Next Article in Journal
Tumor Microenvironment and Metabolism
Next Article in Special Issue
The Dark Side of IFN-γ: Its Role in Promoting Cancer Immunoevasion
Previous Article in Journal
Sphingosine-1-Phosphate Receptor 1, Expressed in Myeloid Cells, Slows Diet-Induced Atherosclerosis and Protects against Macrophage Apoptosis in Ldlr KO Mice
Previous Article in Special Issue
Chemokines as a Conductor of Bone Marrow Microenvironment in Chronic Myeloid Leukemia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Major Challenges and Potential Microenvironment-Targeted Therapies in Glioblastoma

Tumor Angiogenesis laboratory, Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2017, 18(12), 2732; https://doi.org/10.3390/ijms18122732
Submission received: 30 November 2017 / Revised: 13 December 2017 / Accepted: 15 December 2017 / Published: 16 December 2017
(This article belongs to the Special Issue Tumor Microenvironment)

Abstract

:
Glioblastoma (GBM) is considered one of the most malignant, genetically heterogeneous, and therapy-resistant solid tumor. Therapeutic options are limited in GBM and involve surgical resection followed by chemotherapy and/or radiotherapy. Adjuvant therapies, including antiangiogenic treatments (AATs) targeting the VEGF–VEGFR pathway, have witnessed enhanced infiltration of bone marrow-derived myeloid cells, causing therapy resistance and tumor relapse in clinics and in preclinical models of GBM. This review article is focused on gathering previous clinical and preclinical reports featuring major challenges and lessons in GBM. Potential combination therapies targeting the tumor microenvironment (TME) to overcome the myeloid cell-mediated resistance problem in GBM are discussed. Future directions are focused on the use of TME-directed therapies in combination with standard therapy in clinical trials, and the exploration of novel therapies and GBM models for preclinical studies. We believe this review will guide the future of GBM research and therapy.

1. Glioblastoma Statistics

Glioblastoma (GBM) is the most common highly malignant adult primary intracranial neoplasm. GBMs comprise 14.9% of all primary brain and central nervous system (CNS) tumors, 47.1% of the malignant primary brain and CNS tumors, and 56.1% of all gliomas. According to the 2017 Central Brain Tumor Registry of the United States (CBTRUS) report, the average annual age-adjusted incidence rate of GBM is 3.20/100,000 population. The incidence rate ratio is significantly high in older ages [1], in males compared to females (1.58) and in whites compared to blacks (1.93). In past years, GBM had the highest number of cases of all malignant CNS tumors, with 12,500 cases projected in 2017 and 12,760 in 2018 [2]. Overall, GBM has a poor prognosis with quite low relative survival estimates; only 5.5% patients between the age of 55–64 survive five years [2].

2. Overarching Challenges

GBM tumors harbor a large network of blood vessels, have invasive features, and exhibit severe hypoxia with highly complex genetic, molecular and cellular mechanisms. All of these hallmarks contribute to therapy resistance and tumor recurrences, a common outcome was seen in the clinic. In the following sections, we will discuss current challenges in GBM therapy including tumor cells extrinsic characteristics such as the myeloid cell-rich tumor microenvironment (TME) and tumor cell intrinsic properties such as the genetic and molecular heterogeneity that drives therapy resistance. The potential combination of microenvironment-targeted therapies with standard therapies, which could be a key in future GBM therapy, will be discussed at the end. Moreover, this article will highlight critical and translational aspects of GBM.

2.1. Challenges Due to Hypoxia and Hyper-Vasculaturity in the Microenvironment

GBMs are considered to be hypoxic solid tumors. When the tumor grows larger than a critical size (2–3 mm in diameter), it cannot cope with the nutritional demand of the rapidly dividing and growing cancer cells, leading to hypoxia. Hypoxia is one of the major challenges in GBM therapy [3]. In addition, GBM is the most vascularized CNS cancer with the highest degree of vascular proliferation and endothelial cell hyperplasia [4]. One of the classical mechanisms, angiogenesis, which is the formation of new blood vessels, plays a pivotal role in GBMs development and growth. GBM vasculatures are functionally and structurally anomalous; they are characterized by coarse vessel diameter, permeability, tortuosity, and thickened basal lamina that can also lead to more hypoxic regions [5]. Several proteins, cytokines, and factors are known to contribute to the GBM microenvironment (Table 1) [6,7]. Studies have investigated that tumor-associated hypoxia results in upregulation of hypoxia-inducible factor-1α (HIF-1α), which subsequently leads to upregulation of several molecular mediators, e.g., vascular endothelial growth factor (VEGF). VEGF family members signal predominantly through the receptor tyrosine kinases, VEGF receptors (VEGFR)-1, VEGFR-2, and VEGFR-3, in association with the co-receptors [8]. HIF-1α contributes to induction of stromal cell-derived factor 1 (SDF1α) in the TME, which helps in recruiting vascular modulatory bone marrow-derived cells (BMDCs) to stimulate angiogenesis, vasculogenesis, invasion, and immunosuppression mechanisms in GBM tumors [9].
In addition to VEGF and SDF1α, other pro-angiogenic factors upregulated in GBMs include hepatocyte growth factor (HGF), fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), placenta-like growth factor (PLGF), angiopoietin-2 (ANGPT2), interleukin-8 (IL-8), matrix-metalloproteinase (MMP)-2, MMP-9, collagen type I α1 (COLIA1), endothelial markers CD34, Tenascin-C, neuron-glial antigen 2 (NG-2) on pericytes, insulin-like growth factor (IGF), and epidermal growth factor (EGF). VEGF is one of the most crucial growth factors and plays an indispensable role in GBM neovascularization by interacting with a number of signaling pathways to advance GBM growth [10]. These pathways include activation of RAS/RAF/mitogen-activated protein kinase (MAPK) [11,12], phosphatase and tensin homolog (PTEN)/phosphoinositide (PI) 3-kinase/AKT [13], phospholipase C-γ/protein kinase C [13], nitric oxide (NO) [14], platelet-derived growth factor (PDGF)-B [15], and the notch–delta-like ligand (DLL) 4 signaling pathway [16]. GBMs are diagnosed at the advanced stages when they harbor hypoxia and leaky vasculatures. Therefore, we need adjuvant treatments that have capabilities (1) to normalize blood vessels (2) that can penetrate hypoxic regions of the GBM tumor along with current standard therapies and (3) that can block the infiltration of BMDCs and myeloid cells to the GBM tumors, overcoming the challenges in GBM therapies. Interestingly, heterogeneity exists in context to hypoxia and angiogenic areas within the tumors. At the invading front, tumors exhibit hypoxic stress and promote neovascularization. A recent study discovered that tumors display distinct metabolic profile depending on the microenvironment [17]. The dynamic plasticity in metabolism could have implication in therapeutic failures seen in anti-GBM therapies in the past.

2.2. Challenges Due to Microenvironment-Driven Resistance to Antiangiogenic Therapy

For the first time, Folkman et al. demonstrated that tumor growth was dependent on continual blood supply and rapid vascularization, a process called angiogenesis. It was believed that targeting pathological angiogenesis would curb the growth of the tumor by impeding the formation of tumor-associated neovessels [18,19]. The idea of angiogenesis driving tumor growth and metastasis gained momentum and soon the concept of tumor angiogenesis became a hallmark of solid tumors [18,20,21,22]. The hypervascular nature of GBM tumors prompted the implementation of anti-angiogenic therapy (AAT) as an adjuvant to the current standard of care, which included surgical resection of tumors, radiotherapy (RT), and temozolomide (TMZ) chemotherapy. Drugs targeting the VEGF–VEGFR pathway, such as Vatalanib, Sunitinib, and Cediranib found therapeutic application in the treatment of many hypervascular solid tumors including GBM [23,24,25,26,27,28]. GBM tumors are extremely heterogeneous and targeting the endothelial cells was believed to be a viable option for countering the uncontrolled tumor growth. However, GBM tumors developed refractoriness and therapeutic resistance to AAT, which was observed in many GBM cases, thereby limiting the temporal benefits of AAT [29]. Previous magnetic resonance imaging (MRI) studies from our laboratory reported that employing the VEGFR2 inhibitor Vatalanib (PTK787) caused a significantly larger GBM tumor [30]. Vatalanib treatment induced hypoxia and was associated with the increased expression of several pro-angiogenic cytokines and chemokines such as VEGF, SDF-1, HIF-1α, FGF-1, FGF-2, Ephrin (Eph)-A1, Eph-A2, Angiopoietin-1, and their corresponding receptors VEGFR2, VEGFR3 and EGFR at the tumor-invading front of GBM [31]. Vatalanib, Cediranib and Sunitinib, and inhibitors of VEGFR, PDGFR, and c-kit receptors have offered limited therapeutic efficacy and transient benefits and have been associated with higher toxicities in clinical trials [32,33,34,35,36]. The research groups that propagated the idea of AAT did not account for the involvement of BMDCs in the formation of tumor neovasculature through vasculogenesis in GBM.
A recent study from our laboratory demonstrated that Vatalanib treatment increased the number of CD68+ myeloid cells as well as the CD133+, CD34+, and Tie2+ endothelial cell signatures in a novel chimeric mouse model of GBM. As a part of that study, we sought to explore the effects of AAT on the recruitment of BMDCs. We found that GFP+ chimeric bone marrow cells co-localized at the tumor periphery with VEGF, SDF-1α, and PDGF. In addition, our study found a significantly higher number of GFP+ bone marrow cells in close proximity to CD11b+ and F4/80+ cells [37]. In another study, we reported that the paradoxical growth of tumor following Vatalanib treatment was controlled by inhibiting the mobilization of BMDCs and disrupting the CXCR4-SDF-1α interaction using whole body irradiation and AMD3100, respectively [38]. BMDCs orchestrated the therapeutic resistance to AAT in a GBM model. The TME was identified as highly immunosuppressive and pro-angiogenic in GBM. Achyut et al. [39] demonstrated the therapeutic utility of CSF1R inhibitor in controlling AAT resistance in GBM models. The authors established an extracellular signal-regulated kinase (ERK)-mitogen-activated protein kinase (MAPK)-mediated upregulation of CXCL7 following AAT in a GBM model. By employing a CSF1R inhibitor (GW2580), the authors reported a decrease in the levels of CXCL7 thereby establishing a novel cytokine-dependent pathway for controlling AAT resistance in GBM. Further, the authors found that the NFκB pathway was central to the chemokine axis driving GBM growth and conditional deletion of the p65 subunit of the NFκB transcription factor inhibited GBM tumor growth in an immune competent model, suggesting a critical role of inflammation in GBM pathogenesis [40].
Bevacizumab, a humanized monoclonal antibody against VEGF-165, the predominant isoform of VEGF-A, was the first angiogenesis inhibitor to obtain Food and Drug Administration (FDA) approval in 2004 and was the first commercially successful antiangiogenic drug marketed [41]. Although the use of bevacizumab in combination with chemotherapy resulted in some dramatic tumor size reduction with prolonged progression-free survival [42], prolonged use led to deteriorated clinical outcome and development of therapeutic resistance [43,44]. Despite somewhat prolonged progression-free survival, treatment with lomustine plus bevacizumab failed to confer a survival advantage over treatment with lomustine alone in patients with progressive GBM in a European clinical trial [45]. The development of therapeutic resistance in GBM is primarily attributed to the activation of alternative pathways of neovascularization to counter the therapeutic insult [46,47]. Recently, Wang et al. summarized all the AAT clinical trial data and proposed the targeting of alternate pro-angiogenic mechanisms in GBM [5]. Moreover, all the clinical and preclinical data indicate that use of any class of AAT is not an ingenious choice for GBM therapy.

