpression in breast cancer cells promotes tumor aggressiveness and thus became a therapeutic target combined with a companion test [139]. HER2-targeted antibody trastuzumab in breast cancer is a successful example of a targeted therapy.
Even though HER2 expression is low in GBM cells, multitargeted TKI of HER2, EGFR and VEGFR family are being tested in clinical trials.
Lapatinib and neratinib are two treatments used in HER2-positive breast cancer. In GBM, Lapatinib, a dual EGFR and HER2 kinase inhibitor, did not provide therapeutic gain in patients with recurrent GBMs in a Phase II study [140]. This compound together with TMZ and RT in newly diagnosed patients is in clinical trials (NCT01591577) [141].

3.2.2. Multikinase Inhibitors

Series of multikinase inhibitors have been tested in GBM (Table 3 and Table 4). Usually developed initially against one specific target, they proved able to inhibit different RTKs or non-receptor kinases as their ATP/ADP binding pocket revealed similarities. This characteristic may have advantages as simultaneously inhibiting several kinases may limit drug resistance and compensatory pathways [142]. Most of them are able to target EGFR, PDGFR, vascular endothelial growth factor receptors (VEGFR) known targets of GBM or even HER2, a target in breast cancers.
Anlotinib inhibits VEGFR, FGFR, PDGFR and c-kit [143]. Anlotinib is tested in GBM clinical trials as monotherapy or combined with Stupp protocol.
TG02 is an inhibitor of CDKs, JAK2 and FLT3 able to penetrate the blood-brain barrier and is therefore an interesting therapeutic for brain tumors [144]. TG02 is assayed in GBM in combination with TMZ (NCT02942264).
Tesevatinib is an inhibitor of EGFR, HER2, VEGFR and ephrin B4 [145], used in polycystic kidney disease and tested as monotherapy in GBM (NCT02844439).
Vandetanib, an inhibitor of EGFR, VEGFR2 and RET, has shown encouraging preclinical results. A 94% decrease in xenograft tumor size was observed when combined with TMZ and compared to TMZ alone [146]. However, the addition of vandetanib to the Stupp protocol does not prolong the survival of newly diagnosed patients (NCT00441142) [147].
Other multi-kinase inhibitors, such as cabozantinib, TG02, bosutinib are tested in GBM. All clinical trials, ongoing or completed, are listed in Table 3.
(i)
Inhibition of PDGFR
Similar to EGFR, the PDGF receptor is involved in the activation of the PI3K pathway. It is overexpressed or amplified in 75% of GBMs and thus appears as an interesting therapeutic target [150]. PDGFR inhibition has been largely explored in GBM. However, no specific PDGFR inhibitor exists and inhibitors are multikinase inhibitors (Table 4).
Imatinib was the first inhibitor targeting PDGFRα/β, BCR-Abl, c-kit. Although Imatinib has not shown clinical benefit in combination with hydroxyurea [151], it is currently in clinical trials.
Dasatinib, an inhibitor of PDGFRβ, EPHA2, BCR-Abl, c-kit and SRC, was ineffective in a Phase II study in patients with recurrent GBMs [152].
Tandutinib, an inhibitor of PDGFRβ, FLT3, c-Kit, was tested in a Phase II study with Bevacizumab in patients with recurrent GBMs. The results indicated that this combination does not improve patient survival compared to standard therapy (NCT00667394) [153]. Another Phase II study showed similar results and was stopped [154].
Sunitinib, an inhibitor of PDGFRα/β, c-kit, VEGFR1/2/3, FLT3 and RET, also provided disappointing results. A Phase II study did not show any clinical benefit of sunitinib in patients with recurrent GBMs compared to bevacizumab or conventional chemotherapies [166]. Similar results were observed in newly diagnosed non-operable patients [160].
Regorafenib inhibits a mutant isoform of BRAF (BRAFV600E), KIT, RET, angiopoietin 1 receptor, PDGFRα, VEGFR1/2/3 and FGFR1/2 [167]. In GBM, it is evaluated as monotherapy or together with the Stupp protocol.
Crenolanib, an inhibitor PDGFR and FLT3 is evaluated as monotherapy in recurrent GBM with PDGFRα gene amplification (NCT02626364).
Ponatinib (AP24534), a multi-targeted kinase inhibitor of BCR-Abl, PDGFRα, VEGFR2, FGFR1, and Src [168] but also RET, KIT, and FLT1, is assayed as a monotherapy in recurrent GBM refractory to bevacizumab (NCT02478164).
Leflunomide, an antimetabolite and inhibitor of PDGFR, EGFR and FGFR, is used for the treatment of rheumatoid arthritis. In preclinical trials, the active compound inhibited glioma cell proliferation in vitro and in vivo. Now it is evaluated as monotherapy in GBM (NCT00003293).
Besides these multi-target drugs, specific anti PDGFR antibodies have been designed and tested in GBM. A fully human anti-PDGFR antibody (IMC-3G3) blocks ligand binding and receptor activation and is being tested in different solid tumors [169]. A comparative clinical trial between IMC-3G3 monotherapy and ramucirumab (targeting VEGFR2) monotherapy did not show improved survival (NCT00895180).
Another monoclonal anti-PDGFRα antibody, MEDI-575, was well tolerated but showed limited clinical activity in GBM [163].
(ii)
Inhibition of IGFR1 and FGFR
Insulin-like growth factor 1 receptor (IGF1R) activation by its ligand IGF1 promotes GBM cells survival through PI3K/AKT pathway activation. Thus, inhibition of IGF1R may be an interesting strategy to suppress GBM progression [170]. Moreover, IGF1R overexpression in GBM is correlated with a shorter survival and lack of response to TMZ [171]. A phase I/II clinical trial (NCT01721577, Table 4), used AXL1717, an antagonist of IGF1R, as a single agent in the treatment of recurrent malignant astrocytomas. Monotherapy was well tolerated. Further optimizations in dose need to be performed [165].
Mutations of fibroblast growth factor receptor (FGFR) are rare in GBM but signalling through FGFRs impacts GBM progression and patient survival [172]. For example, fusion between FGFR and TACC (transforming acidic coiled-coil containing proteins) enhances tumor-growth and aneuploidy events [173]. FGFR1,2,3 mutations and fusion are targeted by BGJ398 (Table 4) as monotherapy in a phase-II clinical trial in GBM. However, BGJ398 was out licensed and no more studies were performed.
(iii)
Inhibition ALK
Anaplastic lymphoma kinase (ALK), a transmembrane receptor tyrosine kinase that belongs to the insulin receptor superfamily, is expressed in about 60% of GBMs and conveys tumorigenic functions. Second-generation ALK inhibitors, such as alectinib, might be novel therapeutic agents against GBMs, as they induced cell death in various human GBM cell lines with lower concentrations than other ALK inhibitors. The specific anti-tumor mechanism of alectinib is not yet described [174]. Alectinib is currently tested in the N2M2 Phase I/IIa clinical trial (NCT 03158389, Table 4) [71].

3.2.3. Inhibition of the PI3K/AKT Pathway

Table 5 describes the clinical trials concerning the inhibition of the PI3K/AKT pathway.
(i)
Inhibition of mTOR.
Another target in the PI3K/AKT pathway is mTOR. Several mTOR inhibitors are available and tested in clinical trials.
Among them, temsirolimus, which has recently been shown to target GICs [187], is the subject of many clinical trials. Two Phase II studies did not show clinical benefits when combined with bevacizumab [175] or sorafenib (NCT00800917) [188]. More recently, a Phase II study comparing the combination of temsirolimus with RT in newly diagnosed patients did not show any difference in survival compared to the Stupp protocol (NCT01019434) [178]. It is actually tested in the N2M2 (NOA-20) clinical trial (NCT03158389) [71].
Sirolimus (rapamycin) showed promising preclinical results by decreasing 95% tumor mass in vivo [189]. In addition, it also decreased the proliferation of GICs [190] and their differentiation [191]. Despite these results, sirolimus combined with erlotinib is not effective in GBM recurrence (NCT00672243, Table 2) [116].
Similar results were observed with everolimus. A Phase II study showed that the administration of everolimus before the Stupp protocol in newly diagnosed patients does not provide any clinical benefit compared to the standard protocol [181].
ABI-009 is a novel albumin-bound mTOR inhibitor (albumin-bound rapamycin nanoparticles, nab-rapamycin), currently tested as single agent or in combination with standard therapies (NCT03463265) in a Phase II study.
AZD2014, an inhibitor of both mTORC1 and mTORC2, causes radiosensitization of GICs in vitro and in vivo [192]. This compound is currently in a Phase I clinical trial (NCT02619864).
(ii)
Inhibition of PI3K
Several PI3K pan-inhibitors have shown promising in vitro and in vivo results, some of which are being tested in clinical trials.
Pictilisib is an isoform inhibitor of PI3K α/δ. Combined with RT and TMZ, it has a pro-apoptotic action, increases autophagy and decreases the migration capacities of GBMs cell lines. In vivo, it increases sensitivity to RT and TMZ [193]. Pictilisib was compared with pembrolizumab in a phase I/II study but data are not published (NCT02430363).
Buparlisib (BKM120) inhibits cell invasive capacities in vitro and reduces tumor invasion in vivo [194,195]. It is currently being tested in two phase I/II and II studies (NCT01349660 NCT01339052). In the Phase II study (NCT01339052), buparlisib achieved significant brain penetration, but had low efficacy in patients with PI3K-activated recurrent GBM, which was explained by incomplete blockade of PI3K pathway in tumor tissue [196].
Sonolisib (PX-866), an isoform inhibitor of PI3K α, δ and γ reduces the invasive and angiogenic capacities of GBM cells in vitro. In vivo, decreased tumor growth and increased survival of xenografted mice [197] were observed. A Phase II study did not show clinical benefit in the case of recurrent GBMs (NCT01259869) [183].
Paxalisib (GDC-0084) is a brain-penetrant small molecule inhibitor of the PI3K/AKT/mTOR pathway. An interim analysis from Kazia Therapeutics reviewed OS of 17.7 months (nine patients) compared to the median OS for patients treated with TMZ (12.7 months). Final data of the phase II trial (NCT03522298) are expected to be presented in the first half of 2021, but FDA has already granted fast track designation to paxalisib.
(iii)
Inhibition of AKT
Enzastaurin is an inhibitor of AKT and protein kinase C. This molecule was the first to provide clinical benefit in a subgroup of patients with recurrent GBMs according to their MGMT status [185]. Enzastaurin has been compared to lomustine in a Phase III clinical trial (NCT00295815). Median PFS, 6-month PFS rate and OS did not differ significantly between enzastaurin and lomustine. Enzastaurin was well tolerated, had a better hematologic toxicity profile but did not have superior efficacy compared with lomustine in patients with recurrent GBM [184].
Other AKT inhibitors with promising results are being tested in preclinics or Phase I, such as perifosine [198], nelfinavir [199], MK2206 [200].

3.2.4. Inhibition of RAS/MAPK Pathway

RAS/MAPK pathway is activated by many receptors including tyrosine kinase receptors and involved in cell survival and proliferation. RAS/MAPK has been targeted in GBM (Table 6).
One inhibitor of this pathway, TLN-4601, did not demonstrate therapeutic efficacy in monotherapy in a Phase II study in the event of recurrence [201].
Sorafenib is a Raf-1 and p38 inhibitor, involved in the RAS-MAPK, VEGFR, c-kit and PDGFR pathways [206]. Although sorafenib has been shown to potentiate the pro-apoptotic effect in GBMs cells [207], it does not appear to improve sensitivity to radiotherapy and chemotherapy in vivo [208]. For clinical trials, the combination of sorafenib and TMZ in recurrent GBMs provides a PFS of 3.2 months and an OS of 7.4 months [209]. Combined with bevacizumab [204], erlotinib [114] and temsirolimus [188], it does not provide clinical benefit. Disappointing results were also observed in newly diagnosed patients treated with sorafenib and combined to the Stupp protocol in adjuvant therapy [202].
Two Ras-MAPK inhibitors are in Phase II clinical trials: LY2228820 and atorvastatin. The latter molecule could potentiate the effects of TMZ in vitro and in vivo [210]. In a Phase II study (NCT02029573) in combination with standard therapy (RT/TMZ) in newly diagnosed GBM patients, preliminary results are encouraging and met criteria for continued accrual [205].
Dabrafenib is a BRAF inhibitor that binds and inhibits the active conformation of the receptor. Dabrafenib is evaluated in combination with the MEK inhibitor trametinib in newly GBM (NCT03919071).
A very recent study includes binimetinib (a MEK inhibitor) with encorafenib (a BRAF inhibitor) in adults with recurrent BRAF V600-Mutated HGG (NCT03973918).
The lipid proliferation switch led to the discovery of a novel anticancer drug target, the tumor repressor protein sphingomyelin synthase 1 (SGMS1). The activation of SGMS1 by 2OHOA, a synthetic hydroxylated fatty acid, modulates the lipid content of cancer cell membranes, regulates the localization of key signalling proteins, including Ras and PKC at the plasma membrane, leading to inactivation of Ras/MAPK, PI3K/Akt and PKC/cyclin/CDK signalling pathways [211]. The clinical trial in Phase I/IIa NCT01792310 demonstrated its safety and efficacy in humans. 2OHOA was designed as orphan drug by the European Medicines Agency (EMA) for the treatment of glioma and is now tested in a Phase IIb study (NCT04250922).

3.3. Targeting the Cell Cycle and Escape to Cell Death

A major reason for the failure of chemotherapy is the resistance of GBM cells to cell death by apoptosis, necrosis or autophagy [212,213].

3.3.1. Therapies Targeting Apoptosis

Apoptosis can be mediated by the extrinsic and the intrinsic pathways. The extrinsic pathway results from the activation of the TNF-R1, FAS and DR4/DR5 death receptors through their respective ligands TNFα, CD95 and TRAIL [214]. The intrinsic pathway is regulated by proteins of the BCL-2 family and of the inhibitor of aptotosis (IAP) family. Pro and anti-apoptotic members of the BCL2 family regulate mitochondria-dependent cell effects. When apoptosis is triggered mitochondria become permeable and release cytochrome C. The two pathways converge on a series of catalytic cascades involving caspases [105]. The tumor suppressor p53 is implicated in several pro-apoptotic pathways and appears mutated in about 30% of GBM. Restoring apoptosis may be obtained by targeting different apoptosis players (Table 7).
(i)
Activating proteins involved in the extrinsic pathway of apoptosis
The CD95 death receptor is overexpressed in GBMs and mesenchymal GICs. It is also associated with epithelial-mesenchymal transition [219]. APO010 and APG101 are two CD95 agonists. APO010 has significant anti-tumor activity in GICs, increasing their sensitivity to TMZ in vitro. Administered locoregionally, APO010 increases mice survival [220]. A phase II study showed that the combination of the agonist APG101 with re-irradiation in recurrent GBM improves PFS but not OS compared to re-irradiation alone. This therapeutic benefit is more pronounced in mutated IDH tumors [215].
TRAIL/DR5 dependent cell death can be induced by ONC201. ONC201 binds and antagonizes dopamine receptors DRD2 and DRD3 causing p53-independent apoptosis in tumor cells. ONC201 inhibits the phosphorylation of AKT and ERK pathways, leading to the dephosphorylation of transcription factor FOXO3A, and thus transcription of pro-apoptotic death receptor ligand TRAIL. Through a stress response activation ONC201 is involved in EIF2α phosphorylation and increases DR5 expression [221,222] Based on the the first results using ONC201 in monotherapy which showed that the treatment was well tolerated and that ONC201 may have single agent activity in GBM [223], a phase II clinical trial was started on GBM with H3 K27M mutation (NCT02525692). It showed that ONC201 can be used regardless of age or location [216].
(ii)
Activating proteins involved in the intrinsic pathway of apoptosis
The TSPO protein is involved in the permeabilization of the mitochondrial membrane. Its level of expression being correlated with a poor prognosis, it is considered a potential target for apoptosis restoration [224]. Several ligands of TSPO (Translocator protein), derived from pyrazolo[1,5-a]pyrimidine acetamides, are able to specifically reduce the proliferation of GBMs cells [225]. No clinical trials are underway with these new molecules.
(iii)
Targeting proteins involved in the regulation of apoptosis
Due to its role in regulating both pathways of apoptosis, targeting the p53 protein has also been suggested to reactivate its pro-apoptotic functions, by gene therapy or by inhibiting its interaction with MDM2 [226,227].
In a recent study, a tumor-targeting p53 nanodelivery system (SGT53) showed sensitization of resistant GBM cells to TMZ in vitro and increase in the survival of xenografted mice [228]. Gene therapy is currently in a Phase II clinical study (NCT02340156).
Inhibition of MDM2-p53 interaction to trigger apoptosis is an approach that showed encouraging preclinical results. Among these, ISA27 inhibits cell growth in vitro and in vivo [229] while nutlin-3a induces apoptosis and senescence of glioma cells [230]. α5β1 integrin-specific inhibition in association with nutlin-3a also triggered a strong apoptosis in glioma cells expressing a functional p53 [231]. Idasanutlin (RG7388) with more potency, selectivity, and better pharmacokinetic profile than other MDM2 inhibitors appears interesting in preclinical assays, is tested in clinical trials for acute myeloid leukemia and recently in the N2M2 (NOA-20) clinical trials in GBM (NCT01358389) [71]. Finally, the AMG-232 inhibitor has shown encouraging results including inhibition of tumor growth in several xenografts (lung, osteosarcoma, etc.) and tumor regression in mouse models [232]. This agent is currently in Phase I clinical trials (NCT03107780, NCT01723020).
Farnesyltransferase inhibitors (FTI) can induce apoptosis, as they revert cells to a state in which cell-substratum attachment is necessary for viability [233]. Inhibition of farnesyltransferase (FT) by tipifarnib blocks the prenylation of the farnesyltransferase tail CAAX motif, thereby preventing Ras binding to the membrane and its activation. Tipifarnib is tested in four clinical studies in monotherapy or combined with RT or TMZ or other targeted therapies (NCT00050986, NCT00058097, NCT00005859 and NCT00335764). Lonafarnib (SCH66336) is a FTI that blocks farnesylation of cell proliferation proteins, such as RhoB, RAS, laminins and CCAX phosphatase [234,235]. It inhibits in vitro [236] and in vivo [237] cell growth in combination with chemo and/or radiotherapy. A phase II was performed in combination with TMZ (NCT00038493).
Simultaneous reactivation of p53 and TSPO proteins appears to be more effective in promoting apoptosis in GBMs cells but also in reducing the risk of resistance [238]. Reactivating these proteins using molecules with irreversible action has been suggested in order to reduce the risk of recurrence [239].
Another potential approach is to target anti-apoptotic proteins from the BCL-2 family. The compound gossypol binds to the common part of proteins Bcl-2, Bcl-XL and Mcl-1 [240]. Its combination with TMZ was shown to inhibit the invasive and proliferative abilities of GBMs cells and angiogenesis in vitro, and to cause apoptosis in vivo [241]. Gossypol was tested as monotherapy in a phase II (NCT00540722).
Finally, a new therapy targeting the Bcl-2 protein consists of the administration of spherical nucleic acid (SNA). SNA-NU-0129, a formulation containing gold nanoparticles and a siRNA targeting BCL2L12, is involved in the inhibition of this protein and in the induction of cellular apoptosis in vitro [242]. A Phase I study is ongoing in recurrent GBMs and gliosarcomas (NCT03020017).

3.3.2. Therapies Targeting Autophagy

Autophagy is a degradation mechanism that can also induce cell death independently of caspases. It is based on the encapsulation of proteins, cytoplasm and organelles in vesicles that will be degraded in lysosomes. The pro- or anti-tumor function of autophagy in the GBM is still uncertain [243]. Molecules inducing autophagy, such as curcubitacin [244], itraconazole [245], rutin [246], givinostat [247] can have different consequences, but none of them are yet tested in Phase I/II or more.
In addition, chloroquine, inhibiting autophagy via lysosomal protease blockade and fusion between lysosomes and autophagosome [248], provoked a decrease in cell proliferation and migration, and an induction of apoptosis in vivo and in vitro [249]. Chloroquine is in Phase I and II clinical trials in combination with TMZ and/or RT (NCT02378532, NCT02432417, NCT00224978 & NCT00486603) (Table 8).

3.3.3. Targeting Multifaceted Pathways and DNA Modifications

Table 9 details all the clinical trials of this section.
(i)
CDK4/6 inhibitors
Cyclin-dependent kinases 4 and 6 (CDK4/6) signalling regulates cell cycle, cell differentiation, metabolism and apoptosis. In glioma cells, CDK4 is overexpressed which led to glioma cell proliferation and TMZ resistance [260]. CDK4/6 inhibitors (palbociclib/PD 0332991, abemaciclib) specifically blocked the cell cycle at the G1-to-S transition phase, leading to cell cycle arrest and stopped cell proliferation [261]. These inhibitors are approved in combination with anti-oestrogen therapies for the treatment of hormonal breast cancer, and are being studied in GBM upon surgical resection. Palbociclib is one of the drug tested in the GBM phase I/IIa trial NCT03158389 [71].
(ii)
Proteasome inhibitors
The proteasome is a central cellular protein-degradation machinery. It regulates cell homeostasis in normal and cancer cells. Bortezomib, the first-generation proteasome inhibitor, was approved for the treatment of multiple myeloma and mantle cell lymphoma [262]. This therapy is able to increase apoptosis levels in preclinical brain tumor assays. Moreover, clinical trials using proteasome inhibitors in combination strategies are being tested to maximize therapeutic efficacy and limit toxicity [263]. Bortezomib is studied in combination with TMZ and/or radiation, or with an inhibitor of histone deacetylase.
Marizomib, is a second-generation, irreversible proteasome inhibitor with a more lipophilic structure, having the ability to cross the blood-brain barrier [264]. It has been tested in patients with newly diagnosed and recurrent GBM in phase I and phase II studies. In patients with recurrent GBM, marizomib was administered in a Phase I/II study as a single agent or in combination with bevacizumab (NCT02330562) and in a Phase II study as a single agent or in combination with bevacizumab or RT/TMZ or ABI-009, or lomustine (NCT03463265). Based on encouraging observations [265], marizomib combined with RT/TMZ is actually in a Phase III study (NCT0334509).
(iii)
Histone deacetylase inhibitors
Epigenetic alterations in histones control chromatin structure and transcriptional activation. Besides their potential role in onset and progression of cancer, they are generally reversible and thus interesting therapeutic targets. Histone acetylation relaxes chromatin and allows access to DNA and transcription activation. On the other hand, histone deacetylases (HDAC) compacts chromatin and represses transcription [266]. HDACs can be essential for cancer cell survival and growth, showing an epigenetic vulnerability of tumor cells. HDAC inhibition can induce tumour cell cycle arrest, apoptosis, reduction of angiogenesis and enhancement of tumor-mediated immunity [266,267]. HDAC inhibitors [268] in GBM tends to re-establish the balance of histone acetylation and sensitizes tumor-mediated immunity. It can also sensitize tumor cells when used in combination, for example, with radiation therapy [267]. Several clinical trials are testing HDAC inhibitors as monotherapy or in combination in GBM. Vorinostat as a monotherapy had modest activity in patients and did not improve PFS or median OS in association with bevacuzimab (NCT01738646) or bortezomib (NCT00641706). Another HDAC inhibitor, FR901228 (Romidepsin), was ineffective for patients with recurrent GBM (NCT00085540).
(iv)
TGF-β inhibitors
Transforming growth factor-beta (TGF-β) is a cytokine secreted by immune cells, tumor cells, and stromal cells. TGF-β is overexpressed GBM tissues but inexistent in normal brain. TGF-β signalling regulates GBM proliferation, invasion, angiogenesis, immunosuppression, and GSCs stemness [269]. Targeting TGF-β signaling mechanisms is a promising therapeutic strategy [270]. In GBM clinical trials, TGF-β pathway are targeted by antisens oligonucleotide (trabedersen, NCT004331561) and by small molecules, OKN-007 (NCT03649464) [271], and galunisertib (NCT01582269, NCT01220271). Results are available for galunisertib and trabedersen.
Targeting of TGF-β2 signaling through inhibition of TGF-β mRNA translation by using the antisense oligonucleotides trabedersen, injected in the resection cavity, was tested in GBM in a Phase IIb (NCT00431561) but the first results did not show statistically significant differences among the three arms: trabedersen at doses of 10 or 80 mM or standard chemotherapy (TMZ or procarbazine/lomustine/vincristine) [256].
Galunisertib targets the TGF-β1 receptor and selectively inhibits the serine/threonine activity of the receptor, thereby preventing the phosphorylation of downstream proteins, SMAD2 and SMAD3. It demonstrated antitumor effects in preclinical and radiographic responses [272]. But no differences in efficacy, safety or pharmacokinetic variables were observed in a Phase Ib/IIa clinical trial (NCT01220271) between the two treatment arms (TMZ/RT with and without galunisertib) [257].
(v)
PARP inhibitors
Defects in DNA repair pathways are a characteristic feature of cancer cells. They participate in tumour development by promoting genomic instability. For more than 50 years, this characteristic has been exploited as a therapeutic opportunity for the treatment of cancer, with the use of conventional cytotoxic chemotherapies. More recently, the discovery of a synthetic lethality interaction between DNA damage induced by PARP (poly[ADP-ribose] polymerase) inhibitors led to the development of new therapeutic approaches. The PARP proteins use NAD+ as their substrate to modify acceptor proteins with ADP-ribose modifications. Most PARP inhibitors target the NAD+ binding site.
A high expression of PARP-1 mRNA is associated with low survival, particularly in classical GBMs [273]. A few molecules inhibiting PARP-1 are in clinical trials. Among them, iniparib (BSI-201) taken concomitantly with RT and TMZ has shown encouraging results, in human glioma xenografts, resulting in complete tumor regression in 70% of animals [274]. This PARP1 inhibitor plus TMZ was evaluated in a phase I/II in newly-diagnosed GBM (NCT00687765). Other NAD+ mimetics, olaparib (AZD2281), veliparib (ABT-888) and pamiparib (BGB-290) inhibit the catalytic activity of PARP-1 and PARP-2 and are currently being studied in phase I or I/II clinical trials. Only results for veliparib combined with TMZ (NCT01026493) are available [259]. The concept of this study was to exploit methylation at positions N3-adenine and N7-guanine, supposedly independent of the MGMT effect and related more to base excision repair with PARP. But the study did not demonstrate any clinical activity.

3.4. Targeting Angiogenesis

Angiogenesis is a complex process regulated by multiple signaling pathways. Due to a high tumor proliferation, access to oxygen and nutrients decreases in some areas of a tumor, leading to hypoxia and necrosis. GBM are highly angiogenic tumors and blocking neo-angiogenesis has represented an interesting therapeutic way for twenty years.

3.4.1. Targeting VEGF/VEGFR Pathway

Clinical trials for VEGF and VEGFR targeting are described in Table 10.
(i)
Bevacizumab
VEGF is overexpressed in GBMs and plays a major role in angiogenesis by activating its receptor VEGFR [275]. Since 2009, the food and drug administration (FDA) has approved bevacizumab, an anti-VEGF antibody, as a treatment in recurrent GBMs. Indeed, non placebo-controlled Phase II clinical trials highlighted the bevacizumab anti-tumor activity and this molecule is considered effective alone or in combination with Irinotecan, a topoisomerase I DNA inhibitor [276,277]. Based on encouraging results, few clinical trials were conducted to evaluate the efficacy of bevacizumab in comparative studies. However, results of these trials have been estimated insufficient by EMA to approve bevacizumab use in GBM in Europe. This discrepancy between drug authorities lead to huge off-label use of bevacizumab for GBM, mostly at recurrence, since this antibody is also marketed for the treatment of ovarian, lung, breast and colorectal cancer.
For other studies presented in Table 10, bevacizumab is usually the reference treatment of the control arm to be compared to combinations of bevacizumab plus other experimental molecules targeting different pathways.

Clinical Trials in Recurrent GBMs

In a Phase II study, the combination of bevacizumab and TMZ did not show a survival benefit compared to bevacizumab alone [294]. Similar results were observed in several other Phase II studies with bevacizumab in combination with temsirolimus [175], Carboplatin and irinotecan [306]. Only the combination of bevacizumab and lomustine appears to provide encouraging results in terms of survival and quality of life in a Phase II study [307,308]. However, these promising results were not demonstrated in a Phase III study, in which the combination therapy resulted in a PFS benefit but no OS improvement (NCT01290939) [292].
The efficacy of bevacizumab was also studied retrospectively in patients exposed to a second irradiation [309]. This study shows that bevacizumab might be a protective agent against a second irradiation. The improvement in irradiation with an anti-angiogenic agent was explained by the normality of vascularization during VEGFR blockade. Indeed, this “normalization window” allows a temporary increase in tumor oxygenation, which improves the damage induced by irradiation [310].

Clinical Trials in Newly Diagnosed GBMs

No benefit for bevacizumab with or without conventional treatment was obtained in different clinical trials [284,287,297,311,312]. Only one Phase II study, analyzing the combination of RT and bevacizumab followed by an adjuvant therapy combining bevacizumab and irinotecan, showed an improvement in PFS compared to the Stupp protocol in patients with non-methylated MGMT status [291]. A (non-significant) tendency towards an OS gain was also shown when TMZ was combined with bevacizumab in neo-adjuvant Stupp protocol therapy compared to the same protocol without Bevacizumab in non-operable patients [285]. Finally, it was retrospectively shown that proneural GBMs could benefit on the addition of bevacizumab compared to placebo (OS = 17.1 vs. 12.8 months HR = 0.43; p = 0.002) [313].
(ii)
Molecules targeting VEGFR
Pazopanib, a VEGFR1/2/3, PDGFR-α/β, and c-Kit inhibitor, administered as monotherapy, did not show therapeutic benefit in recurrent GBMs [298].
Cediranib is an oral, highly potent VEGFR inhibitor with similar activity against all three VEGF receptors and c-Kit and partial activity against PDGF receptors [314]. Cediranib, as monotherapy, has provided encouraging results in recurrent GBMs [302]. However, in combination with lomustine, cediranib did not show any therapeutic benefit, due to an increase in EGFR levels. Recently, a survival benefit has been reported with the combination of cediranib and gefitinib in recurrent GBMs [299].
Nintedanib, alone, did not show any survival benefit in recurrent GBMs [302]. Note that nintedanib is an inhibitor of VEGFR1/2/3, FGFR1/2/3 and PDGFRα/β.
Dovitinib, an FGFR, PDGFRβ, VEGFR and c-kit inhibitor, currently in clinical trials, sensitizes GBMs cells to TMZ in vitro [315,316].
Vatalanib is a VEGFR1/2/3, PDGFRβ and c-kit inhibitor. Its tolerance and safety were evaluated in a Phase I/II study (NCT00128700) in newly diagnosed patients [304] and in combination with imatinib and hyroxyurea in patients with glioma [317].
Most of these molecules have multiple targets. A few other molecules for which only a few clinical trials are ongoing and for which few results have been published, are listed in Table 10, such as tivozanib, axitinib, semaxanib, CT-322 (a molecule based on an engineered variant of the tenth type III domain of human fibronectin), and the monoclonal antibody tanibirumab (a specific binder to VEGFR2, thereby preventing the binding of its ligand VEGF).

3.4.2. The secondary Pathways of Angiogenesis

Table 11 shows the clinical trials concerning the secondary pathways of angiogenesis.
The failure of anti-VEGF therapies might be explained by compensatory mechanisms, through activation of other factors involved in angiogenesis in response to VEGF inhibition.
(i)
c-MET pathway
The c-MET pathway is deregulated because of an overexpression of (i) the c-MET receptor via mutation or amplification, or (ii) its HGF ligand [322,323]. Activation of this pathway is particularly important in the transformation of endothelial cells into mesenchymal cells, in the induction of aberrant vascularization and in tumor progression [324]. In addition, its activation is associated with a decrease in VEGFR2 expression, which leads to resistance to anti-VEGF therapies [325,326].
Onartuzumab, a monoclonal antibody targeting c-MET, induced a decrease in the growth of GBMs cells. Combined with bevacizumab in recurrent GBMs, ornatuzumab provides a PFS similar to bevacizumab alone. Nevertheless, this study showed a survival benefit in patients with high HGF expression or non-methylated MGMT status [318].
Other c-MET inhibitors have been developed and are currently being investigated. Among these, crizotinib (a c-MET and ALK inhibitor) causes GBMs cells to become sensitive to TMZ [327]. Crizotinib is currently being tested in combination with TMZ in a Phase I study (NCT02270034). Cabozantinib, a c-MET and VEGFR2 inhibitor, was tested in a Phase I study, combined with TMZ during the Stupp protocol [328] and in two Phase II studies as monotherapy in recurrent GBM (NCT01068782 and NCT00704288).
Targeting the c-MET ligand, HGF, is also being investigated. The anti-HGF antibody, rilotumumumab (AMG 102), did not show therapeutic benefit in monotherapy in a Phase II study in patients with recurrent GBMs [320].
(ii)
PIGF pathway
Another factor involved in angiogenesis is PIGF, a member of the VEGF family, binding to VEGFR1 (FLT1) and its neuropilin-1/2 co-receptors (NRP1/2). It is expressed in GBMs and tumor endothelial cells [329]. Aflibercept, also called VEGF-trap, is a recombinant fusion protein mimicking binding domain of VEGFR1 and VEGFR2 and blocking different ligands (VEGF-A, VEGF-B and PlGF). In monotherapy or in combination with bevacizumab in recurrent GBMs, no survival benefit was observed [321,330]. These disappointing results might be explained by a decrease in PIGF expression during tumor progression, in particular after treatment with TMZ. This new therapeutic option seems more relevant in newly diagnosed patients [331].
(iii)
Endoglin
Endoglin (CD105) is strongly expressed in endothelial cells with high proliferation rates [332]. TCR105 is a chimeric antibody targeting endoglin, which enhances the effects of bevacizumab in vivo, tested in two clinical trials (NCT01648348, NCT01564914). The combination of TRC105 and bevacizumab was well tolerated [333], but TRC105 with bevacizumab did not prolong median PFS versus bevacizumab alone in recurrent GBM patients [334].
Endoglin is also studied as a diagnostic marker and to estimate the degree of angiogenesis. The endoglin labelling is more typical of neoplastic endothelial cells and is correlated to Ki67, thus making it specific and sensitive to the evolution of angiogenesis in GBM [335].

3.4.3. Other Pathways of Angiogenesis

Other pathways of angiogenesis are described in Table 12.
Thalidomide is a long-established anti-angiogenic agent that inhibits the angiogenic activity of β-FGF and TNF-α [345]. However, when combined with RT in GBM, no benefit was observed in newly diagnosed GBMs [346]. It has shown limited gastrointestinal toxicity and anti-tumor activity in combination with irinotecan [337], and is currently in clinical trials in combination with the Stupp protocol in newly diagnosed GBMs (NCT00047294).
Integrins αvβ3 and αvβ5 have been proposed as targets of new anti-angiogenic therapies. Promising results have been observed when combining an inhibitor of these integrins, cilengitide, with the Stupp protocol in newly diagnosed patients [342,347]. Nevertheless, in two clinical studies (one phase II and one phase III), this combination did not show survival gains in patients with methylated [340] and non-methylated [341] MGMT status. ATN161 (Ac-PHSCN-NH2) is a selective antagonist for α5β1 integrin. It is a capped five amino-acid peptide derived from the synergy site of fibronectin, a region which enhances the fibronectin’s RGD-mediated binding to the α5β1 integrin. ATN 161 is antiangiogenic and antimetastatic [348] and was evaluated in a phase I/II trial for recurrent malignant glioma (NCT00352313).
Trebananib (AMG-386) is an angiopoietin neutralizing peptibody comprising a peptide with angiopoietin-binding properties that is fused to the Fc region of an antibody with an antiangiogenic effect in solid tumor. It inhibits the interaction between the ligands angiopoietin-1 and angiopoietin-2 with the Tie-2 receptor [349]. Angiopoietins (Ang1 and Ang2) and their RTK (TIE1 and TIE2) are key mediators of tumor angiogenesis. Angiopoietins are overexpressed in GBM and are involved in GBM tumor growth. Moreover, angiopoietin-2 increased in bevacizumab-treated GBM and thus VEGF and angiopoietin-2 combined therapy may overcome bevacizumab resistance. A phase II study used trebananib as monotherapy on patients with recurrent GBM (NCT01290263). Trebananib was also tested in combination with bevacizumab (NCT01609790). However, combination did not significantly improve outcome over bevacizumab alone. Moreover, angiopoietin recombinant humanized monoclonal antibody, PF-04856884, was enrolled on a phase II as monotherapy in patients with recurrent GBM (NCT01225510). This study, which was withdrawn, was not listed in Tables. Until now no further trials were performed in GBM.
Endostatin is a fragment of type XVIII collagen, and one inhibitor of angiogenesis. Endostatin competitively binds to VEGFR-2 and inhibits MAPK signaling pathway and angiogenesis [350]. Recombinant human endostatin improved chemotherapy efficiency in NSCLC, breast cancer and melanoma [351,352,353]. Endostatin is actually tested in GBM in a phase II study with TMZ and irinotecan (NCT04267978).
Prostate-specific membrane antigen (PSMA) expression has been demonstrated in the tumor neovasculature of GBM, by immunohistochemical staining [354]. Although its significance has not been fully determined, PSMA may play a functional role in angiogenesis [355]. It is anchored to the cell membrane, which makes it an ideal promising therapeutic target, and can be internalized making it an appropriate candidate for pro-drug activity. Strong reactivity to the antibody component of PSMA antibody-drug conjugate (ADC), BrUOG 263, was observed in the endothelial cells of new tumor blood vessels in GBM. Following binding and internalization of PSMA ADC, the cytotoxic component of PSMA ADC will be released and destroy the neovasculature that supports tumor growth.
Matrix metalloproteinases (MMPs), especially MMP2 & 9, are thought to play a central role in invasion, owing to their ability to degrade the majority of brain ECM components [356]. Prinomastat and COL-3 are two drugs targeting MMPs that may stop the growth of GBM by stopping blood flow to the tumor. They have been tested in two clinical trials. Prinomastat/TMZ compared to TMZ alone did neither improve the one-year survival rate nor PFS (NCT00004200). The clinical trial (NCT00004147) with COL-3 in progressive and recurrent high-grade gliomas did not warrant further studies and did not reach phase II [357].

4. Discussion-Guidance towards Future GBM Targeted Therapies

Out of 257 Phase I/II to III clinical trials on targeted therapies listed in the tables of this manuscript, almost 70% are phase II studies (62 Phase I/II, 177 Phase II, 4 Phase II/III, 14 Phase III). Of the studies for which results are available, only 37 are comparative studies with statistical data. Comparative trials with a significant difference between two treatments are highlighted in color in the tables, in green and red for those showing a significant and non-significant difference between two treatments, respectively. It is clear that the red color dominates over the green one. Only 12 studies showed improvements mainly of PFS. Most of them (11 out of 12) involve therapies targeting VEGF and VEGFR. Although some specific explanations may be proposed for the high degree of these clinical trial failures (see below), improved clinical trial design is also needed. For exemple, Phase II trials may contain a control arm to assess the efficacy of new therapies and to reduce false positive results which remains difficult to establish in the case of recurrent disease in absence of standard treatment; historical control data became obsolete due to the improvement of patient standard of care in the clinic [358,359]. GBM is a rare disease and enrollment of patients in trials remains too low, promotion of participation must be planned to increase the number of high-quality trials [360]. In addition, the need for stratification of patients at least based on prognostic and predictive biomarkers such as the level of the predictive target is critical. Biomarkers might also help to reduce the development costs through better patient selection. A recent study on the impact of biomarker use in clinical trials shows an overall 5-fold benefit over non-biomarker use by analyzing a collection of 10,000 clinical trials for 745 drugs in four major cancer types (colorectal, lung, melanoma and breast cancer) [361]. The neuro-oncology community must work together to be able to change favorably the guidelines on the treatment of GBM [362].
Many different targeted therapeutic options are investigated. For more recent trials, we identified two main tendencies. First, is underway a clear upward trend towards approaches with multi-kinase inhibitors (i.e., when a kinase inhibitor interacts with multiple members of the protein kinase family). The second trend is towards a multi-targeted therapeutic approach. Drugs able to target multiple critical nodes for GBM development and progression might help to counteract the lack of efficiency and the rapid acquisition of resistance observed with monotherapies [363].
Several factors can explain the therapeutic failure of GBM targeted treatments:
(i)
Performing a full surgical resection is impossible. Eliminating tumor cells that have migrated into the healthy parenchyma without causing neurological or cognitive disorders is not feasible. 35% of newly diagnosed patients are estimated to be non-operable due to the location or size of the tumor. In these cases, a biopsy is recommended in order to establish a diagnosis [364]. When surgery is possible, macroscopic resection is described as a good prognostic factor [365]. A recent meta-analysis showed that out of 27,865 patients diagnosed with GBM between 2004 and 2013, a biopsy (non-operable case), partial resection and massive resection accounted for 28.5%, 34.8% and 36.8% of cases [366].
(ii)
Crossing the BBB is not a turnkey operation, despite its potential destruction by tumor invasion or RT. New approaches proposed, such as nanoparticles or convection-enhanced delivery (CED), [367,368], show encouraging pre-clinical and clinical results.
(iii)
New molecular and genomic data has highlighted the inter- but also intra-tumoral heterogeneity of GBM, with tumors and tumor areas differing in target expression. Intratumoral heterogeneity is described as the root cause of therapy resistance and might explain the failure of targeted therapies specifically targeting tumor biomarkers, including anti-EGFR (cetuximab, gefitinib, erlotinib …), anti-VEGF (bevacizumab) and anti-integrin (cilengitide) therapies. Below, we tried to explain the failure of the therapies targeting these three proteins. These data highlight the need to combine different targeted therapies.

4.1. The Failure of Anti-EGFR Therapies

Besides favourable pre-clinical studies, anti-EGFR therapies barely present any clinical benefit for patients with GBM. Several clinical studies are being carried out in newly diagnosed GBM and recurrent GBM with anti-EGFR therapies as monotherapy or in combination with radiochemotherapy or other targeted agents (Table 2).
Besides the tissue differences between colorectal, head and neck, lung cancers and GBM, EGFR is also molecularly heterogeneous among these cancers. First, EGFR mutations in GBMs (as EGFRvIII) occur within receptor extracellular domain while in lung cancers (as L858R) occur in the kinase domain. Interestingly, EGFRvIII mutation seems to appear at later stages of tumor development. This subclonal EGFR mutation is lost in certain recurrent tumors [369]. However, mutational switch can happen where the initial EGFR mutation is replaced by another in recurrent tumor [370]. EGFRvIII heterogeneity adds another layer of complexity by its location in extrachromosomal double minute structures. Extrachromosomal EGFRvIII loss upon treatment promotes therapy resistance. However, the mutant tends to reappear after TKI withdrawal and resensitizes the tumor [371]. The secondary mutation (T790M) upon TKI treatment provides tumor resistance to therapy, in lung cancer [372]. While, in GBM no EGFR secondary mutation is described as cause of therapy resistance [373].
Tumor heterogeneity can be a reasonable case for GBM resistance to EGFR-targeted therapies. Upregulation of redundant receptor tyrosine kinases and deregulation of EGFR downstream molecules can trigger EGFR therapy resistance.
In GBM, PDGFR and c-MET are also upregulated and contribute to tumor progression. In the same or in other subclones than EGFR, these receptors can mediate an EGFR-inhibition bypass. In vivo, inhibition of EGFR (erlotinib) and c-MET (crizotinib) resulted in decreased tumor growth [374]. Also, in a subcutaneous GBM xenografts, combined inhibition of EGFR and PDGFRβ signaling suppresses tumor growth [375]. Further clinical multi-targeting is needed to test this hypothesis and try to overcome EGFR-therapy resistance in GBM.
In GBM, an EGFR downstream molecule, PTEN, is often loss. PTEN is a suppressor of PI3K/AKT pathway. Simultaneous expression of EGFRvIII and PTEN was associated with patient response to TKI [376]. However, another study showed that even though PTEN is frequently deleted in GBM, it cannot predict therapeutic efficiency of TKI [140].
Moreover, EGFR therapeutic targeting promotes a switch to an angiogenic and mesenchymal tumor phenotype. Mesenchymal switch is associated with GBM therapy resistance [377,378]. GBM resistance to EGFR therapy is still unclear and further studies are needed to improve EGFR-targeting in clinical trials. Although multi-targeted RTK and combinatory therapies have been newly proposed (Table 2, Table 3 and Table 4) [379], there is an urgent need to develop genetic and cellular representative GBM models [380].

4.2. The Failure of Bevacizumab

The lack of efficacy of bevacizumab, a large-size molecule, can be explained by its intravenous route of administration and poor intracerebral bioavailability. Intra-arterial brain administration, after temporary destruction of the BBB by mannitol and followed by intravenous administration, has shown encouraging results in terms of PFS in patients with recurrent GBMs (PFS = 10 months) [381]. Indeed, this route of administration has the advantage of potentiating the cerebral delivery of chemotherapy (local concentration of more than 48.9-fold compared to intravenous administration) [382]. Recent results have confirmed the benefit of this delivery method and are being studied [383,384].
The standard dose of bevacizumab is 10 mg/kg IV, injected every two weeks. Although this dose is clinically well tolerated, it can have adverse biological effects, particularly via the formation of hypoxic areas [321]. The study by Heiland et al., 2016 [385] suggested that a low dose of bevacizumab may decrease the size of cerebral edema and may result in better vascular permeability. This study showed an improvement in PFS when bevacizumab is injected at 5 mg/kg every two weeks and is combined with lomustine, compared to bevacizumab alone at 10 mg/kg every two weeks (PFS = 5 months vs. 3.2 months). This therapeutic benefit was not observed in first-time recurrent patients. Finally, at a dose of 5 mg/kg/week, no gain in PFS or survival was observed [288].

4.3. The Failure of Cilengitide

Although preclinical studies nicely demonstrated that cilengitide may affect both tumoral cells and endothelial cells, failure to improve GBM patient survival of the first antagonist of integrins reaching the clinic was really disappointing. The reasons of this failure can only be guessed, but different factors may be included [386,387,388].
First, the short half-life (a few hours) and pharmacokinetics of cilengitide restricts its properties in patients. Second, the use of cilengitide at low dose has been shown to stimulate angiogenesis in preclinical models [389]. This point has been addressed in patients [390] where no cilengitide-specific pattern of progression has been detected. Third, no reliable biomarker of cilengitide activity has been identified for stratification of patients. For the CENTRIC assay (the phase III clinical trial), patients were stratified according to the MGMT promoter methylation status, i.e., inclusion concerned only patients with a methylated promoter [340]. A phase II clinical trial (CORE) was conducted concomitantly with patients exhibiting a non-methylated MGMT promoter. Interestingly, a retrospective analysis of both cohorts regarding the expression of the cilengitide targets (αvβ3/β5 integrins) expression, concluded that cilengitide was the most effective in the CORE patients with high level of αvβ3 expression in the tumoral cells and not in the endothelial cells [391]. These results highlight the need for stratification of patients at least based on the level of the predictive target. In line with this, it was recently shown in an elegant work from the Cheresh group, that GBM sensitivity to αvβ3 integrin blockade is not simply related to the overexpression of the integrin but rather to an addiction to glucose uptake by the glucose transporteur Glut3 [392,393]. A fourth point could be added concerning the redundancy of integrin targets; in fact, other integrins (such as α5β1 integrin) may remain active after cilengitide relaying pro-tumoral effects. The story of cilengitide highlights some pitfalls in the transfer of preclinical results towards the clinic but also the need to stratify patients according to pertinent biomarkers.
(iv)
The plasticity of GBM cells complicates heterogeneity. It has been shown a bidirectional plasticity between glioma stem cell and their more differentiated counterparts either to form the tumor mass or in answer to therapies. These two types of cells will have different sensitivity to radio/chemotherapies but also to targeted therapies. Recent data emphasized that differentiated tumoral cells may contribute to GIC-dependent tumor progression [394,395]. These results indicate that targeting both cell populations will be needed to eradicate GBM. In a given tumor, glioma stem cells may vary from a proneuronal to a mesenchymal phenotype with intermediary states and thus acquiring new targets. Plasticity occurs also at the metabolic level when GBM cells adapt to the microenvironment to survive (for example from hypoxic to normoxic area) leading to new resistances. Treatments by themselves induce phenotypic and genomic modifications of tumor areas provoking secondary resistance. For example, bevacizumab has been shown to become ineffective due to the activation of secondary pathways involved in angiogenesis (c-MET, PIGF …).
(v)
It is increasingly recognized that preclinical models have to be improved to reflect the clinical reality. In vitro, from 2D long term established cell lines grown on flat surface, 3D spheroids or cells embedded in several matrices, we now go through investigations on patient-derived primary cell lines either as glioma stem cell culture or as organoids. This last model certainly will recapitulate at best the tumoral and environmental heterogeneity of GBM. The deal for the following years will be to test therapies on such personalized models in a time framework which will allow to return towards the patient as rapidly as possible. Majority of in vivo models still are based on nude mice where immunological networks are absent. Even if syngeneic mice models of glioma can be useful, they lack the human specificities and complexities. Success of targeted therapies may be in part dependent on the development of reliable modeling of GBM.
Although targeting the immune system is not the subject of this review, this strategy is also part of many ongoing clinical investigations. Moreover, targeted therapy also mediates immunostimulatory and immunosuppressive effects [396]. While early results of checkpoint inhibitors or others immune-targeting drugs have been disappointing when used as monotherapy, likely because of the overwhelming immunosuppressive contribution of the immune tumor microenvironment (iTME), new combinatorial approach might overcome this issue. Interestingly, targeting microglia which is believed to be a major regulator of this iTME, has been suggested in combination with targeted or antiangiogenic therapies responsible of iTME modulation [397]. Indeed, VEGF and TGF-β signaling and abnormal vasculature, all belonging to the selected targets presented in this review has been implicated in fostering immunosuppression [398]. Their inhibition have been already shown to improved immunotherapies clinical outcomes in various cancer [399]. Although the impact of targeted therapies on iTME is still unclear, ongoing clinical trials combining bevacizumab or others targeted therapies to check-point inhibitors (for instance: NCT03743662, NCT03661723, NCT04704154) open new perspectives for GBM treatment.

5. Conclusions

Within molecular targeted therapies, the most frequently reported are those targeting (i) EGFR, which gene is amplified or over-expressed in more than 50% of GBMs (40 clinical trials), and more generally tyrosine kinase receptors (85 clinical trials) and (ii) VEGF/VEGFR (75 clinical trials of which 53 involving bevacizumab). Besides diagnostic and prognostic relevance, some markers can be of predictive interest (therapeutic decision making) or even constitute a molecular target that can be activated by a specific therapy (theranostic marker). It seems that new approaches aim to counter heterogeneity by targeting, not specifically certain tumor markers expressed irregularly, but the potential cause of the heterogeneity. New and combined approaches (targeted-, chemo-, immuno-, radiotherapies) may result in reduced secondary resistance because they target the whole tumor. Indeed, the discovery of GBM stem cells gave new hope for the treatment of GBM. Their likely significance in tumor initiation, and therefore in the heterogeneity of the GBM, makes them relevant targets but their differentiated counterparts need to be considered as well as their crosstalk only begin to be understood.
The 257 clinical trials described in tables of this manuscript reveal that many different options are explored and raised questions still unanswered about targeted therapies. However, they led to the accumulation of new fundamental knowledge, which will definitely help to understand the mechanisms of resistance and advance research. The results obtained in recent years highlight the need to better stratify patients, by providing more personalized treatment corresponding to the genetic composition and evolution of GBMs. In that way, initiatives such as N2M2 (NOA-20) phase I/II trial (NCT03158389) of molecularly matched targeted therapies plus radiotherapy in GBM patients, with an unmethylated MGMT promoter, appears of great interest [71]. In this trial, molecular profile characterization of tumors allows allocation of patients to first line targeted therapies according to their mode of action. Indeed, complex molecular diagnostics will translate in clinical decision and may be the future for GBM treatment.

Author Contributions

Conceptualization, M.-C.M. and L.C.; formal analysis, M.-C.M., E.C.D.S. and L.C.; data curation, resources and original data preparation, E.C.D.S. and L.C.; supervision and project administration: L.C.; All authors have contributed to writing and reviewing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by ITI InnoVec (IdEx-ANR-10-IDEX-0002, SFRI-ANR-20-SFRI-0012).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

CNScentral nervous system
GBMglioblastoma
HRhazard ratio
OSoverall survival
PFSprogression-free survival