2.3. Challenges Due to Microenvironment-Driven Alternative Vascularization

The solid tumors have the capability to sustain and grow through complex networks of neovascularization. The tumor vasculature was believed to encompass angiogenesis process of endothelial sprouting and proliferation as a principal route of new blood vessel formation. Recent data suggest that tumors have several distinct mechanisms of neovascularization to drive tumor growth, such as vasculogenesis and vascular mimicry (VM) [48]. VM phenomenon was identified as one of the key tumor-inherent mechanisms to drive AAT resistance in GBM tumors [49,50,51]. VM is the uncanny ability of tumor cells to transdifferentiate into endothelial-like phenotypes and form neovascular structures to irrigate the hypoxic tumors to meet the nutritional and metabolic demands [52,53]. Tumor cells switch to adaptive mechanisms of neovascularization under a prolonged hypoxic environment at the tumor center to counter the selective pressure exerted upon the tumor cells induced by AAT. These mechanisms include upregulation of pro-angiogenic growth factors, chemokines, and cytokines necessary for sustaining the growth of tumor cells as well as for the recruitment of endothelial progenitor cells (EPCs), endothelial cells (ECs), angiogenic myeloid cells, and vasculogenic leukocyte cells [50]. Apart from the creation of a niche favorable for the growth of the tumor, a myriad of invasive and metastatic gene signatures are also initiated in the tumor and the tumor-associated stromal cells to ensure sustainable conditions for tumor survival during AAT [54,55]. The growing tumor resorts to the development of novel tumor-dependent pathological neovascularization mechanisms for vascular perfusion independent of the endothelial and vasculogenic systems as the dogma states [47,56].
Emerging literature supports that GBM stem-like cells (GSCs) are instrumental in GBM microenvironment [57,58,59,60]. During VM, transdifferentiated GSCs acquire endothelial characteristics and form a pool of cells that initiate and sustain VM. The trans-differentiation of tumor cells into endothelial-like cells, which were CD45− CD31+ CD34+ (termed tumor-derived endothelial cells, TDECs) [61] and GSCs that were CD133+CD144+ [62] initiated and promoted VM in GBM models. GSCs also transdifferentiate into pericytes and targeting GSCs enhanced chemotherapeutic efficacy through disrupting the blood-tumor barrier, indicating a critical role of GSCs in establishing alternative vasculature in growing GBM [57]. VM in gliomas positively correlates with high grades of tumor malignancy and invasiveness is associated with extremely poor prognosis [63,64]. In light of these numerous reports and evidence, delineating the mechanisms of VM to better understand this new facet of AAT resistance in GBM is the need of the hour. Therefore, it becomes imperative for us to look into not just the tumor-associated stromal contribution but also tumor cell-dependent heterogeneous mechanisms that confer GBM a paradoxical growth and sustenance advantage along with the refractoriness and relapse associated with the development of AAT resistance.

2.4. Challenges Due to an Immune Suppressive Microenvironment Following Standard Therapy

GBM is an invasive neoplasm with a median survival of three months if untreated [2,65] and approximately only 15 months with standard therapies [66]. GBM’s ability to severely invade and infiltrate normal surrounding tissue oftentimes in integral areas of the brain, including areas that control speech and motor functions make comprehensive resection impossible. Infiltrating tumor cells invariably remain within the surrounding brain, and prompt to later disease progression or recurrence. Tumors smaller than 5–6 cm and those that do not cross the mid-line, and supratentorial (cerebrum) and cerebellar tumors (persuadable to surgical resection) have been associated with favorable outcomes [67,68]. Current standard therapy for GBMs encompasses maximally secured surgical resection followed by concomitant RT and TMZ chemotherapy [69,70].
Until 2005, postoperative RT alone was the prevailing treatment. Stupp et al. in 2005 [66] showed that RT plus concomitant TMZ chemotherapy, an oral second-generation imidazotetrazine derivative or a DNA alkylating agent that exerted its cytotoxic effects by methylation of specific DNA sites, was more effective than RT alone for improving overall survival of patients with GBM tumors. TMZ is the most successful drug that has added several months to the life expectancy of GBM patients. In some cases, despite aggressive treatment, tumors inevitably recur due to the infiltrative nature of GBM, resulting in poor overall survival [71,72,73,74]. Therapy outcomes are even poorer in elderly patients [75,76]. Reviews of molecular-targeted therapies for primary and recurrent GBM have indicated modest benefits in overall survival of 5 to 8 months [77].
Chronic inflammation and the release of cytokines following radiation and TMZ therapies cause a recurrence of GBM [78,79,80]. Recurrent GBMs have an increased infiltration of BMDCs in the tumor bulk and infiltrative regions after therapy [81]. The enhanced myeloid cell infiltration in the TME following chemotherapy was associated with the activation of the CSF1–CSF1R pathway [82,83]. After the TMZ chemotherapy, some tumors displayed aggressive immune suppressive features in the tumor [84,85]. Moreover, tumor cell-secreted factors provide a gradient that helps myeloid cell infiltration into the tumor. The infiltrated myeloid cells, in turn, are capable of impairing chemotherapeutic responses [86,87]. In the autonomous mechanism, several tumor p53 (TP53) mutations in the tumor cell compartment also contributed to tumor progression and resistance to TMZ [88,89]. In some cases, the survival advantage conferred by TMZ is associated with methylation of the promoter region of the gene encoding O6-methylguanine DNA-methyl transferase (MGMT) [90]. Presence of TP53 mutations in the tumor cell compartment may decrease the TMZ sensitivity by increasing MGMT expression [88]. Thus, one of the current challenges is to enhance the effectiveness of the available standard therapies against GBM.

2.5. Challenges Due to Molecular and Genetic Heterogeneity in GBM Tumors

GBM originates from astrocytes or astroglia that reside in the brain and spinal cord. Astrocytes make up the supportive tissue of the CNS. Initially, GBMs were thought to be derived solely from glial cells; however, several pieces of evidence suggest that GBMs usually contain a mix of cell types at multiple stages of differentiation with significant phenotypic variations [91]. GBM is primarily detected in the cerebral hemisphere of the brain but can be found anywhere in the brain or spinal cord. Recent genetic and molecular advances have contributed to a better understanding of GBM pathophysiology and disease subgroups. Although all GBMs are categorized as World Health Organization (WHO) grade IV astrocytoma, they display strong genetic discrepancy and tumor subtypes with genetic alterations.
Gene expression profiling of GBMs identified distinct transcriptional subgroups such as proneural, mesenchymal, neural, and classical [92,93]. In addition to the transcriptional subgrouping of GBMs, GBMs were grouped based on inactivation pattern of tumor suppressor genes and activation of oncogenes. In the 2016 WHO classification of CNS tumors, GBMs were divided into three main groups based on their shared genetic driver mutations in the gene encoding isocitrate dehydrogenase enzyme 1/2 (IDH1/2), which drives distinct growth pattern and behaviors. This granted a persuasive classification based on both phenotype and genotype: (1) IDH-wild-type GBMs (about 90% of cases), (2) IDH-mutant GBMs (about 10% of cases), and (3) not otherwise specified (NOS) GBMs, for those tumors for which full IDH evaluation cannot be performed [94,95,96,97]. A new variant of GBM (IDH-wild-type), called epithelioid GBM, has been added to the classification and has been found mostly in children and young adults [95,98,99].
In addition, several other molecular genetic alterations have been found in GBMs. Primary GBMs show overexpression of epidermal growth factor receptor (EGFR), mouse double-minute 2 (MDM2), mutations of phosphatase and tensin homolog gene (PTEN). High frequency of telomerase reverse transcriptase (hTERT) promoter and absence of IDH1 mutation have also been seen in primary GBMs. The hallmark of secondary GBMs is a mutation in the IDH1, TP53, and α thalassemia/mental retardation syndrome X-linked (ATRX) genes. Recently, the WHO added a rare and controversial subtype of GBM termed “with oligodendroglioma component” (GBM-O) that occurs in younger patients. This subtype often contains the TP53 and IDH1 mutation, lack of EGFR amplification, and lower frequency of PTEN deletions. In GBM-O group, observed genetic heterogeneity displayed longer survival compared to patients with another group of GBMs [1,100,101,102]. In GBM-O tumors, the combined loss of heterozygosity (LOH) on chromosomes 1 p and 19 q were correlated with classic oligodendroglioma morphology and was associated with IDH mutations, TP53 expression, and MGMT promoter methylation status [103,104,105]. Moreover, these reports indicate that GBM is highly heterogeneous in the context of genetic and molecular alterations. This heterogeneous nature of GBM may have a critical role in regulating therapeutic outcomes.

3. Potential Adjuvant Therapies against GBM

Initially, AAT targeting the VEGF–VEGFR pathway was proposed against GBM tumors due to their hypervascular nature. However, AAT resulted in leaky blood vessels, which enhanced hypoxia and activated alternate mechanisms through recruiting BMDCs and myeloid precursors to the tumor. Some reports including publications from our laboratory identified that AAT enhanced GBM growth after transient benefits [39,50]. These results indicate that targeting the TME through myeloid inhibitor and other immune therapies is a better option that could block alternative mechanisms and will provide a benefit in anti-tumor responses. Some of these potential therapies are under investigations at the preclinical level (e.g., anti-cytochrome P450 (CYP) 4A or HET0016), early phases of clinical trials (e.g., anti-CSF1R), and others are advancing to late phase (e.g., immune therapies). There are upcoming therapies against GSCs and pericytes, key mediators of TME. However, this article only focuses on immune therapies with the special interest in targeting protumor myeloid cells in GBM tumors. Immunotherapies including anti-myeloid treatments mainly rely on polarizing the protumor microenvironment into antitumor phenotypes without disrupting the microenvironment, which is prone to hypoxia and vasculature leakiness. Moreover, this section is dedicated to potential emerging approaches and therapeutic modalities for the combination therapy options with the available standard therapies against GBM (Figure 1).