References

  1. Stoyanov, G.S.; Dzhenkov, D.L. On the Concepts and History of Glioblastoma Multiforme—Morphology, Genetics and Epigenetics. Folia Med. 2018, 60, 48–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Lee, S.C. Diffuse Gliomas for Nonneuropathologists: The New Integrated Molecular Diagnostics. Arch. Pathol. Lab. Med. 2018, 142, 804–814. [Google Scholar] [CrossRef]
  3. Stupp, R.; Hegi, M.E.; Mason, W.P.; van den Bent, M.J.; 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 Stud y: 5-Year Analysis of the EORTC-NCIC Trial. Lancet Oncol. 2009, 10, 459–466. [Google Scholar] [CrossRef]
  4. Zhang, J.; Stevens, M.F.G.; Bradshaw, T.D. Temozolomide: Mechanisms of Action, Repair and Resistance. Curr. Mol. Pharmacol. 2012, 5, 102–114. [Google Scholar] [CrossRef]
  5. Prados, M.D.; Byron, S.A.; Tran, N.L.; Phillips, J.J.; Molinaro, A.M.; Ligon, K.L.; Wen, P.Y.; Kuhn, J.G.; Mellinghoff, I.K.; de Groot, J.F.; et al. Toward Precision Medicine in Glioblastoma: The Promise and the Challenges. Neuro-Oncology 2015, 17, 1051–1063. [Google Scholar] [CrossRef] [Green Version]
  6. Franceschi, E.; Tosoni, A.; Bartolini, S.; Mazzocchi, V.; Fioravanti, A.; Brandes, A.A. Treatment Options for Recurrent Glioblastoma: Pitfalls and Future Trends. Expert Rev. Anticancer Ther. 2009, 9, 613–619. [Google Scholar] [CrossRef] [PubMed]
  7. Gallego, O. Nonsurgical Treatment of Recurrent Glioblastoma. Curr. Oncol. 2015, 22, e273–e281. [Google Scholar] [CrossRef] [Green Version]
  8. Barani, I.J.; Larson, D.A. Radiation Therapy of Glioblastoma. Cancer Treat. Res. 2015, 163, 49–73. [Google Scholar] [CrossRef] [PubMed]
  9. Lawrie, T.A.; Evans, J.; Gillespie, D.; Erridge, S.; Vale, L.; Kernohan, A.; Grant, R. Long-term Side Effects of Radiotherapy, with or without Chemotherapy, for Glioma. Cochrane Database Syst. Rev. 2019, 8, CD013047. [Google Scholar] [CrossRef] [PubMed]
  10. Sun, H.; Du, S.; Liao, G.; Xie, X.; Ren, C.; Yuan, Y.W. Do Glioma Patients Derive Any Therapeutic Benefit from Taking a Higher Cumulative Dose of Temozolomide Regimens?: A Meta-Analysis. Medicine 2015, 94, e827. [Google Scholar] [CrossRef] [PubMed]
  11. Stupp, R.; Brada, M.; van den Bent, M.J.; Tonn, J.-C.; Pentheroudakis, G. High-Grade Glioma: ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2014, 25 (Suppl. 3), iii93–iii101. [Google Scholar] [CrossRef]
  12. Maschio, M. Brain Tumor-Related Epilepsy. Curr. Neuropharmacol. 2012, 10, 124–133. [Google Scholar] [CrossRef] [PubMed]
  13. Geraldo, L.H.M.; Garcia, C.; da Fonseca, A.C.C.; Dubois, L.G.F.; de Sampaio e Spohr, T.C.L.; Matias, D.; de Camargo Magalhães, E.S.; do Amaral, R.F.; da Rosa, B.G.; Grimaldi, I.; et al. Glioblastoma Therapy in the Age of Molecular Medicine. Trends Cancer 2019, 5, 46–65. [Google Scholar] [CrossRef]
  14. Jain, K.K. A Critical Overview of Targeted Therapies for Glioblastoma. Front. Oncol. 2018, 8, 419. [Google Scholar] [CrossRef] [PubMed]
  15. Mooney, J.; Bernstock, J.D.; Ilyas, A.; Ibrahim, A.; Yamashita, D.; Markert, J.M.; Nakano, I. Current Approaches and Challenges in the Molecular Therapeutic Targeting of Glioblastoma. World Neurosurg. 2019, 129, 90–100. [Google Scholar] [CrossRef] [PubMed]
  16. Rajaratnam, V.; Islam, M.M.; Yang, M.; Slaby, R.; Ramirez, H.M.; Mirza, S.P. Glioblastoma: Pathogenesis and Current Status of Chemotherapy and Other Novel Treatments. Cancers 2020, 12, 937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Zanders, E.D.; Svensson, F.; Bailey, D.S. Therapy for Glioblastoma: Is It Working? Drug Discov. Today 2019, 24, 1193–1201. [Google Scholar] [CrossRef] [PubMed]
  18. Alemany, M.; Velasco, R.; Simó, M.; Bruna, J. Late Effects of Cancer Treatment: Consequences for Long-Term Brain Cancer Survivors. Neuro-Oncol. Pract. 2020, 8, 18–30. [Google Scholar] [CrossRef]
  19. Bobo, R.H.; Laske, D.W.; Akbasak, A.; Morrison, P.F.; Dedrick, R.L.; Oldfield, E.H. Convection-Enhanced Delivery of Macromolecules in the Brain. Proc. Natl. Acad. Sci. USA 1994, 91, 2076–2080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Akiyama, Y.; Kimura, Y.; Enatsu, R.; Mikami, T.; Wanibuchi, M.; Mikuni, N. Advantages and Disadvantages of Combined Chemotherapy with Carmustine Wafer and Bevacizumab in Patients with Newly Diagnosed Glioblastoma: A Single-Institutional Experience. World Neurosurg. 2018, 113, e508–e514. [Google Scholar] [CrossRef] [PubMed]
  21. Vellimana, A.K.; Recinos, V.R.; Hwang, L.; Fowers, K.D.; Li, K.W.; Zhang, Y.; Okonma, S.; Eberhart, C.G.; Brem, H.; Tyler, B.M. Combination of Paclitaxel Thermal Gel Depot with Temozolomide and Radiotherapy Significantly Prolongs Survival in an Experimental Rodent Glioma Model. J. Neurooncol. 2013, 111, 229–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Westphal, M.; Hilt, D.C.; Bortey, E.; Delavault, P.; Olivares, R.; Warnke, P.C.; Whittle, I.R.; Jääskeläinen, J.; Ram, Z. A Phase 3 Trial of Local Chemotherapy with Biodegradable Carmustine (BCNU) Wafers (Gliadel Wafers) in Patients with Primary Malignant Glioma. Neuro-Oncology 2003, 5, 79–88. [Google Scholar] [CrossRef] [PubMed]
  23. Wolinsky, J.B.; Colson, Y.L.; Grinstaff, M.W. Local Drug Delivery Strategies for Cancer Treatment: Gels, Nanoparticles, Polymeric Films, Rods, and Wafers. J. Control. Release Off. J. Control. Release Soc. 2012, 159, 14–26. [Google Scholar] [CrossRef] [Green Version]
  24. Xing, W.; Shao, C.; Qi, Z.; Yang, C.; Wang, Z. The Role of Gliadel Wafers in the Treatment of Newly Diagnosed GBM: A Meta-Analysis. Drug Des. Devel. Ther. 2015, 9, 3341–3348. [Google Scholar] [CrossRef] [Green Version]
  25. Zhang, Y.-D.; Dai, R.-Y.; Chen, Z.; Zhang, Y.-H.; He, X.-Z.; Zhou, J. Efficacy and Safety of Carmustine Wafers in the Treatment of Glioblastoma Multiforme: A Systematic Review. Turk. Neurosurg. 2014, 24, 639–645. [Google Scholar] [CrossRef] [PubMed]
  26. Karim, R.; Palazzo, C.; Evrard, B.; Piel, G. Nanocarriers for the Treatment of Glioblastoma Multiforme: Current State-of-the-Art. J. Control. Release Off. J. Control. Release Soc. 2016, 227, 23–37. [Google Scholar] [CrossRef] [PubMed]
  27. Gutkin, A.; Cohen, Z.R.; Peer, D. Harnessing Nanomedicine for Therapeutic Intervention in Glioblastoma. Expert Opin. Drug Deliv. 2016, 13, 1573–1582. [Google Scholar] [CrossRef]
  28. Kim, S.-S.; Harford, J.B.; Pirollo, K.F.; Chang, E.H. Effective Treatment of Glioblastoma Requires Crossing the Blood-Brain Barrier and Targeting Tumors Including Cancer Stem Cells: The Promise of Nanomedicine. Biochem. Biophys. Res. Commun. 2015, 468, 485–489. [Google Scholar] [CrossRef] [Green Version]
  29. Avril, T.; Vauleon, E.; Tanguy-Royer, S.; Mosser, J.; Quillien, V. Mechanisms of Immunomodulation in Human Glioblastoma. Immunotherapy 2011, 3, 42–44. [Google Scholar] [CrossRef] [Green Version]
  30. Brown, N.F.; Carter, T.J.; Ottaviani, D.; Mulholland, P. Harnessing the Immune System in Glioblastoma. Br. J. Cancer 2018, 119, 1171–1181. [Google Scholar] [CrossRef] [Green Version]
  31. McGranahan, T.; Therkelsen, K.E.; Ahmad, S.; Nagpal, S. Current State of Immunotherapy for Treatment of Glioblastoma. Curr. Treat. Options Oncol. 2019, 20, 24. [Google Scholar] [CrossRef] [Green Version]
  32. Miyazaki, T.; Ishikawa, E.; Sugii, N.; Matsuda, M. Therapeutic Strategies for Overcoming Immunotherapy Resistance Mediated by Immunosuppressive Factors of the Glioblastoma Microenvironment. Cancers 2020, 12, 1960. [Google Scholar] [CrossRef]
  33. Romani, M.; Pistillo, M.P.; Carosio, R.; Morabito, A.; Banelli, B. Immune Checkpoints and Innovative Therapies in Glioblastoma. Front. Oncol. 2018, 8, 464. [Google Scholar] [CrossRef]
  34. Fuchs, Q.; Pierrevelcin, M.; Messe, M.; Lhermitte, B.; Blandin, A.-F.; Papin, C.; Coca, A.; Dontenwill, M.; Entz-Werlé, N. Hypoxia Inducible Factors’ Signaling in Pediatric High-Grade Gliomas: Role, Modelization and Innovative Targeted Approaches. Cancers 2020, 12, 979. [Google Scholar] [CrossRef] [Green Version]
  35. Fan, Y.; Potdar, A.A.; Gong, Y.; Eswarappa, S.M.; Donnola, S.; Lathia, J.D.; Hambardzumyan, D.; Rich, J.N.; Fox, P.L. Profilin-1 Phosphorylation Directs Angiocrine Expression and Glioblastoma Progression through HIF-1α Accumulation. Nat. Cell Biol. 2014, 16, 445–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Wang, E.; Zhang, C.; Polavaram, N.; Liu, F.; Wu, G.; Schroeder, M.A.; Lau, J.S.; Mukhopadhyay, D.; Jiang, S.-W.; O’Neill, B.P.; et al. The Role of Factor Inhibiting HIF (FIH-1) in Inhibiting HIF-1 Transcriptional Activity in Glioblastoma Multiforme. PLoS ONE 2014, 9, e86102. [Google Scholar] [CrossRef] [Green Version]
  37. Covello, K.L.; Kehler, J.; Yu, H.; Gordan, J.D.; Arsham, A.M.; Hu, C.-J.; Labosky, P.A.; Simon, M.C.; Keith, B. HIF-2alpha Regulates Oct-4: Effects of Hypoxia on Stem Cell Function, Embryonic Development, and Tumor Growth. Genes Dev. 2006, 20, 557–570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Strowd, R.E.; Ellingson, B.M.; Wen, P.Y.; Ahluwalia, M.S.; Piotrowski, A.F.; Desai, A.S.; Clarke, J.L.; Lieberman, F.S.; Desideri, S.; Nabors, L.B.; et al. Safety and Activity of a First-in-Class Oral HIF2-Alpha Inhibitor, PT2385, in Patients with First Recurrent Glioblastoma (GBM). J. Clin. Oncol. 2019, 37, 2027. [Google Scholar] [CrossRef]
  39. Clark, P.M.; Mai, W.X.; Cloughesy, T.F.; Nathanson, D.A. Emerging Approaches for Targeting Metabolic Vulnerabilities in Malignant Glioma. Curr. Neurol. Neurosci. Rep. 2016, 16, 17. [Google Scholar] [CrossRef] [PubMed]
  40. Woolf, E.C.; Scheck, A.C. The Ketogenic Diet for the Treatment of Malignant Glioma. J. Lipid Res. 2015, 56, 5–10. [Google Scholar] [CrossRef] [Green Version]
  41. Hay, N. Reprogramming Glucose Metabolism in Cancer: Can It Be Exploited for Cancer Therapy? Nat. Rev. Cancer 2016, 16, 635–649. [Google Scholar] [CrossRef] [Green Version]
  42. Ghiaseddin, A.P.; Shin, D.; Melnick, K.; Tran, D.D. Tumor Treating Fields in the Management of Patients with Malignant Gliomas. Curr. Treat. Options Oncol. 2020, 21, 76. [Google Scholar] [CrossRef] [PubMed]
  43. 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]
  44. Perrin, S.L.; Samuel, M.S.; Koszyca, B.; Brown, M.P.; Ebert, L.M.; Oksdath, M.; Gomez, G.A. Glioblastoma Heterogeneity and the Tumour Microenvironment: Implications for Preclinical Research and Development of New Treatments. Biochem. Soc. Trans. 2019, 47, 625–638. [Google Scholar] [CrossRef]
  45. Verhaak, R.G.W.; 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]
  46. White, K.; Connor, K.; Clerkin, J.; Murphy, B.M.; Salvucci, M.; O’Farrell, A.C.; Rehm, M.; O’Brien, D.; Prehn, J.H.M.; Niclou, S.P.; et al. New Hints towards a Precision Medicine Strategy for IDH Wild-Type Glioblastoma. Ann. Oncol. 2020, 31, 1679–1692. [Google Scholar] [CrossRef] [PubMed]
  47. Aldape, K.; Zadeh, G.; Mansouri, S.; Reifenberger, G.; von Deimling, A. Glioblastoma: Pathology, Molecular Mechanisms and Markers. Acta Neuropathol. 2015, 129, 829–848. [Google Scholar] [CrossRef]
  48. Behnan, J.; Finocchiaro, G.; Hanna, G. The Landscape of the Mesenchymal Signature in Brain Tumours. Brain J. Neurol. 2019, 142, 847–866. [Google Scholar] [CrossRef] [Green Version]
  49. Tirosh, I.; Suvà, M.L. Tackling the Many Facets of Glioblastoma Heterogeneity. Cell Stem Cell 2020, 26, 303–304. [Google Scholar] [CrossRef]
  50. 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]
  51. Bailey, P.; Cushing, H. A Classification of Tumors of the Glioma Groupo on a Histogenetic Basis with a Correlation Study of Prognosis; Philadelphia, J.B Lippincott Co.: Philadelphia, PA, USA, 1926. [Google Scholar]
  52. Kernohan, J.W.; Mabon, R.F. A Simplified Classification of the Gliomas. Proc. Staff Meet. Mayo Clin. 1949, 24, 71–75. [Google Scholar] [PubMed]
  53. Zülch, K.J. Types Histologiques des Tumeurs du Système Nerveux Central; Organisation mondiale de la santé: Genève, Switzerland, 1979; ISBN 978-92-4-276021-7. [Google Scholar]
  54. Kleihues, P.; Burger, P.C.; Scheithauer, B.W. The New WHO Classification of Brain Tumours. Brain Pathol. 1993, 3, 255–268. [Google Scholar] [CrossRef]
  55. Louis, D.N.; Perry, A.; Reifenberger, G.; von Deimling, A.; Figarella-Branger, D.; Cavenee, W.K.; Ohgaki, H.; Wiestler, O.D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A Summary. Acta Neuropathol. 2016, 131, 803–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Louis, D.N.; Ohgaki, H.; Wiestler, O.D.; Cavenee, W.K.; Burger, P.C.; Jouvet, A.; Scheithauer, B.W.; Kleihues, P. The 2007 WHO Classification of Tumours of the Central Nervous System. Acta Neuropathol. 2007, 114, 97–109. [Google Scholar] [CrossRef] [Green Version]
  57. Louis, D.N.; Holland, E.C.; Cairncross, J.G. Glioma Classification: A Molecular Reappraisal. Am. J. Pathol. 2001, 159, 779–786. [Google Scholar] [CrossRef]
  58. Louis, D.N.; Perry, A.; Burger, P.; Ellison, D.W.; Reifenberger, G.; von Deimling, A.; Aldape, K.; Brat, D.; Collins, V.P.; Eberhart, C.; et al. International Society of Neuropathology—Haarlem Consensus Guidelines for Nervous System Tumor Classification and Grading. Brain Pathol. 2014, 24, 429–435. [Google Scholar] [CrossRef]
  59. Singh, S.K.; Clarke, I.D.; Terasaki, M.; Bonn, V.E.; Hawkins, C.; Squire, J.; Dirks, P.B. Identification of a Cancer Stem Cell in Human Brain Tumors. Cancer Res. 2003, 63, 5821–5828. [Google Scholar]
  60. Singh, S.K.; Clarke, I.D.; Hide, T.; Dirks, P.B. Cancer Stem Cells in Nervous System Tumors. Oncogene 2004, 23, 7267–7273. [Google Scholar] [CrossRef] [Green Version]
  61. Cruceru, M.L.; Neagu, M.; Demoulin, J.-B.; Constantinescu, S.N. Therapy Targets in Glioblastoma and Cancer Stem Cells: Lessons from Haematopoietic Neoplasms. J. Cell. Mol. Med. 2013, 17, 1218–1235. [Google Scholar] [CrossRef] [PubMed]
  62. Ignatova, T.N.; Kukekov, V.G.; Laywell, E.D.; Suslov, O.N.; Vrionis, F.D.; Steindler, D.A. Human Cortical Glial Tumors Contain Neural Stem-like Cells Expressing Astroglial and Neuronal Markers in Vitro. Glia 2002, 39, 193–206. [Google Scholar] [CrossRef]
  63. Yi, Y.; Hsieh, I.-Y.; Huang, X.; Li, J.; Zhao, W. Glioblastoma Stem-Like Cells: Characteristics, Microenvironment, and Therapy. Front. Pharmacol. 2016, 7, 477. [Google Scholar] [CrossRef] [Green Version]
  64. Koso, H.; Takeda, H.; Yew, C.C.K.; Ward, J.M.; Nariai, N.; Ueno, K.; Nagasaki, M.; Watanabe, S.; Rust, A.G.; Adams, D.J.; et al. Transposon Mutagenesis Identifies Genes That Transform Neural Stem Cells into Glioma-Initiating Cells. Proc. Natl. Acad. Sci. USA 2012, 109, E2998–E3007. [Google Scholar] [CrossRef] [Green Version]
  65. Iwadate, Y. Plasticity in Glioma Stem Cell Phenotype and Its Therapeutic Implication. Neurol. Med. Chir. 2018, 58, 61–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Chen, W.; Dong, J.; Haiech, J.; Kilhoffer, M.-C.; Zeniou, M. Cancer Stem Cell Quiescence and Plasticity as Major Challenges in Cancer Therapy. Stem Cells Int. 2016, 2016, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Jackson, M.; Hassiotou, F.; Nowak, A. Glioblastoma Stem-like Cells: At the Root of Tumor Recurrence and a Therapeutic Target. Carcinogenesis 2015, 36, 177–185. [Google Scholar] [CrossRef]
  68. Sørensen, M.D.; Fosmark, S.; Hellwege, S.; Beier, D.; Kristensen, B.W.; Beier, C.P. Chemoresistance and Chemotherapy Targeting Stem-like Cells in Malignant Glioma. Adv. Exp. Med. Biol. 2015, 853, 111–138. [Google Scholar] [CrossRef]
  69. Penas-Prado, M.; Hess, K.R.; Fisch, M.J.; Lagrone, L.W.; Groves, M.D.; Levin, V.A.; De Groot, J.F.; Puduvalli, V.K.; Colman, H.; Volas-Redd, G.; et al. Randomized Phase II Adjuvant Factorial Study of Dose-Dense Temozolomide Alone and in Combination with Isotretinoin, Celecoxib, and/or Thalidomide for Glioblastoma. Neuro-Oncology 2015, 17, 266–273. [Google Scholar] [CrossRef]
  70. Grossman, S.A.; Olson, J.; Batchelor, T.; Peereboom, D.; Lesser, G.; Desideri, S.; Ye, X.; Hammour, T.; Supko, J.G. New Approaches to Brain Tumor Therapy CNS Consortium Effect of Phenytoin on Celecoxib Pharmacokinetics in Patients with Glioblastoma. Neuro-Oncology 2008, 10, 190–198. [Google Scholar] [CrossRef] [Green Version]
  71. Wick, W.; Dettmer, S.; Berberich, A.; Kessler, T.; Karapanagiotou-Schenkel, I.; Wick, A.; Winkler, F.; Pfaff, E.; Brors, B.; Debus, J.; et al. N2M2 (NOA-20) Phase I/II Trial of Molecularly Matched Targeted Therapies plus Radiotherapy in Patients with Newly Diagnosed Non-MGMT Hypermethylated Glioblastoma. Neuro-Oncology 2019, 21, 95–105. [Google Scholar] [CrossRef] [Green Version]
  72. Nusse, R. Wnt Signaling and Stem Cell Control. Cell Res. 2008, 18, 523–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Yu, C.-Y.; Liang, G.-B.; Du, P.; Liu, Y.-H. Lgr4 Promotes Glioma Cell Proliferation through Activation of Wnt Signaling. Asian Pac. J. Cancer Prev. APJCP 2013, 14, 4907–4911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Bhuvanalakshmi, G.; Arfuso, F.; Millward, M.; Dharmarajan, A.; Warrier, S. Secreted Frizzled-Related Protein 4 Inhibits Glioma Stem-like Cells by Reversing Epithelial to Mesenchymal Transition, Inducing Apoptosis and Decreasing Cancer Stem Cell Properties. PLoS ONE 2015, 10, e0127517. [Google Scholar] [CrossRef]
  75. Sareddy, G.R.; Kesanakurti, D.; Kirti, P.B.; Babu, P.P. Nonsteroidal Anti-Inflammatory Drugs Diclofenac and Celecoxib Attenuates Wnt/β-Catenin/Tcf Signaling Pathway in Human Glioblastoma Cells. Neurochem. Res. 2013, 38, 2313–2322. [Google Scholar] [CrossRef]
  76. Korur, S.; Huber, R.M.; Sivasankaran, B.; Petrich, M.; Morin, P.; Hemmings, B.A.; Merlo, A.; Lino, M.M. GSK3beta Regulates Differentiation and Growth Arrest in Glioblastoma. PLoS ONE 2009, 4, e7443. [Google Scholar] [CrossRef]
  77. Lee, Y.; Lee, J.-K.; Ahn, S.H.; Lee, J.; Nam, D.-H. WNT Signaling in Glioblastoma and Therapeutic Opportunities. Lab. Investig. J. Tech. Methods Pathol. 2016, 96, 137–150. [Google Scholar] [CrossRef] [Green Version]
  78. Ramachandran, I.; Ganapathy, V.; Gillies, E.; Fonseca, I.; Sureban, S.M.; Houchen, C.W.; Reis, A.; Queimado, L. Wnt Inhibitory Factor 1 Suppresses Cancer Stemness and Induces Cellular Senescence. Cell Death Dis. 2014, 5, e1246. [Google Scholar] [CrossRef] [PubMed]
  79. Kanabur, P.; Guo, S.; Simonds, G.R.; Kelly, D.F.; Gourdie, R.G.; Verbridge, S.S.; Sheng, Z. Patient-Derived Glioblastoma Stem Cells Respond Differentially to Targeted Therapies. Oncotarget 2016, 7, 86406–86419. [Google Scholar] [CrossRef]
  80. Li, J.-L.; Sainson, R.C.A.; Oon, C.E.; Turley, H.; Leek, R.; Sheldon, H.; Bridges, E.; Shi, W.; Snell, C.; Bowden, E.T.; et al. DLL4-Notch Signaling Mediates Tumor Resistance to Anti-VEGF Therapy in Vivo. Cancer Res. 2011, 71, 6073–6083. [Google Scholar] [CrossRef] [Green Version]
  81. Yu, J.-B.; Jiang, H.; Zhan, R.-Y. Aberrant Notch Signaling in Glioblastoma Stem Cells Contributes to Tumor Recurrence and Invasion. Mol. Med. Rep. 2016, 14, 1263–1268. [Google Scholar] [CrossRef] [Green Version]
  82. Shih, I.-M.; Wang, T.-L. Notch Signaling, Gamma-Secretase Inhibitors, and Cancer Therapy. Cancer Res. 2007, 67, 1879–1882. [Google Scholar] [CrossRef] [Green Version]
  83. Hovinga, K.E.; Shimizu, F.; Wang, R.; Panagiotakos, G.; Van Der Heijden, M.; Moayedpardazi, H.; Correia, A.S.; Soulet, D.; Major, T.; Menon, J.; et al. Inhibition of Notch Signaling in Glioblastoma Targets Cancer Stem Cells via an Endothelial Cell Intermediate. Stem Cells Dayt. Ohio 2010, 28, 1019–1029. [Google Scholar] [CrossRef] [Green Version]
  84. Saito, N.; Fu, J.; Zheng, S.; Yao, J.; Wang, S.; Liu, D.D.; Yuan, Y.; Sulman, E.P.; Lang, F.F.; Colman, H.; et al. A High Notch Pathway Activation Predicts Response to γ Secretase Inhibitors in Proneural Subtype of Glioma Tumor-Initiating Cells: Targeting Proneural GBM with Notch Inhibition. Stem Cells 2014, 32, 301–312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Tanaka, S.; Nakada, M.; Yamada, D.; Nakano, I.; Todo, T.; Ino, Y.; Hoshii, T.; Tadokoro, Y.; Ohta, K.; Ali, M.A.E.; et al. Strong Therapeutic Potential of γ-Secretase Inhibitor MRK003 for CD44-High and CD133-Low Glioblastoma Initiating Cells. J. Neurooncol. 2015, 121, 239–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Yahyanejad, S.; King, H.; Iglesias, V.S.; Granton, P.V.; Barbeau, L.M.O.; van Hoof, S.J.; Groot, A.J.; Habets, R.; Prickaerts, J.; Chalmers, A.J.; et al. NOTCH Blockade Combined with Radiation Therapy and Temozolomide Prolongs Survival of Orthotopic Glioblastoma. Oncotarget 2016, 7, 41251–41264. [Google Scholar] [CrossRef] [Green Version]
  87. Opačak-Bernardi, T.; Ryu, J.S.; Raucher, D. Effects of Cell Penetrating Notch Inhibitory Peptide Conjugated to Elastin-like Polypeptide on Glioblastoma Cells. J. Drug Target. 2017, 25, 523–531. [Google Scholar] [CrossRef]
  88. Pan, E.; Supko, J.G.; Kaley, T.J.; Butowski, N.A.; Cloughesy, T.; Jung, J.; Desideri, S.; Grossman, S.; Ye, X.; Park, D.M. Phase I Study of RO4929097 with Bevacizumab in Patients with Recurrent Malignant Glioma. J. Neurooncol. 2016, 130, 571–579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Auffinger, B.; Spencer, D.; Pytel, P.; Ahmed, A.U.; Lesniak, M.S. The Role of Glioma Stem Cells in Chemotherapy Resistance and Glioblastoma Multiforme Recurrence. Expert Rev. Neurother. 2015, 15, 741–752. [Google Scholar] [CrossRef] [Green Version]
  90. Lee, Y.; Kim, K.H.; Kim, D.G.; Cho, H.J.; Kim, Y.; Rheey, J.; Shin, K.; Seo, Y.J.; Choi, Y.-S.; Lee, J.-I.; et al. FoxM1 Promotes Stemness and Radio-Resistance of Glioblastoma by Regulating the Master Stem Cell Regulator Sox2. PLoS ONE 2015, 10, e0137703. [Google Scholar] [CrossRef] [Green Version]
  91. Tu, Y.; Niu, M.; Xie, P.; Yue, C.; Liu, N.; Qi, Z.; Gao, S.; Liu, H.; Shi, Q.; Yu, R.; et al. Smoothened Is a Poor Prognosis Factor and a Potential Therapeutic Target in Glioma. Sci. Rep. 2017, 7, 42630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Rimkus, T.K.; Carpenter, R.L.; Qasem, S.; Chan, M.; Lo, H.-W. Targeting the Sonic Hedgehog Signaling Pathway: Review of Smoothened and GLI Inhibitors. Cancers 2016, 8, 22. [Google Scholar] [CrossRef] [Green Version]
  93. Lin, T.L.; Matsui, W. Hedgehog Pathway as a Drug Target: Smoothened Inhibitors in Development. OncoTargets Ther. 2012, 5, 47–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Bar, E.E.; Chaudhry, A.; Lin, A.; Fan, X.; Schreck, K.; Matsui, W.; Piccirillo, S.; Vescovi, A.L.; DiMeco, F.; Olivi, A.; et al. Cyclopamine-Mediated Hedgehog Pathway Inhibition Depletes Stem-like Cancer Cells in Glioblastoma. Stem Cells Dayt. Ohio 2007, 25, 2524–2533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Bensalma, S.; Chadeneau, C.; Legigan, T.; Renoux, B.; Gaillard, A.; de Boisvilliers, M.; Pinet-Charvet, C.; Papot, S.; Muller, J.M. Evaluation of Cytotoxic Properties of a Cyclopamine Glucuronide Prodrug in Rat Glioblastoma Cells and Tumors. J. Mol. Neurosci. 2015, 55, 51–61. [Google Scholar] [CrossRef] [PubMed]
  96. Liu, Y.-J.; Ma, Y.-C.; Zhang, W.-J.; Yang, Z.-Z.; Liang, D.-S.; Wu, Z.-F.; Qi, X.-R. Combination Therapy with Micellarized Cyclopamine and Temozolomide Attenuate Glioblastoma Growth through Gli1 Down-Regulation. Oncotarget 2017, 8, 42495–42509. [Google Scholar] [CrossRef] [Green Version]
  97. de la Iglesia, N.; Puram, S.V.; Bonni, A. STAT3 Regulation of Glioblastoma Pathogenesis. Curr. Mol. Med. 2009, 9, 580–590. [Google Scholar] [CrossRef] [Green Version]
  98. Reguera-Nuñez, E.; Roca, C.; Hardy, E.; de la Fuente, M.; Csaba, N.; Garcia-Fuentes, M. Implantable Controlled Release Devices for BMP-7 Delivery and Suppression of Glioblastoma Initiating Cells. Biomaterials 2014, 35, 2859–2867. [Google Scholar] [CrossRef]
  99. Rampazzo, E.; Dettin, M.; Maule, F.; Scabello, A.; Calvanese, L.; D’Auria, G.; Falcigno, L.; Porcù, E.; Zamuner, A.; Della Puppa, A.; et al. A Synthetic BMP-2 Mimicking Peptide Induces Glioblastoma Stem Cell Differentiation. Biochim. Biophys. Acta 2017, 1861, 2282–2292. [Google Scholar] [CrossRef]
  100. Garrido, W.; Rocha, J.D.; Jaramillo, C.; Fernandez, K.; Oyarzun, C.; San Martin, R.; Quezada, C. Chemoresistance in High-Grade Gliomas: Relevance of Adenosine Signalling in Stem-like Cells of Glioblastoma Multiforme. Curr. Drug Targets 2014, 15, 931–942. [Google Scholar] [CrossRef]
  101. Uribe, D.; Torres, Á.; Rocha, J.D.; Niechi, I.; Oyarzún, C.; Sobrevia, L.; San Martín, R.; Quezada, C. Multidrug Resistance in Glioblastoma Stem-like Cells: Role of the Hypoxic Microenvironment and Adenosine Signaling. Mol. Asp. Med. 2017, 55, 140–151. [Google Scholar] [CrossRef]
  102. Quezada, C.; Garrido, W.; Oyarzún, C.; Fernández, K.; Segura, R.; Melo, R.; Casanello, P.; Sobrevia, L.; San Martín, R. 5′-Ectonucleotidase Mediates Multiple-Drug Resistance in Glioblastoma Multiforme Cells. J. Cell. Physiol. 2013, 228, 602–608. [Google Scholar] [CrossRef]
  103. Torres, A.; Vargas, Y.; Uribe, D.; Jaramillo, C.; Gleisner, A.; Salazar-Onfray, F.; López, M.N.; Melo, R.; Oyarzún, C.; San Martín, R.; et al. Adenosine A3 Receptor Elicits Chemoresistance Mediated by Multiple Resistance-Associated Protein-1 in Human Glioblastoma Stem-like Cells. Oncotarget 2016, 7, 67373–67386. [Google Scholar] [CrossRef]
  104. Daniele, S.; Zappelli, E.; Natali, L.; Martini, C.; Trincavelli, M.L. Modulation of A1 and A2B Adenosine Receptor Activity: A New Strategy to Sensitise Glioblastoma Stem Cells to Chemotherapy. Cell Death Dis. 2014, 5, e1539. [Google Scholar] [CrossRef] [Green Version]
  105. Nørøxe, D.S.; Poulsen, H.S.; Lassen, U. Hallmarks of Glioblastoma: A Systematic Review. ESMO Open 2016, 1, e000144. [Google Scholar] [CrossRef]
  106. Majewska, E.; Szeliga, M. AKT/GSK3β Signaling in Glioblastoma. Neurochem. Res. 2017, 42, 918–924. [Google Scholar] [CrossRef] [Green Version]
  107. Normanno, N.; De Luca, A.; Bianco, C.; Strizzi, L.; Mancino, M.; Maiello, M.R.; Carotenuto, A.; De Feo, G.; Caponigro, F.; Salomon, D.S. Epidermal Growth Factor Receptor (EGFR) Signaling in Cancer. Gene 2006, 366, 2–16. [Google Scholar] [CrossRef]
  108. Frederick, L.; Wang, X.Y.; Eley, G.; James, C.D. Diversity and Frequency of Epidermal Growth Factor Receptor Mutations in Human Glioblastomas. Cancer Res. 2000, 60, 1383–1387. [Google Scholar]
  109. Combs, S.E.; Schulz-Ertner, D.; Hartmann, C.; Welzel, T.; Timke, C.; Herfarth, K.; von Deimling, A.; Edler, L.; Platten, M.; Wick, W.; et al. Phase I/II Study of Cetuximab plus Temozolomide as Radiochemotherapy for Primary Glioblastoma (GERT)—Eudract Number 2005–003911–63; NCT00311857. J. Clin. Oncol. 2008, 26, 2077. [Google Scholar] [CrossRef]
  110. Hasselbalch, B.; Lassen, U.; Hansen, S.; Holmberg, M.; Sørensen, M.; Kosteljanetz, M.; Broholm, H.; Stockhausen, M.-T.; Poulsen, H.S. Cetuximab, Bevacizumab, and Irinotecan for Patients with Primary Glioblastoma and Progression after Radiation Therapy and Temozolomide: A Phase II Trial. Neuro-Oncology 2010, 12, 508–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Westphal, M.; Heese, O.; Steinbach, J.P.; Schnell, O.; Schackert, G.; Mehdorn, M.; Schulz, D.; Simon, M.; Schlegel, U.; Senft, C.; et al. A Randomised, Open Label Phase III Trial with Nimotuzumab, an Anti-Epidermal Growth Factor Receptor Monoclonal Antibody in the Treatment of Newly Diagnosed Adult Glioblastoma. Eur. J. Cancer 2015, 51, 522–532. [Google Scholar] [CrossRef]
  112. Brown, P.D.; Krishnan, S.; Sarkaria, J.N.; Wu, W.; Jaeckle, K.A.; Uhm, J.H.; Geoffroy, F.J.; Arusell, R.; Kitange, G.; Jenkins, R.B.; et al. Phase I/II Trial of Erlotinib and Temozolomide with Radiation Therapy in the Treatment of Newly Diagnosed Glioblastoma Multiforme: North Central Cancer Treatment Group Study N0177. J. Clin. Oncol. 2008, 26, 5603–5609. [Google Scholar] [CrossRef] [PubMed]
  113. Peereboom, D.M.; Ahluwalia, M.S.; Ye, X.; Supko, J.G.; Hilderbrand, S.L.; Phuphanich, S.; Nabors, L.B.; Rosenfeld, M.R.; Mikkelsen, T.; Grossman, S.A. NABTT 0502: A Phase II and Pharmacokinetic Study of Erlotinib and Sorafenib for Patients with Progressive or Recurrent Glioblastoma Multiforme. Neuro-Oncology 2013, 15, 490–496. [Google Scholar] [CrossRef] [Green Version]
  114. Raizer, J.J.; Giglio, P.; Hu, J.; Groves, M.; Merrell, R.; Conrad, C.; Phuphanich, S.; Puduvalli, V.K.; Loghin, M.; Paleologos, N.; et al. A Phase II Study of Bevacizumab and Erlotinib after Radiation and Temozolomide in MGMT Unmethylated GBM Patients. J. Neurooncol. 2016, 126, 185–192. [Google Scholar] [CrossRef] [Green Version]
  115. Peereboom, D.M.; Shepard, D.R.; Ahluwalia, M.S.; Brewer, C.J.; Agarwal, N.; Stevens, G.H.J.; Suh, J.H.; Toms, S.A.; Vogelbaum, M.A.; Weil, R.J.; et al. Phase II Trial of Erlotinib with Temozolomide and Radiation in Patients with Newly Diagnosed Glioblastoma Multiforme. J. Neurooncol. 2010, 98, 93–99. [Google Scholar] [CrossRef]
  116. Reardon, D.A.; Desjardins, A.; Vredenburgh, J.J.; Gururangan, S.; Friedman, A.H.; Herndon, J.E.; Marcello, J.; Norfleet, J.A.; McLendon, R.E.; Sampson, J.H.; et al. Phase 2 Trial of Erlotinib plus Sirolimus in Adults with Recurrent Glioblastoma. J. Neurooncol. 2010, 96, 219–230. [Google Scholar] [CrossRef] [Green Version]
  117. Sathornsumetee, S.; Desjardins, A.; Vredenburgh, J.J.; McLendon, R.E.; Marcello, J.; Herndon, J.E.; Mathe, A.; Hamilton, M.; Rich, J.N.; Norfleet, J.A.; et al. Phase II Trial of Bevacizumab and Erlotinib in Patients with Recurrent Malignant Glioma. Neuro-Oncology 2010, 12, 1300–1310. [Google Scholar] [CrossRef] [Green Version]
  118. van den Bent, M.J.; Brandes, A.A.; Rampling, R.; Kouwenhoven, M.C.M.; Kros, J.M.; Carpentier, A.F.; Clement, P.M.; Frenay, M.; Campone, M.; Baurain, J.-F.; et al. Randomized Phase II Trial of Erlotinib versus Temozolomide or Carmustine in Recurrent Glioblastoma: EORTC Brain Tumor Group Study 26034. J. Clin. Oncol. 2009, 27, 1268–1274. [Google Scholar] [CrossRef] [Green Version]
  119. Kesavabhotla, K.; Schlaff, C.D.; Shin, B.; Mubita, L.; Kaplan, R.; Tsiouris, A.J.; Pannullo, S.C.; Christos, P.; Lavi, E.; Scheff, R.; et al. Phase I/II Study of Oral Erlotinib for Treatment of Relapsed/Refractory Glioblastoma Multiforme and Anaplastic Astrocytoma. J. Exp. Ther. Oncol. 2012, 10, 71–81. [Google Scholar] [PubMed]
  120. Wen, P.Y.; Chang, S.M.; Lamborn, K.R.; Kuhn, J.G.; Norden, A.D.; Cloughesy, T.F.; Robins, H.I.; Lieberman, F.S.; Gilbert, M.R.; Mehta, M.P.; et al. Phase I/II Study of Erlotinib and Temsirolimus for Patients with Recurrent Malignant Gliomas: North American Brain Tumor Consortium Trial 04-02. Neuro-Oncology 2014, 16, 567–578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Raizer, J.J.; Abrey, L.E.; Lassman, A.B.; Chang, S.M.; Lamborn, K.R.; Kuhn, J.G.; Yung, W.K.A.; Gilbert, M.R.; Aldape, K.A.; Wen, P.Y.; et al. A Phase II Trial of Erlotinib in Patients with Recurrent Malignant Gliomas and Nonprogressive Glioblastoma Multiforme Postradiation Therapy. Neuro-Oncology 2010, 12, 95–103. [Google Scholar] [CrossRef]
  122. Hegi, M.E.; Diserens, A.-C.; Bady, P.; Kamoshima, Y.; Kouwenhoven, M.C.M.; Delorenzi, M.; Lambiv, W.L.; Hamou, M.-F.; Matter, M.S.; Koch, A.; et al. Pathway Analysis of Glioblastoma Tissue after Preoperative Treatment with the EGFR Tyrosine Kinase Inhibitor Gefitinib--a Phase II Trial. Mol. Cancer Ther. 2011, 10, 1102–1112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Rich, J.N.; Reardon, D.A.; Peery, T.; Dowell, J.M.; Quinn, J.A.; Penne, K.L.; Wikstrand, C.J.; Van Duyn, L.B.; Dancey, J.E.; McLendon, R.E.; et al. Phase II Trial of Gefitinib in Recurrent Glioblastoma. J. Clin. Oncol. 2004, 22, 133–142. [Google Scholar] [CrossRef] [PubMed]
  124. Chakravarti, A.; Wang, M.; Robins, H.I.; Lautenschlaeger, T.; Curran, W.J.; Brachman, D.G.; Schultz, C.J.; Choucair, A.; Dolled-Filhart, M.; Christiansen, J.; et al. RTOG 0211: A Phase 1/2 Study of Radiation Therapy with Concurrent Gefitinib for Newly Diagnosed Glioblastoma Patients. Int. J. Radiat. Oncol. Biol. Phys. 2013, 85, 1206–1211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Reardon, D.A.; Nabors, L.B.; Mason, W.P.; Perry, J.R.; Shapiro, W.; Kavan, P.; Mathieu, D.; Phuphanich, S.; Cseh, A.; Fu, Y.; et al. Phase I/Randomized Phase II Study of Afatinib, an Irreversible ErbB Family Blocker, with or without Protracted Temozolomide in Adults with Recurrent Glioblastoma. Neuro-Oncology 2015, 17, 430–439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Sepúlveda-Sánchez, J.M.; Vaz, M.Á.; Balañá, C.; Gil-Gil, M.; Reynés, G.; Gallego, Ó.; Martínez-García, M.; Vicente, E.; Quindós, M.; Luque, R.; et al. Phase II Trial of Dacomitinib, a Pan–Human EGFR Tyrosine Kinase Inhibitor, in Recurrent Glioblastoma Patients with EGFR Amplification. Neuro-Oncology 2017, 19, 1522–1531. [Google Scholar] [CrossRef] [Green Version]
  127. Reardon, D.A.; Groves, M.D.; Wen, P.Y.; Nabors, L.; Mikkelsen, T.; Rosenfeld, S.; Raizer, J.; Barriuso, J.; McLendon, R.E.; Suttle, A.B.; et al. A Phase I/II Trial of Pazopanib in Combination with Lapatinib in Adult Patients with Relapsed Malignant Glioma. Clin. Cancer Res. 2013, 19, 900–908. [Google Scholar] [CrossRef] [Green Version]
  128. Neyns, B.; Sadones, J.; Joosens, E.; Bouttens, F.; Verbeke, L.; Baurain, J.-F.; D’Hondt, L.; Strauven, T.; Chaskis, C.; In’t Veld, P.; et al. Stratified Phase II Trial of Cetuximab in Patients with Recurrent High-Grade Glioma. Ann. Oncol. 2009, 20, 1596–1603. [Google Scholar] [CrossRef]
  129. Solomon, M.T.; Miranda, N.; Jorrín, E.; Chon, I.; Marinello, J.J.; Alert, J.; Lorenzo-Luaces, P.; Crombet, T. Nimotuzumab in Combination with Radiotherapy in High Grade Glioma Patients: A Single Institution Experience. Cancer Biol. Ther. 2014, 15, 504–509. [Google Scholar] [CrossRef] [Green Version]
  130. Nitta, Y.; Shimizu, S.; Shishido-Hara, Y.; Suzuki, K.; Shiokawa, Y.; Nagane, M. Nimotuzumab Enhances Temozolomide-Induced Growth Suppression of Glioma Cells Expressing Mutant EGFR in Vivo. Cancer Med. 2016, 5, 486–499. [Google Scholar] [CrossRef] [Green Version]
  131. Lim, Y.; Yoo, J.; Kim, M.-S.; Hur, M.; Lee, E.H.; Hur, H.-S.; Lee, J.-C.; Lee, S.-N.; Park, T.W.; Lee, K.; et al. GC1118, an Anti-EGFR Antibody with a Distinct Binding Epitope and Superior Inhibitory Activity against High-Affinity EGFR Ligands. Mol. Cancer Ther. 2016, 15, 251–263. [Google Scholar] [CrossRef] [Green Version]
  132. Iida, M.; Brand, T.M.; Starr, M.M.; Li, C.; Huppert, E.J.; Luthar, N.; Pedersen, M.W.; Horak, I.D.; Kragh, M.; Wheeler, D.L. Sym004, a Novel EGFR Antibody Mixture, Can Overcome Acquired Resistance to Cetuximab. Neoplasia 2013, 15, 1196–1206. [Google Scholar] [CrossRef]
  133. Montagut, C.; Argilés, G.; Ciardiello, F.; Poulsen, T.T.; Dienstmann, R.; Kragh, M.; Kopetz, S.; Lindsted, T.; Ding, C.; Vidal, J.; et al. Efficacy of Sym004 in Patients with Metastatic Colorectal Cancer with Acquired Resistance to Anti-EGFR Therapy and Molecularly Selected by Circulating Tumor DNA Analyses: A Phase 2 Randomized Clinical Trial. JAMA Oncol. 2018, 4, e175245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Phillips, A.C.; Boghaert, E.R.; Vaidya, K.S.; Mitten, M.J.; Norvell, S.; Falls, H.D.; DeVries, P.J.; Cheng, D.; Meulbroek, J.A.; Buchanan, F.G.; et al. ABT-414, an Antibody-Drug Conjugate Targeting a Tumor-Selective EGFR Epitope. Mol. Cancer Ther. 2016, 15, 661–669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Van Den Bent, M.; Eoli, M.; Sepulveda, J.M.; Smits, M.; Walenkamp, A.; Frenel, J.-S.; Franceschi, E.; Clement, P.M.; Chinot, O.; De Vos, F.; et al. INTELLANCE 2/EORTC 1410 Randomized Phase II Study of Depatux-M Alone and with Temozolomide vs Temozolomide or Lomustine in Recurrent EGFR Amplified Glioblastoma. Neuro-Oncology 2020, 22, 684–693. [Google Scholar] [CrossRef]
  136. Clarke, J.L.; Molinaro, A.M.; Phillips, J.J.; Butowski, N.A.; Chang, S.M.; Perry, A.; Costello, J.F.; DeSilva, A.A.; Rabbitt, J.E.; Prados, M.D. A Single-Institution Phase II Trial of Radiation, Temozolomide, Erlotinib, and Bevacizumab for Initial Treatment of Glioblastoma. Neuro-Oncology 2014, 16, 984–990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Uhm, J.H.; Ballman, K.V.; Wu, W.; Giannini, C.; Krauss, J.C.; Buckner, J.C.; James, C.D.; Scheithauer, B.W.; Behrens, R.J.; Flynn, P.J.; et al. Phase II Evaluation of Gefitinib in Patients with Newly Diagnosed Grade 4 Astrocytoma: Mayo/North Central Cancer Treatment Group Study N0074. Int. J. Radiat. Oncol. Biol. Phys. 2011, 80, 347–353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Arkhipov, A.; Shan, Y.; Kim, E.T.; Dror, R.O.; Shaw, D.E. Her2 Activation Mechanism Reflects Evolutionary Preservation of Asymmetric Ectodomain Dimers in the Human EGFR Family. eLife 2013, 2, e00708. [Google Scholar] [CrossRef] [PubMed]
  139. Iqbal, N.; Iqbal, N. Human Epidermal Growth Factor Receptor 2 (HER2) in Cancers: Overexpression and Therapeutic Implications. Mol. Biol. Int. 2014, 2014. [Google Scholar] [CrossRef]
  140. Thiessen, B.; Stewart, C.; Tsao, M.; Kamel-Reid, S.; Schaiquevich, P.; Mason, W.; Easaw, J.; Belanger, K.; Forsyth, P.; McIntosh, L.; et al. A Phase I/II Trial of GW572016 (Lapatinib) in Recurrent Glioblastoma Multiforme: Clinical Outcomes, Pharmacokinetics and Molecular Correlation. Cancer Chemother. Pharmacol. 2010, 65, 353–361. [Google Scholar] [CrossRef]
  141. Yu, A.; Faiq, N.; Green, S.; Lai, A.; Green, R.; Hu, J.; Cloughesy, T.F.; Mellinghoff, I.; Nghiemphu, P.L. Report of Safety of Pulse Dosing of Lapatinib with Temozolomide and Radiation Therapy for Newly-Diagnosed Glioblastoma in a Pilot Phase II Study. J. Neurooncol. 2017, 134, 357–362. [Google Scholar] [CrossRef]
  142. Cicenas, J.; Cicenas, E. Multi-Kinase Inhibitors, AURKs and Cancer. Med. Oncol. 2016, 33, 43. [Google Scholar] [CrossRef]
  143. Shen, G.; Zheng, F.; Ren, D.; Du, F.; Dong, Q.; Wang, Z.; Zhao, F.; Ahmad, R.; Zhao, J. Anlotinib: A Novel Multi-Targeting Tyrosine Kinase Inhibitor in Clinical Development. J. Hematol. Oncol. 2018, 11, 120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Goh, K.C.; Novotny-Diermayr, V.; Hart, S.; Ong, L.C.; Loh, Y.K.; Cheong, A.; Tan, Y.C.; Hu, C.; Jayaraman, R.; William, A.D.; et al. TG02, a Novel Oral Multi-Kinase Inhibitor of CDKs, JAK2 and FLT3 with Potent Anti-Leukemic Properties. Leukemia 2012, 26, 236–243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Du, E.; Li, X.; He, S.; Li, X.; He, S. The Critical Role of the Interplays of EphrinB2/EphB4 and VEGF in the Induction of Angiogenesis. Mol. Biol. Rep. 2020, 47, 4681–4690. [Google Scholar] [CrossRef] [PubMed]
  146. Jo, M.-Y.; Kim, Y.G.; Kim, Y.; Lee, S.J.; Kim, M.H.; Joo, K.M.; Kim, H.H.; Nam, D.-H. Combined Therapy of Temozolomide and ZD6474 (Vandetanib) Effectively Reduces Glioblastoma Tumor Volume through Anti-Angiogenic and Anti-Proliferative Mechanisms. Mol. Med. Rep. 2012, 6, 88–92. [Google Scholar] [CrossRef] [PubMed]
  147. Lee, E.Q.; Kaley, T.J.; Duda, D.G.; Schiff, D.; Lassman, A.B.; Wong, E.T.; Mikkelsen, T.; Purow, B.W.; Muzikansky, A.; Ancukiewicz, M.; et al. A Multicenter, Phase II, Randomized, Noncomparative Clinical Trial of Radiation and Temozolomide with or without Vandetanib in Newly Diagnosed Glioblastoma Patients. Clin. Cancer Res. 2015, 21, 3610–3618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. McNeill, K.; Iwamoto, F.; Kreisl, T.; Sul, J.; Shih, J.; Fine, H. AT-39A randomized phase II trial of vandetanib (ZD6474) in combination with carboplatin versus carboplatin alone in adults with recurrent glioblastoma. Neuro-Oncology 2014, 16, v17. [Google Scholar] [CrossRef] [Green Version]
  149. Taylor, J.W.; Dietrich, J.; Gerstner, E.R.; Norden, A.D.; Rinne, M.L.; Cahill, D.P.; Stemmer-Rachamimov, A.; Wen, P.Y.; Betensky, R.A.; Giorgio, D.H.; et al. Phase 2 Study of Bosutinib, a Src Inhibitor, in Adults with Recurrent Glioblastoma. J. Neurooncol. 2015, 121, 557–563. [Google Scholar] [CrossRef] [Green Version]
  150. Joensuu, H.; Puputti, M.; Sihto, H.; Tynninen, O.; Nupponen, N.N. Amplification of Genes Encoding KIT, PDGFRalpha and VEGFR2 Receptor Tyrosine Kinases Is Frequent in Glioblastoma Multiforme. J. Pathol. 2005, 207, 224–231. [Google Scholar] [CrossRef]
  151. Reardon, D.A.; Dresemann, G.; Taillibert, S.; Campone, M.; van den Bent, M.; Clement, P.; Blomquist, E.; Gordower, L.; Schultz, H.; Raizer, J.; et al. Multicentre Phase II Studies Evaluating Imatinib plus Hydroxyurea in Patients with Progressive Glioblastoma. Br. J. Cancer 2009, 101, 1995–2004. [Google Scholar] [CrossRef]
  152. Lassman, A.B.; Pugh, S.L.; Gilbert, M.R.; Aldape, K.D.; Geinoz, S.; Beumer, J.H.; Christner, S.M.; Komaki, R.; DeAngelis, L.M.; Gaur, R.; et al. Phase 2 Trial of Dasatinib in Target-Selected Patients with Recurrent Glioblastoma (RTOG 0627). Neuro-Oncology 2015, 17, 992–998. [Google Scholar] [CrossRef] [Green Version]
  153. Odia, Y.; Sul, J.; Shih, J.H.; Kreisl, T.N.; Butman, J.A.; Iwamoto, F.M.; Fine, H.A. A Phase II Trial of Tandutinib (MLN 518) in Combination with Bevacizumab for Patients with Recurrent Glioblastoma. CNS Oncol. 2016, 5, 59–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Batchelor, T.T.; Gerstner, E.R.; Ye, X.; Desideri, S.; Duda, D.G.; Peereboom, D.; Lesser, G.J.; Chowdhary, S.; Wen, P.Y.; Grossman, S.; et al. Feasibility, Phase I, and Phase II Studies of Tandutinib, an Oral Platelet-Derived Growth Factor Receptor-β Tyrosine Kinase Inhibitor, in Patients with Recurrent Glioblastoma. Neuro-Oncology 2017, 19, 567–575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Dresemann, G.; Weller, M.; Rosenthal, M.A.; Wedding, U.; Wagner, W.; Engel, E.; Heinrich, B.; Mayer-Steinacker, R.; Karup-Hansen, A.; Fluge, O.; et al. Imatinib in Combination with Hydroxyurea versus Hydroxyurea Alone as Oral Therapy in Patients with Progressive Pretreated Glioblastoma Resistant to Standard Dose Temozolomide. J. Neurooncol. 2010, 96, 393–402. [Google Scholar] [CrossRef] [Green Version]
  156. Wen, P.Y.; Yung, W.K.A.; Lamborn, K.R.; Dahia, P.L.; Wang, Y.; Peng, B.; Abrey, L.E.; Raizer, J.; Cloughesy, T.F.; Fink, K.; et al. Phase I/II Study of Imatinib Mesylate for Recurrent Malignant Gliomas: North American Brain Tumor Consortium Study 99-08. Clin. Cancer Res. 2006, 12, 4899–4907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Raymond, E.; Brandes, A.A.; Dittrich, C.; Fumoleau, P.; Coudert, B.; Clement, P.M.J.; Frenay, M.; Rampling, R.; Stupp, R.; Kros, J.M.; et al. Phase II Study of Imatinib in Patients with Recurrent Gliomas of Various Histologies: A European Organisation for Research and Treatment of Cancer Brain Tumor Group Study. J. Clin. Oncol. 2008, 26, 4659–4665. [Google Scholar] [CrossRef] [Green Version]
  158. Galanis, E.; Anderson, S.K.; Twohy, E.L.; Carrero, X.W.; Dixon, J.G.; Tran, D.D.; Jeyapalan, S.A.; Anderson, D.M.; Kaufmann, T.J.; Feathers, R.W.; et al. A Phase 1 and Randomized, Placebo-Controlled Phase 2 Trial of Bevacizumab plus Dasatinib in Patients with Recurrent Glioblastoma: Alliance/North Central Cancer Treatment Group N0872. Cancer 2019, 125, 3790–3800. [Google Scholar] [CrossRef]
  159. Franceschi, E.; Stupp, R.; van den Bent, M.J.; van Herpen, C.; Laigle Donadey, F.; Gorlia, T.; Hegi, M.; Lhermitte, B.; Strauss, L.C.; Allgeier, A.; et al. EORTC 26083 Phase I/II Trial of Dasatinib in Combination with CCNU in Patients with Recurrent Glioblastoma. Neuro-Oncology 2012, 14, 1503–1510. [Google Scholar] [CrossRef] [Green Version]
  160. Balaña, C.; Gil, M.J.; Perez, P.; Reynes, G.; Gallego, O.; Ribalta, T.; Capellades, J.; Gonzalez, S.; Verger, E. Sunitinib Administered Prior to Radiotherapy in Patients with Non-Resectable Glioblastoma: Results of a Phase II Study. Target. Oncol. 2014, 9, 321–329. [Google Scholar] [CrossRef] [PubMed]
  161. Hutterer, M.; Nowosielski, M.; Haybaeck, J.; Embacher, S.; Stockhammer, F.; Gotwald, T.; Holzner, B.; Capper, D.; Preusser, M.; Marosi, C.; et al. A Single-Arm Phase II Austrian/German Multicenter Trial on Continuous Daily Sunitinib in Primary Glioblastoma at First Recurrence (SURGE 01-07). Neuro-Oncology 2014, 16, 92–102. [Google Scholar] [CrossRef] [Green Version]
  162. Pan, E.; Yu, D.; Yue, B.; Potthast, L.; Chowdhary, S.; Smith, P.; Chamberlain, M. A Prospective Phase II Single-Institution Trial of Sunitinib for Recurrent Malignant Glioma. J. Neurooncol. 2012, 110, 111–118. [Google Scholar] [CrossRef] [Green Version]
  163. Phuphanich, S.; Raizer, J.; Chamberlain, M.; Canelos, P.; Narwal, R.; Hong, S.; Miday, R.; Nade, M.; Laubscher, K. Phase II Study of MEDI-575, an Anti-Platelet-Derived Growth Factor-α Antibody, in Patients with Recurrent Glioblastoma. J. Neurooncol. 2017, 131, 185–191. [Google Scholar] [CrossRef]
  164. Lee, E.Q.; Muzikansky, A.; Reardon, D.A.; Dietrich, J.; Nayak, L.; Duda, D.G.; Chukwueke, U.N.; Beroukhim, R.; Doherty, L.M.; Kane, C.; et al. Phase II Trial of Ponatinib in Patients with Bevacizumab-Refractory Glioblastoma. J. Clin. Oncol. 2018, 36, 2032. [Google Scholar] [CrossRef]
  165. Aiken, R.; Axelson, M.; Harmenberg, J.; Klockare, M.; Larsson, O.; Wassberg, C. Phase I Clinical Trial of AXL1717 for Treatment of Relapsed Malignant Astrocytomas: Analysis of Dose and Response. Oncotarget 2017, 8, 81501–81510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Grisanti, S.; Ferrari, V.D.; Buglione, M.; Agazzi, G.M.; Liserre, R.; Poliani, L.; Buttolo, L.; Gipponi, S.; Pedersini, R.; Consoli, F.; et al. Second Line Treatment of Recurrent Glioblastoma with Sunitinib: Results of a Phase II Study and Systematic Review of Literature. J. Neurosurg. Sci. 2016, 63, 458–467. [Google Scholar] [CrossRef] [PubMed]
  167. Fondevila, F.; Méndez-Blanco, C.; Fernández-Palanca, P.; González-Gallego, J.; Mauriz, J.L. Anti-Tumoral Activity of Single and Combined Regorafenib Treatments in Preclinical Models of Liver and Gastrointestinal Cancers. Exp. Mol. Med. 2019, 51, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. O’Hare, T.; Shakespeare, W.C.; Zhu, X.; Eide, C.A.; Rivera, V.M.; Wang, F.; Adrian, L.T.; Zhou, T.; Huang, W.-S.; Xu, Q.; et al. AP24534, a Pan-BCR-ABL Inhibitor for Chronic Myeloid Leukemia, Potently Inhibits the T315I Mutant and Overcomes Mutation-Based Resistance. Cancer Cell 2009, 16, 401–412. [Google Scholar] [CrossRef] [Green Version]
  169. Shah, G.D.; Loizos, N.; Youssoufian, H.; Schwartz, J.D.; Rowinsky, E.K. Rationale for the Development of IMC-3G3, a Fully Human Immunoglobulin G Subclass 1 Monoclonal Antibody Targeting the Platelet-Derived Growth Factor Receptor Alpha. Cancer 2010, 116, 1018–1026. [Google Scholar] [CrossRef]
  170. Zhang, M.; Liu, J.; Li, M.; Zhang, S.; Lu, Y.; Liang, Y.; Zhao, K.; Li, Y. Insulin-like Growth Factor 1/Insulin-like Growth Factor 1 Receptor Signaling Protects against Cell Apoptosis through the PI3K/AKT Pathway in Glioblastoma Cells. Exp. Ther. Med. 2018, 16, 1477–1482. [Google Scholar] [CrossRef]
  171. Maris, C.; D’Haene, N.; Trépant, A.-L.; Le Mercier, M.; Sauvage, S.; Allard, J.; Rorive, S.; Demetter, P.; Decaestecker, C.; Salmon, I. IGF-IR: A New Prognostic Biomarker for Human Glioblastoma. Br. J. Cancer 2015, 113, 729–737. [Google Scholar] [CrossRef]
  172. Jimenez-Pascual, A.; Siebzehnrubl, F.A. Fibroblast Growth Factor Receptor Functions in Glioblastoma. Cells 2019, 8, 715. [Google Scholar] [CrossRef] [Green Version]
  173. Lasorella, A.; Sanson, M.; Iavarone, A. FGFR-TACC Gene Fusions in Human Glioma. Neuro-Oncology 2017, 19, 475–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Kawauchi, D.; Takahashi, M.; Yamamuro, S.; Kobayashi, T.; Uchida, E.; Iwadate, Y.; Ichimura, K.; Tomiyama, A. ET-05 Alectinib and Ceritinib, the Second-Generation ALK Inhibitors, Effectively Induce Glioblastoma Cell Death. Neuro-Oncol. Adv. 2020, 2, ii6. [Google Scholar] [CrossRef]
  175. Lassen, U.; Sorensen, M.; Gaziel, T.B.; Hasselbalch, B.; Poulsen, H.S. Phase II Study of Bevacizumab and Temsirolimus Combination Therapy for Recurrent Glioblastoma Multiforme. Anticancer Res. 2013, 33, 1657–1660. [Google Scholar]
  176. Galanis, E.; Buckner, J.C.; Maurer, M.J.; Kreisberg, J.I.; Ballman, K.; Boni, J.; Peralba, J.M.; Jenkins, R.B.; Dakhil, S.R.; Morton, R.F.; et al. Phase II Trial of Temsirolimus (CCI-779) in Recurrent Glioblastoma Multiforme: A North Central Cancer Treatment Group Study. J. Clin. Oncol. 2005, 23, 5294–5304. [Google Scholar] [CrossRef]
  177. Schiff, D.; Jaeckle, K.A.; Anderson, S.K.; Galanis, E.; Giannini, C.; Buckner, J.C.; Stella, P.; Flynn, P.J.; Erickson, B.J.; Schwerkoske, J.F.; et al. Phase I/II Trial of Temsirolimus and Sorafenib in Treatment of Patients with Recurrent Glioblastoma: North Central Cancer Treatment Group Study/Alliance N0572. Cancer 2018, 124, 1455–1463. [Google Scholar] [CrossRef] [PubMed]
  178. Wick, W.; Gorlia, T.; Bady, P.; Platten, M.; van den Bent, M.J.; Taphoorn, M.J.B.; Steuve, J.; Brandes, A.A.; Hamou, M.-F.; Wick, A.; et al. Phase II Study of Radiotherapy and Temsirolimus versus Radiochemotherapy with Temozolomide in Patients with Newly Diagnosed Glioblastoma without MGMT Promoter Hypermethylation (EORTC 26082). Clin. Cancer Res. 2016, 22, 4797–4806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Chang, S.M.; Wen, P.; Cloughesy, T.; Greenberg, H.; Schiff, D.; Conrad, C.; Fink, K.; Robins, H.I.; De Angelis, L.; Raizer, J.; et al. Phase II Study of CCI-779 in Patients with Recurrent Glioblastoma Multiforme. Investig. New Drugs 2005, 23, 357–361. [Google Scholar] [CrossRef]
  180. Hainsworth, J.D.; Shih, K.C.; Shepard, G.C.; Tillinghast, G.W.; Brinker, B.T.; Spigel, D.R. Phase II Study of Concurrent Radiation Therapy, Temozolomide, and Bevacizumab Followed by Bevacizumab/Everolimus as First-Line Treatment for Patients with Glioblastoma. Clin. Adv. Hematol. Oncol. 2012, 10, 240–246. [Google Scholar] [PubMed]
  181. Ma, D.J.; Galanis, E.; Anderson, S.K.; Schiff, D.; Kaufmann, T.J.; Peller, P.J.; Giannini, C.; Brown, P.D.; Uhm, J.H.; McGraw, S.; et al. A Phase II Trial of Everolimus, Temozolomide, and Radiotherapy in Patients with Newly Diagnosed Glioblastoma: NCCTG N057K. Neuro-Oncology 2015, 17, 1261–1269. [Google Scholar] [CrossRef]
  182. Chinnaiyan, P.; Won, M.; Wen, P.Y.; Rojiani, A.M.; Werner-Wasik, M.; Shih, H.A.; Ashby, L.S.; Michael Yu, H.-H.; Stieber, V.W.; Malone, S.C.; et al. A Randomized Phase II Study of Everolimus in Combination with Chemoradiation in Newly Diagnosed Glioblastoma: Results of NRG Oncology RTOG 0913. Neuro-Oncology 2018, 20, 666–673. [Google Scholar] [CrossRef] [Green Version]
  183. Pitz, M.W.; Eisenhauer, E.A.; MacNeil, M.V.; Thiessen, B.; Easaw, J.C.; Macdonald, D.R.; Eisenstat, D.D.; Kakumanu, A.S.; Salim, M.; Chalchal, H.; et al. Phase II Study of PX-866 in Recurrent Glioblastoma. Neuro-Oncology 2015, 17, 1270–1274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Wick, W.; Puduvalli, V.K.; Chamberlain, M.C.; van den Bent, M.J.; Carpentier, A.F.; Cher, L.M.; Mason, W.; Weller, M.; Hong, S.; Musib, L.; et al. Phase III Study of Enzastaurin Compared with Lomustine in the Treatment of Recurrent Intracranial Glioblastoma. J. Clin. Oncol. 2010, 28, 1168–1174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Wick, W.; Steinbach, J.P.; Platten, M.; Hartmann, C.; Wenz, F.; von Deimling, A.; Shei, P.; Moreau-Donnet, V.; Stoffregen, C.; Combs, S.E. Enzastaurin before and Concomitant with Radiation Therapy, Followed by Enzastaurin Maintenance Therapy, in Patients with Newly Diagnosed Glioblastoma without MGMT Promoter Hypermethylation. Neuro-Oncology 2013, 15, 1405–1412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Odia, Y.; Iwamoto, F.M.; Moustakas, A.; Fraum, T.J.; Salgado, C.A.; Li, A.; Kreisl, T.N.; Sul, J.; Butman, J.A.; Fine, H.A. A Phase II Trial of Enzastaurin (LY317615) in Combination with Bevacizumab in Adults with Recurrent Malignant Gliomas. J. Neurooncol. 2016, 127, 127–135. [Google Scholar] [CrossRef] [PubMed]
  187. Chandrika, G.; Natesh, K.; Ranade, D.; Chugh, A.; Shastry, P. Mammalian Target of Rapamycin Inhibitors, Temsirolimus and Torin 1, Attenuate Stemness-Associated Properties and Expression of Mesenchymal Markers Promoted by Phorbol-Myristate-Acetate and Oncostatin-M in Glioblastoma Cells. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2017, 39. [Google Scholar] [CrossRef] [Green Version]
  188. Lee, E.Q.; Kuhn, J.; Lamborn, K.R.; Abrey, L.; DeAngelis, L.M.; Lieberman, F.; Robins, H.I.; Chang, S.M.; Yung, W.K.A.; Drappatz, J.; et al. Phase I/II Study of Sorafenib in Combination with Temsirolimus for Recurrent Glioblastoma or Gliosarcoma: North American Brain Tumor Consortium Study 05-02. Neuro-Oncology 2012, 14, 1511–1518. [Google Scholar] [CrossRef] [Green Version]
  189. Arcella, A.; Biagioni, F.; Antonietta Oliva, M.; Bucci, D.; Frati, A.; Esposito, V.; Cantore, G.; Giangaspero, F.; Fornai, F. Rapamycin Inhibits the Growth of Glioblastoma. Brain Res. 2013, 1495, 37–51. [Google Scholar] [CrossRef]
  190. Mendiburu-Eliçabe, M.; Gil-Ranedo, J.; Izquierdo, M. Efficacy of Rapamycin against Glioblastoma Cancer Stem Cells. Clin. Transl. Oncol. 2014, 16, 495–502. [Google Scholar] [CrossRef]
  191. Ferrucci, M.; Biagioni, F.; Lenzi, P.; Gambardella, S.; Ferese, R.; Calierno, M.T.; Falleni, A.; Grimaldi, A.; Frati, A.; Esposito, V.; et al. Rapamycin Promotes Differentiation Increasing βIII-Tubulin, NeuN, and NeuroD While Suppressing Nestin Expression in Glioblastoma Cells. Oncotarget 2017, 8, 29574–29599. [Google Scholar] [CrossRef] [Green Version]
  192. Kahn, J.; Hayman, T.J.; Jamal, M.; Rath, B.H.; Kramp, T.; Camphausen, K.; Tofilon, P.J. The MTORC1/MTORC2 Inhibitor AZD2014 Enhances the Radiosensitivity of Glioblastoma Stem-like Cells. Neuro-Oncology 2014, 16, 29–37. [Google Scholar] [CrossRef] [Green Version]
  193. Shi, F.; Guo, H.; Zhang, R.; Liu, H.; Wu, L.; Wu, Q.; Liu, J.; Liu, T.; Zhang, Q. The PI3K Inhibitor GDC-0941 Enhances Radiosensitization and Reduces Chemoresistance to Temozolomide in GBM Cell Lines. Neuroscience 2017, 346, 298–308. [Google Scholar] [CrossRef]
  194. Netland, I.A.; Førde, H.E.; Sleire, L.; Leiss, L.; Rahman, M.A.; Skeie, B.S.; Miletic, H.; Enger, P.Ø.; Goplen, D. Treatment with the PI3K Inhibitor Buparlisib (NVP-BKM120) Suppresses the Growth of Established Patient-Derived GBM Xenografts and Prolongs Survival in Nude Rats. J. Neurooncol. 2016, 129, 57–66. [Google Scholar] [CrossRef] [Green Version]
  195. Speranza, M.-C.; Nowicki, M.O.; Behera, P.; Cho, C.-F.; Chiocca, E.A.; Lawler, S.E. BKM-120 (Buparlisib): A Phosphatidyl-Inositol-3 Kinase Inhibitor with Anti-Invasive Properties in Glioblastoma. Sci. Rep. 2016, 6, 20189. [Google Scholar] [CrossRef] [Green Version]
  196. Wen, P.Y.; Touat, M.; Alexander, B.M.; Mellinghoff, I.K.; Ramkissoon, S.; McCluskey, C.S.; Pelton, K.; Haidar, S.; Basu, S.S.; Gaffey, S.C.; et al. Buparlisib in Patients with Recurrent Glioblastoma Harboring Phosphatidylinositol 3-Kinase Pathway Activation: An Open-Label, Multicenter, Multi-Arm, Phase II Trial. J. Clin. Oncol. 2019, 37, 741–750. [Google Scholar] [CrossRef]
  197. Koul, D.; Shen, R.; Kim, Y.-W.; Kondo, Y.; Lu, Y.; Bankson, J.; Ronen, S.M.; Kirkpatrick, D.L.; Powis, G.; Yung, W.K.A. Cellular and in Vivo Activity of a Novel PI3K Inhibitor, PX-866, against Human Glioblastoma. Neuro-Oncology 2010, 12, 559–569. [Google Scholar] [CrossRef]
  198. Ramezani, S.; Vousooghi, N.; Ramezani Kapourchali, F.; Joghataei, M.T. Perifosine Enhances Bevacizumab-Induced Apoptosis and Therapeutic Efficacy by Targeting PI3K/AKT Pathway in a Glioblastoma Heterotopic Model. Apoptosis Int. J. Program. Cell Death 2017, 22, 1025–1034. [Google Scholar] [CrossRef] [PubMed]
  199. Alonso-Basanta, M.; Fang, P.; Maity, A.; Hahn, S.M.; Lustig, R.A.; Dorsey, J.F. A Phase I Study of Nelfinavir Concurrent with Temozolomide and Radiotherapy in Patients with Glioblastoma Multiforme. J. Neurooncol. 2014, 116, 365–372. [Google Scholar] [CrossRef] [PubMed]