3.1. Anti-Myeloid Therapies

There is a significant role of TME in the modulation of therapeutic responses [106,107]. The TME in GBM tumors is characterized by active immunosuppressive mechanisms. Previously, our laboratory reported that AAT induces marked hypoxia in the invasive tumor [31], which promoted accumulation of BMDCs in GBM models [39,86]. The myeloid subpopulations of BMDCs not only suppress immune responses but also enhance neovascularization and modulate cancer stem cells (CSCs) [108,109,110,111,112,113,114]. One class of the critical tumor-associated myeloid cells is the immunosuppressive myeloid-derived suppressor cells (MDSCs), which are abundant in the GBM. MDSCs inhibit T-cell-mediated anti-tumor immunity and contribute to therapy resistance [86,115,116,117]. There are several studies showing the role of tumor-associated myeloid cells (TAMCs) in resistance to a different class of adjuvant therapies [86]. Studies noticed that tumor refractoriness to AATs was mediated by immune suppressive myeloid cells [118,119,120]. The mechanistic study identified that T helper type 17 cells and MDSCs induce the expression of granulocyte colony-stimulating factor CSF (G-CSF) in the stromal compartment, which in turn attracts MDSCs to drive anti-VEGFA resistance [121]. Similarly, we also found that myeloid cells mediate escape from AAT in a preclinical chimeric mouse model of GBM [39]. Key chemokines, such as macrophage colony stimulating factor-1 (M-CSF/CSF1) and monocyte chemotactic protein-1 (MCP1/CCL2), are known to contribute to the recruitment of myeloid cells to the tumors due to the presence of CSF1R [116,122,123]. The CSF1R expression has been reported on MDSCs, tumor-associated macrophages (TAMs), and dendritic cells [124,125,126] and regulates survival, differentiation, and proliferation of monocytes and macrophages [127,128] and has a critical role in angiogenesis and tumor progression [129,130]. Studies indicated that TME-accumulated TAMs can be targeted through anti-CSF1R with short-term treatment protocol to inhibit GBM progression via inhibiting chemokines such as CXCL7 (human IL8 variant) in animal models [39,86,131]. The CSF1R blockade has been shown to reverse macrophage polarization, inhibited GBM progression [131], and improved efficacy of RT [125]. A recent study identified that although overall survival was significantly prolonged in response to long-term CSF-1R inhibition, a subset of tumors recurred in a GBM model [132]. Consequently, combining IGF-1R or PI3K blockade with continuous CSF-1R inhibition in recurrent tumors significantly prolonged overall survival [132]. Several anti-myeloid therapies are undergoing clinical trials [133]. Therefore, targeting of tumor-recruited myeloid cells is a creative choice to target GBMs.

3.2. Immune Therapies

Immune therapies are a powerful strategy that inhibits tumor growth through enhancing immune responses inhibit tumor growth [134,135]. Immune therapy has two components referred to as active and passive approaches. The active immune therapy approach uses peptide or cellular vaccine to enhance the Th1 responses. However, the passive immune therapy uses the adoptive transfer of effector immune cells to induce antitumor responses [135]. Recently, several immune therapies against malignant cancers have been approved by the US FDA against malignant cancers [136]. Surprisingly, this class of therapy also showed resistance characterized by lack of durable and sustained immune responses in tumors [137,138,139] and indicated the use of the combinatorial approach, discovery of next-generation immune therapies, and anti-inflammatory approaches. Since GBM harbors a significant immune component, several immune therapy clinical trials are underway such as monotherapy or combination therapies of immune checkpoint inhibitors, peptide and dendritic cell vaccines, and adoptive T cell therapy in new or recurrent GBM tumors. Recently, Huang et al. reviewed current advances and discussed ongoing potential trials involving GBM patients [140].

3.3. Anti-CYP4A Therapy

Several alternative therapies are possible to overcome AAT resistance. Our research group has been studying the role of N-hydroxy-N′-(4-butyl-2 methyl phenyl) formamidine (HET0016) as a selective inhibitor of 20-HETE synthesis. 20-HETE, an arachidonic acid metabolite, can be synthesized by enzymes from the CYP4A and CYP4F families and promotes tumor neovascularization [49,141], proliferation [142], migration [143,144] and regulation of EPCs [145]. Previously, we demonstrated that the overexpression of 20-HETE can increase the GBM tumor volume by 10 fold [141]. Moreover, we observed that HET0016 controls tumor growth and migration in a time-dependent manner and attenuates the resistance of AATs [143]. Recently, we observed that VM can drive AAT resistance in GBM and the HET0016 treatment reduced the incidence of VM in a human GBM animal model [49] by altering the tumor vascular kinetics and permeability [146]. HET0016 decreased the periodic acid Schiff (PAS)-positive VM structures both at the core and the periphery of tumors thereby opening new avenues to counter AAT resistance in GBM [49].
Several other studies have shown that resistance to AAT also has a strong contribution of immune cells [81,147,148,149]. In particular, myeloid cells recruited to the TME can acquire endothelial signatures (CD202b and CD34) after vatalanib treatment [39], increasing the tumoral angiogenic potential and the resistance to treatment. In glioma, AATs were associated with the increased myeloid cell infiltration and stem cell accumulation [120]. Our laboratory has recently shown that HET0016 decreased the granulocytic MDSC (gMDSC) population in the metastatic niche by inhibiting the polarization of the MDSCs to a granulocytic phenotype [144]. A novel flavonoid, FLA-16, normalized the tumor vasculature through inhibiting CYP4A pathways and improved survival. It was accompanied by the decreased secretion of 20-HETE, VEGF and transforming growth factor (TGF)-β molecules in stroma [150]. Altogether, CYP4A expression in TAMCs is crucial for tumor dependent macrophage phenotype shift, and its inhibition by HET0016 or FLA-16 decreased tumor-associated phenotypes in GBM [150,151]. Numerous mechanisms might be involved in the 20-HETE inhibition. We previously demonstrated that HET0016 decreased proliferation of 9L gliosarcoma cells by 55% by reducing the phosphorylation of protein kinases and growth factors such as ERK1/2, stress-activated protein kinase (SAPK), cJUN, epidermal growth factor (EGF) and platelet-derived growth factor receptor (PDGFR) [142]. Moreover, we observed that HET0016 decreases expression of VEGF via ERK1/2 [141], and MMP2 and MMP9 via PI3K/AKT pathways [144]. A schematic of the signaling pathway presented by Shankar et al. [143] from our research group summarizes the possible therapeutic actions of HET0016. Given the involvement of HET0016 in decreasing the gMDSC population and phenotype shift in macrophages from M2 to M1, it is interesting to consider the role of the CYP4A/20-HETE axis in the recruitment, proliferation, and polarization of the aforementioned pro-tumorigenic immune cells. Moreover, our observations strongly support the critical role of the CYP4A/20-HETE axis in mediating AAT resistance by favoring the acquisition of endothelial signatures by myeloid cells in the TME. This evidence encourages us to exploit the CYP4A/20-HETE axis as a potential target for future therapeutic interventions [152].

4. Conclusions and Future Directions

Therapy resistance is an emerging hallmark and daunting outcome for any adjuvant treatment in the clinic, and involves (1) targeting tumor cells through chemotherapies [86,153], (2) targeting endothelial cells through AATs [5,86,154], and (3) improving anti-tumor immunity through immune-therapies [138,139]. Most of the adjuvant treatments have witnessed resistance through the involvement of BMDCs and tumor-promoting myeloid cells [86,116]. Involvement of BMDCs in cancer therapy warrants a need for a careful therapeutic strategy that will not activate alternative signaling pathways of vasculogenesis via increased infiltration of myeloid cells to the tumor or tumor cell-derived VM in GBM [50,51,155]. Therapies targeted against the TME represent a promising approach for GBM therapy. Targeting the TME may have decreased likelihood of acquired resistance through mutations in the cellular microenvironment, as is frequently observed with tumor cell-targeted therapies. Since several therapies targeting the TME are undergoing clinical trials such as anti-CSF1R [133,140,156], the combination of TME-targeted therapy with available standard therapies is an ingenious choice at present to target new and recurrent GBMs. Some novel treatments that have given successful results and survival benefits in preclinical models of GBM, such as anti-CYP4A or HET0016, could be the perfect adjuvant therapy following standard therapies in GBM clinical trials in the near future [146]. Above all, GBM research laboratories are rare around the world and there is a great demand for GBM-dedicated laboratories to develop better preclinical in vivo models with the intact immune system to test novel therapies before they progress to clinical trials [157,158,159].

Acknowledgments

The authors acknowledge the American Cancer Society grant IRG-14-193-01 to Bhagelu R. Achyut and the Summerville-Georgia Cancer Center Collaborative Research Award, Cancer Biorepository Use Award, and National Institutes of Health grants R01CA160216 and R01CA172048 to Ali S. Arbab. The authors acknowledge Rhea-Beth Markowitz for correcting grammatical errors in the manuscript.

Author Contributions

Bhagelu R. Achyut searched the recent references, wrote anti-myeloid, immune therapy, future directions, compiled the writings of all authors, and finalized the manuscript draft; Mohammad H. Rashid searched the recent references and wrote the section on overarching challenges, the challenge of genetic and molecular heterogeneity, and challenges in standard therapy sections. Kartik Angara searched the recent references and wrote the section on tumor microenvironment and challenges of alternative vascularization in GBM; Thaiz F. Borin searched the recent references and wrote the section on anti-CYP4A therapy; Meenu Jain searched the recent references and wrote the section on challenges due to hypoxia and hyper vasculature; Ali S. Arbab and Ping-Chang Lin reviewed the manuscript, advised the correction, wrote the section on antiangiogenic resistance, and participated in manuscript revision.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

20-HETE20-Hydroxyeicosatetraenoic acid
CD202bAngiopoietin-1 receptor
AGPT2Angiopoietin-2
AATAntiangiogenic therapy
BMDCsBone marrow-derived cells
CSCCancer stem cell
CBTRUS Central Brain Tumor Registry of the United States
CNSCentral nervous system
CXCL7Chemokine (C-X-C motif) ligand 7
CXCL8Chemokine (C-X-C motif) ligand 7
CDClusters of differentiation
COL1A1Collagen, type 1, alpha 1
CSFColony-stimulating factor
CSF1RColony-stimulating factor receptor 1
CD45Common leukocyte antigen
CXCR4C-X-C Motif Chemokine Receptor 4
CYP4A and CYP4FCytochromes P450 family enzymes
DLLDelta-like ligand
ECsEndothelial cells
EPCsEndothelial progenitor cells
EphEphrin
EGFEpidermal growth factor
EGFREpidermal growth factor receptor
FGFFibroblast growth factor
GBMGlioblastoma
GBM-OGlioblastoma with oligodendroglioma component
GSCsGlioma stem cell-like cells
G-CIMPGlioma-CpG island methylator phenotype
G-CSFGranulocyte-colony stimulating factor
gMDSCGranulocytic myeloid-derived suppressor cells
GFPGreen fluorescent protein
CD34Hematopoietic progenitor cell antigen
HGFHepatocyte growth factor
hTERThuman telomerase reverse transcriptase
HIF1-αHypoxia-inducible factor- alpha
IGFInsulin-like growth factor
IL8Interleukin 8
IDHIsocitrate dehydrogenase
MMPsMatrix metalloproteinases
MAPKMitogen-activated protein kinase
MCP-1Monocyte chemoattractant protein 1
MDM2Mouse double minute 2 homolog
MDSCMyeloid-derived suppressor cells
NG2Neuron-glial antigen 2
HET0016N-Hydroxy-N′-(4-butyl-2-methylphenyl)-formamidine
NONitric oxide
NFκBNuclear factor-kappa beta
MGMTO-6-Methylguanine-DNA Methyltransferase
PASPeriodic Acid Schiff
PTENPhosphatase and tensin homolog
PI3KPhosphatidylinositide 3-kinase
PLGFPlacental growth factor
CD31/PECAM-1Platelet endothelial cell adhesion molecule
PDGFPlatelet-derived growth factor
PDGFRPlatelet-derived growth factor receptor
RTRadiotherapy
CD133Stem-cell marker (Prominin-1)
SDF-1αStromal-derived factor 1 alpha
TMZTemozolomide
TN-CTenascin C
TGFβTransforming growth factor-beta
TME Tumor microenvironment
TP53Tumor p53
TAMTumor-associated macrophage
TAMCsTumor-associated myeloid cells
TDECTumor-derived endothelial cells
US FDAUnited States Food and Drug Administration
VEGFVascular endothelial growth factor
VEGFRVascular endothelial growth factor receptor
VMVascular mimicry
WHOWorld Health Organization

References

  1. Thakkar, J.P.; Dolecek, T.A.; Horbinski, C.; Ostrom, Q.T.; Lightner, D.D.; Barnholtz-Sloan, J.S.; Villano, J.L. Epidemiologic and molecular prognostic review of glioblastoma. Cancer Epidemiol. Biomarkers Prev. 2014, 23, 1985–1996. [Google Scholar] [CrossRef] [PubMed]
  2. Ostrom, Q.T.; Gittleman, H.; Liao, P.; Vecchione-Koval, T.; Wolinsky, Y.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary brain and other central nervous system tumors diagnosed in the United States in 2010–2014. Neuro Oncol. 2017, 19 (Suppl. 5), v1–v88. [Google Scholar] [CrossRef] [PubMed]
  3. Oliver, L.; Olivier, C.; Marhuenda, F.B.; Campone, M.; Vallette, F.M. Hypoxia and the malignant glioma microenvironment: Regulation and implications for therapy. Curr. Mol. Pharmacol. 2009, 2, 263–284. [Google Scholar] [CrossRef] [PubMed]
  4. Brem, S.; Cotran, R.; Folkman, J. Tumor angiogenesis: A quantitative method for histologic grading. J. Natl. Cancer Inst. 1972, 48, 347–356. [Google Scholar] [PubMed]
  5. Wang, N.; Jain, R.K.; Batchelor, T.T. New Directions in Anti-Angiogenic Therapy for Glioblastoma. Neurotherapeutics 2017, 14, 321–332. [Google Scholar] [CrossRef] [PubMed]
  6. Hanash, S.; Schliekelman, M. Proteomic profiling of the tumor microenvironment: Recent insights and the search for biomarkers. Genome Med. 2014, 6, 12. [Google Scholar] [CrossRef] [PubMed]
  7. Charles, N.A.; Holland, E.C.; Gilbertson, R.; Glass, R.; Kettenmann, H. The brain tumor microenvironment. Glia 2011, 59, 1169–1180. [Google Scholar] [CrossRef] [PubMed]
  8. Koch, S.; Tugues, S.; Li, X.; Gualandi, L.; Claesson-Welsh, L. Signal transduction by vascular endothelial growth factor receptors. Biochem. J. 2011, 437, 169–183. [Google Scholar] [CrossRef] [PubMed]
  9. Du, R.; Lu, K.V.; Petritsch, C.; Liu, P.; Ganss, R.; Passegue, E.; Song, H.; Vandenberg, S.; Johnson, R.S.; Werb, Z.; et al. HIF1alpha induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell 2008, 13, 206–220. [Google Scholar] [CrossRef] [PubMed]
  10. Plate, K.H.; Breier, G.; Weich, H.A.; Risau, W. Vascular endothelial growth factor is a potential tumour angiogenesis factor in human gliomas in vivo. Nature 1992, 359, 845–848. [Google Scholar] [CrossRef] [PubMed]
  11. Jones, M.K.; Itani, R.M.; Wang, H.; Tomikawa, M.; Sarfeh, I.J.; Szabo, S.; Tarnawski, A.S. Activation of VEGF and Ras genes in gastric mucosa during angiogenic response to ethanol injury. Am. J. Physiol. 1999, 276, G1345–G1355. [Google Scholar] [CrossRef]
  12. Gomez-Manzano, C.; Fueyo, J.; Jiang, H.; Glass, T.L.; Lee, H.Y.; Hu, M.; Liu, J.L.; Jasti, S.L.; Liu, T.J.; Conrad, C.A.; et al. Mechanisms underlying PTEN regulation of vascular endothelial growth factor and angiogenesis. Ann. Neurol. 2003, 53, 109–117. [Google Scholar] [CrossRef] [PubMed]
  13. Takahashi, T.; Shibuya, M. The 230 kDa mature form of KDR/Flk-1 (VEGF receptor-2) activates the PLC-gamma pathway and partially induces mitotic signals in NIH3T3 fibroblasts. Oncogene 1997, 14, 2079–2089. [Google Scholar] [CrossRef] [PubMed]
  14. Saino, M.; Maruyama, T.; Sekiya, T.; Kayama, T.; Murakami, Y. Inhibition of angiogenesis in human glioma cell lines by antisense RNA from the soluble guanylate cyclase genes, GUCY1A3 and GUCY1B3. Oncol. Rep. 2004, 12, 47–52. [Google Scholar] [CrossRef] [PubMed]
  15. Tsai, J.C.; Goldman, C.K.; Gillespie, G.Y. Vascular endothelial growth factor in human glioma cell lines: Induced secretion by EGF, PDGF-BB, and bFGF. J. Neurosurg. 1995, 82, 864–873. [Google Scholar] [CrossRef] [PubMed]
  16. Kerbel, R.S. Tumor angiogenesis. N. Engl. J. Med. 2008, 358, 2039–2049. [Google Scholar] [CrossRef] [PubMed]
  17. Talasila, K.M.; Rosland, G.V.; Hagland, H.R.; Eskilsson, E.; Flones, I.H.; Fritah, S.; Azuaje, F.; Atai, N.; Harter, P.N.; Mittelbronn, M.; et al. The angiogenic switch leads to a metabolic shift in human glioblastoma. Neuro Oncol. 2017, 19, 383–393. [Google Scholar] [CrossRef] [PubMed]
  18. Folkman, J. Tumor angiogenesis: Therapeutic implications. N. Engl. J. Med. 1971, 285, 1182–1186. [Google Scholar] [PubMed]
  19. Folkman, J.; Shing, Y. Angiogenesis. J. Biol. Chem. 1992, 267, 10931–10934. [Google Scholar] [PubMed]
  20. Ferrara, N. VEGF and Intraocular Neovascularization: From Discovery to Therapy. Transl. Vis. Sci. Technol. 2016, 5, 10. [Google Scholar] [CrossRef] [PubMed]
  21. Folkman, J.; Long, D.M., Jr.; Becker, F.F. Growth and metastasis of tumor in organ culture. Cancer 1963, 16, 453–467. [Google Scholar] [CrossRef]
  22. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
  23. Mittal, K.; Ebos, J.; Rini, B. Angiogenesis and the tumor microenvironment: Vascular endothelial growth factor and beyond. Semin. Oncol. 2014, 41, 235–251. [Google Scholar] [CrossRef] [PubMed]
  24. Bruno, A.; Pagani, A.; Magnani, E.; Rossi, T.; Noonan, D.M.; Cantelmo, A.R.; Albini, A. Inflammatory angiogenesis and the tumor microenvironment as targets for cancer therapy and prevention. Cancer Treat. Res. 2014, 159, 401–426. [Google Scholar] [PubMed]
  25. Samples, J.; Willis, M.; Klauber-Demore, N. Targeting angiogenesis and the tumor microenvironment. Surg. Oncol. Clin. N. Am. 2013, 22, 629–639. [Google Scholar] [CrossRef] [PubMed]
  26. Saharinen, P.; Eklund, L.; Pulkki, K.; Bono, P.; Alitalo, K. VEGF and angiopoietin signaling in tumor angiogenesis and metastasis. Trends Mol. Med. 2011, 17, 347–362. [Google Scholar] [CrossRef] [PubMed]
  27. Rahman, R.; Smith, S.; Rahman, C.; Grundy, R. Antiangiogenic therapy and mechanisms of tumor resistance in malignant glioma. J. Oncol. 2010, 2010, 251231. [Google Scholar] [CrossRef] [PubMed]
  28. Dietrich, J.; Norden, A.D.; Wen, P.Y. Emerging antiangiogenic treatments for gliomas—Efficacy and safety issues. Curr. Opin. Neurol. 2008, 21, 736–744. [Google Scholar] [CrossRef] [PubMed]
  29. Miller, K.D.; Sweeney, C.J.; Sledge, G.W., Jr. Can tumor angiogenesis be inhibited without resistance? In Mechanisms of Angiogenesis; Matthias, C., Georg, B., Eds.; Birkhäuser Basel: Basel, Switzerland; pp. 95–112.
  30. Ali, M.M.; Kumar, S.; Shankar, A.; Varma, N.R.; Iskander, A.S.; Janic, B.; Chwang, W.B.; Jain, R.; Babajeni-Feremi, A.; Borin, T.F.; et al. Effects of tyrosine kinase inhibitors and CXCR4 antagonist on tumor growth and angiogenesis in rat glioma model: MRI and protein analysis study. Transl. Oncol. 2013, 6, 660–669. [Google Scholar] [CrossRef] [PubMed]
  31. Ali, M.M.; Janic, B.; Babajani-Feremi, A.; Varma, N.R.; Iskander, A.S.; Anagli, J.; Arbab, A.S. Changes in vascular permeability and expression of different angiogenic factors following anti-angiogenic treatment in rat glioma. PLoS ONE 2010, 5, e8727. [Google Scholar] [CrossRef] [PubMed]
  32. 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]
  33. Kamoun, W.S.; Ley, C.D.; Farrar, C.T.; Duyverman, A.M.; Lahdenranta, J.; Lacorre, D.A.; Batchelor, T.T.; di Tomaso, E.; Duda, D.G.; Munn, L.L.; et al. Edema control by cediranib, a vascular endothelial growth factor receptor-targeted kinase inhibitor, prolongs survival despite persistent brain tumor growth in mice. J. Clin. Oncol. 2009, 27, 2542–2552. [Google Scholar] [CrossRef] [PubMed]
  34. Neyns, B.; Sadones, J.; Chaskis, C.; Dujardin, M.; Everaert, H.; Lv, S.; Duerinck, J.; Tynninen, O.; Nupponen, N.; Michotte, A.; et al. Phase II study of sunitinib malate in patients with recurrent high-grade glioma. J. Neurooncol. 2011, 103, 491–501. [Google Scholar] [CrossRef] [PubMed]
  35. Reardon, D.A.; Vredenburgh, J.J.; Coan, A.; Desjardins, A.; Peters, K.B.; Gururangan, S.; Sathornsumetee, S.; Rich, J.N.; Herndon, J.E.; Friedman, H.S. Phase I study of sunitinib and irinotecan for patients with recurrent malignant glioma. J. Neurooncol. 2011, 105, 621–627. [Google Scholar] [CrossRef] [PubMed]
  36. Reardon, D.A.; Egorin, M.J.; Desjardins, A.; Vredenburgh, J.J.; Beumer, J.H.; Lagattuta, T.F.; Gururangan, S.; Herndon, J.E., 2nd; 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]
  37. Achyut, B.R.; Shankar, A.; Iskander, A.S.; Ara, R.; Knight, R.A.; Scicli, A.G.; Arbab, A.S. Chimeric Mouse model to track the migration of bone marrow derived cells in glioblastoma following anti-angiogenic treatments. Cancer Biol. Ther. 2016, 17, 280–290. [Google Scholar] [CrossRef] [PubMed]