  200. Narayan, R.S.; Fedrigo, C.A.; Brands, E.; Dik, R.; Stalpers, L.J.A.; Baumert, B.G.; Slotman, B.J.; Westerman, B.A.; Peters, G.J.; Sminia, P. The Allosteric AKT Inhibitor MK2206 Shows a Synergistic Interaction with Chemotherapy and Radiotherapy in Glioblastoma Spheroid Cultures. BMC Cancer 2017, 17, 204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Mason, W.P.; Belanger, K.; Nicholas, G.; Vallières, I.; Mathieu, D.; Kavan, P.; Desjardins, A.; Omuro, A.; Reymond, D. A Phase II Study of the Ras-MAPK Signaling Pathway Inhibitor TLN-4601 in Patients with Glioblastoma at First Progression. J. Neurooncol. 2012, 107, 343–349. [Google Scholar] [CrossRef] [PubMed]
  202. Hainsworth, J.D.; Ervin, T.; Friedman, E.; Priego, V.; Murphy, P.B.; Clark, B.L.; Lamar, R.E. Concurrent Radiotherapy and Temozolomide Followed by Temozolomide and Sorafenib in the First-Line Treatment of Patients with Glioblastoma Multiforme. Cancer 2010, 116, 3663–3669. [Google Scholar] [CrossRef] [PubMed]
  203. Reardon, D.A.; Vredenburgh, J.J.; Desjardins, A.; Peters, K.; Gururangan, S.; Sampson, J.H.; Marcello, J.; Herndon, J.E.; McLendon, R.E.; Janney, D.; et al. Effect of CYP3A-Inducing Anti-Epileptics on Sorafenib Exposure: Results of a Phase II Study of Sorafenib plus Daily Temozolomide in Adults with Recurrent Glioblastoma. J. Neurooncol. 2011, 101, 57–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Galanis, E.; Anderson, S.K.; Lafky, J.M.; Uhm, J.H.; Giannini, C.; Kumar, S.K.; Kimlinger, T.K.; Northfelt, D.W.; Flynn, P.J.; Jaeckle, K.A.; et al. Phase II Study of Bevacizumab in Combination with Sorafenib in Recurrent Glioblastoma (N0776): A North Central Cancer Treatment Group Trial. Clin. Cancer Res. 2013, 19, 4816–4823. [Google Scholar] [CrossRef] [Green Version]
  205. Altwairgi, A.K.; Alghareeb, W.; Alnajjar, F.; Alsaeed, E.; Balbaid, A.; Alhussain, H.; Aldanan, S.; Orz, Y.; Lari, A.; Alsharm, A. Phase II Study of Atorvastatin in Combination with Radiotherapy and Temozolomide In Patients with Glioblastoma (ART): Interim Analysis Report. Ann. Oncol. 2016, 27, vi103–vi113. [Google Scholar] [CrossRef]
  206. Wilhelm, S.; Carter, C.; Lynch, M.; Lowinger, T.; Dumas, J.; Smith, R.A.; Schwartz, B.; Simantov, R.; Kelley, S. Discovery and Development of Sorafenib: A Multikinase Inhibitor for Treating Cancer. Nat. Rev. Drug Discov. 2006, 5, 835–844. [Google Scholar] [CrossRef] [PubMed]
  207. Jakubowicz-Gil, J.; Bądziul, D.; Langner, E.; Wertel, I.; Zając, A.; Rzeski, W. Temozolomide and Sorafenib as Programmed Cell Death Inducers of Human Glioma Cells. Pharmacol. Rep. PR 2017, 69, 779–787. [Google Scholar] [CrossRef] [PubMed]
  208. Riedel, M.; Struve, N.; Müller-Goebel, J.; Köcher, S.; Petersen, C.; Dikomey, E.; Rothkamm, K.; Kriegs, M. Sorafenib Inhibits Cell Growth but Fails to Enhance Radio- and Chemosensitivity of Glioblastoma Cell Lines. Oncotarget 2016, 7, 61988–61995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Zustovich, F.; Landi, L.; Lombardi, G.; Porta, C.; Galli, L.; Fontana, A.; Amoroso, D.; Galli, C.; Andreuccetti, M.; Falcone, A.; et al. Sorafenib plus Daily Low-Dose Temozolomide for Relapsed Glioblastoma: A Phase II Study. Anticancer Res. 2013, 33, 3487–3494. [Google Scholar] [CrossRef]
  210. Peng, P.; Wei, W.; Long, C.; Li, J. Atorvastatin Augments Temozolomide’s Efficacy in Glioblastoma via Prenylation-Dependent Inhibition of Ras Signaling. Biochem. Biophys. Res. Commun. 2017, 489, 293–298. [Google Scholar] [CrossRef]
  211. Azaro, A.; Plummer, E.R.; Urruticoechea, A.; Rodon, J.; Haris, N.R.M.; Veal, G.; Perier, A.; Tur, V.; Escriba, P.V.; Busquets, X.; et al. Final Report of a Phase I Study of 2-Hydroxyoleic Acid (2OHOA) a Novel Sphingomyelin Synthase Activator in Patients (Pt) with Advanced Solid Tumors (AST) Including Recurrent High Grade Gliomas (RHGG). J. Clin. Oncol. 2017, 35, 2554. [Google Scholar] [CrossRef]
  212. Escamilla-Ramírez, A.; Castillo-Rodríguez, R.A.; Zavala-Vega, S.; Jimenez-Farfan, D.; Anaya-Rubio, I.; Briseño, E.; Palencia, G.; Guevara, P.; Cruz-Salgado, A.; Sotelo, J.; et al. Autophagy as a Potential Therapy for Malignant Glioma. Pharmaceuticals 2020, 13, 156. [Google Scholar] [CrossRef]
  213. Kroemer, G.; Galluzzi, L.; Vandenabeele, P.; Abrams, J.; Alnemri, E.S.; Baehrecke, E.H.; Blagosklonny, M.V.; El-Deiry, W.S.; Golstein, P.; Green, D.R.; et al. Classification of Cell Death: Recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ. 2009, 16, 3–11. [Google Scholar] [CrossRef]
  214. Peter, M.E.; Krammer, P.H. The CD95(APO-1/Fas) DISC and Beyond. Cell Death Differ. 2003, 10, 26–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Wick, W.; Fricke, H.; Junge, K.; Kobyakov, G.; Martens, T.; Heese, O.; Wiestler, B.; Schliesser, M.G.; von Deimling, A.; Pichler, J.; et al. A Phase II, Randomized, Study of Weekly APG101+reirradiation versus Reirradiation in Progressive Glioblastoma. Clin. Cancer Res. 2014, 20, 6304–6313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Chi, A.S.; Tarapore, R.S.; Hall, M.D.; Shonka, N.; Gardner, S.; Umemura, Y.; Sumrall, A.; Khatib, Z.; Mueller, S.; Kline, C.; et al. Pediatric and Adult H3 K27M-Mutant Diffuse Midline Glioma Treated with the Selective DRD2 Antagonist ONC201. J. Neurooncol. 2019, 145, 97–105. [Google Scholar] [CrossRef] [PubMed]
  217. Lustig, R.; Mikkelsen, T.; Lesser, G.; Grossman, S.; Ye, X.; Desideri, S.; Fisher, J.; Wright, J. Phase II Preradiation R115777 (Tipifarnib) in Newly Diagnosed GBM with Residual Enhancing Disease. Neuro-Oncology 2008, 10, 1004–1009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Cloughesy, T.F.; Wen, P.Y.; Robins, H.I.; Chang, S.M.; Groves, M.D.; Fink, K.L.; Junck, L.; Schiff, D.; Abrey, L.; Gilbert, M.R.; et al. Phase II Trial of Tipifarnib in Patients with Recurrent Malignant Glioma Either Receiving or Not Receiving Enzyme-Inducing Antiepileptic Drugs: A North American Brain Tumor Consortium Study. J. Clin. Oncol. 2006, 24, 3651–3656. [Google Scholar] [CrossRef]
  219. Drachsler, M.; Kleber, S.; Mateos, A.; Volk, K.; Mohr, N.; Chen, S.; Cirovic, B.; Tüttenberg, J.; Gieffers, C.; Sykora, J.; et al. CD95 Maintains Stem Cell-like and Non-Classical EMT Programs in Primary Human Glioblastoma Cells. Cell Death Dis. 2016, 7, e2209. [Google Scholar] [CrossRef]
  220. Eisele, G.; Roth, P.; Hasenbach, K.; Aulwurm, S.; Wolpert, F.; Tabatabai, G.; Wick, W.; Weller, M. APO010, a Synthetic Hexameric CD95 Ligand, Induces Human Glioma Cell Death in Vitro and in Vivo. Neuro-Oncology 2011, 13, 155–164. [Google Scholar] [CrossRef]
  221. Ralff, M.D.; Lulla, A.R.; Wagner, J.; El-Deiry, W.S. ONC201: A New Treatment Option Being Tested Clinically for Recurrent Glioblastoma. Transl. Cancer Res. 2017, 6, S1239–S1243. [Google Scholar] [CrossRef]
  222. Kline, C.L.B.; Van den Heuvel, A.P.J.; Allen, J.E.; Prabhu, V.V.; Dicker, D.T.; El-Deiry, W.S. ONC201 Kills Solid Tumor Cells by Triggering an Integrated Stress Response Dependent on ATF4 Activation by Specific EIF2α Kinases. Sci. Signal. 2016, 9, ra18. [Google Scholar] [CrossRef] [Green Version]
  223. Arrillaga-Romany, I.; Chi, A.S.; Allen, J.E.; Oster, W.; Wen, P.Y.; Batchelor, T.T. A Phase 2 Study of the First Imipridone ONC201, a Selective DRD2 Antagonist for Oncology, Administered Every Three Weeks in Recurrent Glioblastoma. Oncotarget 2017, 8, 79298–79304. [Google Scholar] [CrossRef] [Green Version]
  224. Werry, E.L.; Barron, M.L.; Kassiou, M. TSPO as a Target for Glioblastoma Therapeutics. Biochem. Soc. Trans. 2015, 43, 531–536. [Google Scholar] [CrossRef] [PubMed]
  225. Werry, E.L.; King, V.A.; Barron, M.L.; Banister, S.D.; Sokias, R.; Kassiou, M. Derivatives of the Pyrazolo[1,5-a]Pyrimidine Acetamide DPA-713 as Translocator Protein (TSPO) Ligands and pro-Apoptotic Agents in Human Glioblastoma. Eur. J. Pharm. Sci. 2017, 96, 186–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. England, B.; Huang, T.; Karsy, M. Current Understanding of the Role and Targeting of Tumor Suppressor P53 in Glioblastoma Multiforme. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2013, 34, 2063–2074. [Google Scholar] [CrossRef]
  227. Hong, B.; van den Heuvel, A.P.J.; Prabhu, V.V.; Zhang, S.; El-Deiry, W.S. Targeting Tumor Suppressor P53 for Cancer Therapy: Strategies, Challenges and Opportunities. Curr. Drug Targets 2014, 15, 80–89. [Google Scholar] [CrossRef] [PubMed]
  228. Kim, S.-S.; Rait, A.; Kim, E.; Pirollo, K.F.; Chang, E.H. A Tumor-Targeting P53 Nanodelivery System Limits Chemoresistance to Temozolomide Prolonging Survival in a Mouse Model of Glioblastoma Multiforme. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 301–311. [Google Scholar] [CrossRef] [Green Version]
  229. Costa, B.; Bendinelli, S.; Gabelloni, P.; Da Pozzo, E.; Daniele, S.; Scatena, F.; Vanacore, R.; Campiglia, P.; Bertamino, A.; Gomez-Monterrey, I.; et al. Human Glioblastoma Multiforme: P53 Reactivation by a Novel MDM2 Inhibitor. PLoS ONE 2013, 8, e72281. [Google Scholar] [CrossRef]
  230. Villalonga-Planells, R.; Coll-Mulet, L.; Martínez-Soler, F.; Castaño, E.; Acebes, J.-J.; Giménez-Bonafé, P.; Gil, J.; Tortosa, A. Activation of P53 by Nutlin-3a Induces Apoptosis and Cellular Senescence in Human Glioblastoma Multiforme. PLoS ONE 2011, 6, e18588. [Google Scholar] [CrossRef] [Green Version]
  231. Renner, G.; Janouskova, H.; Noulet, F.; Koenig, V.; Guerin, E.; Bär, S.; Nuesch, J.; Rechenmacher, F.; Neubauer, S.; Kessler, H.; et al. Integrin A5β1 and P53 Convergent Pathways in the Control of Anti-Apoptotic Proteins PEA-15 and Survivin in High-Grade Glioma. Cell Death Differ. 2016, 23, 640–653. [Google Scholar] [CrossRef] [Green Version]
  232. Canon, J.; Osgood, T.; Olson, S.H.; Saiki, A.Y.; Robertson, R.; Yu, D.; Eksterowicz, J.; Ye, Q.; Jin, L.; Chen, A.; et al. The MDM2 Inhibitor AMG 232 Demonstrates Robust Antitumor Efficacy and Potentiates the Activity of P53-Inducing Cytotoxic Agents. Mol. Cancer Ther. 2015, 14, 649–658. [Google Scholar] [CrossRef] [Green Version]
  233. Lebowitz, P.F.; Sakamuro, D.; Prendergast, G.C. Farnesyl Transferase Inhibitors Induce Apoptosis of Ras-Transformed Cells Denied Substratum Attachment. Cancer Res. 1997, 57, 708–713. [Google Scholar]
  234. Basso, A.D.; Kirschmeier, P.; Bishop, W.R. Lipid Posttranslational Modifications. Farnesyl Transferase Inhibitors. J. Lipid Res. 2006, 47, 15–31. [Google Scholar] [CrossRef] [Green Version]
  235. Sebti, S.M.; Hamilton, A.D. Farnesyltransferase Inhibitors in Cancer Therapy. Available online: https://www.buecher.de/shop/mund/farnesyltransferase-inhibitors-in-cancer-therapy/sebti-sad-m-hamilton-andrew-d-eds-/products_products/detail/prod_id/20963971/ (accessed on 22 September 2020).
  236. Glass, T.L.; Liu, T.J.; Yung, W.K. Inhibition of Cell Growth in Human Glioblastoma Cell Lines by Farnesyltransferase Inhibitor SCH66336. Neuro-Oncology 2000, 2, 151–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Chaponis, D.; Barnes, J.W.; Dellagatta, J.L.; Kesari, S.; Fast, E.; Sauvageot, C.; Panagrahy, D.; Greene, E.R.; Ramakrishna, N.; Wen, P.Y.; et al. Lonafarnib (SCH66336) Improves the Activity of Temozolomide and Radiation for Orthotopic Malignant Gliomas. J. Neurooncol. 2011, 104, 179–189. [Google Scholar] [CrossRef] [Green Version]
  238. Daniele, S.; Taliani, S.; Da Pozzo, E.; Giacomelli, C.; Costa, B.; Trincavelli, M.L.; Rossi, L.; La Pietra, V.; Barresi, E.; Carotenuto, A.; et al. Apoptosis Therapy in Cancer: The First Single-Molecule Co-Activating P53 and the Translocator Protein in Glioblastoma. Sci. Rep. 2014, 4, 4749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Daniele, S.; Barresi, E.; Zappelli, E.; Marinelli, L.; Novellino, E.; Da Settimo, F.; Taliani, S.; Trincavelli, M.L.; Martini, C. Long Lasting MDM2/Translocator Protein Modulator: A New Strategy for Irreversible Apoptosis of Human Glioblastoma Cells. Oncotarget 2016, 7, 7866–7884. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  240. Qiu, J.; Levin, L.R.; Buck, J.; Reidenberg, M.M. Different Pathways of Cell Killing by Gossypol Enantiomers. Exp. Biol. Med. 2002, 227, 398–401. [Google Scholar] [CrossRef]
  241. Jarzabek, M.A.; Amberger-Murphy, V.; Callanan, J.J.; Gao, C.; Zagozdzon, A.M.; Shiels, L.; Wang, J.; Ligon, K.L.; Rich, B.E.; Dicker, P.; et al. Interrogation of Gossypol Therapy in Glioblastoma Implementing Cell Line and Patient-Derived Tumour Models. Br. J. Cancer 2014, 111, 2275–2286. [Google Scholar] [CrossRef] [Green Version]
  242. Jensen, S.A.; Day, E.S.; Ko, C.H.; Hurley, L.A.; Luciano, J.P.; Kouri, F.M.; Merkel, T.J.; Luthi, A.J.; Patel, P.C.; Cutler, J.I.; et al. Spherical Nucleic Acid Nanoparticle Conjugates as an RNAi-Based Therapy for Glioblastoma. Sci. Transl. Med. 2013, 5, 209ra152. [Google Scholar] [CrossRef] [Green Version]
  243. Würstle, S.; Schneider, F.; Ringel, F.; Gempt, J.; Lämmer, F.; Delbridge, C.; Wu, W.; Schlegel, J. Temozolomide Induces Autophagy in Primary and Established Glioblastoma Cells in an EGFR Independent Manner. Oncol. Lett. 2017, 14, 322–328. [Google Scholar] [CrossRef] [Green Version]
  244. Yuan, G.; Yan, S.-F.; Xue, H.; Zhang, P.; Sun, J.-T.; Li, G. Cucurbitacin I Induces Protective Autophagy in Glioblastoma in Vitro and in Vivo. J. Biol. Chem. 2014, 289, 10607–10619. [Google Scholar] [CrossRef] [Green Version]
  245. Liu, R.; Li, J.; Zhang, T.; Zou, L.; Chen, Y.; Wang, K.; Lei, Y.; Yuan, K.; Li, Y.; Lan, J.; et al. Itraconazole Suppresses the Growth of Glioblastoma through Induction of Autophagy: Involvement of Abnormal Cholesterol Trafficking. Autophagy 2014, 10, 1241–1255. [Google Scholar] [CrossRef] [Green Version]
  246. Zhang, P.; Sun, S.; Li, N.; Ho, A.S.W.; Kiang, K.M.Y.; Zhang, X.; Cheng, Y.S.; Poon, M.W.; Lee, D.; Pu, J.K.S.; et al. Rutin Increases the Cytotoxicity of Temozolomide in Glioblastoma via Autophagy Inhibition. J. Neurooncol. 2017, 132, 393–400. [Google Scholar] [CrossRef]
  247. Angeletti, F.; Fossati, G.; Pattarozzi, A.; Würth, R.; Solari, A.; Daga, A.; Masiello, I.; Barbieri, F.; Florio, T.; Comincini, S. Inhibition of the Autophagy Pathway Synergistically Potentiates the Cytotoxic Activity of Givinostat (ITF2357) on Human Glioblastoma Cancer Stem Cells. Front. Mol. Neurosci. 2016, 9, 107. [Google Scholar] [CrossRef] [Green Version]
  248. Maycotte, P.; Aryal, S.; Cummings, C.T.; Thorburn, J.; Morgan, M.J.; Thorburn, A. Chloroquine Sensitizes Breast Cancer Cells to Chemotherapy Independent of Autophagy. Autophagy 2012, 8, 200–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  249. Liu, X.; Sun, K.; Wang, H.; Dai, Y. Inhibition of Autophagy by Chloroquine Enhances the Antitumor Efficacy of Sorafenib in Glioblastoma. Cell. Mol. Neurobiol. 2016, 36, 1197–1208. [Google Scholar] [CrossRef] [PubMed]
  250. Sotelo, J.; Briceño, E.; López-González, M.A. Adding Chloroquine to Conventional Treatment for Glioblastoma Multiforme: A Randomized, Double-Blind, Placebo-Controlled Trial. Ann. Intern. Med. 2006, 144, 337–343. [Google Scholar] [CrossRef] [PubMed]
  251. 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]
  252. Taylor, J.W.; Parikh, M.; Phillips, J.J.; James, C.D.; Molinaro, A.M.; Butowski, N.A.; Clarke, J.L.; Oberheim-Bush, N.A.; Chang, S.M.; Berger, M.S.; et al. Phase-2 Trial of Palbociclib in Adult Patients with Recurrent RB1-Positive Glioblastoma. J. Neurooncol. 2018, 140, 477–483. [Google Scholar] [CrossRef]
  253. Friday, B.B.; Anderson, S.K.; Buckner, J.; Yu, C.; Giannini, C.; Geoffroy, F.; Schwerkoske, J.; Mazurczak, M.; Gross, H.; Pajon, E.; et al. Phase II Trial of Vorinostat in Combination with Bortezomib in Recurrent Glioblastoma: A North Central Cancer Treatment Group Study. Neuro-Oncology 2012, 14, 215–221. [Google Scholar] [CrossRef] [Green Version]
  254. Galanis, E.; Jaeckle, K.A.; Maurer, M.J.; Reid, J.M.; Ames, M.M.; Hardwick, J.S.; Reilly, J.F.; Loboda, A.; Nebozhyn, M.; Fantin, V.R.; et al. Phase II Trial of Vorinostat in Recurrent Glioblastoma Multiforme: A North Central Cancer Treatment Group Study. J. Clin. Oncol. 2009, 27, 2052–2058. [Google Scholar] [CrossRef] [Green Version]
  255. Iwamoto, F.M.; Lamborn, K.R.; Kuhn, J.G.; Wen, P.Y.; Yung, W.K.A.; Gilbert, M.R.; Chang, S.M.; Lieberman, F.S.; Prados, M.D.; Fine, H.A. A Phase I/II Trial of the Histone Deacetylase Inhibitor Romidepsin for Adults with Recurrent Malignant Glioma: North American Brain Tumor Consortium Study 03-03. Neuro-Oncology 2011, 13, 509–516. [Google Scholar] [CrossRef]
  256. Bogdahn, U.; Hau, P.; Stockhammer, G.; Venkataramana, N.K.; Mahapatra, A.K.; Suri, A.; Balasubramaniam, A.; Nair, S.; Oliushine, V.; Parfenov, V.; et al. Targeted Therapy for High-Grade Glioma with the TGF-Β2 Inhibitor Trabedersen: Results of a Randomized and Controlled Phase IIb Study. Neuro-Oncology 2011, 13, 132–142. [Google Scholar] [CrossRef] [Green Version]
  257. Wick, A.; Desjardins, A.; Suarez, C.; Forsyth, P.; Gueorguieva, I.; Burkholder, T.; Cleverly, A.L.; Estrem, S.T.; Wang, S.; Lahn, M.M.; et al. Phase 1b/2a Study of Galunisertib, a Small Molecule Inhibitor of Transforming Growth Factor-Beta Receptor I, in Combination with Standard Temozolomide-Based Radiochemotherapy in Patients with Newly Diagnosed Malignant Glioma. Investig. New Drugs 2020, 38, 1570–1579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  258. Blakeley, J.O.; Grossman, S.A.; Chi, A.S.; Mikkelsen, T.; Rosenfeld, M.R.; Ahluwalia, M.S.; Nabors, L.B.; Eichler, A.; Ribas, I.G.; Desideri, S.; et al. Phase II Study of Iniparib with Concurrent Chemoradiation in Patients with Newly Diagnosed Glioblastoma. Clin. Cancer Res. 2019, 25, 73–79. [Google Scholar] [CrossRef] [Green Version]
  259. Robins, H.I.; Zhang, P.; Gilbert, M.R.; Chakravarti, A.; de Groot, J.F.; Grimm, S.A.; Wang, F.; Lieberman, F.S.; Krauze, A.; Trotti, A.M.; et al. A Randomized Phase I/II Study of ABT-888 in Combination with Temozolomide in Recurrent Temozolomide Resistant Glioblastoma: An NRG Oncology RTOG Group Study. J. Neurooncol. 2016, 126, 309–316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  260. Cao, Y.; Li, X.; Kong, S.; Shang, S.; Qi, Y. CDK4/6 Inhibition Suppresses Tumour Growth and Enhances the Effect of Temozolomide in Glioma Cells. J. Cell. Mol. Med. 2020, 24, 5135–5145. [Google Scholar] [CrossRef] [Green Version]
  261. Sobhani, N.; D’Angelo, A.; Pittacolo, M.; Roviello, G.; Miccoli, A.; Corona, S.P.; Bernocchi, O.; Generali, D.; Otto, T. Updates on the CDK4/6 Inhibitory Strategy and Combinations in Breast Cancer. Cells 2019, 8, 321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Roeten, M.S.F.; Cloos, J.; Jansen, G. Positioning of Proteasome Inhibitors in Therapy of Solid Malignancies. Cancer Chemother. Pharmacol. 2018, 81, 227–243. [Google Scholar] [CrossRef] [Green Version]
  263. Huang, W.-J.; Chen, W.-W.; Zhang, X. Proteasome Inhibitors in Glioblastoma. Oncol. Lett. 2017, 13, 1058–1062. [Google Scholar] [CrossRef] [Green Version]
  264. Di, K.; Lloyd, G.K.; Abraham, V.; MacLaren, A.; Burrows, F.J.; Desjardins, A.; Trikha, M.; Bota, D.A. Marizomib Activity as a Single Agent in Malignant Gliomas: Ability to Cross the Blood-Brain Barrier. Neuro-Oncology 2016, 18, 840–848. [Google Scholar] [CrossRef] [PubMed]
  265. Roth, P.; Mason, W.P.; Richardson, P.G.; Weller, M. Proteasome Inhibition for the Treatment of Glioblastoma. Expert Opin. Investig. Drugs 2020, 29, 1133–1141. [Google Scholar] [CrossRef] [PubMed]
  266. Ceccacci, E.; Minucci, S. Inhibition of Histone Deacetylases in Cancer Therapy: Lessons from Leukaemia. Br. J. Cancer 2016, 114, 605–611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  267. Lee, P.; Murphy, B.; Miller, R.; Menon, V.; Banik, N.L.; Giglio, P.; Lindhorst, S.M.; Varma, A.K.; Vandergrift, W.A.; Patel, S.J.; et al. Mechanisms and Clinical Significance of Histone Deacetylase Inhibitors: Epigenetic Glioblastoma Therapy. Anticancer Res. 2015, 35, 615–625. [Google Scholar]
  268. Chen, R.; Zhang, M.; Zhou, Y.; Guo, W.; Yi, M.; Zhang, Z.; Ding, Y.; Wang, Y. The Application of Histone Deacetylases Inhibitors in Glioblastoma. J. Exp. Clin. Cancer Res. CR 2020, 39, 138. [Google Scholar] [CrossRef]
  269. Han, J.; Alvarez-Breckenridge, C.A.; Wang, Q.-E.; Yu, J. TGF-β Signaling and Its Targeting for Glioma Treatment. Am. J. Cancer Res. 2015, 5, 945–955. [Google Scholar]
  270. Birch, J.L.; Coull, B.J.; Spender, L.C.; Watt, C.; Willison, A.; Syed, N.; Chalmers, A.J.; Hossain-Ibrahim, M.K.; Inman, G.J. Multifaceted Transforming Growth Factor-Beta (TGFβ) Signalling in Glioblastoma. Cell. Signal. 2020, 72, 109638. [Google Scholar] [CrossRef]
  271. Towner, R.A.; Zalles, M.; Saunders, D.; Smith, N. Novel Approaches to Combat Chemoresistance against Glioblastomas. Cancer Drug Resist. 2020, 3, 686–698. [Google Scholar] [CrossRef]
  272. Sawyer, J.S.; Anderson, B.D.; Beight, D.W.; Campbell, R.M.; Jones, M.L.; Herron, D.K.; Lampe, J.W.; McCowan, J.R.; McMillen, W.T.; Mort, N.; et al. Synthesis and Activity of New Aryl- and Heteroaryl-Substituted Pyrazole Inhibitors of the Transforming Growth Factor-Beta Type I Receptor Kinase Domain. J. Med. Chem. 2003, 46, 3953–3956. [Google Scholar] [CrossRef]
  273. Murnyák, B.; Kouhsari, M.C.; Hershkovitch, R.; Kálmán, B.; Marko-Varga, G.; Klekner, Á.; Hortobágyi, T. PARP1 Expression and Its Correlation with Survival Is Tumour Molecular Subtype Dependent in Glioblastoma. Oncotarget 2017, 8, 46348–46362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Blakeley, J.O.; Grossman, S.A.; Mikkelsen, T.; Rosenfeld, M.R.; Peereboom, D.; Nabors, L.B.; Chi, A.S.; Emmons, G.; Ribas, I.G.; Supko, J.G.; et al. Phase I Study of Iniparib Concurrent with Monthly or Continuous Temozolomide Dosing Schedules in Patients with Newly Diagnosed Malignant Gliomas. J. Neurooncol. 2015, 125, 123–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Onishi, M.; Kurozumi, K.; Ichikawa, T.; Date, I. Mechanisms of Tumor Development and Anti-Angiogenic Therapy in Glioblastoma Multiforme. Neurol. Med. Chir. 2013, 53, 755–763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  276. Friedman, H.S.; Prados, M.D.; Wen, P.Y.; Mikkelsen, T.; Schiff, D.; Abrey, L.E.; Yung, W.K.A.; Paleologos, N.; Nicholas, M.K.; Jensen, R.; 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]
  277. 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] [Green Version]
  278. Brandes, A.A.; Gil-Gil, M.; Saran, F.; Carpentier, A.F.; Nowak, A.K.; Mason, W.; Zagonel, V.; Dubois, F.; Finocchiaro, G.; Fountzilas, G.; et al. A Randomized Phase II Trial (TAMIGA) Evaluating the Efficacy and Safety of Continuous Bevacizumab Through Multiple Lines of Treatment for Recurrent Glioblastoma. Oncologist 2019, 24, 521–528. [Google Scholar] [CrossRef] [Green Version]
  279. Peters, K.B.; Lou, E.; Desjardins, A.; Reardon, D.A.; Lipp, E.S.; Miller, E.; Herndon, J.E.; McSherry, F.; Friedman, H.S.; Vredenburgh, J.J. Phase II Trial of Upfront Bevacizumab, Irinotecan, and Temozolomide for Unresectable Glioblastoma. Oncologist 2015, 20, 727–728. [Google Scholar] [CrossRef] [Green Version]
  280. Wirsching, H.-G.; Tabatabai, G.; Roelcke, U.; Hottinger, A.F.; Jörger, F.; Schmid, A.; Plasswilm, L.; Schrimpf, D.; Mancao, C.; Capper, D.; et al. Bevacizumab plus Hypofractionated Radiotherapy versus Radiotherapy Alone in Elderly Patients with Glioblastoma: The Randomized, Open-Label, Phase II ARTE Trial. Ann. Oncol. 2018, 29, 1423–1430. [Google Scholar] [CrossRef]
  281. Reyes-Botero, G.; Cartalat-Carel, S.; Chinot, O.L.; Barrie, M.; Taillandier, L.; Beauchesne, P.; Catry-Thomas, I.; Barrière, J.; Guillamo, J.-S.; Fabbro, M.; et al. Temozolomide Plus Bevacizumab in Elderly Patients with Newly Diagnosed Glioblastoma and Poor Performance Status: An ANOCEF Phase II Trial (ATAG). Oncologist 2018, 23, 524.e44. [Google Scholar] [CrossRef] [Green Version]
  282. Ghiaseddin, A.; Reardon, D.; Massey, W.; Mannerino, A.; Lipp, E.S.; Herndon, J.E.; McSherry, F.; Desjardins, A.; Randazzo, D.; Friedman, H.S.; et al. Phase II Study of Bevacizumab and Vorinostat for Patients with Recurrent World Health Organization Grade 4 Malignant Glioma. Oncologist 2018, 23, 157-e21. [Google Scholar] [CrossRef] [Green Version]
  283. Vredenburgh, J.J.; Desjardins, A.; Herndon, J.E.; Dowell, J.M.; Reardon, D.A.; Quinn, J.A.; Rich, J.N.; Sathornsumetee, S.; Gururangan, S.; Wagner, M.; et al. Phase II Trial of Bevacizumab and Irinotecan in Recurrent Malignant Glioma. Clin. Cancer Res. 2007, 13, 1253–1259. [Google Scholar] [CrossRef] [Green Version]
  284. Ney, D.E.; Carlson, J.A.; Damek, D.M.; Gaspar, L.E.; Kavanagh, B.D.; Kleinschmidt-DeMasters, B.K.; Waziri, A.E.; Lillehei, K.O.; Reddy, K.; Chen, C. Phase II Trial of Hypofractionated Intensity-Modulated Radiation Therapy Combined with Temozolomide and Bevacizumab for Patients with Newly Diagnosed Glioblastoma. J. Neurooncol. 2015, 122, 135–143. [Google Scholar] [CrossRef] [PubMed]
  285. Balana, C.; De Las Penas, R.; Sepúlveda, J.M.; Gil-Gil, M.J.; Luque, R.; Gallego, O.; Carrato, C.; Sanz, C.; Reynes, G.; Herrero, A.; et al. Bevacizumab and Temozolomide versus Temozolomide Alone as Neoadjuvant Treatment in Unresected Glioblastoma: The GENOM 009 Randomized Phase II Trial. J. Neurooncol. 2016, 127, 569–579. [Google Scholar] [CrossRef] [PubMed]
  286. Chauffert, B.; Feuvret, L.; Bonnetain, F.; Taillandier, L.; Frappaz, D.; Taillia, H.; Schott, R.; Honnorat, J.; Fabbro, M.; Tennevet, I.; et al. Randomized Phase II Trial of Irinotecan and Bevacizumab as Neo-Adjuvant and Adjuvant to Temozolomide-Based Chemoradiation Compared with Temozolomide-Chemoradiation for Unresectable Glioblastoma: Final Results of the TEMAVIR Study from ANOCEF. Ann. Oncol. 2014, 25, 1442–1447. [Google Scholar] [CrossRef] [PubMed]
  287. 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]
  288. Weathers, S.-P.; Han, X.; Liu, D.D.; Conrad, C.A.; Gilbert, M.R.; Loghin, M.E.; O’Brien, B.J.; Penas-Prado, M.; Puduvalli, V.K.; Tremont-Lukats, I.; et al. A Randomized Phase II Trial of Standard Dose Bevacizumab versus Low Dose Bevacizumab plus Lomustine (CCNU) in Adults with Recurrent Glioblastoma. J. Neurooncol. 2016, 129, 487–494. [Google Scholar] [CrossRef] [PubMed]
  289. Badruddoja, M.A.; Pazzi, M.; Sanan, A.; Schroeder, K.; Kuzma, K.; Norton, T.; Scully, T.; Mahadevan, D.; Ahmadi, M.M. Phase II Study of Bi-Weekly Temozolomide plus Bevacizumab for Adult Patients with Recurrent Glioblastoma. Cancer Chemother. Pharmacol. 2017, 80, 715–721. [Google Scholar] [CrossRef]
  290. Brandes, A.A.; Finocchiaro, G.; Zagonel, V.; Reni, M.; Caserta, C.; Fabi, A.; Clavarezza, M.; Maiello, E.; Eoli, M.; Lombardi, G.; et al. AVAREG: A Phase II, Randomized, Noncomparative Study of Fotemustine or Bevacizumab for Patients with Recurrent Glioblastoma. Neuro-Oncology 2016, 18, 1304–1312. [Google Scholar] [CrossRef]
  291. Herrlinger, U.; Schäfer, N.; Steinbach, J.P.; Weyerbrock, A.; Hau, P.; Goldbrunner, R.; Friedrich, F.; Rohde, V.; Ringel, F.; Schlegel, U.; et al. Bevacizumab Plus Irinotecan Versus Temozolomide in Newly Diagnosed O6-Methylguanine-DNA Methyltransferase Nonmethylated Glioblastoma: The Randomized GLARIUS Trial. J. Clin. Oncol. 2016, 34, 1611–1619. [Google Scholar] [CrossRef] [PubMed]
  292. 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]
  293. Cloughesy, T.F.; Brenner, A.; de Groot, J.F.; Butowski, N.A.; Zach, L.; Campian, J.L.; Ellingson, B.M.; Freedman, L.S.; Cohen, Y.C.; Lowenton-Spier, N.; et al. A Randomized Controlled Phase III Study of VB-111 Combined with Bevacizumab vs Bevacizumab Monotherapy in Patients with Recurrent Glioblastoma (GLOBE). Neuro-Oncology 2020, 22, 705–717. [Google Scholar] [CrossRef]
  294. Desjardins, A.; Reardon, D.A.; Coan, A.; Marcello, J.; Herndon, J.E.; Bailey, L.; Peters, K.B.; Friedman, H.S.; Vredenburgh, J.J. Bevacizumab and Daily Temozolomide for Recurrent Glioblastoma. Cancer 2012, 118, 1302–1312. [Google Scholar] [CrossRef]
  295. Vredenburgh, J.J.; Desjardins, A.; Reardon, D.A.; Peters, K.B.; Herndon, J.E.; Marcello, J.; Kirkpatrick, J.P.; Sampson, J.H.; Bailey, L.; Threatt, S.; et al. The Addition of Bevacizumab to Standard Radiation Therapy and Temozolomide Followed by Bevacizumab, Temozolomide and Irinotecan for Newly Diagnosed Glioblastoma. Clin. Cancer Res. 2011, 17, 4119–4124. [Google Scholar] [CrossRef] [Green Version]