  38. Shaaban, S.; Alsulami, M.; Arbab, S.A.; Ara, R.; Shankar, A.; Iskander, A.; Angara, K.; Jain, M.; Bagher-Ebadian, H.; Achyut, B.R.; et al. Targeting Bone Marrow to Potentiate the Anti-Tumor Effect of Tyrosine Kinase Inhibitor in Preclinical Rat Model of Human Glioblastoma. Int. J. Cancer Res. 2016, 12, 69–81. [Google Scholar] [PubMed]
  39. Achyut, B.R.; Shankar, A.; Iskander, A.S.; Ara, R.; Angara, K.; Zeng, P.; Knight, R.A.; Scicli, A.G.; Arbab, A.S. Bone marrow derived myeloid cells orchestrate antiangiogenic resistance in glioblastoma through coordinated molecular networks. Cancer Lett. 2015, 369, 416–426. [Google Scholar] [CrossRef] [PubMed]
  40. Achyut, B.R.; Angara, K.; Jain, M.; Borin, T.F.; Rashid, M.H.; Iskander, A.S.M.; Ara, R.; Kolhe, R.; Howard, S.; Venugopal, N.; et al. Canonical NFkappaB signaling in myeloid cells is required for the glioblastoma growth. Sci. Rep. 2017, 7, 13754. [Google Scholar] [CrossRef] [PubMed]
  41. Rosen, L.S. Clinical experience with angiogenesis signaling inhibitors: Focus on vascular endothelial growth factor (VEGF) blockers. Cancer Control 2002, 9 (Suppl. 2), 36–44. [Google Scholar] [CrossRef] [PubMed]
  42. Norden, A.D.; Young, G.S.; Setayesh, K.; Muzikansky, A.; Klufas, R.; Ross, G.L.; Ciampa, A.S.; Ebbeling, L.G.; Levy, B.; Drappatz, J.; et al. Bevacizumab for recurrent malignant gliomas: Efficacy, toxicity, and patterns of recurrence. Neurology 2008, 70, 779–787. [Google Scholar] [CrossRef] [PubMed]
  43. Kreisl, T.N.; Zhang, W.; Odia, Y.; Shih, J.H.; Butman, J.A.; Hammoud, D.; Iwamoto, F.M.; Sul, J.; Fine, H.A. A phase II trial of single-agent bevacizumab in patients with recurrent anaplastic glioma. Neuro Oncol. 2011, 13, 1143–1150. [Google Scholar] [CrossRef] [PubMed]
  44. Reardon, D.A.; Desjardins, A.; Peters, K.B.; Gururangan, S.; Sampson, J.H.; McLendon, R.E.; Herndon, J.E., 2nd; Bulusu, A.; Threatt, S.; Friedman, A.H.; et al. Phase II study of carboplatin, irinotecan, and bevacizumab for bevacizumab naive, recurrent glioblastoma. J. Neurooncol. 2012, 107, 155–164. [Google Scholar] [CrossRef] [PubMed]
  45. Wick, W.; Gorlia, T.; Bendszus, M.; Taphoorn, M.; Sahm, F.; Harting, I.; Brandes, A.A.; Taal, W.; Domont, J.; Idbaih, A.; et al. Lomustine and Bevacizumab in Progressive Glioblastoma. N. Engl. J. Med. 2017, 377, 1954–1963. [Google Scholar] [CrossRef] [PubMed]
  46. Kumar, S.; Arbab, A.S. Neovascularization in Glioblastoma: Current Pitfall in Anti-angiogenic therapy. Zhong Liu Za Zhi 2013, 1, 16–19. [Google Scholar] [PubMed]
  47. Hardee, M.E.; Zagzag, D. Mechanisms of glioma-associated neovascularization. Am. J. Pathol. 2012, 181, 1126–1141. [Google Scholar] [CrossRef] [PubMed]
  48. Carmeliet, P.; Jain, R.K. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat. Rev. Drug Discov. 2011, 10, 417–427. [Google Scholar] [CrossRef] [PubMed]
  49. Angara, K.; Rashid, M.H.; Shankar, A.; Ara, R.; Iskander, A.; Borin, T.F.; Jain, M.; Achyut, B.R.; Arbab, A.S. Vascular mimicry in glioblastoma following anti-angiogenic and anti-20-HETE therapies. Histol. Histopathol. 2017, 32, 917–928. [Google Scholar] [PubMed]
  50. Angara, K.; Borin, T.F.; Arbab, A.S. Vascular Mimicry: A Novel Neovascularization Mechanism Driving Anti-Angiogenic Therapy (AAT) Resistance in Glioblastoma. Transl. Oncol. 2017, 10, 650–660. [Google Scholar] [CrossRef] [PubMed]
  51. Arbab, A.S.; Jain, M.; Achyut, B.R. Vascular Mimicry: The Next Big Glioblastoma Target. Biochem. Physiol. 2015, 4, e410. [Google Scholar] [PubMed]
  52. Folberg, R.; Hendrix, M.J.; Maniotis, A.J. Vasculogenic mimicry and tumor angiogenesis. Am. J. Pathol. 2000, 156, 361–381. [Google Scholar] [CrossRef]
  53. Maniotis, A.J.; Folberg, R.; Hess, A.; Seftor, E.A.; Gardner, L.M.; Pe’er, J.; Trent, J.M.; Meltzer, P.S.; Hendrix, M.J. Vascular channel formation by human melanoma cells in vivo and in vitro: Vasculogenic mimicry. Am. J. Pathol. 1999, 155, 739–752. [Google Scholar] [CrossRef]
  54. Sullivan, J.P.; Nahed, B.V.; Madden, M.W.; Oliveira, S.M.; Springer, S.; Bhere, D.; Chi, A.S.; Wakimoto, H.; Rothenberg, S.M.; Sequist, L.V.; et al. Brain tumor cells in circulation are enriched for mesenchymal gene expression. Cancer Discov. 2014, 4, 1299–1309. [Google Scholar] [CrossRef] [PubMed]
  55. Ortensi, B.; Setti, M.; Osti, D.; Pelicci, G. Cancer stem cell contribution to glioblastoma invasiveness. Stem Cell Res. Ther. 2013, 4, 18. [Google Scholar] [CrossRef] [PubMed]
  56. Carmeliet, P.; Jain, R.K. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011, 473, 298–307. [Google Scholar] [CrossRef] [PubMed]
  57. Zhou, W.; Chen, C.; Shi, Y.; Wu, Q.; Gimple, R.C.; Fang, X.; Huang, Z.; Zhai, K.; Ke, S.Q.; Ping, Y.F.; et al. Targeting Glioma Stem Cell-Derived Pericytes Disrupts the Blood-Tumor Barrier and Improves Chemotherapeutic Efficacy. Cell Stem Cell 2017, 21, 591–603. [Google Scholar] [CrossRef] [PubMed]
  58. 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] [PubMed]
  59. Liebelt, B.D.; Shingu, T.; Zhou, X.; Ren, J.; Shin, S.A.; Hu, J. Glioma Stem Cells: Signaling, Microenvironment, and Therapy. Stem Cells Int. 2016, 2016, 7849890. [Google Scholar] [CrossRef] [PubMed]
  60. Audia, A.; Conroy, S.; Glass, R.; Bhat, K.P.L. The Impact of the Tumor Microenvironment on the Properties of Glioma Stem-Like Cells. Front. Oncol. 2017, 7, 143. [Google Scholar] [CrossRef] [PubMed]
  61. Soda, Y.; Marumoto, T.; Friedmann-Morvinski, D.; Soda, M.; Liu, F.; Michiue, H.; Pastorino, S.; Yang, M.; Hoffman, R.M.; Kesari, S.; et al. Transdifferentiation of glioblastoma cells into vascular endothelial cells. Proc. Natl. Acad. Sci. USA 2011, 108, 4274–4280. [Google Scholar] [CrossRef] [PubMed]
  62. Wang, R.; Chadalavada, K.; Wilshire, J.; Kowalik, U.; Hovinga, K.E.; Geber, A.; Fligelman, B.; Leversha, M.; Brennan, C.; Tabar, V. Glioblastoma stem-like cells give rise to tumour endothelium. Nature 2010, 468, 829–833. [Google Scholar] [CrossRef] [PubMed]
  63. Wang, S.Y.; Ke, Y.Q.; Lu, G.H.; Song, Z.H.; Yu, L.; Xiao, S.; Sun, X.L.; Jiang, X.D.; Yang, Z.L.; Hu, C.C. Vasculogenic mimicry is a prognostic factor for postoperative survival in patients with glioblastoma. J. Neurooncol. 2013, 112, 339–345. [Google Scholar] [CrossRef] [PubMed]
  64. Liu, X.M.; Zhang, Q.P.; Mu, Y.G.; Zhang, X.H.; Sai, K.; Pang, J.C.; Ng, H.K.; Chen, Z.P. Clinical significance of vasculogenic mimicry in human gliomas. J. Neurooncol. 2011, 105, 173–179. [Google Scholar] [CrossRef] [PubMed]
  65. Malmstrom, A.; Gronberg, B.H.; Marosi, C.; Stupp, R.; Frappaz, D.; Schultz, H.; Abacioglu, U.; Tavelin, B.; Lhermitte, B.; Hegi, M.E.; et al. Temozolomide versus standard 6-week radiotherapy versus hypofractionated radiotherapy in patients older than 60 years with glioblastoma: The Nordic randomised, phase 3 trial. Lancet Oncol. 2012, 13, 916–926. [Google Scholar] [CrossRef]
  66. Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef] [PubMed]
  67. Ellor, S.V.; Pagano-Young, T.A.; Avgeropoulos, N.G. Glioblastoma: Background, standard treatment paradigms, and supportive care considerations. J. Law Med. Ethics 2014, 42, 171–182. [Google Scholar] [CrossRef] [PubMed]
  68. Walid, M.S. Prognostic factors for long-term survival after glioblastoma. Perm. J. 2008, 12, 45–48. [Google Scholar] [CrossRef] [PubMed]
  69. Stupp, R.; Hegi, M.E.; Mason, W.P.; van den Bent, M.J.; Taphoorn, M.J.; Janzer, R.C.; Ludwin, S.K.; Allgeier, A.; Fisher, B.; Belanger, K.; et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009, 10, 459–466. [Google Scholar] [CrossRef]
  70. Van Meir, E.G.; Hadjipanayis, C.G.; Norden, A.D.; Shu, H.K.; Wen, P.Y.; Olson, J.J. Exciting new advances in neuro-oncology: The avenue to a cure for malignant glioma. CA Cancer J. Clin. 2010, 60, 166–193. [Google Scholar] [CrossRef] [PubMed]
  71. Woehrer, A.; Bauchet, L.; Barnholtz-Sloan, J.S. Glioblastoma survival: Has it improved? Evidence from population-based studies. Curr. Opin. Neurol. 2014, 27, 666–674. [Google Scholar] [CrossRef] [PubMed]
  72. Swartz, A.M.; Li, Q.J.; Sampson, J.H. Rindopepimut: A promising immunotherapeutic for the treatment of glioblastoma multiforme. Immunotherapy 2014, 6, 679–690. [Google Scholar] [CrossRef] [PubMed]
  73. Swartz, A.M.; Batich, K.A.; Fecci, P.E.; Sampson, J.H. Peptide vaccines for the treatment of glioblastoma. J. Neurooncol. 2014, 123, 433–440. [Google Scholar] [CrossRef] [PubMed]