  296. Reardon, D.A.; Desjardins, A.; Vredenburgh, J.J.; Gururangan, S.; Sampson, J.H.; Sathornsumetee, S.; McLendon, R.E.; Herndon, J.E.; Marcello, J.E.; Norfleet, J.; et al. Metronomic Chemotherapy with Daily, Oral Etoposide plus Bevacizumab for Recurrent Malignant Glioma: A Phase II Study. Br. J. Cancer 2009, 101, 1986–1994. [Google Scholar] [CrossRef]
  297. Gilbert, M.R.; Dignam, J.J.; Armstrong, T.S.; Wefel, J.S.; Blumenthal, D.T.; Vogelbaum, M.A.; Colman, H.; Chakravarti, A.; Pugh, S.; Won, M.; et al. A Randomized Trial of Bevacizumab for Newly Diagnosed Glioblastoma. N. Engl. J. Med. 2014, 370, 699–708. [Google Scholar] [CrossRef] [Green Version]
  298. Iwamoto, F.M.; Lamborn, K.R.; Robins, H.I.; Mehta, M.P.; Chang, S.M.; Butowski, N.A.; Deangelis, L.M.; Abrey, L.E.; Zhang, W.-T.; Prados, M.D.; et al. Phase II Trial of Pazopanib (GW786034), an Oral Multi-Targeted Angiogenesis Inhibitor, for Adults with Recurrent Glioblastoma (North American Brain Tumor Consortium Study 06-02). Neuro-Oncology 2010, 12, 855–861. [Google Scholar] [CrossRef] [Green Version]
  299. Brown, N.; McBain, C.; Nash, S.; Hopkins, K.; Sanghera, P.; Saran, F.; Phillips, M.; Dungey, F.; Clifton-Hadley, L.; Wanek, K.; et al. Multi-Center Randomized Phase II Study Comparing Cediranib plus Gefitinib with Cediranib plus Placebo in Subjects with Recurrent/Progressive Glioblastoma. PLoS ONE 2016, 11, e0156369. [Google Scholar] [CrossRef]
  300. Batchelor, T.T.; Mulholland, P.; Neyns, B.; Nabors, L.B.; Campone, M.; Wick, A.; Mason, W.; Mikkelsen, T.; Phuphanich, S.; Ashby, L.S.; et al. Phase III Randomized Trial Comparing the Efficacy of Cediranib as Monotherapy, and in Combination with Lomustine, versus Lomustine Alone in Patients with Recurrent Glioblastoma. J. Clin. Oncol. 2013, 31, 3212–3218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  301. Batchelor, T.T.; Duda, D.G.; di Tomaso, E.; Ancukiewicz, M.; Plotkin, S.R.; Gerstner, E.; Eichler, A.F.; Drappatz, J.; Hochberg, F.H.; Benner, T.; et al. Phase II Study of Cediranib, an Oral Pan-Vascular Endothelial Growth Factor Receptor Tyrosine Kinase Inhibitor, in Patients with Recurrent Glioblastoma. J. Clin. Oncol. 2010, 28, 2817–2823. [Google Scholar] [CrossRef] [PubMed]
  302. Muhic, A.; Poulsen, H.S.; Sorensen, M.; Grunnet, K.; Lassen, U. Phase II Open-Label Study of Nintedanib in Patients with Recurrent Glioblastoma Multiforme. J. Neurooncol. 2013, 111, 205–212. [Google Scholar] [CrossRef]
  303. Norden, A.D.; Schiff, D.; Ahluwalia, M.S.; Lesser, G.J.; Nayak, L.; Lee, E.Q.; Rinne, M.L.; Muzikansky, A.; Dietrich, J.; Purow, B.; et al. Phase II Trial of Triple Tyrosine Kinase Receptor Inhibitor Nintedanib in Recurrent High-Grade Gliomas. J. Neurooncol. 2015, 121, 297–302. [Google Scholar] [CrossRef] [PubMed]
  304. Gerstner, E.R.; Eichler, A.F.; Plotkin, S.R.; Drappatz, J.; Doyle, C.L.; Xu, L.; Duda, D.G.; Wen, P.Y.; Jain, R.K.; Batchelor, T.T. Phase I Trial with Biomarker Studies of Vatalanib (PTK787) in Patients with Newly Diagnosed Glioblastoma Treated with Enzyme Inducing Anti-Epileptic Drugs and Standard Radiation and Temozolomide. J. Neurooncol. 2011, 103, 325–332. [Google Scholar] [CrossRef] [Green Version]
  305. Kalpathy-Cramer, J.; Chandra, V.; Da, X.; Ou, Y.; Emblem, K.E.; Muzikansky, A.; Cai, X.; Douw, L.; Evans, J.G.; Dietrich, J.; et al. Phase II Study of Tivozanib, an Oral VEGFR Inhibitor, in Patients with Recurrent Glioblastoma. J. Neurooncol. 2017, 131, 603–610. [Google Scholar] [CrossRef]
  306. Reardon, D.A.; Desjardins, A.; Peters, K.B.; Gururangan, S.; Sampson, J.H.; McLendon, R.E.; Herndon, J.E.; Bulusu, A.; Threatt, S.; Friedman, A.H.; et al. Phase II Study of Carboplatin, Irinotecan, and Bevacizumab for Bevacizumab Naïve, Recurrent Glioblastoma. J. Neurooncol. 2012, 107, 155–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  307. Dirven, L.; van den Bent, M.J.; Bottomley, A.; van der Meer, N.; van der Holt, B.; Vos, M.J.; Walenkamp, A.M.E.; Beerepoot, L.V.; Hanse, M.C.J.; Reijneveld, J.C.; et al. The Impact of Bevacizumab on Health-Related Quality of Life in Patients Treated for Recurrent Glioblastoma: Results of the Randomised Controlled Phase 2 BELOB Trial. Eur. J. Cancer 2015, 51, 1321–1330. [Google Scholar] [CrossRef]
  308. Taal, W.; Oosterkamp, H.M.; Walenkamp, A.M.E.; Dubbink, H.J.; Beerepoot, L.V.; Hanse, M.C.J.; Buter, J.; Honkoop, A.H.; Boerman, D.; de Vos, F.Y.F.; et al. Single-Agent Bevacizumab or Lomustine versus a Combination of Bevacizumab plus Lomustine in Patients with Recurrent Glioblastoma (BELOB Trial): A Randomised Controlled Phase 2 Trial. Lancet Oncol. 2014, 15, 943–953. [Google Scholar] [CrossRef]
  309. Schnell, O.; Thorsteinsdottir, J.; Fleischmann, D.F.; Lenski, M.; Abenhardt, W.; Giese, A.; Tonn, J.-C.; Belka, C.; Kreth, F.W.; Niyazi, M. Re-Irradiation Strategies in Combination with Bevacizumab for Recurrent Malignant Glioma. J. Neurooncol. 2016, 130, 591–599. [Google Scholar] [CrossRef] [PubMed]
  310. Niyazi, M.; Harter, P.N.; Hattingen, E.; Rottler, M.; von Baumgarten, L.; Proescholdt, M.; Belka, C.; Lauber, K.; Mittelbronn, M. Bevacizumab and Radiotherapy for the Treatment of Glioblastoma: Brothers in Arms or Unholy Alliance? Oncotarget 2016, 7, 2313–2328. [Google Scholar] [CrossRef] [PubMed]
  311. Chinot, O.L.; de La Motte Rouge, T.; Moore, N.; Zeaiter, A.; Das, A.; Phillips, H.; Modrusan, Z.; Cloughesy, T. AVAglio: Phase 3 Trial of Bevacizumab plus Temozolomide and Radiotherapy in Newly Diagnosed Glioblastoma Multiforme. Adv. Ther. 2011, 28, 334–340. [Google Scholar] [CrossRef] [Green Version]
  312. van Linde, M.E.; Verhoeff, J.J.C.; Richel, D.J.; van Furth, W.R.; Reijneveld, J.C.; Verheul, H.M.W.; Stalpers, L.J.A. Bevacizumab in Combination with Radiotherapy and Temozolomide for Patients with Newly Diagnosed Glioblastoma Multiforme. Oncologist 2015, 20, 107–108. [Google Scholar] [CrossRef] [Green Version]
  313. Sandmann, T.; Bourgon, R.; Garcia, J.; Li, C.; Cloughesy, T.; Chinot, O.L.; Wick, W.; Nishikawa, R.; Mason, W.; Henriksson, R.; et al. Patients with Proneural Glioblastoma May Derive Overall Survival Benefit From the Addition of Bevacizumab to First-Line Radiotherapy and Temozolomide: Retrospective Analysis of the AVAglio Trial. J. Clin. Oncol. 2015, 33, 2735–2744. [Google Scholar] [CrossRef]
  314. Brave, S.R.; Ratcliffe, K.; Wilson, Z.; James, N.H.; Ashton, S.; Wainwright, A.; Kendrew, J.; Dudley, P.; Broadbent, N.; Sproat, G.; et al. Assessing the Activity of Cediranib, a VEGFR-2/3 Tyrosine Kinase Inhibitor, against VEGFR-1 and Members of the Structurally Related PDGFR Family. Mol. Cancer Ther. 2011, 10, 861–873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  315. Schäfer, N.; Gielen, G.H.; Kebir, S.; Wieland, A.; Till, A.; Mack, F.; Schaub, C.; Tzaridis, T.; Reinartz, R.; Niessen, M.; et al. Phase I Trial of Dovitinib (TKI258) in Recurrent Glioblastoma. J. Cancer Res. Clin. Oncol. 2016, 142, 1581–1589. [Google Scholar] [CrossRef] [PubMed]
  316. Thanasupawat, T.; Natarajan, S.; Rommel, A.; Glogowska, A.; Bergen, H.; Krcek, J.; Pitz, M.; Beiko, J.; Krawitz, S.; Verma, I.M.; et al. Dovitinib Enhances Temozolomide Efficacy in Glioblastoma Cells. Mol. Oncol. 2017, 11, 1078–1098. [Google Scholar] [CrossRef] [Green Version]
  317. Reardon, D.A.; Egorin, M.J.; Desjardins, A.; Vredenburgh, J.J.; Beumer, J.H.; Lagattuta, T.F.; Gururangan, S.; Herndon, J.E.; Salvado, A.J.; Friedman, H.S. Phase I Pharmacokinetic Study of the Vascular Endothelial Growth Factor Receptor Tyrosine Kinase Inhibitor Vatalanib (PTK787) plus Imatinib and Hydroxyurea for Malignant Glioma. Cancer 2009, 115, 2188–2198. [Google Scholar] [CrossRef] [PubMed]
  318. Cloughesy, T.; Finocchiaro, G.; Belda-Iniesta, C.; Recht, L.; Brandes, A.A.; Pineda, E.; Mikkelsen, T.; Chinot, O.L.; Balana, C.; Macdonald, D.R.; et al. Randomized, Double-Blind, Placebo-Controlled, Multicenter Phase II Study of Onartuzumab Plus Bevacizumab Versus Placebo Plus Bevacizumab in Patients with Recurrent Glioblastoma: Efficacy, Safety, and Hepatocyte Growth Factor and O6-Methylguanine-DNA Methyltransferase Biomarker Analyses. J. Clin. Oncol. 2017, 35, 343–351. [Google Scholar] [CrossRef] [Green Version]
  319. Wen, P.Y.; Drappatz, J.; de Groot, J.; Prados, M.D.; Reardon, D.A.; Schiff, D.; Chamberlain, M.; Mikkelsen, T.; Desjardins, A.; Holland, J.; et al. Phase II Study of Cabozantinib in Patients with Progressive Glioblastoma: Subset Analysis of Patients Naive to Antiangiogenic Therapy. Neuro-Oncology 2018, 20, 249–258. [Google Scholar] [CrossRef]
  320. Wen, P.Y.; Schiff, D.; Cloughesy, T.F.; Raizer, J.J.; Laterra, J.; Smitt, M.; Wolf, M.; Oliner, K.S.; Anderson, A.; Zhu, M.; et al. A Phase II Study Evaluating the Efficacy and Safety of AMG 102 (Rilotumumab) in Patients with Recurrent Glioblastoma. Neuro-Oncology 2011, 13, 437–446. [Google Scholar] [CrossRef] [Green Version]
  321. de Groot, J.F.; Lamborn, K.R.; Chang, S.M.; Gilbert, M.R.; Cloughesy, T.F.; Aldape, K.; Yao, J.; Jackson, E.F.; Lieberman, F.; Robins, H.I.; et al. Phase II Study of Aflibercept in Recurrent Malignant Glioma: A North American Brain Tumor Consortium Study. J. Clin. Oncol. 2011, 29, 2689–2695. [Google Scholar] [CrossRef]
  322. Blumenschein, G.R.; Mills, G.B.; Gonzalez-Angulo, A.M. Targeting the Hepatocyte Growth Factor-CMET Axis in Cancer Therapy. J. Clin. Oncol. 2012, 30, 3287–3296. [Google Scholar] [CrossRef] [Green Version]
  323. Garnett, J.; Chumbalkar, V.; Vaillant, B.; Gururaj, A.E.; Hill, K.S.; Latha, K.; Yao, J.; Priebe, W.; Colman, H.; Elferink, L.A.; et al. Regulation of HGF Expression by ΔEGFR-Mediated c-Met Activation in Glioblastoma Cells. Neoplasia 2013, 15, 73–84. [Google Scholar] [CrossRef] [Green Version]
  324. Huang, M.; Liu, T.; Ma, P.; Mitteer, R.A.; Zhang, Z.; Kim, H.J.; Yeo, E.; Zhang, D.; Cai, P.; Li, C.; et al. C-Met-Mediated Endothelial Plasticity Drives Aberrant Vascularization and Chemoresistance in Glioblastoma. J. Clin. Investig. 2016, 126, 1801–1814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  325. Jahangiri, A.; De Lay, M.; Miller, L.M.; Carbonell, W.S.; Hu, Y.-L.; Lu, K.; Tom, M.W.; Paquette, J.; Tokuyasu, T.A.; Tsao, S.; et al. Gene Expression Profile Identifies Tyrosine Kinase C-Met as a Targetable Mediator of Antiangiogenic Therapy Resistance. Clin. Cancer Res. 2013, 19, 1773–1783. [Google Scholar] [CrossRef] [Green Version]
  326. Lu, K.V.; Bergers, G. Mechanisms of Evasive Resistance to Anti-VEGF Therapy in Glioblastoma. CNS Oncol. 2013, 2, 49–65. [Google Scholar] [CrossRef]
  327. Das, A.; Cheng, R.R.; Hilbert, M.L.T.; Dixon-Moh, Y.N.; Decandio, M.; Vandergrift, W.A.; Banik, N.L.; Lindhorst, S.M.; Cachia, D.; Varma, A.K.; et al. Synergistic Effects of Crizotinib and Temozolomide in Experimental FIG-ROS1 Fusion-Positive Glioblastoma. Cancer Growth Metastasis 2015, 8, 51–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  328. Schiff, D.; Desjardins, A.; Cloughesy, T.; Mikkelsen, T.; Glantz, M.; Chamberlain, M.C.; Reardon, D.A.; Wen, P.Y. Phase 1 Dose Escalation Trial of the Safety and Pharmacokinetics of Cabozantinib Concurrent with Temozolomide and Radiotherapy or Temozolomide after Radiotherapy in Newly Diagnosed Patients with High-Grade Gliomas. Cancer 2016, 122, 582–587. [Google Scholar] [CrossRef]
  329. Schneider, K.; Weyerbrock, A.; Doostkam, S.; Plate, K.; Machein, M.R. Lack of Evidence for PlGF Mediating the Tumor Resistance after Anti-Angiogenic Therapy in Malignant Gliomas. J. Neurooncol. 2015, 121, 269–278. [Google Scholar] [CrossRef] [PubMed]
  330. Lassen, U.; Chinot, O.L.; McBain, C.; Mau-Sørensen, M.; Larsen, V.A.; Barrie, M.; Roth, P.; Krieter, O.; Wang, K.; Habben, K.; et al. Phase 1 Dose-Escalation Study of the Antiplacental Growth Factor Monoclonal Antibody RO5323441 Combined with Bevacizumab in Patients with Recurrent Glioblastoma. Neuro-Oncology 2015, 17, 1007–1015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  331. Tabouret, E.; Denicolai, E.; Delfino, C.; Graillon, T.; Boucard, C.; Nanni, I.; Padovani, L.; Figarella-Branger, D.; Chinot, O. Changes in PlGF and MET-HGF Expressions in Paired Initial and Recurrent Glioblastoma. J. Neurooncol. 2016, 130, 431–437. [Google Scholar] [CrossRef]
  332. Rosen, L.S.; Gordon, M.S.; Robert, F.; Matei, D.E. Endoglin for Targeted Cancer Treatment. Curr. Oncol. Rep. 2014, 16, 365. [Google Scholar] [CrossRef] [PubMed]
  333. Ahluwalia, M.S.; Rogers, L.R.; Chaudhary, R.T.; Newton, H.B.; Seon, B.K.; Jivani, M.A.; Adams, B.J.; Shazer, R.L.; Theuer, C.P. A Phase 2 Trial of TRC105 with Bevacizumab for Bevacizumab Refractory Glioblastoma. J. Clin. Oncol. 2016, 34, 2035. [Google Scholar] [CrossRef]
  334. Galanis, E.; Anderson, S.K.; Butowski, N.A.; Hormigo, A.; Schiff, D.; Tran, D.D.; Omuro, A.M.P.; Jaeckle, K.A.; Kumar, S.; Kaufmann, T.J.; et al. NCCTG N1174: Phase I/Comparative Randomized Phase (Ph) II Trial of TRC105 plus Bevacizumab versus Bevacizumab in Recurrent Glioblastoma (GBM) (Alliance). J. Clin. Oncol. 2017, 35, 2023. [Google Scholar] [CrossRef]
  335. Afshar Moghaddam, N.; Mahsuni, P.; Taheri, D. Evaluation of Endoglin as an Angiogenesis Marker in Glioblastoma. Iran. J. Pathol. 2015, 10, 89–96. [Google Scholar]
  336. Puduvalli, V.K.; Giglio, P.; Groves, M.D.; Hess, K.R.; Gilbert, M.R.; Mahankali, S.; Jackson, E.F.; Levin, V.A.; Conrad, C.A.; Hsu, S.H.; et al. Phase II Trial of Irinotecan and Thalidomide in Adults with Recurrent Glioblastoma Multiforme. Neuro-Oncology 2008, 10, 216–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  337. Fadul, C.E.; Kingman, L.S.; Meyer, L.P.; Cole, B.F.; Eskey, C.J.; Rhodes, C.H.; Roberts, D.W.; Newton, H.B.; Pipas, J.M. A Phase II Study of Thalidomide and Irinotecan for Treatment of Glioblastoma Multiforme. J. Neurooncol. 2008, 90, 229–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  338. Kesari, S.; Schiff, D.; Henson, J.W.; Muzikansky, A.; Gigas, D.C.; Doherty, L.; Batchelor, T.T.; Longtine, J.A.; Ligon, K.L.; Weaver, S.; et al. Phase II Study of Temozolomide, Thalidomide, and Celecoxib for Newly Diagnosed Glioblastoma in Adults. Neuro-Oncology 2008, 10, 300–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  339. Groves, M.D.; Puduvalli, V.K.; Chang, S.M.; Conrad, C.A.; Gilbert, M.R.; Tremont-Lukats, I.W.; Liu, T.-J.; Peterson, P.; Schiff, D.; Cloughesy, T.F.; et al. A North American Brain Tumor Consortium (NABTC 99-04) Phase II Trial of Temozolomide plus Thalidomide for Recurrent Glioblastoma Multiforme. J. Neurooncol. 2007, 81, 271–277. [Google Scholar] [CrossRef]
  340. Stupp, R.; Hegi, M.E.; Gorlia, T.; Erridge, S.C.; Perry, J.; Hong, Y.-K.; Aldape, K.D.; Lhermitte, B.; Pietsch, T.; Grujicic, D.; et al. Cilengitide Combined with Standard Treatment for Patients with Newly Diagnosed Glioblastoma with Methylated MGMT Promoter (CENTRIC EORTC 26071-22072 Study): A Multicentre, Randomised, Open-Label, Phase 3 Trial. Lancet Oncol. 2014, 15, 1100–1108. [Google Scholar] [CrossRef] [Green Version]
  341. Nabors, L.B.; Fink, K.L.; Mikkelsen, T.; Grujicic, D.; Tarnawski, R.; Nam, D.H.; Mazurkiewicz, M.; Salacz, M.; Ashby, L.; Zagonel, V.; et al. Two Cilengitide Regimens in Combination with Standard Treatment for Patients with Newly Diagnosed Glioblastoma and Unmethylated MGMT Gene Promoter: Results of the Open-Label, Controlled, Randomized Phase II CORE Study. Neuro-Oncology 2015, 17, 708–717. [Google Scholar] [CrossRef] [PubMed]
  342. Nabors, L.B.; Mikkelsen, T.; Hegi, M.E.; Ye, X.; Batchelor, T.; Lesser, G.; Peereboom, D.; Rosenfeld, M.R.; Olsen, J.; Brem, S.; et al. A Safety Run-in and Randomized Phase 2 Study of Cilengitide Combined with Chemoradiation for Newly Diagnosed Glioblastoma (NABTT 0306). Cancer 2012, 118, 5601–5607. [Google Scholar] [CrossRef]
  343. Reardon, D.A.; Fink, K.L.; Mikkelsen, T.; Cloughesy, T.F.; O’Neill, A.; Plotkin, S.; Glantz, M.; Ravin, P.; Raizer, J.J.; Rich, K.M.; et al. Randomized Phase II Study of Cilengitide, an Integrin-Targeting Arginine-Glycine-Aspartic Acid Peptide, in Recurrent Glioblastoma Multiforme. J. Clin. Oncol. 2008, 26, 5610–5617. [Google Scholar] [CrossRef]
  344. Elinzano, H.; Hebda, N.; Luppe, D.; Turchetti, W.; Rosati, K.; Sikov, W.M.; Safran, H. PSMA ADC for Progressive Glioblastoma: Phase II Brown University Oncology Research Group Study. J. Clin. Oncol. 2016, 34, 2065. [Google Scholar] [CrossRef]
  345. Riva, M.; Imbesi, F.; Beghi, E.; Galli, C.; Citterio, A.; Trapani, P.; Sterzi, R.; Collice, M. Temozolomide and Thalidomide in the Treatment of Glioblastoma Multiforme. Anticancer Res. 2007, 27, 1067–1071. [Google Scholar]
  346. Alexander, B.M.; Wang, M.; Yung, W.K.A.; Fine, H.A.; Donahue, B.A.; Tremont, I.W.; Richards, R.S.; Kerlin, K.J.; Hartford, A.C.; Curran, W.J.; et al. A Phase II Study of Conventional Radiation Therapy and Thalidomide for Supratentorial, Newly-Diagnosed Glioblastoma (RTOG 9806). J. Neurooncol. 2013, 111, 33–39. [Google Scholar] [CrossRef] [PubMed]
  347. Stupp, R.; Hegi, M.E.; Neyns, B.; Goldbrunner, R.; Schlegel, U.; Clement, P.M.J.; Grabenbauer, G.G.; Ochsenbein, A.F.; Simon, M.; Dietrich, P.-Y.; et al. Phase I/IIa Study of Cilengitide and Temozolomide with Concomitant Radiotherapy Followed by Cilengitide and Temozolomide Maintenance Therapy in Patients with Newly Diagnosed Glioblastoma. J. Clin. Oncol. 2010, 28, 2712–2718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  348. Cianfrocca, M.E.; Kimmel, K.A.; Gallo, J.; Cardoso, T.; Brown, M.M.; Hudes, G.; Lewis, N.; Weiner, L.; Lam, G.N.; Brown, S.C.; et al. Phase 1 Trial of the Antiangiogenic Peptide ATN-161 (Ac-PHSCN-NH(2)), a Beta Integrin Antagonist, in Patients with Solid Tumours. Br. J. Cancer 2006, 94, 1621–1626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  349. Neal, J.; Wakelee, H. AMG-386, a Selective Angiopoietin-1/-2-Neutralizing Peptibody for the Potential Treatment of Cancer. Curr. Opin. Mol. Ther. 2010, 12, 487–495. [Google Scholar] [PubMed]
  350. Walia, A.; Yang, J.F.; Huang, Y.-H.; Rosenblatt, M.I.; Chang, J.-H.; Azar, D.T. Endostatin’s Emerging Roles in Angiogenesis, Lymphangiogenesis, Disease, and Clinical Applications. Biochim. Biophys. Acta 2015, 1850, 2422–2438. [Google Scholar] [CrossRef] [Green Version]
  351. Chen, J.; Yao, Q.; Li, D.; Zhang, J.; Wang, T.; Yu, M.; Zhou, X.; Huan, Y.; Wang, J.; Wang, L. Neoadjuvant Rh-Endostatin, Docetaxel and Epirubicin for Breast Cancer: Efficacy and Safety in a Prospective, Randomized, Phase II Study. BMC Cancer 2013, 13, 248. [Google Scholar] [CrossRef] [Green Version]
  352. Cui, C.; Mao, L.; Chi, Z.; Si, L.; Sheng, X.; Kong, Y.; Li, S.; Lian, B.; Gu, K.; Tao, M.; et al. A Phase II, Randomized, Double-Blind, Placebo-Controlled Multicenter Trial of Endostar in Patients with Metastatic Melanoma. Mol. Ther. 2013, 21, 1456–1463. [Google Scholar] [CrossRef] [Green Version]
  353. Zhao, X.; Su, Y.; You, J.; Gong, L.; Zhang, Z.; Wang, M.; Zhao, Z.; Zhang, Z.; Li, X.; Wang, C. Combining Antiangiogenic Therapy with Neoadjuvant Chemotherapy Increases Treatment Efficacy in Stage IIIA (N2) Non-Small Cell Lung Cancer without Increasing Adverse Effects. Oncotarget 2016, 7, 62619–62626. [Google Scholar] [CrossRef] [Green Version]
  354. Saffar, H.; Noohi, M.; Tavangar, S.M.; Saffar, H.; Azimi, S. Expression of Prostate-Specific Membrane Antigen (PSMA) in Brain Glioma and Its Correlation with Tumor Grade. Iran. J. Pathol. 2018, 13, 45–53. [Google Scholar] [PubMed] [Green Version]
  355. Anilkumar, G.; Barwe, S.P.; Christiansen, J.J.; Rajasekaran, S.A.; Kohn, D.B.; Rajasekaran, A.K. Association of Prostate-Specific Membrane Antigen with Caveolin-1 and Its Caveolae-Dependent Internalization in Microvascular Endothelial Cells: Implications for Targeting to Tumor Vasculature. Microvasc. Res. 2006, 72, 54–61. [Google Scholar] [CrossRef] [PubMed]
  356. Hatoum, A.; Mohammed, R.; Zakieh, O. The Unique Invasiveness of Glioblastoma and Possible Drug Targets on Extracellular Matrix. Available online: https://www.dovepress.com/the-unique-invasiveness-of-glioblastoma-and-possible-drug-targets-on-e-peer-reviewed-fulltext-article-CMAR (accessed on 19 June 2020).
  357. Rempe, R.G.; Hartz, A.M.; Bauer, B. Matrix Metalloproteinases in the Brain and Blood–Brain Barrier: Versatile Breakers and Makers. J. Cereb. Blood Flow Metab. 2016, 36, 1481–1507. [Google Scholar] [CrossRef] [Green Version]
  358. Mandel, J.J.; Yust-Katz, S.; Patel, A.J.; Cachia, D.; Liu, D.; Park, M.; Yuan, Y.; Kent, T.A.; de Groot, J.F. Inability of Positive Phase II Clinical Trials of Investigational Treatments to Subsequently Predict Positive Phase III Clinical Trials in Glioblastoma. Neuro-Oncology 2018, 20, 113–122. [Google Scholar] [CrossRef] [Green Version]
  359. Ventz, S.; Lai, A.; Cloughesy, T.F.; Wen, P.Y.; Trippa, L.; Alexander, B.M. Design and Evaluation of an External Control Arm Using Prior Clinical Trials and Real-World Data. Clin. Cancer Res. 2019, 25, 4993–5001. [Google Scholar] [CrossRef]
  360. Lee, E.Q.; Chukwueke, U.N.; Hervey-Jumper, S.L.; de Groot, J.F.; Leone, J.P.; Armstrong, T.S.; Chang, S.M.; Arons, D.; Oliver, K.; Verble, K.; et al. Barriers to Accrual and Enrollment in Brain Tumor Trials. Neuro-Oncology 2019, 21, 1100–1117. [Google Scholar] [CrossRef]
  361. Parker, J.L.; Kuzulugil, S.S.; Pereverzev, K.; Mac, S.; Lopes, G.; Shah, Z.; Weerasinghe, A.; Rubinger, D.; Falconi, A.; Bener, A.; et al. Does Biomarker Use in Oncology Improve Clinical Trial Failure Risk? A Large-Scale Analysis. Cancer Med. 2021, 10, 1955–1963. [Google Scholar] [CrossRef]
  362. Weller, M.; van den Bent, M.; Preusser, M.; Le Rhun, 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. 2021, 18, 170–186. [Google Scholar] [CrossRef]
  363. Sestito, S.; Runfola, M.; Tonelli, M.; Chiellini, G.; Rapposelli, S. New Multitarget Approaches in the War Against Glioblastoma: A Mini-Perspective. Front. Pharmacol. 2018, 9, 874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  364. Wolbers, J.G. Novel Strategies in Glioblastoma Surgery Aim at Safe, Supra-Maximum Resection in Conjunction with Local Therapies. Chin. J. Cancer 2014, 33, 8–15. [Google Scholar] [CrossRef]
  365. Sanai, N.; Polley, M.-Y.; McDermott, M.W.; Parsa, A.T.; Berger, M.S. An Extent of Resection Threshold for Newly Diagnosed Glioblastomas: Clinical Article. J. Neurosurg. 2011, 115, 3–8. [Google Scholar] [CrossRef] [Green Version]
  366. Trifiletti, D.M.; Alonso, C.; Grover, S.; Fadul, C.E.; Sheehan, J.P.; Showalter, T.N. Prognostic Implications of Extent of Resection in Glioblastoma: Analysis from a Large Database. World Neurosurg. 2017, 103, 330–340. [Google Scholar] [CrossRef] [PubMed]
  367. Jena, L.; McErlean, E.; McCarthy, H. Delivery across the Blood-Brain Barrier: Nanomedicine for Glioblastoma Multiforme. Drug Deliv. Transl. Res. 2020, 10, 304–318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  368. Shi, M.; Sanche, L. Convection-Enhanced Delivery in Malignant Gliomas: A Review of Toxicity and Efficacy. J. Oncol. 2019, 2019, 9342796. [Google Scholar] [CrossRef]
  369. van den Bent, M.J.; Gao, Y.; Kerkhof, M.; Kros, J.M.; Gorlia, T.; van Zwieten, K.; Prince, J.; van Duinen, S.; Sillevis Smitt, P.A.; Taphoorn, M.; et al. Changes in the EGFR Amplification and EGFRvIII Expression between Paired Primary and Recurrent Glioblastomas. Neuro-Oncology 2015, 17, 935–941. [Google Scholar] [CrossRef] [Green Version]
  370. Wang, J.; Cazzato, E.; Ladewig, E.; Frattini, V.; Rosenbloom, D.I.S.; Zairis, S.; Abate, F.; Liu, Z.; Elliott, O.; Shin, Y.-J.; et al. Clonal Evolution of Glioblastoma under Therapy. Nat. Genet. 2016, 48, 768–776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  371. Nathanson, D.A.; Gini, B.; Mottahedeh, J.; Visnyei, K.; Koga, T.; Gomez, G.; Eskin, A.; Hwang, K.; Wang, J.; Masui, K.; et al. Targeted Therapy Resistance Mediated by Dynamic Regulation of Extrachromosomal Mutant EGFR DNA. Science 2014, 343, 72–76. [Google Scholar] [CrossRef] [Green Version]
  372. Huang, L.; Fu, L. Mechanisms of Resistance to EGFR Tyrosine Kinase Inhibitors. Acta Pharm. Sin. B 2015, 5, 390–401. [Google Scholar] [CrossRef] [Green Version]
  373. An, Z.; Aksoy, O.; Zheng, T.; Fan, Q.-W.; Weiss, W.A. Epidermal Growth Factor Receptor and EGFRvIII in Glioblastoma: Signaling Pathways and Targeted Therapies. Oncogene 2018, 37, 1561–1575. [Google Scholar] [CrossRef]
  374. Goodwin, C.R.; Rath, P.; Oyinlade, O.; Lopez, H.; Mughal, S.; Xia, S.; Li, Y.; Kaur, H.; Zhou, X.; Ahmed, A.K.; et al. Crizotinib and Erlotinib Inhibits Growth of C-Met+/EGFRvIII+ Primary Human Glioblastoma Xenografts. Clin. Neurol. Neurosurg. 2018, 171, 26–33. [Google Scholar] [CrossRef]
  375. Akhavan, D.; Pourzia, A.L.; Nourian, A.A.; Williams, K.J.; Nathanson, D.; Babic, I.; Villa, G.R.; Tanaka, K.; Nael, A.; Yang, H.; et al. De-Repression of PDGFRβ Transcription Promotes Acquired Resistance to EGFR Tyrosine Kinase Inhibitors in Glioblastoma Patients. Cancer Discov. 2013, 3, 534–547. [Google Scholar] [CrossRef] [Green Version]
  376. Mellinghoff, I.K.; Wang, M.Y.; Vivanco, I.; Haas-Kogan, D.A.; Zhu, S.; Dia, E.Q.; Lu, K.V.; Yoshimoto, K.; Huang, J.H.Y.; Chute, D.J.; et al. Molecular Determinants of the Response of Glioblastomas to EGFR Kinase Inhibitors. N. Engl. J. Med. 2005, 353, 2012–2024. [Google Scholar] [CrossRef] [Green Version]
  377. Bhat, K.P.L.; Balasubramaniyan, V.; Vaillant, B.; Ezhilarasan, R.; Hummelink, K.; Hollingsworth, F.; Wani, K.; Heathcock, L.; James, J.D.; Goodman, L.D.; et al. Mesenchymal Differentiation Mediated by NF-ΚB Promotes Radiation Resistance in Glioblastoma. Cancer Cell 2013, 24, 331–346. [Google Scholar] [CrossRef] [Green Version]
  378. Halliday, J.; Helmy, K.; Pattwell, S.S.; Pitter, K.L.; LaPlant, Q.; Ozawa, T.; Holland, E.C. In Vivo Radiation Response of Proneural Glioma Characterized by Protective P53 Transcriptional Program and Proneural-Mesenchymal Shift. Proc. Natl. Acad. Sci. USA 2014, 111, 5248–5253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  379. Ghosh, D.; Nandi, S.; Bhattacharjee, S. Combination Therapy to Checkmate Glioblastoma: Clinical Challenges and Advances. Clin. Transl. Med. 2018, 7, 33. [Google Scholar] [CrossRef] [Green Version]
  380. Eskilsson, E.; Røsland, G.V.; Solecki, G.; Wang, Q.; Harter, P.N.; Graziani, G.; Verhaak, R.G.W.; Winkler, F.; Bjerkvig, R.; Miletic, H. EGFR Heterogeneity and Implications for Therapeutic Intervention in Glioblastoma. Neuro-Oncology 2018, 20, 743–752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  381. Burkhardt, J.-K.; Riina, H.; Shin, B.J.; Christos, P.; Kesavabhotla, K.; Hofstetter, C.P.; Tsiouris, A.J.; Boockvar, J.A. Intra-Arterial Delivery of Bevacizumab after Blood-Brain Barrier Disruption for the Treatment of Recurrent Glioblastoma: Progression-Free Survival and Overall Survival. World Neurosurg. 2012, 77, 130–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  382. Boockvar, J.A.; Tsiouris, A.J.; Hofstetter, C.P.; Kovanlikaya, I.; Fralin, S.; Kesavabhotla, K.; Seedial, S.M.; Pannullo, S.C.; Schwartz, T.H.; Stieg, P.; et al. Safety and Maximum Tolerated Dose of Superselective Intraarterial Cerebral Infusion of Bevacizumab after Osmotic Blood-Brain Barrier Disruption for Recurrent Malignant Glioma. Clinical Article. J. Neurosurg. 2011, 114, 624–632. [Google Scholar] [CrossRef] [PubMed]
  383. Alter, R.A.; White, T.G.; Fanous, A.A.; Chakraborty, S.; Filippi, C.G.; Pisapia, D.J.; Tsiouris, A.J.; Boockvar, J.A. Long-Term Benefit of Intra-Arterial Bevacizumab for Recurrent Glioblastoma. J. Exp. Ther. Oncol. 2017, 12, 67–71. [Google Scholar]
  384. Chakraborty, S.; Filippi, C.G.; Burkhardt, J.-K.; Fralin, S.; Ray, A.; Wong, T.; Ortiz, R.; Langer, D.J.; Boockvar, J.A. Durability of Single Dose Intra-Arterial Bevacizumab after Blood/Brain Barrier Disruption for Recurrent Glioblastoma. J. Exp. Ther. Oncol. 2016, 11, 261–267. [Google Scholar] [PubMed]