  74. Venur, V.A.; Peereboom, D.M.; Ahluwalia, M.S. Current medical treatment of glioblastoma. Cancer Treat. Res. 2015, 163, 103–115. [Google Scholar] [PubMed]
  75. Hoffermann, M.; Bruckmann, L.; Kariem Mahdy, A.; Asslaber, M.; Payer, F.; von Campe, G. Treatment results and outcome in elderly patients with glioblastoma multiforme—A retrospective single institution analysis. Clin. Neurol. Neurosurg. 2015, 128, 60–69. [Google Scholar] [CrossRef] [PubMed]
  76. Tsang, D.S.; Khan, L.; Perry, J.R.; Soliman, H.; Sahgal, A.; Keith, J.L.; Mainprize, T.G.; Das, S.; Zhang, L.; Tsao, M.N. Survival Outcomes in Elderly Patients with Glioblastoma. Clin. Oncol. 2015, 27, 176–183. [Google Scholar] [CrossRef] [PubMed]
  77. Lau, D.; Magill, S.T.; Aghi, M.K. Molecularly targeted therapies for recurrent glioblastoma: Current and future targets. Neurosurg. Focus 2014, 37, E15. [Google Scholar] [CrossRef] [PubMed]
  78. Multhoff, G.; Radons, J. Radiation, Inflammation, and Immune Responses in Cancer. Front. Oncol. 2012, 2, 58. [Google Scholar] [CrossRef] [PubMed]
  79. Yeung, Y.T.; McDonald, K.L.; Grewal, T.; Munoz, L. Interleukins in glioblastoma pathophysiology: Implications for therapy. Br. J. Pharmacol. 2013, 168, 591–606. [Google Scholar] [CrossRef] [PubMed]
  80. Hardee, M.E.; Marciscano, A.E.; Medina-Ramirez, C.M.; Zagzag, D.; Narayana, A.; Lonning, S.M.; Barcellos-Hoff, M.H. Resistance of Glioblastoma-Initiating Cells to Radiation Mediated by the Tumor Microenvironment Can Be Abolished by Inhibiting Transforming Growth Factor-β. Cancer Res. 2012, 72, 4119–4129. [Google Scholar] [CrossRef] [PubMed]
  81. Lu-Emerson, C.; Snuderl, M.; Kirkpatrick, N.D.; Goveia, J.; Davidson, C.; Huang, Y.; Riedemann, L.; Taylor, J.; Ivy, P.; Duda, D.G.; et al. Increase in tumor-associated macrophages after antiangiogenic therapy is associated with poor survival among patients with recurrent glioblastoma. Neuro Oncol. 2013, 15, 1079–1087. [Google Scholar] [CrossRef] [PubMed]
  82. Allavena, P.; Garlanda, C.; Borrello, M.G.; Sica, A.; Mantovani, A. Pathways connecting inflammation and cancer. Curr. Opin. Genet. Dev. 2008, 18, 3–10. [Google Scholar] [CrossRef] [PubMed]
  83. DeNardo, D.G.; Brennan, D.J.; Rexhepaj, E.; Ruffell, B.; Shiao, S.L.; Madden, S.F.; Gallagher, W.M.; Wadhwani, N.; Keil, S.D.; Junaid, S.A.; et al. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov. 2011, 1, 54–67. [Google Scholar] [CrossRef] [PubMed]
  84. Fadul, C.E.; Fisher, J.L.; Gui, J.; Hampton, T.H.; Cote, A.L.; Ernstoff, M.S. Immune modulation effects of concomitant temozolomide and radiation therapy on peripheral blood mononuclear cells in patients with glioblastoma multiforme. Neuro Oncol. 2011, 13, 393–400. [Google Scholar] [CrossRef] [PubMed]
  85. Sengupta, S.; Marrinan, J.; Frishman, C.; Sampath, P. Impact of temozolomide on immune response during malignant glioma chemotherapy. Clin. Dev. Immunol. 2012, 2012, 831090. [Google Scholar] [CrossRef] [PubMed]
  86. Achyut, B.R.; Arbab, A.S. Myeloid cell signatures in tumor microenvironment predicts therapeutic response in cancer. OncoTargets Ther. 2016, 9, 1047–1055. [Google Scholar]
  87. Bruchard, M.; Mignot, G.; Derangere, V.; Chalmin, F.; Chevriaux, A.; Vegran, F.; Boireau, W.; Simon, B.; Ryffel, B.; Connat, J.L.; et al. Chemotherapy-triggered cathepsin B release in myeloid-derived suppressor cells activates the Nlrp3 inflammasome and promotes tumor growth. Nat. Med. 2013, 19, 57–64. [Google Scholar] [CrossRef] [PubMed]
  88. Wang, X.; Chen, J.X.; Liu, J.P.; You, C.; Liu, Y.H.; Mao, Q. Gain of function of mutant TP53 in glioblastoma: Prognosis and response to temozolomide. Ann. Surg. Oncol. 2014, 21, 1337–1344. [Google Scholar] [CrossRef] [PubMed]
  89. Brazdova, M.; Quante, T.; Togel, L.; Walter, K.; Loscher, C.; Tichy, V.; Cincarova, L.; Deppert, W.; Tolstonog, G.V. Modulation of gene expression in U251 glioblastoma cells by binding of mutant p53 R273H to intronic and intergenic sequences. Nucleic Acids Res. 2009, 37, 1486–1500. [Google Scholar] [CrossRef] [PubMed]
  90. Hegi, M.E.; Diserens, A.C.; Gorlia, T.; Hamou, M.F.; de Tribolet, N.; Weller, M.; Kros, J.M.; Hainfellner, J.A.; Mason, W.; Mariani, L.; et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N. Engl. J. Med. 2005, 352, 997–1003. [Google Scholar] [CrossRef] [PubMed]
  91. Davis, M.E. Glioblastoma: Overview of Disease and Treatment. Clin. J. Oncol. Nurs. 2016, 20, S2–S8. [Google Scholar] [CrossRef] [PubMed]
  92. Olar, A.; Aldape, K.D. Using the molecular classification of glioblastoma to inform personalized treatment. J. Pathol. 2014, 232, 165–177. [Google Scholar] [CrossRef] [PubMed]
  93. Verhaak, R.G.; Hoadley, K.A.; Purdom, E.; Wang, V.; Qi, Y.; Wilkerson, M.D.; Miller, C.R.; Ding, L.; Golub, T.; Mesirov, J.P.; et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010, 17, 98–110. [Google Scholar] [CrossRef] [PubMed]
  94. 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]
  95. Hoshide, R.; Jandial, R. 2016 World Health Organization Classification of Central Nervous System Tumors: An Era of Molecular Biology. World Neurosurg. 2016, 94, 561–562. [Google Scholar] [CrossRef] [PubMed]
  96. Yan, H.; Parsons, D.W.; Jin, G.; McLendon, R.; Rasheed, B.A.; Yuan, W.; Kos, I.; Batinic-Haberle, I.; Jones, S.; Riggins, G.J.; et al. IDH1 and IDH2 mutations in gliomas. N. Engl. J. Med. 2009, 360, 765–773. [Google Scholar] [CrossRef] [PubMed]
  97. Ohgaki, H.; Kleihues, P. The definition of primary and secondary glioblastoma. Clin. Cancer Res. 2013, 19, 764–772. [Google Scholar] [CrossRef] [PubMed]
  98. Broniscer, A.; Tatevossian, R.G.; Sabin, N.D.; Klimo, P., Jr.; Dalton, J.; Lee, R.; Gajjar, A.; Ellison, D.W. Clinical, radiological, histological and molecular characteristics of paediatric epithelioid glioblastoma. Neuropathol. Appl. Neurobiol. 2014, 40, 327–336. [Google Scholar] [CrossRef] [PubMed]
  99. Kleinschmidt-DeMasters, B.K.; Aisner, D.L.; Foreman, N.K. BRAF VE1 immunoreactivity patterns in epithelioid glioblastomas positive for BRAF V600E mutation. Am. J. Surg. Pathol. 2015, 39, 528–540. [Google Scholar] [CrossRef] [PubMed]
  100. Parsons, D.W.; Jones, S.; Zhang, X.; Lin, J.C.; Leary, R.J.; Angenendt, P.; Mankoo, P.; Carter, H.; Siu, I.M.; Gallia, G.L.; et al. An integrated genomic analysis of human glioblastoma multiforme. Science 2008, 321, 1807–1812. [Google Scholar] [CrossRef] [PubMed]
  101. Appin, C.L.; Gao, J.; Chisolm, C.; Torian, M.; Alexis, D.; Vincentelli, C.; Schniederjan, M.J.; Hadjipanayis, C.; Olson, J.J.; Hunter, S.; et al. Glioblastoma with oligodendroglioma component (GBM-O): Molecular genetic and clinical characteristics. Brain Pathol. 2013, 23, 454–461. [Google Scholar] [CrossRef] [PubMed]
  102. Hinrichs, B.H.; Newman, S.; Appin, C.L.; Dunn, W.; Cooper, L.; Pauly, R.; Kowalski, J.; Rossi, M.R.; Brat, D.J. Farewell to GBM-O: Genomic and transcriptomic profiling of glioblastoma with oligodendroglioma component reveals distinct molecular subgroups. Acta Neuropathol. Commun. 2016, 4, 4. [Google Scholar] [CrossRef] [PubMed]
  103. Labussiere, M.; Idbaih, A.; Wang, X.W.; Marie, Y.; Boisselier, B.; Falet, C.; Paris, S.; Laffaire, J.; Carpentier, C.; Criniere, E.; et al. All the 1p19q codeleted gliomas are mutated on IDH1 or IDH2. Neurology 2010, 74, 1886–1890. [Google Scholar] [CrossRef] [PubMed]
  104. Xiong, J.; Liu, Y.; Wang, Y.; Ke, R.H.; Mao, Y.; Ye, Z.R. Chromosome 1p/19q status combined with expression of p53 protein improves the diagnostic and prognostic evaluation of oligodendrogliomas. Chin. Med. J. (Engl.) 2010, 123, 3566–3573. [Google Scholar] [PubMed]
  105. Eoli, M.; Menghi, F.; Bruzzone, M.G.; De Simone, T.; Valletta, L.; Pollo, B.; Bissola, L.; Silvani, A.; Bianchessi, D.; D’Incerti, L.; et al. Methylation of O6-methylguanine DNA methyltransferase and loss of heterozygosity on 19q and/or 17p are overlapping features of secondary glioblastomas with prolonged survival. Clin. Cancer Res. 2007, 13, 2606–2613. [Google Scholar] [CrossRef] [PubMed]
  106. Achyut, B.R. Impact of Microenvironment in Therapy-Induced Neovascularization of Glioblastoma. Biochem. Physiol. 2013, 2, e121. [Google Scholar] [CrossRef]
  107. Achyut, B.R.; Yang, L. Transforming growth factor-beta in the gastrointestinal and hepatic tumor microenvironment. Gastroenterology 2011, 141, 1167–1178. [Google Scholar] [CrossRef] [PubMed]