  385. Heiland, D.H.; Masalha, W.; Franco, P.; Machein, M.R.; Weyerbrock, A. Progression-Free and Overall Survival in Patients with Recurrent Glioblastoma Multiforme Treated with Last-Line Bevacizumab versus Bevacizumab/Lomustine. J. Neurooncol. 2016, 126, 567–575. [Google Scholar] [CrossRef]
  386. Chinot, O.L. Cilengitide in Glioblastoma: When Did It Fail? Lancet Oncol. 2014, 15, 1044–1045. [Google Scholar] [CrossRef]
  387. Stupp, R.; Picard, M.; Weller, M. Does Cilengitide Deserve Another Chance?-Authors’ Reply. Lancet Oncol. 2014, 15, e585–e586. [Google Scholar] [CrossRef]
  388. Tucci, M.; Stucci, S.; Silvestris, F. Does Cilengitide Deserve Another Chance? Lancet Oncol. 2014, 15, e584–e585. [Google Scholar] [CrossRef]
  389. Reynolds, A.R.; Hart, I.R.; Watson, A.R.; Welti, J.C.; Silva, R.G.; Robinson, S.D.; Da Violante, G.; Gourlaouen, M.; Salih, M.; Jones, M.C.; et al. Stimulation of Tumor Growth and Angiogenesis by Low Concentrations of RGD-Mimetic Integrin Inhibitors. Nat. Med. 2009, 15, 392–400. [Google Scholar] [CrossRef]
  390. Eisele, G.; Wick, A.; Eisele, A.-C.; Clément, P.M.; Tonn, J.; Tabatabai, G.; Ochsenbein, A.; Schlegel, U.; Neyns, B.; Krex, D.; et al. Cilengitide Treatment of Newly Diagnosed Glioblastoma Patients Does Not Alter Patterns of Progression. J. Neurooncol. 2014, 117, 141–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  391. Weller, M.; Nabors, L.B.; Gorlia, T.; Leske, H.; Rushing, E.; Bady, P.; Hicking, C.; Perry, J.; Hong, Y.-K.; Roth, P.; et al. Cilengitide in Newly Diagnosed Glioblastoma: Biomarker Expression and Outcome. Oncotarget 2016, 7, 15018–15032. [Google Scholar] [CrossRef]
  392. Cosset, É.; Weis, S.M.; Cheresh, D.A. Re-Thinking the Preclinical Development of GBM Therapeutics. Oncoscience 2018, 5, 11–12. [Google Scholar] [CrossRef] [PubMed]
  393. Cosset, É.; Ilmjärv, S.; Dutoit, V.; Elliott, K.; von Schalscha, T.; Camargo, M.F.; Reiss, A.; Moroishi, T.; Seguin, L.; Gomez, G.; et al. Glut3 Addiction Is a Druggable Vulnerability for a Molecularly Defined Subpopulation of Glioblastoma. Cancer Cell 2017, 32, 856–868.e5. [Google Scholar] [CrossRef] [Green Version]
  394. Uneda, A.; Kurozumi, K.; Fujimura, A.; Fujii, K.; Ishida, J.; Shimazu, Y.; Otani, Y.; Tomita, Y.; Hattori, Y.; Matsumoto, Y.; et al. Differentiated Glioblastoma Cells Accelerate Tumor Progression by Shaping the Tumor Microenvironment via CCN1-Mediated Macrophage Infiltration. Acta Neuropathol. Commun. 2021, 9, 29. [Google Scholar] [CrossRef] [PubMed]
  395. Wang, X.; Prager, B.C.; Wu, Q.; Kim, L.J.Y.; Gimple, R.C.; Shi, Y.; Yang, K.; Morton, A.R.; Zhou, W.; Zhu, Z.; et al. Reciprocal Signaling between Glioblastoma Stem Cells and Differentiated Tumor Cells Promotes Malignant Progression. Cell Stem Cell 2018, 22, 514–528.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  396. Petroni, G.; Buqué, A.; Zitvogel, L.; Kroemer, G.; Galluzzi, L. Immunomodulation by Targeted Anticancer Agents. Cancer Cell 2021, 39, 310–345. [Google Scholar] [CrossRef] [PubMed]
  397. Martins, T.A.; Schmassmann, P.; Shekarian, T.; Boulay, J.-L.; Ritz, M.-F.; Zanganeh, S.; Vom Berg, J.; Hutter, G. Microglia-Centered Combinatorial Strategies Against Glioblastoma. Front. Immunol. 2020, 11, 571951. [Google Scholar] [CrossRef]
  398. Fukumura, D.; Kloepper, J.; Amoozgar, Z.; Duda, D.G.; Jain, R.K. Enhancing Cancer Immunotherapy Using Antiangiogenics: Opportunities and Challenges. Nat. Rev. Clin. Oncol. 2018, 15, 325–340. [Google Scholar] [CrossRef] [PubMed]
  399. Song, Y.; Fu, Y.; Xie, Q.; Zhu, B.; Wang, J.; Zhang, B. Anti-Angiogenic Agents in Combination with Immune Checkpoint Inhibitors: A Promising Strategy for Cancer Treatment. Front. Immunol. 2020, 11, 1956. [Google Scholar] [CrossRef]
Figure 1. Timeline showing the principal dates of the histological and molecular classifications of gliomas. Classifying brain tumors has been the subject of many studies for several years. The first classification published in 1926 by Bailey and Cushing was based on histogenetics [51]. According to this classification, the presence of embryonic cells would be at the origin of tumor cells. The second classification proposed in 1949 by Kernohan JW, Mabon [52], includes grades of malignancy. The WHO proposed a new classification of gliomas in 1979 [53], which is internationally recognized and was revised in 1993, 2000, 2007 and 2016 [54,55,56,57]. These classifications are based on anatomopathological analysis of a representative glioma fragment (from biopsy or surgical resection) and “grading” elements. The International Society of Neuropathology was held from 1–3 May 2014 in Haarlem, the Netherlands [58]. The meeting reached consensus regarding the incorporation of non-histological data, such as molecular information, into the next WHO classification [55].
Figure 1. Timeline showing the principal dates of the histological and molecular classifications of gliomas. Classifying brain tumors has been the subject of many studies for several years. The first classification published in 1926 by Bailey and Cushing was based on histogenetics [51]. According to this classification, the presence of embryonic cells would be at the origin of tumor cells. The second classification proposed in 1949 by Kernohan JW, Mabon [52], includes grades of malignancy. The WHO proposed a new classification of gliomas in 1979 [53], which is internationally recognized and was revised in 1993, 2000, 2007 and 2016 [54,55,56,57]. These classifications are based on anatomopathological analysis of a representative glioma fragment (from biopsy or surgical resection) and “grading” elements. The International Society of Neuropathology was held from 1–3 May 2014 in Haarlem, the Netherlands [58]. The meeting reached consensus regarding the incorporation of non-histological data, such as molecular information, into the next WHO classification [55].
Cancers 13 01795 g001
Figure 2. Flowchart.
Figure 2. Flowchart.
Cancers 13 01795 g002
Figure 3. Principal biomarkers and drugs in GBM targeted therapies. The color code corresponds to the four sections of the Results section. The targeting of stem cells and stem cell pathways is represented in green, the targeting of growth autonomy and migration in blue, the targeting of the cell cycle & escape to cell death in black and the targeting of angiogenesis in red. Acronyms are defined in the text.
Figure 3. Principal biomarkers and drugs in GBM targeted therapies. The color code corresponds to the four sections of the Results section. The targeting of stem cells and stem cell pathways is represented in green, the targeting of growth autonomy and migration in blue, the targeting of the cell cycle & escape to cell death in black and the targeting of angiogenesis in red. Acronyms are defined in the text.
Cancers 13 01795 g003
Table 1. Clinical studies analyzing therapies targeting the self-renewal of GICs.
Table 1. Clinical studies analyzing therapies targeting the self-renewal of GICs.
TargetMoleculeDateProtocolPhasePatients
Wnt pathwayCelecoxib
NCT0011250206/2005–09/2014Combined with TMZIIN
Results (43 patients): PFS 10.5 months vs. 13.4 months; TMZ vs. TMZ + celecoxib (p = 0.97) [69]
NCT0004728101/2003–07/2017Combined with thalidomide, etoposide and Cyclophosphamide. Unpublished dataIIR
NCT0277037805/2016–10/2019Combined with TMZ and eight repurposed drugsI/IIR
Results: ongoing studies (no recruitment)
NCT0006877009/2003–03/2015Combined with RT and anticonvulsant drugs (p450 inhibitor)IIN undergoing RT and anticonvulsant treatment
Results (35 patients): OS 11.5 months vs. 16 months (p = 0.11; HR = 2.7); p450 inhibitor vs. no p450 inhibitor [70]
NCT0004729410/2002–06/2017Thalidomide combined with the Stupp protocol and celecoxibIIN
See Thalidomide
Notch pathwayRO4929097
NCT0112290111/2010–03/2017MonotherapyIIR
Results (47 patients): PFS 1.7 vs. 1.7 months; OS 6.6 months vs. 6.7 months; RO4929097 after vs. before resection (No statistical data)
Hedgehog pathwayVismodegib GDC-0449
NCT0098034309/2009–08/2017MonotherapyIIR resectable
Results (44 patients): PFS-6 0% vs. 0%; OS 7.8 vs. 7.6 months. Before surgical resection vs. without surgery (No statistical data)
NCT0315838905/2017–02/2020Molecularly Matched Targeted Therapies (APG101, alectinib, idasanutlin, atezolizumab, vismodegib, temsirolimus, palbociclib) combined with RT [71]I/IIN without MGMT promoter methylation
Results (350 patients): ongoing studies (recruitment)
Glasdegib (PF-04449913)
NCT0346645003/2018–04/2020Combined with TMZI/IIN
Results: ongoing studies (recruitment)
STAT3 pathwayNapabucasin (BBI608)
NCT0231553412/2014–10/2019Combined with TMZI/IIR
Unpublished data
R: recurrent GBM; N: newly diagnosed GBM; PFS: progression-free survival; PFS-6: 6-month survival; OS: overall survival. In red, not significant comparative tests. In italics, clinical trials listed in other tables (as mentioned). Results obtained from Clinicaltrials.com (accessed on 1 April 2020) and/or in cited references. Dates correspond to first posted and last update posted.
Table 2. Clinical studies analyzing therapies targeting EGFR and HER2.
Table 2. Clinical studies analyzing therapies targeting EGFR and HER2.
TargetMoleculeDateProtocolPhasePatients
EGFRCetuximab
NCT0104422501/2010–03/2012Combined with RT/TMZ and cilengitide (non-comparative)IIN with MGMT-promoter unmethylated
Unpublished data
NCT0031185704/2006–09/2006Combined with RT/TMZI/IIN
Results (77 patients): PFS6 = 81%; PFS12 = 37%; OS12 = 87%; [109]
NCT0046307304/2007–12/2008Combined with bevacizumab and irinotecanIIR
Results (43 patients): PFS 16 weeks; OS 30 weeks [110]
NCT0280048605/2016–01/2017Intracranial monotherapyIIN
Results: ongoing studies (recruitment)
NCT0188474006/2013–01/2017Combined with bevacizumab and intracranial administrationI/IIN aged under 22
Results: ongoing studies (recruitment)
NCT0286189808/2016–05/2019Intra-arterial combined with STUPP protocolI/IIN
Results: ongoing studies (recruitment)
Panitumumab
NCT0101765311/2009–07/2016Combined with irinotecanIIR
Results (16 patients): PFS-6 12.5%; OS 4.6 months
Nimotuzumab
NCT0075324611/2007–11/2012Combined with RT/TMZ vs. RT/TMZIIIN
Results (142 patients): PFS = 7.7 months vs. 5.8 months (p = 0.7989); OS = 22.3 months vs. 19.6 months (p = 0.485) Nimotuzumab + RT/TMZ vs. RT/TMZ [111]
NCT0338837208/2010–01/2018Combined with RT/TMZIIN
Unpublished data
Depatuxizumab-mafodotin
NCT0341940302/2018–04/2020Combined with RT/TMZ and ophthalmologic prophylactic treatmentIII
Unpublished data
NCT0257332410/2015–04/2020Combined with RT/TMZII/IIIN with EGFR amplification
Results: ongoing studies (no recruitment)
NCT0259026310/2015–05/2019Monotherapy or combined with RT/TMZI/IIN/R
Results: ongoing studies (no recruitment)
NCT0234340601/2015–05/2020Monotherapy or combined with TMZIIR
Results (260 patients): PFS = 2.7 vs. 1.9 vs. 1.9 months; OS = 9.6 vs. 7.9 vs. 8.2 months Depatux-M + TMZ vs. Depatux-M vs. Lomustine or TMZ
GC1118
NCT0361866708/2018–08/2018MonotherapyIIR with high EGFR amplification
Results: ongoing studies (recruitment)
Sym004
NCT0254016109/2015–08/2019MonotherapyIIR
Results: ongoing studies (no recruitment)
Erlotinib
NCT0033788306/2006–03/2014MonotherapyIIR first
Unpublished data
NCT0003949401/2003–08/2013Combined with TMZ/RTI/IIN
Results (100 patients): PFS 7.2 months; OS 15.3 months [112]
NCT0044558803/2007–03/2016Combined with sorafenibIIR
Results (56 patients): PFS 2.5 months; OS 5.7 months [113]
NCT0052552509/2007–05/2014Combined with bevacizumab. TMZ in adjuvant therapyIIN
Results (150 patients): PFS 9.2 months; OS 13.6 months [114]
NCT0018748609/2005–08/2012Combined with TMZ during the Stupp protocolIIN
Results (28 patients): PFS 2.8 months; OS 8.6 months [115]
NCT0072035606/2008–10/2018Combined with bevacizumab. in adjuvant therapy after RT/TMZIIN
Results (48 patients): PFS-12 32%; OS 13.2 months
NCT0067224301/2008–08/2013Combined with sirolimusIIR
Results (32 patients): PFS 6.9 weeks; OS 33.8 weeks [116]
NCT0067197001/2008–03/2013Combined with bevacizumabIIR
Results (25 patients): PFS-6 28%; OS = 42 weeks [117]
NCT0008687906/2004–09/2017Monotherapy compared to TMZ or BCNUIIR
Results (110 patients): PFS 1.8 months vs. 2.4 months; OS 7.7 months vs. 7.3 months (No statistical data); Erlotinib vs. BCNU/TMZ [118]
NCT00301418 03/2006–02/2016MonotherapyI/IIR
Results (11 patients): PFS 1.9 months; OS 6.9 months [119]
NCT0027483301/2006–12/2012Combined with TMZ/RTIIN
Unpublished data
NCT0038789410/2006–06/2013MonotherapyIIR
Results (6 patients): Terminated because ongoing literature at the time confirmed that the selection process was not likely to enrich for a patient population expected to benefit, and rapid disease progression in the first 6 patients.
NCT0005449602/2003–01/2014MonotherapyIIR
Results: ongoing studies (recruitment unknown)
NCT0011273606/2005–06/2015Combined with temsirolimusI/IIR
Results (47 patients): PFS-6 13% [120]
NCT0111087604/2010–11/2014Combined with vorinostat and TMZI/IIR
Unpublished data
NCT0004511001/2003–08/2017MonotherapyI/IIR/N
Results (96 patients): PFS 2 months GBM R; OS 14 months GBM N Post RT [121]
NCT0033576404/2006–07/2018Sorafenib combined with erlotinib. tipifarnib or temsirolimusI/IIR
See Sorafenib
Gefitinib
NCT0023879710/2005–01/2011Combined with RTII-
Unpublished data
NCT0025088711/2005–10/2007Pre- and post-surgery (second surgery)IIR
Results (22 patients): OS 8.8 months [122]
NCT0001417004/2001–07/2013MonotherapyIIN
Unpublished data
NCT0001699106/2001–06/2013MonotherapyIIR first
Results (53 patients): PFS 8.1 weeks; OS 39.4 weeks [123]
HER2NCT0005220801/2003–06/2013Combined with RTI/IIN
Results (147 patients): PFS 4.9 months; OS 11.0 months [124]
NCT0002567501/2003–06/2018MonotherapyIIR
No results posted
NCT0131085503/2011–05/2017Cediranib combined with gefitinib, compared to cediranib and placeboIIR
See Cediranib
Afatinib
NCT0072750606/2008–06/2017Monotherapy ± TMZ and compared with TMZIIR
Results (119 patients): PFS 0.99 months vs. 1.53 months (p = 0.032) vs. 1.87 months (p = 0.204); 9.8 months vs. 8 months (p = 0.386) vs. 10.6 months (p = 0.119); Afatinib vs. Afatinib + TMZ vs. TMZ [125]
Dacomitinib
NCT0152087001/2012–03/2018MonotherapyIIR with EGFR Amplification or EGFRvIII Mutation
Results (49 patients): PFS-6 s 10.6%; PFS 2.7 months; OS 7.4 months [126]
NCT0111252704/2010–08/2018MonotherapyIIR
Unpublished data
Lapatinib
NCT0159157705/2012–09/2016Combined with or non- combined with RT/TMZ. Unpublished dataIIN
NCT0009906012/2004–01/2014Monotherapy.
Unpublished data
I/IIR
NCT0010700304/2005–07/22018Pre-operatory monotherapy. IIR
Unpublished data
NCT0035072707/2006–04/2013Combined with pazopanibIIR
Results (41 patients): PFS 62 vs. 56 days; PFS-6 0 vs. 15%; Patients positive vs. negative for EGFRvIII and/or PTEN [127]
Neratinib
NCT0297778011/2016–02/2020Combined with TMZ vs. TMZIIN
Results: ongoing studies (recruitment)
R: recurrent GBM; N: newly diagnosed GBM; PFS: progression-free survival; PFS-6: 6-month survival; OS: overall sur-vival. In red, not significant comparative tests. In italics, clinical trials listed in other tables (as mentioned). Results obtained from Clinicaltrials.com (accessed on 1 April 2020) and/or in cited references. Dates correspond to first posted and last update posted.
Table 3. Clinical studies analyzing multi-kinase inhibitors.
Table 3. Clinical studies analyzing multi-kinase inhibitors.
MoleculeDateProtocolPhasePatients
Anlotinib
NCT0415747811/2019–11/2019Combined with Stupp protocol compared to Stupp protocol aloneIIN
Not yet recruiting
NCT0400497507/2019–07/2019MonotherapyI/IIR
Results: ongoing studies (recruitment)
NCT0411967410/2019–10/2019Combined with Stupp protocolI/IIN
Results: ongoing studies (recruitment)
Tesevatinib
NCT0284443907/2016–02/2020MonotherapyIIR
Unpublished data
Dacomitinib/Afatinib (see EGFR)
Cabozantinib
NCT0106878202/2010–07/2014MonotherapyIIR first or second
Unpublished data
TG02
NCT0294226410/2016–01/2020Combined with TMZ and compared with TMW aloneI/IIR
Results: ongoing studies (recruitment)
Vandetamib
NCT0044114202/2007–03/2019Combined with TMZ during Stupp protocol compared to Stupp protocol (non- comparative)I/IIN
Results (106 patients): OS 15.9 months vs. 16.6 months (p = 0.75); PFS 6.2 vs. 7.7 months; RT/TMZ vs. vandetanib + RT/TMZ (p = 0.61) [147]
NCT0099500710/2009–03/2016Combined with carboplatin and then monotherapy compared to carboplatin aloneIIR
Results (64 patients): PFS-6 1.7% vs. 0.9%s; OS 5.6 months vs. 5.2 months carboplatin + vandetanib vs. carboplatin (No statistical data) [148]
Bosutinib
NCT0133129104/2011–07/2016MonotherapyIIR
Results (9 patients): PFS 7.71 weeks; OS 50 weeks [149]
R: recurrent GBM; N: newly diagnosed GBM; PFS: progression-free survival; PFS-6: 6-month survival; OS: overall sur-vival. Results obtained from Clinicaltrials.com (accessed on 1 April 2020) and/or in cited references. Dates correspond to first posted and last update posted.
Table 4. Clinical studies analyzing therapies targeting, PDGFR, IGFR, FGFR, ALK.
Table 4. Clinical studies analyzing therapies targeting, PDGFR, IGFR, FGFR, ALK.
TargetMoleculeDateProtocolPhasePatients
PDGFRImatinib
NCT0029077104/2006–04/2011Combined with hydroxyureaIIR
Results (231 patients): PFS 5.6 weeks; OS 26 weeks [151]
NCT0017193809/2005–02/2017Monotherapy in case of impossible re-operationIIR
Unresectable with PDGFR positive
Unpublished data
NCT0015437509/2005–04/2011Combined with hydroxyurea compared with hydroxyurea aloneIIIR
Results (240 patients): PFS 6 weeks vs. 6 weeks (HR = 0.93); OS 21 weeks vs. 19 weeks (HR = 0.92); imatinib + hydroxyurea vs. hydroxyurea alone [155]
NCT0001004901/2003–06/2018MonotherapyI/IIR
Results (34 patients): PFS-6 3% [156]
NCT0003936401/2003–07/2012MonotherapyIIR
Results (51 patients): PFS-6 16% [157]
Dasatinib
NCT0089217705/2009–10/2019Combined with bevacizumab and compared with bevacizumab aloneIIR
Results (121 patients): PFS 3.3 months vs. 3.5 months (p = 0.52; HR = 1.14); OS 7.3 months vs. 7.9 months (p = 0.7; HR = 0.92) bevacizumab + dasatinib vs. bevacizumab + placebo [158]
NCT0042373501/2007–04/2017MonotherapyIIR
Results (77 patients): PFS 1.7 vs. 1.8 months; OS = 6.5 vs. 8.9 months; 200 mg/j vs. 400 mg/j (No statistical data) [152]
NCT0094838906/2008–08/2012Combined with lomustine I/IIR
Results (28 patients): PFS 1.35 months; OS 6.4 months [159]
NCT0086940103/2009–02/2020Combined with RT/TMZ compared to placeboI/IIN
Results (196 patients): OS 15.6 vs. 19.3 months; PFS: 6.2 vs. 7.8 months; dasatinib vs. placebo
Tandutinib
NCT0037908009/2006–04/2017MonotherapyI/IIR
Results (31 patients): PFS-6 16%; OS 8.8 months [154]
NCT0066739404/2008–10/2015Combined with bevacizumabIIR
Results (41 patients): PFS 4.1 months; OS 11 months [153]
Crenolanib
NCT0262636411/2015–06/2017MonotherapyIIR PDGFRA Gene Amplification
Results: ongoing studies (recruitment)
Sunitinib
NCT0110017704/2010–03/2013Monotherapy before and during RTIIN unresectable
Results:(12 patients): PFS 7.7 weeks; OS 12.8 weeks [160]
NCT0092311707/2009–09/2015Monotherapy with or without bevacizumabIIR
Results (87 patients): PFS-6 0.92 vs. 1.08 months Bevacizumab resistant vs. naïve patients
NCT0053537909/2007–08/2010MonotherapyIIR
Results (40 patients): PFS 2.2 months; OS 9.2 months [161]
NCT0292857501/2016–10/2016Combined with TMZ/RTIIN
Results: ongoing studies (recruitment unknown)
NCT0060600801/2008–11/2012MonotherapyIIR
Results (16 patients): PFS 1.4 months; OS 12.6 months [162]
NCT0302589301/2017–06/2017Monotherapy (high dose)II/IIIR
Results: ongoing studies (recruitment)
NCT0049947307/2007–02/2016MonotherapyIIR
Results (25 patients): OS 5.7 vs. 12.3 months; Patients non-EIAC (enzyme-inducing anticonvulsants) vs. EIAC
Regorafenib
NCT0397044705/2019–03/2020Combined with RT/TMZII/IIIN/R
Results: ongoing studies (recruitment)
NCT0405160608/2019–02/2020MonotherapyIIR
Results: ongoing studies (recruitment)
NCT0292622210/2016–09/2018MonotherapyIIR
Results: ongoing studies (recruitment)
MEDI-575
NCT0126856612/2010–04/2017MonotherapyIIR
Results (56 patients): PFS-6 15.4%; PFS 1.4 months; OS 9.7 months [163]
Olaratumab (IMC-3G3)
NCT0089518005/2009–12/2017Monotherapy compared to ramucirumabIIR
Results (80 patients): PFS-6 12.5% vs. 7.5%; OS 49.5 vs. 34.3 weeks; ramucirumab vs. olaratumab
Ponatinib
NCT0247816406/2015–07/2018MonotherapyIIR Bevacizumab-Refractory
Results (15 patients): PFS 28 days; OS 98 days [164]
Leflunomide
NCT0000329306/2004–09/2012Monotherapy compared to procarbazineIIIR
Unpublished data
IGFRAxl1717
NCT0172157711/2012–01/2015MonotherapyI/IIR
Results (8 patients): PFS 8 weeks; OS 15 weeks [165]
FGFRBGJ398
NCT0197570111/2013–12/2019MonotherapyIIR
Results (26 patients): PFS 1.7 months; OS 6.74 months
ALKAlectinib
NCT0315838905/2017–02/2020Molecularly Matched Targeted Therapies (APG101, alectinib, idasanutlin, atezolizumab, vismodegib, temsirolimus, palbociclib) combined with RT [71]I/IIN without MGMT promoter methylation
See Vismodegib
R: recurrent GBM; N: newly diagnosed GBM; PFS: progression-free survival; PFS-6: 6-month survival; OS: overall sur-vival. In italics, clinical trials listed in other tables (as mentioned). Results obtained from Clinicaltrials.com (accessed on 1 April 2020) and/or in cited references. Dates correspond to first posted and last update posted.
Table 5. Clinical studies analyzing therapies targeting mTOR, PI3K/mTOR, Akt & protein kinase c.
Table 5. Clinical studies analyzing therapies targeting mTOR, PI3K/mTOR, Akt & protein kinase c.
TargetMoleculeDateProtocolPhasePatients
mTORTemsirolimus
NCT0080091712/2008–01/2010Combined with bevacizumabIIR
Results (13 patients): PFS 8 weeks; OS 15 weeks [175]
NCT0001632805/2001–07/2013MonotherapyIIR
Results (65 patients): PFS 2.3 weeks; OS 4.4 months [176]
NCT0032971905/2006–10/2018Combined with sorafenib ± surgeryI/IIR
Results (102 patients): PFS 2.71 vs. 4.34 vs. 1.87 months; OS 6.55 vs. 6.74 vs. 3.93 months. Temsirolimus + sorafenib vs. temsirolimus + sorafenib + surgery vs. temsirolimus + sorafenib in patients treated with anti-VEGF (No statistical data) [177]
NCT0101943411/2009–10/2016Combined with RT, compared with RT/TMZIIN. unmethylated MGMT
Results (111 patients): PFS 5.4 months vs. 6.0 months (p = 0.24; HR = 1.26); OS 14.8 months vs. 16.0 months (p = 0.47; HR = 1.2) temsirolimus/RT vs. TMZ/RT [178]
NCT0002272401/2003–06/2018MonotherapyI/IIR
Results (43 patients): 9 weeks [179]
NCT0011273606/2005–06/2015Combined with erlotinibI/IIR
See Erlotinib
NCT0033576404/2006–07/2018Sorafenib combined with erlotinib, tipifarnib or temsirolimusI/IIR
See Sorafenib
NCT0315838905/2017–02/2020Molecularly Matched Targeted Therapies (APG101, alectinib, idasanutlin, atezolizumab, vismodegib, temsirolimus, palbociclib) combined with RT [71]I/IIN without MGMT promoter methylation
See Vismodegib
Sirolimus
NCT0067224301/2008–02/2013Combined with erlotinibIIR
See Erlotinib
Everolimus
NCT0051508608/2007–09/2011MonotherapyIIR
Unpublished data
NCT0010723704/2005–06/2013Combined with AEE788 (inhibitor of the EGFR, HER-2, VEGFR family)IIR
Unpublished data
NCT01434602 09/2011–07/2017Combined with sorafenibIIR
Results: ongoing studies
NCT0080596112/2008–08/2013Combined with Bevacizumab in adjuvant therapy after RT/TMZIIN
Results (68 patients): PFS 11.3 months; OS 13.9 months [180]
NCT0055315011/2007–02/2020Combination of RT/TMZ then TMZ/everolimusIIN
Results (100 patients): PFS-12 6.4 months; OS-12 15.8 months [181]
NCT0106239902/2010–05/2019Combined with RT/TMZI/IIN
Results (171 patients): PFS: 8.2 vs. 10.2 months (p = 0.79); OS: 16.5 vs. 21.2 months (p = 0.008); Patients with or without everolimus [182]
ABI-009 (nab-Rapamycin)
NCT0346326508/2018–12/2020Monotherapy or in combination with bevacuzimab or RT/TMZ or marizomib, or lomustineIIR/N
Results: ongoing studies (recruitment)
PI3KPictilisib
NCT0243036303/2013–01/2016Monotherapy compared with pembrolizumabI/IIR
Unpublished data
Buparlisib (BKM120)
NCT0134966004/2011–01/2017Combined with bevacizumabI/IIR
Preliminary data (76 patients): PFS 2.8 vs. 5.3 months; OS 6.5 vs. 10.8 months; buparlisib + bevacizumab vs. bevacizumab alone (No statistical data)
NCT0133905204/2011–03/2019Monotherapy combined or not combined with surgeryIIR
Results (65 patients): PFS 1.7 months; OS 9.8 months; Patients not submitted to surgery [165]
Sonolisib (PX-866)
NCT0125986904/2015–02/2015MonotherapyIIR first
Results (17 patients): PFS6 = 17% [183]
Paxalisib (GDC-0084)
NCT0352229805/2018–03/2020MonotherapyIIN
Results: ongoing studies (no recruitment)
PI3K/mTORBimiralisib (PQR309)
NCT0285074408/2016–10/2018MonotherapyIIN
Unpublished data
Akt & protein kinase cEnzastaurin
NCT0029581502/2006–11/2016Compared with lomustineIIIR
Results (293 patients): PFS 1.51 months vs. 1.64 months (p = 0.08; HR = 1.28); OS 6.60 months vs. 7.13 months (p = 0.25; HR = 1.20) enzastaurin vs. lomustine [184]
NCT0050982106/2007–04/2016Combined with RT (before, during, after)IIN
Results (60 patients): PFS 6.6 months; OS 15.0 months [185]
NCT0040211611/2006–10/2010Combined with the Stupp protocolI/IIN
Unpublished Phase II results
NCT0058650812/2007–10/2013Combined with bevacizumabIIN
Results (40 patients): PFS 2.0 months; OS = 7.5 months [186]
NCT0377607112/2018–05/2019Combined with RT/TMZIIN
Results: ongoing studies (recruitment)
R: recurrent GBM; N: newly diagnosed GBM; PFS: progression-free survival; PFS-6: 6-month survival; OS: overall sur-vival. In red, not significant comparative tests. In italics, clinical trials listed in other tables (as mentioned). Results obtained from Clinicaltrials.com (accessed on 1 April 2020) and/or in cited references. Dates correspond to first posted and last update posted.
Table 6. Clinical studies analyzing therapies targeting Ras/MAPK/MEK.
Table 6. Clinical studies analyzing therapies targeting Ras/MAPK/MEK.
TargetMoleculeDateProtocolPhasePatients
Ras/MAPKTLN-4601
NCT0073026208/2008–12/2017MonotherapyIIR
Results (20 patients): PFS-6 0%; OS 130 days [201]
Sorafenib
NCT0054481710/2007–06/2016Combined with the Stupp protocol in adjuvant therapyIIN
Results (47 patients): PFS 6 months; OS 12 months [202]
NCT0059749301/2008–03/2013Combined with TMZIIR
Results (32 patients): PFS 6.4 weeks; OS 41.5 weeks [203]
NCT0032971905/2006–11/2016Combined with temsirolimusIIR
See Temsirolimus
NCT0033576404/2006–07/2018Combined with erlotinib. tipifarnib or temsirolimusI/IIR
Results not fully available
NCT0044558803/2007–03/2016Combined with erlotinibIIR
See Erlotinib
NCT0062168602/2008–01/2017Combined with bevacizumabIIR
Results (54 patients): PFS 2.9 months; OS 5.6 months [204]
NCT0143460209/2011–06/2017Combined with everolimusIIR
See Everolimus
NCT0181775103/2013–05/2017Combined with valproic acid and sildenafilIIR
Results: ongoing studies (recruitment)
LY2228820
NCT0236420602/2015–08/2019Combined with the Stupp protocolIIN
Unpublished data
Atorvastatin
NCT02029573 01/2014–08/2017Combined with RT/TMZII/
Results (20 patients): PFS 9.1 months [205]
Dabrafenib
NCT0391907104/2019–03/2020Combined with trametinib (MEK inhibitor) post-RTIIN
Results: ongoing studies (recruitment)
2-OHOA
NCT0425092201/2020–01/2020Combined with RT/TMZIIR
Results: ongoing studies (recruitment)
MEKBinimetinib
NCT0397391806/2019–03/2020Combined with encorafenibIIR BRAF V600-Mutated HGG
Results: ongoing studies (recruitment)
Trametinib
NCT0391907104/2019–03/2020Combined with dabrafenib post-RTIIN
See Dabrafenib
R: recurrent GBM; N: newly diagnosed GBM; PFS: progression-free survival; PFS-6: 6-month survival; OS: overall sur-vival. In italics, clinical trials listed in other tables (as mentioned). Results obtained from Clinicaltrials.com (accessed on 1 April 2020) and/or in cited references. Dates correspond to first posted and last update posted.
Table 7. Clinical studies analyzing therapies targeting apoptosis.
Table 7. Clinical studies analyzing therapies targeting apoptosis.
TargetMoleculeDateProtocolPhasePatients