  108. Lahmar, Q.; Keirsse, J.; Laoui, D.; Movahedi, K.; Van Overmeire, E.; Van Ginderachter, J.A. Tissue-resident versus monocyte-derived macrophages in the tumor microenvironment. Biochim. Biophys. Acta 2016, 1865, 23–34. [Google Scholar] [CrossRef] [PubMed]
  109. El Gazzar, M. microRNAs as potential regulators of myeloid-derived suppressor cell expansion. Innate Immun. 2014, 20, 227–238. [Google Scholar] [CrossRef] [PubMed]
  110. Nakasone, E.S.; Askautrud, H.A.; Kees, T.; Park, J.H.; Plaks, V.; Ewald, A.J.; Fein, M.; Rasch, M.G.; Tan, Y.X.; Qiu, J.; et al. Imaging tumor-stroma interactions during chemotherapy reveals contributions of the microenvironment to resistance. Cancer Cell 2012, 21, 488–503. [Google Scholar] [CrossRef] [PubMed]
  111. De Visser, K.E.; Eichten, A.; Coussens, L.M. Paradoxical roles of the immune system during cancer development. Nat. Rev. Cancer 2006, 6, 24–37. [Google Scholar] [CrossRef] [PubMed]
  112. Campbell, M.; Tonlaar, N.; Garwood, E.; Huo, D.; Moore, D.; Khramtsov, A.; Au, A.; Baehner, F.; Chen, Y.; Malaka, D.; et al. Proliferating macrophages associated with high grade, hormone receptor negative breast cancer and poor clinical outcome. Breast Cancer Res. Treat. 2011, 128, 703–711. [Google Scholar] [CrossRef] [PubMed]
  113. Jinushi, M.; Baghdadi, M.; Chiba, S.; Yoshiyama, H. Regulation of cancer stem cell activities by tumor-associated macrophages. Am. J. Cancer Res. 2012, 2, 529–539. [Google Scholar] [PubMed]
  114. Shostak, K.; Chariot, A. NF-κB, stem cells and breast cancer: The links get stronger. Breast Cancer Res. BCR 2011, 13, 214. [Google Scholar] [CrossRef] [PubMed]
  115. Andrade, J.; Ge, S.; Symbatyan, G.; Rosol, M.S.; Olch, A.J.; Crooks, G.M. Effects of sublethal irradiation on patterns of engraftment after murine bone marrow transplantation. Biol. Blood Marrow Transplant. 2011, 17, 608–619. [Google Scholar] [CrossRef] [PubMed]
  116. Achyut, B.R.; Arbab, A.S. Myeloid Derived Suppressor Cells: Fuel the Fire. Biochem. Physiol. 2014, 3, e123. [Google Scholar] [PubMed]
  117. Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Bronte, V. Coordinated regulation of myeloid cells by tumours. Nat. Rev. Immunol. 2012, 12, 253–268. [Google Scholar] [CrossRef] [PubMed]
  118. Shojaei, F.; Wu, X.; Qu, X.; Kowanetz, M.; Yu, L.; Tan, M.; Meng, Y.G.; Ferrara, N. G-CSF-initiated myeloid cell mobilization and angiogenesis mediate tumor refractoriness to anti-VEGF therapy in mouse models. Proc. Natl. Acad. Sci. USA 2009, 106, 6742–6747. [Google Scholar] [CrossRef] [PubMed]
  119. Shojaei, F.; Wu, X.; Malik, A.K.; Zhong, C.; Baldwin, M.E.; Schanz, S.; Fuh, G.; Gerber, H.P.; Ferrara, N. Tumor refractoriness to anti-VEGF treatment is mediated by CD11b+Gr1+ myeloid cells. Nat. Biotechnol. 2007, 25, 911–920. [Google Scholar] [CrossRef] [PubMed]
  120. Piao, Y.; Liang, J.; Holmes, L.; Zurita, A.J.; Henry, V.; Heymach, J.V.; de Groot, J.F. Glioblastoma resistance to anti-VEGF therapy is associated with myeloid cell infiltration, stem cell accumulation, and a mesenchymal phenotype. Neuro Oncol. 2012, 14, 1379–1392. [Google Scholar] [CrossRef] [PubMed]
  121. Chung, A.S.; Wu, X.; Zhuang, G.; Ngu, H.; Kasman, I.; Zhang, J.; Vernes, J.M.; Jiang, Z.; Meng, Y.G.; Peale, F.V.; et al. An interleukin-17-mediated paracrine network promotes tumor resistance to anti-angiogenic therapy. Nat. Med. 2013, 19, 1114–1123. [Google Scholar] [CrossRef] [PubMed]
  122. Sawanobori, Y.; Ueha, S.; Kurachi, M.; Shimaoka, T.; Talmadge, J.E.; Abe, J.; Shono, Y.; Kitabatake, M.; Kakimi, K.; Mukaida, N.; et al. Chemokine-mediated rapid turnover of myeloid-derived suppressor cells in tumor-bearing mice. Blood 2008, 111, 5457–5466. [Google Scholar] [CrossRef] [PubMed]
  123. Lin, E.Y.; Nguyen, A.V.; Russell, R.G.; Pollard, J.W. Colony-stimulating factor 1 promotes progression of mammary tumors to malignancy. J. Exp. Med. 2001, 193, 727–740. [Google Scholar] [CrossRef] [PubMed]
  124. Movahedi, K.; Guilliams, M.; Van den Bossche, J.; Van den Bergh, R.; Gysemans, C.; Beschin, A.; De Baetselier, P.; Van Ginderachter, J.A. Identification of discrete tumor-induced myeloid-derived suppressor cell subpopulations with distinct T cell-suppressive activity. Blood 2008, 111, 4233–4244. [Google Scholar] [CrossRef] [PubMed]
  125. Xu, J.; Escamilla, J.; Mok, S.; David, J.; Priceman, S.; West, B.; Bollag, G.; McBride, W.; Wu, L. CSF1R signaling blockade stanches tumor-infiltrating myeloid cells and improves the efficacy of radiotherapy in prostate cancer. Cancer Res. 2013, 73, 2782–2794. [Google Scholar] [CrossRef] [PubMed]
  126. MacDonald, K.P.; Rowe, V.; Bofinger, H.M.; Thomas, R.; Sasmono, T.; Hume, D.A.; Hill, G.R. The colony-stimulating factor 1 receptor is expressed on dendritic cells during differentiation and regulates their expansion. J. Immunol. 2005, 175, 1399–1405. [Google Scholar] [CrossRef] [PubMed]
  127. Hamilton, J.A. Colony-stimulating factors in inflammation and autoimmunity. Nat. Rev. Immunol. 2008, 8, 533–544. [Google Scholar] [CrossRef] [PubMed]
  128. Hume, D.A.; MacDonald, K.P. Therapeutic applications of macrophage colony-stimulating factor-1 (CSF-1) and antagonists of CSF-1 receptor (CSF-1R) signaling. Blood 2012, 119, 1810–1820. [Google Scholar] [CrossRef] [PubMed]
  129. Priceman, S.J.; Sung, J.L.; Shaposhnik, Z.; Burton, J.B.; Torres-Collado, A.X.; Moughon, D.L.; Johnson, M.; Lusis, A.J.; Cohen, D.A.; Iruela-Arispe, M.L.; et al. Targeting distinct tumor-infiltrating myeloid cells by inhibiting CSF-1 receptor: Combating tumor evasion of antiangiogenic therapy. Blood 2010, 115, 1461–1471. [Google Scholar] [CrossRef] [PubMed]
  130. Zhu, Y.; Knolhoff, B.L.; Meyer, M.A.; Nywening, T.M.; West, B.L.; Luo, J.; Wang-Gillam, A.; Goedegebuure, S.P.; Linehan, D.C.; DeNardo, D.G. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 2014, 74, 5057–5069. [Google Scholar] [CrossRef] [PubMed]
  131. Pyonteck, S.M.; Akkari, L.; Schuhmacher, A.J.; Bowman, R.L.; Sevenich, L.; Quail, D.F.; Olson, O.C.; Quick, M.L.; Huse, J.T.; Teijeiro, V.; et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat. Med. 2013, 19, 1264–1272. [Google Scholar] [CrossRef] [PubMed]
  132. Quail, D.F.; Bowman, R.L.; Akkari, L.; Quick, M.L.; Schuhmacher, A.J.; Huse, J.T.; Holland, E.C.; Sutton, J.C.; Joyce, J.A. The tumor microenvironment underlies acquired resistance to CSF-1R inhibition in gliomas. Science 2016, 352, aad3018. [Google Scholar] [CrossRef] [PubMed]
  133. Quail, D.F.; Joyce, J.A. Molecular Pathways: Deciphering Mechanisms of Resistance to Macrophage-Targeted Therapies. Clin. Cancer Res. 2017, 23, 876–884. [Google Scholar] [CrossRef] [PubMed]
  134. Couzin-Frankel, J. Breakthrough of the year 2013. Cancer immunotherapy. Science 2013, 342, 1432–1433. [Google Scholar] [CrossRef] [PubMed]
  135. Farkona, S.; Diamandis, E.P.; Blasutig, I.M. Cancer immunotherapy: The beginning of the end of cancer? BMC Med. 2016, 14, 73. [Google Scholar] [CrossRef] [PubMed]
  136. Liu, K.G.; Gupta, S.; Goel, S. Immunotherapy: Incorporation in the evolving paradigm of renal cancer management and future prospects. Oncotarget 2017, 8, 17313–17327. [Google Scholar] [CrossRef] [PubMed]
  137. Thallinger, C.; Fureder, T.; Preusser, M.; Heller, G.; Mullauer, L.; Holler, C.; Prosch, H.; Frank, N.; Swierzewski, R.; Berger, W.; et al. Review of cancer treatment with immune checkpoint inhibitors: Current concepts, expectations, limitations and pitfalls. Wien. Klin. Wochenschr. 2017, 1–7. [Google Scholar] [CrossRef] [PubMed]
  138. Restifo, N.P.; Smyth, M.J.; Snyder, A. Acquired resistance to immunotherapy and future challenges. Nat. Rev. Cancer 2016, 16, 121–126. [Google Scholar] [CrossRef] [PubMed]
  139. Zaretsky, J.M.; Garcia-Diaz, A.; Shin, D.S.; Escuin-Ordinas, H.; Hugo, W.; Hu-Lieskovan, S.; Torrejon, D.Y.; Abril-Rodriguez, G.; Sandoval, S.; Barthly, L.; et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N. Engl. J. Med. 2016, 375, 819–829. [Google Scholar] [CrossRef] [PubMed]
  140. Huang, B.; Zhang, H.; Gu, L.; Ye, B.; Jian, Z.; Stary, C.; Xiong, X. Advances in Immunotherapy for Glioblastoma Multiforme. J. Immunol. Res. 2017, 2017, 3597613. [Google Scholar] [CrossRef] [PubMed]
  141. Guo, A.M.; Sheng, J.; Scicli, G.M.; Arbab, A.S.; Lehman, N.L.; Edwards, P.A.; Falck, J.R.; Roman, R.J.; Scicli, A.G. Expression of CYP4A1 in U251 human glioma cell induces hyperproliferative phenotype in vitro and rapidly growing tumors in vivo. J. Pharmacol. Exp. Ther. 2008, 327, 10–19. [Google Scholar] [CrossRef] [PubMed]