CD95APG101
NCT0107183702/2010–06/2015Combined with re-irradiation compared to re-irradiation aloneIIR
Results (91 patients): PFS 2.5 months vs. 4.5 months (p = 0.0162; HR = 0.49); OS 11.5 months vs. 11.5 months; reirradation vs. reirradiation + APG101 [215]
NCT0315838905/2017–02/2020Molecularly Matched Targeted Therapies (APG101, alectinib, idasanutlin, atezolizumab, vismodegib, temsirolimus, palbociclib) combined with RT [71]I/IIN without MGMT promoter methylation
See Vismodegib
DRD2/3ONC201
NCT0252569208/2015–01/2020MonotherapyIIR H3 K27M positive
Results: (14 patients): OS 17 weeks; PFS 14 weeks [216]
p53Gene therapy (SGT-53)
NCT0234015612/2014–03/2020Combined with TMZIIR
Unpublished data
p53-MDM2Idasanutlin (RG7388)
NCT0315838905/2017–02/2020Molecularly Matched Targeted Therapies (APG101, alectinib, idasanutlin, atezolizumab, vismodegib, temsirolimus, palbociclib) combined with RT [71]I/IIN without MGMT promoter methylation
See Vismodegib
Bcl-2Gossypol
NCT0054072210/2007–03/2017MonotherapyIIR
Results (56 patients): PFS 1.87 months; OS = 5.9 months
Farnesyl transferaseTipifarnib
NCT00050986 01/2003–08/2012Combined with TMZI/IIR
No published results
NCT0005809704/2003–04/2013Combined with RTIIN
Results (28 patients): PFS 42 days; OS 234.5 days [217]
NCT0000585901/2003–06/2018MonotherapyI/IIR
Results (67 patients): PFS 8 vs. 6 weeks (p = 0.01) patients non- EIAED vs. patients EIAED [218]
NCT0033576404/2006–07/2018Sorafenib combined with erlotinib. tipifarnib or temsirolimusI/IIR
See Sorafenib
Lonafarnib
NCT0003849306/2002–10/2018Combined with TMZIIR
Unpublished data
R: recurrent GBM; N: newly diagnosed GBM; PFS: progression-free survival; PFS-6: 6-month survival; OS: overall sur-vival. In red, not significant comparative tests. In green, significant comparative tests. In italics, clinical trials listed in other tables (as mentioned). Results obtained from Clinicaltrials.com (accessed on 1 April 2020) and/or in cited references. Dates correspond to first posted and last update posted.
Table 8. Clinical studies analyzing therapies targeting autophagy.
Table 8. Clinical studies analyzing therapies targeting autophagy.
TargetMoleculeDateProtocolPhasePatients
AutophagyChloroquine
NCT0243241704/2015–06/2019Combined with the Stupp protocolIIN
Results: ongoing studies
NCT0022497809/2005–11/2009MonotherapyIIIN
Results (30 patients): OS 24 vs. 11 months; chloroquine-treated patients vs. controls [250]
NCT0048660306/2007–07/2019Combined with RT/TMZI/IIN
Results (76 patients): OS 15.6 months [251]
N: newly diagnosed GBM; OS: overall sur-vival. Results obtained from Clinicaltrials.com (accessed on 1 April 2020) and/or in cited references. Dates correspond to first posted and last update posted.
Table 9. Clinical studies analyzing therapies targeting the cell cycle (CDK4/6), multifaceted pathways (proteasome, histone deacetylase, TGFβ) and DNA repair (PARP).
Table 9. Clinical studies analyzing therapies targeting the cell cycle (CDK4/6), multifaceted pathways (proteasome, histone deacetylase, TGFβ) and DNA repair (PARP).
TargetMoleculeDateProtocolPhasePatients
CDK4/6Palbociclib (PD 0332991)
NCT0122743410/2010–07/2015Monotherapy combined or not combined to surgeryIIR
Rb positif
Results (22 patients): PFS 5.14 weeks; OS 15.4 weeks [252]
NCT0315838905/2017–02/2020Molecularly Matched Targeted Therapies (APG101, alectinib, idasanutlin, atezolizumab, vismodegib, temsirolimus, palbociclib) combined with RT [71]I/IIN without MGMT promoter methylation
See Vismodegib
Abemaciclib
NCT0298194012/2016–03/2020Monotherapy combined or not combined to surgeryIIR
Results: ongoing studies (no recruitment)
ProteasomeBortezomib
NCT0364354908/2018–02/2020Combined with TMZI/IIR
MGMT unmethylated
Results: ongoing studies (recruitment)
NCT0064170603/2008–05/2014Combined with vorinostatIIR
Results (37 patients): PFS 1.5 mois; OS 3.2 mois [253]
NCT0099801010/2009–05/2019Combined with TMZ/RTIIN
Unpublished data
NCT0061132502/2008–03/2014Combined with bevacizumabIIR
See Bevacizumab
Marizomib
NCT0334509511/2017–06/2019Combined with TMZ/RTIIIN
Results (749 patients): ongoing studies (recruitment)
NCT0346326508/2018–12/2020Monotherapy (ABI-009) or in combination with bevacuzimab or RT/TMZ or ABI-009, or lomustineIIR/N
See ABI-009
NCT0233056201/2015–03/2020Combined with bevacuzimabI/IIR
See Bevacizumab
Histone desacetylaseVorinostat
NCT00555399 11/2007–12/2019Combined with Isotretinoin and temozolomideI/IIR
Results: ongoing studies (no recruitment)
NCT00731731 08/2008–03/2020Combined with TMZ/RTIIN
Preliminary results (107 patients): OS-15 months 54.6%; PFS 8.05 months
NCT00238303 10/2005–05/2014Combined with surgeryIIR
Results (68 patients): PFS 1.9 months; OS 5.7 months [254]
NCT0111087604/2010–11/2014Combined with erlotinib and TMZI/IIR
See Erlotinib
NCT0064170603/2008–05/2014Combined with bortezomibIIR
See Bortezomib
NCT0126603112/2010–07/2018Bevacizumab in monotherapy vs. combined with vorinostatI/IIR
See Bevacizumab
NCT0173864611/2012–02/2017Combined with bevacizumabIIR
See Bevacizumab
NCT0093999107/2009–06/2013Combined with bevacizumab and TMZI/IIR
See Bevacizumab
Panobinostat (LBH589)
NCT0084852302/2009–07/2010MonotherapyIIR
Unpublished data
FR901228
NCT0008554006/2004–01/2017MonotherapyI/IIR
Results (35 patients): PFS 8 weeks [255]
TGFβ & TGFβRTrabedersen (AP12009)
NCT00431561 02/2007–12/2013Monotherapy vs. TMZ or PVC (procarbazine/lomustine/vincristine)IIbR
Results (145 patients): In GBM patients, response and survival results were comparable among the 3 arms [256]
Galunisertib (LY2157299)
NCT0158226904/2012–12/2019Monotherapy or combined with lomustineIIR
Results: ongoing studies (no recruitment)
NCT0122027110/2010–02/2017Combined with TMZ/RT vs. TMZ/RTI/IIN
Results (56 patients): OS 18.2 vs. 17.9 months (HR = 1.2), PFS 7.6 vs. 11.5 months (HR = 1.8), patients treated with galunisertib combined with TMZ/RT vs. TMZ/RT [257]
OKN-007
NCT0364946408/2018–03/2020MonotherapyI/IIR
Not yet recruiting
PARPIniparib (BSI-201)
NCT0068776506/2008–07/2015Combined with TMZI/IIN
Results (81 patients): OS 22 months [258]
Veliparib
NCT0215298206/2014–03/2020Combined with TMZII/IIIN
Results: ongoing studies (no recruitment)
NCT0358129207/2018–03/2020Combined with RT/TMZIIN
Negative H3 K27M or BRAFV600
Results: ongoing studies (recruitment)
NCT0102649312/2009-/07/2017Combined with TMZI/IIR
Results (215 patients): OS 10.3 vs. 10.7 months (p = 0.95; HR = 0.99) patients BEV-naïve low vs. high TMZ dose; OS 4.7 vs. 4.7 months (p = 0.93; HR = 0.93) patients BEV-failure low vs. high TMZ dose; PFS-6 17 vs. 4.4% patients BEV-naïve vs. BEV-failure [259]
Olaparib
NCT0321227407/2017–03/2020MonotherapyIIIDH1/2 mutations
Results: ongoing studies (recruitment)
NCT0297462111/2016–03/2020Cediranib combined with olaparib and compared to bevacizumabIIR
See Cediranib
Pamiparib
NCT0315086205/2017–11/2019Combined with RT/TMZI/IIR/N
Results: ongoing studies (no recruitment)
NCT0391474204/2019-/2020Combined with TMZI/IIR IDH1/2 mutations
Results: ongoing studies (recruitment)
R: recurrent GBM; N: newly diagnosed GBM; PFS: progression-free survival; PFS-6: 6-month survival; OS: overall sur-vival. In red, not significant comparative tests. In italics, clinical trials listed in other tables (as mentioned). Results obtained from Clinicaltrials.com (accessed on 1 April 2020) and/or in cited references. Dates correspond to first posted and last update posted.
Table 10. Clinical studies analyzing therapies targeting VEGF and VEGFR.
Table 10. Clinical studies analyzing therapies targeting VEGF and VEGFR.
TargetMoleculeDateProtocolPhasePatients
VEGFBevacizumab
NCT0160979006/2012–03/2020Combined with trebananibIIR
Preliminary results (116 patients): OS 11.5 vs. 7.5 months (p = 0.09; HR = 1.46); PFS 4.8 vs. 4.2% (p = 0.04; HR = 1.51)
NCT0081728401/2009–11/2011Combined with RT/TMZ or RT/irinotecanIIN
Unpublished data
NCT0186063805/2013–04/2018Continuous treatment with Stupp, followed with Lomustine in first disease progression (PD1) and with chemotherapy in second progression (PD2)IIR
Results (296 patients): OS 6.4 vs. 5.5 months (HR = 1.04); PFS 2.3 vs. 1.8 months (HR = 0.70) PD1 lomustine bevacizumab vs. lomustine alone; PFS 2 vs. 2.2 months (HR = 0.70) PD2 bevacizumab chemotherapy vs. chemotherapy alone. No p values were reported [278]
NCT0111549105/2010–12/2014Combined with TMZIIR
Results (32 patients): PFS 18.29 weeks; OS 31.43 weeks
NCT0059068101/2008–09/2015Combined with TMZIIN
Unpublished data
NCT0097901709/2009–03/2014Combined with TMZ and irinotecanIIN unresectable and multifocal
Results (41 patients): OS 12 months; PFS 8.6 months [279]
NCT0118640608/2010–02/2019Combined with gliadel, TMZ and RTIIN
Results (41 patients): OS 19.4 months; PFS 11.3 months
NCT0190333007/2013–11/2019Combined with ERC1671 (vaccine) and granulocyte-macrophage colony-stimulating factor (GM-CSF) compared to combination with placeboIIR
Results: ongoing studies (recruitment)
NCT0144367609/2011–11/2016Combined with RT compared to RT aloneIIN in elderly
Results (75 patients): PFS 7.6 vs. 4.8 months (p = 0.003); OS 12.1 vs. 12.2 months (p = 0.77); bevacizumab + RT vs. RT [280]
NCT0289801209/2016–09/2016Combined with TMZIIN age over 70
Results (66 patients): OS 23.9 weeks; PFS 15.3 weeks [281]
NCT0114985006/2010–02/2020Combined with TMZIIN in elderly
Results: ongoing studies (no recruitment)
NCT0100487410/2009–02/2020Combined with RT/TMZ followed by combination with TMZ/popotecanII/
Preliminary results (80 patients): OS 17.2 months; PFS 11.1 months
NCT0073543608/2008–02/2013Combined with gliadel and irinotecanIIN
Results (18 patients): PFS 8 months; OS 13.5 months
NCT0269828003/2016–07/2018Combined with nimustineIIR
Unpublished data
NCT0126603112/2010–07/2018Monotherapy vs. combined with vorinostatI/IIR
Results (patients): OS 9.24 vs. 7.8 months; bevacizumab vs. bevacizumab + vorinostat
NCT0101328511/2009–01/2016Combined with TMZ and RTIIN
Results: ongoing studies (recruitment unknown)
NCT0173864611/2012–02/2017Combined with vorinostatIIR
Results (38 patients): PFS 3.7 months; OS 10.4 months; PFS-6 30% [282]
NCT0093999107/2009–06/2013Combined with vorinostat and TMZI/IIR
Results (39 patients): PFS 6.7 months; OS 12.5 months; PFS-6 53.8%
NCT0033720706/2006–02/2020MonotherapyIIR
Results (54 patients): PFS-6 24%
NCT0026835912/2005–07/2014Combined with irinotecanIIR
Results (32 patients): PFS 23 weeks; PFS-6 38% OS-6 72% [283]
NCT0079566511/2008–03/2020Combined with carmustineIIR
Unpublished data
NCT0233056201/2015–03/2020Combined with marizomibI/IIR
Results: ongoing studies (no recruitment)
NCT0092116706/2009–12/2013Combined with irinotecanIIR
Results: completed, no results posted
NCT0215710306/2014–05/2018Subcutaneous monotherapyIIR
Results (3 patients): 66.7% decrease in radiation-related edema
NCT01209442 09/2010–04/2019Combined with hypofractionated RT and TMZIIN
Results (30 patients): PFS 14.3 months; OS 16.3 months [284]
NCT02120287 04/2014–05/2019Combined with radiosurgeryIIR
Results (16 patients): OS 11.73 months
NCT01102595 04/2010–08/2015Combined with TMZ in neoadjuvant therapy of the Stupp protocol compared to the Stupp protocolIIN, unresectable
Results (102 patients): PFS 2.2 vs. 4.8 months (p = 0.10; HR = 0.70); OS 7.7 vs. 10.6 months (p = 0.07; HR = 0.68); TMZ vs. TMZ + bevacizumab [285]
NCT01022918 12/2009–09/2012Combined with irinotecan in neoadjuvant and adjuvant therapy with TMZ, compared to neoadjuvant TMZ and StuppIIN, unresectable
Results: (120 patients): PFS = 7.1 vs. 5.2 months (HR = 0.82); OS = 11.1 vs. 11.1 months; bevacizumab/Irinotecan vs. ctrl [286]
NCT00943826 07/2009–09/2017Combined with TMZ during the Stupp protocol, compared to the Stupp protocolIIIN
Results (921 patients): PFS 10.6 vs. 6.2 months (p < 0.001; HR = 0.64); OS 16.8 vs. 16.7 months (p = 0.1; HR = 0.88); bevacizumab + Stupp vs. Stupp [287]
NCT01067469 02/2010–03/2020Low dose and combined with lomustine, compared to high dose bevacizumab aloneIIR
Results (69 patients): PFS 4.34 vs. 4.11 months (p = 0.19); OS 9.6 vs. 8.3 months (p = 0.75); bevacizumab + lomustine vs. bevacizumab [288]
NCT00883298 04/2009–03/2017Combined with TMZ twice a weekIIR
Results (30 patients): PFS 5.5 months; OS 51 weeks [289]
NCT00345163 06/2006–05/2017Combined with or not combined with irinotecanIIR
Results (167 patients): PFS-6 42.6% vs. 50.3% (p < 0.0001); PFS 4.2 vs. 5.6 months; OS 9.2 months vs. 8.7 months; bevacizumab alone vs. bevacizumab + irinotecan [276]
NCT01474239 11/2011–03/2016Compared with fotemustineIIR
Results (91 patients): PFS 3.38 vs. 3.45 months; OS 7.3 vs. 8.7 months; bevacizumab vs. fotemustine (no statistical data) [290]
NCT02761070 05/2016–02/2019Combined with high-dose TMZ compared to bevacizumab aloneIIIR
Results: ongoing studies (recruitment)
NCT02743078 04/2016–11/2019Combined with Optune®IIR Beva refractory or resistant to Beva
Unpublished data
NCT01894061 07/2013–03/2020Combined with NovoTTFIIR
Unpublished data
NCT01814813 03/2013–06/2019Combined with vaccination (HSPPC-96) compared to bevacizumab aloneIIR
Preliminary results (90 patients): PFS 3.7 vs. 2.5 vs. 5.3 months (p < 0.01); OS 6.6 vs. 9.2 vs. 10.7 months (p = 0.16); HSPPC-96 + Bevacizumab concomitant vs. HSPPC-96 + bevacizumab on progression vs. bevacizumab alone
NCT01730950 11/2012–03/2020Combined with re-irradiation, compared to bevacizumab aloneIIR
Preliminary results (170 patients): PFS 8.9 vs. 7.9% (p = 0.05; HR = 0.73); OS 25.1 vs. 21.6% (p = 0.46; HR = 0.98); bevacizumab alone vs. bevacizumab + RT
NCT0096733008/2009–11/2015Combined with RT, then in adjuvant therapy combined with Irinotecan compared to the Stupp protocolIIN. MGMT non methylated
Results (182 patients): PFS 5.99 vs. 9.7 months (HR = 0.57; p < 0.001); OS 16.6 vs. 17.5 months (HR = 1.02; p = 0.55); TMZ vs. bevacizumab + irinotecan [291]
NCT0234354901/2015–07/2019Combined with Optune® and TMZIIN
Results: ongoing studies (recruitment)
NCT01290939 02/2011–02/2018Combined with lomustineIIIR
Results (437 patients): PFS 4.2 vs. 1.5 months (HR = 0.49; p < 0.001); OS 9.1 vs. 8.6 months (HR = 0.95; p = 0.65); bevacizumab + lomustine vs. lomustine alone [292]
NCT00611325 02/2008–03/2014Combined with bortezomibIIR
Results (56 patients): PFS 2 vs. 2.5 months; OS 8 vs. 6 moonths; PFS-6 25 vs. 28.6%; EIAED vs. non-EIAED
NCT01269853 01/2011–05/2019Intracerebral administrationI/IIR
Results: ongoing studies (recruitment)
NCT01811498 03/2013–05/2019Intracerebral administrationI/IIN
Results: ongoing studies (recruitment)
NCT02511405 07/2015–10/2018Combined with VB-111 (antiangiogenic), compared to bevacizumab aloneIIIR
Results (256 patients): OS 6.8 vs. 7.9 months (p = 0.19; HR = 1.20) combined vs. bevacizumab alone [293]
NCT00612339 02/2008–05/2013Combined with TMZIINon resectable
Results (41 patients): RR 24.4%
NCT03149003 05/2017–01/2020Combined with DSP-7888 (peptide vaccine) compared to bevacizumab aloneIIR
Results: ongoing studies (no recruitment)
NCT00501891 07/2007–05/2013Combined with TMZIIR
Results (32 patients): PFS 15.8 weeks; OS 37.1 weeks [294]
NCT00597402 01/2008–05/2014Combined with RT/TMZ, then combined with irinotecanIIN
Results (75 patients): PFS 14.2 months; OS 21.2 months [295]
NCT00433381 02/2007–09/2018Combined with irinotecan or combined with TMZIIR
Unpublished data
NCT00613028 02/2008–06/2013Combined with etoposide or TMZIIR Resistant to Beva/Irinotecan
Results (22 patients): PFS 4.1 vs. 8.1 weeks; OS 12.6 vs. 19 weeks; PFS-6 0 vs. 7.7%; bevacizumab + TMZ vs. bevacizumab + etoposide
NCT00612430 02/2008–08/2013Combined with etoposideIIR
Results (27 GBM et 32 grade III glioma patients): PFS6 40.6% & 44,4%; OS 63.1 & 44.4 weeks [296]
NCT00884741 04/2009–07/2019Combined with adjuvant TMZ compared to the Stupp protocolIIIN
Results (621 patients): PFS 10.7 months vs. 7.3 months (HR 0.79; p 0.007); OS 15.7 months vs. 16.1 months (HR 1.13; p 0.21) (bevacizumab + Stupp vs. Stupp + placebo) [297]
NCT0046307304/2007–12/2008Combined with cetuximab and irinotecanIIR
See Cetuximab
NCT0188474006/2013–01/2017Combined with cetuximab and intracranial administrationI/IIN aged under 22
See Cetuximab
NCT0052552509/2007–05/2014Combined with erlotinib, TMZ in adjuvant therapyIIN
See Erlotinib
NCT0072035606/2008–10/2018Combined with erlotinib, in adjuvant therapy after RT/TMZIIN
See Erlotinib
NCT0067197001/2008–03/2013Combined with erlotinibIIR
See Erlotinib
NCT0089217705/2009–10/2019Combined with dasatinib and compared with bevacizumab aloneIIR
See Dasatinib
NCT0066739404/2008–10/2015Combined with tandutinibIIR
See Tandutinib
NCT0092311707/2009–09/2015Sunitinib in monotherapy with or without bevacizumabIIR
See Sunitinib
NCT0080091712/2008–01/2010Combined with temsirolimusIIR
See Temsirolimus
NCT0080596112/2008–08/2013Combined with everolimus in adjuvant therapy after RT/TMZ IIN
See Everolimus
NCT0346326508/2018–12/2020Monotherapy (ABI-009) or in combination with bevacuzimab or RT/TMZ or marizomib, or lomustineIIR/N
See ABI-009
NCT0134966004/2011–01/2017Combined with buparlisibI/IIR
See Buparlisib
NCT0058650812/2007–10/2013Combined with enzastaurinIIN
See Enzastaurin
NCT0062168602/2008–01/2017Combined with sorafenibIIR
See Sorafenib
NCT0163222806/2012–02/2018Onartuzumab combined or not with bevacizumab, compared to bevacizumab aloneIIR
See Onartuzumab
NCT0111339804/2010–12/2015Rilotumumab combined with bevacizumabIIR
See Rilotumumab
NCT0164834806/2012–05/2018TRC105 combined with bevacizumab, compared to bevacizumab aloneIIR
See TRC105
NCT0156491403/2012–06/2019TRC105 combined with bevacizumabIIR treated with Bevacizumab
See TRC105
NCT0129026302/2011–07/2017Trebananib combined or not with bevacizumabI/IIR
See Trebananib
VEGFRPazopanib
NCT0233149811/2014–07/2019Combined with the Stupp protocolI/IIN
Results: ongoing studies (recruitment)
NCT0045938104/2007–03/2017MonotherapyIIR
Results (35 patients): PFS 12 weeks; OS 35 weeks; PFS-6 3% [298]
NCT0193109808/2013–03/2020Combined with topotecanIIR
Results (35 patients): OS 42 weeks; PFS 24 weeks; PFS-6 46%; OS-6 77% [277]
NCT0035072707/2006–04/2013Combined with lapatinibIIR
See Lapatinib
Cediranib
NCT0131085503/2011–05/2017Combined with Gefitinib, compared to cediranib and placeboIIR
Results (97 patients): PFS 3.6 vs. 2.8 months (p = 0.17; HR = 0.72); OS 7.2 months vs. 5.5 months (HR = 0.68); cediranib + gefetinib vs. cediranib + placebo [299]
NCT0077715310/2008–12/2016Monotherapy or combination with lomustine, compared with lomustine alone IIIR
Results (325 patients): PFS 92 vs. 125 vs. 44 days (p = 0.90; 0.16; HR = 1.05; 0.76); OS 8 vs. 9.4 vs. 9.8 months (p = 0.10; 0.50; HR = 1.43; 1.15); cediranib vs. cediranib + lomustine vs. lomustine + placebo [300]
NCT0297462111/2016–03/2020Combined with olaparib and compared to bevacizumabIIR
Results: ongoing studies (no recruitment)
NCT0106242502/2010–03/2020Combined with TMZ in the Stupp protocol, compared to the Stupp protocolIIN
Preliminary data (149 patients): PFS 2.7 vs. 6.2 months (p = 0.03; HR = 0.67); OS 13.8 vs. 14.5 months (p = 0.44; HR = 0.87);
Stupp vs. cediranib + Stupp
NCT0066250604/2008–09/2017Combined with TMZ/RTIIN
Unpublished data
NCT0030565603/2006–08/2013MonotherapyIIR
Results (31 patients): PFS 117 days; OS 227 days [301]
Nintedanib
NCT0125148412/2010–10/2012Monotherapy (after treatment with the Stupp protocol or with bevacizumab)IIR
Results (25 patients): PFS 1 vs. 1 month; OS 10 vs. 2 months (p < 0.02); previous treatment with Stupp vs. bevacizumab [302]
NCT0166660006/2012–11/2017Combined with RT, compared to RT aloneI/IIR
Unpublished data
NCT0138078206/2011–08/2014MonotherapyIIR whether or not treated with Bevacizumab
Results (36 patients): PFS 28 vs. 28 days; OS 6.9 vs. 2.6 months; not treated with bevacizumab vs. 1st line with bevacizumab (No statistical data) [303]
Dovitinib
NCT0175371312/2012–12/2017MonotherapyIIR whether or not treated with Bevacizumab
Results (33 patients): PFS 2 vs. 1.8 months; OS 8 vs. 4.3 months; bevacizumab-naive vs. 1st line with bevacizumab
Vandetanib (see Multikinase inhibitors)
NCT0044114202/2007–04/2017Combined with the TMZ of the Stupp protocolI/IIN
See Multikinase inhibitors
NCT0099500710/2009–02/2016Combined with carpoblatin and compared to carboplatin aloneIIR
See Multikinase inhibitors
Vatalanib
NCT0012870008/2005–09/2012Combined with TMZ/RTI/IIN
Results (20 patients): PFS 7.2 months; OS 16.2 months [304]
Tivozanib
NCT01846871 03/2013–01/2019MonotherapyIIR
Results (10 patients): PFS-6 10%; PFS 2.3 months; OS 8.1 months [305]
Axitinib
NCT0156219703/2012–01/2019Monotherapy or combined with lomustineIIR
Unpublished data
NCT0150811701/2012–09/2017Combined with RTIIN elderly
Results (1 patient): OS 0.2 years
NCT0366076109/2018–04/2019Combined with TMZIIR
Unpublished data
CT-322
NCT0056241911/2007–10/2010Combined with irinotecanIIR
Results: ongoing studies (recruitment unknown)
Semaxanib (SU5416)
NCT0000486803/2003–06/2018MonotherapyI/IIR RT non-responder
Unpublished data
Tanibirumab
NCT0385609902/2019–03/2020MonotherapyIIR
Results: ongoing studies (recruitment)
NCT0303352401/2017–01/2017MonotherapyIIR
Results: ongoing studies (recruitment unknown)
R: recurrent GBM; N: newly diagnosed GBM; PFS: progression-free survival; PFS-6: 6-month survival; OS: overall sur-vival. In red, not significant comparative tests. In green, significant comparative tests. In italics, clinical trials listed in other tables (as mentioned). Results obtained from Clinicaltrials.com (accessed on 1 April 2020) and/or in cited references. Dates correspond to first posted and last update posted.
Table 11. Phase I/II clinical studies analyzing therapies targeting c-MET and its ligand HGF, PIGF and Endoglin (CD105).
Table 11. Phase I/II clinical studies analyzing therapies targeting c-MET and its ligand HGF, PIGF and Endoglin (CD105).
TargetMoleculeDateProtocolPhasePatients
c-METOnartuzumab
NCT0163222806/2012–02/2018Combined or not with bevacizumab, compared to bevacizumab aloneIIR
Results (129 patients): PFS 3.9 months vs. 2.9 months (p = 0.7444; HR = 1.06); OS 8.8 months vs. 12.9 months (p = 0.1389; HR = 1.45); ornatuzumab + bevacizumab vs. placebo + bevacizumab [318]
Cabozantinib
NCT0070428806/2008–06/2014MonotherapyIIR
Results (152 patients): PFS 3.7 vs. 3.7 months; OS 7.7 months vs. 10.4 months; 140 mg/j vs. 100 mg/j (No statistical data) [319]
HGFRilotumumab
NCT0111339804/2010–12/2015Combined with bevacizumabIIR
Results (60 patients):
PFS 4 weeks vs. 4.1 weeks (10 mg/kg vs. 20 mg/kg); OS = 3.6 months vs. 3.4 months in patients previously treated with bevacizumab
PFS 4.1 weeks vs. 4.7 weeks; OS 10.9 months vs. 11.4 months in patients previously untreated with bevacizumab [320]
PIGFAflibercept
NCT0036959008/2006–08/2015MonotherapyIIR
Results (42 patients): PFS 12 weeks; OS 39 weeks [321]
CD105TRC105
NCT0164834806/2012–05/2018Combined with bevacizumab, compared to bevacizumab aloneIIR
Results (101 patients): OS 9.7 vs. 7.4 months (HR = 1.06; p = 0.82); PFS-6 25 vs. 30.2%
NCT0156491403/2012–06/2019Combined with bevacizumabIIR treated with Bevacizumab
Results (22 patients): OS 5.75 months; PFS 1.81 vs. 1.30 patients receiving or not simultaneously bevacizumab
R: recurrent GBM; N: newly diagnosed GBM; PFS: progression-free survival; PFS-6: 6-month survival; OS: overall sur-vival. In red, not significant comparative tests. In italics, clinical trials listed in other tables (as mentioned). Results obtained from Clinicaltrials.com (accessed on 1 April 2020) and/or in cited references. Dates correspond to first posted and last update posted.
Table 12. Clinical studies analyzing therapies targeting secondary pathways of angiogenesis.
Table 12. Clinical studies analyzing therapies targeting secondary pathways of angiogenesis.
TargetMoleculeDateProtocolPhasePatients
β-FGF & TNThalidomide
NCT0041254212/2006–02/2012Combined with irinotecanIIR
Results (33 patients): PFS-6 25%; PFS 13 weeks; OS 36 weeks [336]
NCT0003946806/2002–10/2011Combined with irinotecan and RTII-
Results (26 patients): PFS6 19% vs. 40%; recurrent vs. new (No statistical data) [337]
NCT0004729410/2002–06/2017Combined with the Stupp protocol and celecoxibIIN
Results (50 patients): PFS 5.9 months; OS 12.6 months [338]
NCT0052148208/2007–08/2007Combined with TMZ and compared TMZ aloneIIR
Results: ongoing studies (recruitment unknown)
NCT0007909203/2004–04/2017Combined with procarbazineIIR
Unpublished data
NCT0000635805/2004–06/2018Combined with TMZIIR
Results (44 patients): PFS 15 weeks [339]
NCT0004728101/2003–07/2017combined with celecoxib, etoposide and cyclophosphamideIIR
See Celecoxib
IntegrinsCilengitide
NCT00689221 06/2008–11/2014Combined with the Stupp protocolIIIN methylated MGMT status
Results (926 patients): PFS 13.5 months vs. 10.7 months; Investigator (p = 0.46; HR = 0.93); PFS 10.6 months vs. 7.9 months (p = 0.41; HR = 0.92); Independent; OS 26.3 months vs. 26.3 months; cilengitide + Stupp vs. Stupp (p = 0.86; HR = 1.02) [340]
NCT00813943 12/2008–01/2017Combined with the Stupp protocolIIN non-methylated MGMT status
Results (265 patients): PFS 5.6 vs. 5.9 (HR = 0.822) vs. 4.1 months (HR = 0.794); Independent PFS 6.4 vs. 7.5 (HR = 0.772) vs. 6.0 months (HR = 0.720) Investigator
OS 16.3 vs. 14.5 (p = 0.32; HR = 0.686) vs. 13.4 months (p = 0.3771; HR = 0.822);
cilengitide 2x/week vs. cilengitide 5x/week vs. Stupp [341]
NCT0104422501/2010–03/2012Combined with the Stupp protocolIIN non-methylated MGMT status
See Cetuximab
NCT00085254 06/2004–02/2016Combined with RT/TMZIIN
Results (112 patients): OS 19.7 months; OS 17.4 months (cilengitide 500 mg); OS 20.7 months (cilengitide 2000 mg);
OS 30 months (methylated MGMT); OS 17.4 months (non-methylated MGMT) [342]
NCT00112866 10/2004–10/2017MonotherapyIIR
Results (26 patients): PFS-6 12%; PFS 8 weeks
NCT01124240 05/2010–07/2011Combined with TMZ, RT and procarbazineIIN Non Methylated
Results: ongoing studies (recruitment unknown)
NCT0009396410/2004–04/2019MonotherapyIIR
Results (81 patients): PFS-6 7.5 vs. 15%; PFS 1.81 vs. 1.91 months; OS 6.54 vs. 9.91 months; Patients receiving 500 mg vs. 2000 mg [343]
NCT0000609301/2003–06/2013MonotherapyI/IIR
Unpublished data
ATN-161
NCT0035231307/2006–05/2012Combined with carboplatinI/IIR
Unpublished data
AngiopoietinTrebananib (AMG-386)
NCT01290263 02/2011–07/2017Combined or not with bevacizumabI/IIR
Results (48 patients): OS 285 vs. 341 days; PFS 108 vs. 21 days; AMG-386 + bevacizumab vs. AMG-386 alone
NCT0160979006/2012–03/2020Combined with bevacizumabIIR
See Bevacizumab
Target not clearly identifiedRecombinant Human Endostatin
NCT0426797802/2020–03/2020Combined with TMZ and irinotecanIIR
Results: ongoing studies (recruitment)
PSMAProstate Specific Membrane Antigen (PSMA) ADC
NCT0185693305/2013–04/2019MonotherapyIIR
Results (6 patients): No objective responses noted [344]
MMPPrinomastat
NCT0000420005/2004–08/2012Combined with TMZ/RTIIN
Unpublished data
R: recurrent GBM; N: newly diagnosed GBM; PFS: progression-free survival; PFS-6: 6-month survival; OS: overall sur-vival. In red, not significant comparative tests. In italics, clinical trials listed in other tables (as mentioned). Results obtained from Clinicaltrials.com (accessed on 1 April 2020) and/or in cited references. Dates correspond to first posted and last update posted.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cruz Da Silva, E.; 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. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13081795

AMA Style

Cruz Da Silva E, 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(8):1795. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13081795

Chicago/Turabian Style

Cruz Da Silva, Elisabete, Marie-Cécile Mercier, Nelly Etienne-Selloum, Monique Dontenwill, and Laurence Choulier. 2021. "A Systematic Review of Glioblastoma-Targeted Therapies in Phases II, III, IV Clinical Trials" Cancers 13, no. 8: 1795. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13081795

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