  142. Guo, M.; Roman, R.J.; Fenstermacher, J.D.; Brown, S.L.; Falck, J.R.; Arbab, A.S.; Edwards, P.A.; Scicli, A.G. 9L gliosarcoma cell proliferation and tumor growth in rats are suppressed by N-hydroxy-N’-(4-butyl-2-methylphenol) formamidine (HET0016), a selective inhibitor of CYP4A. J. Pharmacol. Exp. Ther. 2006, 317, 97–108. [Google Scholar] [CrossRef] [PubMed]
  143. Shankar, A.; Borin, T.F.; Iskander, A.; Varma, N.R.; Achyut, B.R.; Jain, M.; Mikkelsen, T.; Guo, A.M.; Chwang, W.B.; Ewing, J.R.; et al. Combination of vatalanib and a 20-HETE synthesis inhibitor results in decreased tumor growth in an animal model of human glioma. OncoTargets Ther. 2016, 9, 1205–1219. [Google Scholar]
  144. Borin, T.F.; Shankar, A.; Angara, K.; Rashid, M.H.; Jain, M.; Iskander, A.; Ara, R.; Lebedyeva, I.; Korkaya, H.; Achyut, B.R.; et al. HET0016 decreases lung metastasis from breast cancer in immune-competent mouse model. PLoS ONE 2017, 12, e0178830. [Google Scholar] [CrossRef] [PubMed]
  145. Guo, A.M.; Janic, B.; Sheng, J.; Falck, J.R.; Roman, R.J.; Edwards, P.A.; Arbab, A.S.; Scicli, A.G. The cytochrome P450 4A/F-20-hydroxyeicosatetraenoic acid system: A regulator of endothelial precursor cells derived from human umbilical cord blood. J. Pharmacol. Exp. Ther. 2011, 338, 421–429. [Google Scholar] [CrossRef] [PubMed]
  146. Jain, M.; Gamage, N.H.; Alsulami, M.; Shankar, A.; Achyut, B.R.; Angara, K.; Rashid, M.H.; Iskander, A.; Borin, T.F.; Wenbo, Z.; et al. Intravenous Formulation of HET0016 Decreased Human Glioblastoma Growth and Implicated Survival Benefit in Rat Xenograft Models. Sci. Rep. 2017, 7, 41809. [Google Scholar] [CrossRef] [PubMed]
  147. Achyut, B.R.; Bader, D.A.; Robles, A.I.; Wangsa, D.; Harris, C.C.; Ried, T.; Yang, L. Inflammation-mediated genetic and epigenetic alterations drive cancer development in the neighboring epithelium upon stromal abrogation of TGF-beta signaling. PLoS Genet. 2013, 9, e1003251. [Google Scholar] [CrossRef] [PubMed]
  148. Dondossola, E.; Rangel, R.; Guzman-Rojas, L.; Barbu, E.M.; Hosoya, H.; St John, L.S.; Molldrem, J.J.; Corti, A.; Sidman, R.L.; Arap, W.; et al. CD13-positive bone marrow-derived myeloid cells promote angiogenesis, tumor growth, and metastasis. Proc. Natl. Acad. Sci. USA 2013, 110, 20717–20722. [Google Scholar] [CrossRef] [PubMed]
  149. Seton-Rogers, S. Tumour microenvironment: Means of resistance. Nat. Rev. Cancer 2013, 13, 607. [Google Scholar] [CrossRef] [PubMed]
  150. Wang, C.; Li, Y.; Chen, H.; Zhang, J.; Zhang, J.; Qin, T.; Duan, C.; Chen, X.; Liu, Y.; Zhou, X.; et al. Inhibition of CYP4A by a novel flavonoid FLA-16 prolongs survival and normalizes tumor vasculature in glioma. Cancer Lett. 2017, 402, 131–141. [Google Scholar] [CrossRef] [PubMed]
  151. Chen, X.W.; Yu, T.J.; Zhang, J.; Li, Y.; Chen, H.L.; Yang, G.F.; Yu, W.; Liu, Y.Z.; Liu, X.X.; Duan, C.F.; et al. CYP4A in tumor-associated macrophages promotes pre-metastatic niche formation and metastasis. Oncogene 2017, 36, 5045–5057. [Google Scholar] [CrossRef] [PubMed]
  152. Borin, T.F.A.K.; Rashid, M.H.; Achyut, B.R.; Arbab, A.S. Arachidonic Acid Metabolite as a Novel Therapeutic Target in Breast Cancer Metastasis. Int. J. Mol. Sci. 2017, 18, 2661. [Google Scholar] [CrossRef]
  153. Acharyya, S.; Oskarsson, T.; Vanharanta, S.; Malladi, S.; Kim, J.; Morris, P.G.; Manova-Todorova, K.; Leversha, M.; Hogg, N.; Seshan, V.E.; et al. A CXCL1 paracrine network links cancer chemoresistance and metastasis. Cell 2012, 150, 165–178. [Google Scholar] [CrossRef] [PubMed]
  154. Fakhrejahani, E.; Toi, M. Antiangiogenesis therapy for breast cancer: An update and perspectives from clinical trials. Jpn. J. Clin. Oncol. 2014, 44, 197–207. [Google Scholar] [CrossRef] [PubMed]
  155. Arbab, A.S. Activation of alternative pathways of angiogenesis and involvement of stem cells following anti-angiogenesis treatment in glioma. Histol. Histopathol. 2012, 27, 549–557. [Google Scholar] [PubMed]
  156. Polivka, J., Jr.; Polivka, J.; Holubec, L.; Kubikova, T.; Priban, V.; Hes, O.; Pivovarcikova, K.; Treskova, I. Advances in Experimental Targeted Therapy and Immunotherapy for Patients with Glioblastoma Multiforme. Anticancer Res. 2017, 37, 21–33. [Google Scholar] [CrossRef] [PubMed]
  157. Lenting, K.; Verhaak, R.; Ter Laan, M.; Wesseling, P.; Leenders, W. Glioma: Experimental models and reality. Acta Neuropathol. 2017, 133, 263–282. [Google Scholar] [CrossRef] [PubMed]
  158. Chen, L.; Zhang, Y.; Yang, J.; Hagan, J.P.; Li, M. Vertebrate animal models of glioma: Understanding the mechanisms and developing new therapies. Biochim. Biophys. Acta 2013, 1836, 158–165. [Google Scholar] [CrossRef] [PubMed]
  159. Ben-David, U.; Ha, G.; Tseng, Y.Y.; Greenwald, N.F.; Oh, C.; Shih, J.; McFarland, J.M.; Wong, B.; Boehm, J.S.; Beroukhim, R.; et al. Patient-derived xenografts undergo mouse-specific tumor evolution. Nat. Genet. 2017, 49, 1567–1575. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of GBM tumor microenvironment. Bone marrow-derived cells (BMDCs) are recruited to the tumor during the GBM growth. Recent data suggest that BMDCs recruitment was enhanced following therapies, e.g., anti-tumor chemotherapy or antiangiogenic therapy. The tumor-promoting myeloid cells are the subpopulations of recruited BMDCs in the microenvironment. Here, we propose the combination of anti-CSF1R, anti-CYP4A, or immune therapy with standard therapies to overcome the myeloid cell-mediated resistance in GBM.
Figure 1. Schematic representation of GBM tumor microenvironment. Bone marrow-derived cells (BMDCs) are recruited to the tumor during the GBM growth. Recent data suggest that BMDCs recruitment was enhanced following therapies, e.g., anti-tumor chemotherapy or antiangiogenic therapy. The tumor-promoting myeloid cells are the subpopulations of recruited BMDCs in the microenvironment. Here, we propose the combination of anti-CSF1R, anti-CYP4A, or immune therapy with standard therapies to overcome the myeloid cell-mediated resistance in GBM.
Ijms 18 02732 g001
Table 1. A list of critical protein molecules involved in the GBM microenvironment.
Table 1. A list of critical protein molecules involved in the GBM microenvironment.
Key ProteinFull NameCategoryKey Function(s)
ANGPT2Angiopoietin-2Growth factorTumor neovascularization, metastasis, and inflammation
COL1A1Collagen, type 1, alpha (α) 1Structural protein, part of connective tissueTumor neovascularization
CD31/PECAM-1Platelet endothelial cell adhesion moleculeEndothelial cell markerLeukocyte transmigration, neovascularization, and integrin activation
CD34Hematopoietic progenitor cell antigenHematopoietic stem cell markerAttachment of stem cells to bone marrow ECM, stromal cells, facilitates cell migration
CD45Protein tyrosine phosphatase, receptor type, C (also known as Common leukocyte antigen)Pan-leukocyte markerSignal transduction in hematopoiesis
CD133Prominin-1Stem-cell markerCancer stem cells with CD133 undergo self-renewal and differentiation
CD202bAngiopoietin-1 receptorEndothelial-cell markerPromotes neovascularization
CSFColony-stimulating factor 1/Macrophage colony-stimulating factorCytokineproliferation, differentiation, and survival of monocytes, macrophages, and bone marrow progenitor cells
CSF-1RColony-stimulating factor receptor-1Cytokine receptorCytokine receptor that facilitates the actions of CSF-1
CYP4A and CYP4FCytochromes P450 family of enzymesEnzymes involved in arachidonic acid metabolismProduction of 20-HETE, an eicosanoid metabolite that promotes neovascularization, migration, inflammation, and metastasis
CXCL7Chemokine (C-X-C motif) ligand 7Cytokinemitogenesis, synthesis of extracellular matrix, glucose metabolism and synthesis of plasminogen activator, recruitment of CXCR2+ myeloid cells
CXCL8 (IL-8)Chemokine (C-X-C motif) ligand 8 (Interleukin-8)ChemokineNeutrophil chemotactic factor, chemotaxis of other granulocytic cells and CXCR2+ myeloid cells, potent pro-neovasculogenic chemokine
EGFEpithelial Growth FatorGrowth factorCellular proliferation, differentiation, and survival
Eph A1 and A2Ephrin A1 and A2Receptor tyrosine kinaseEmbryonic development, post-natal angiogenesis, stem cell differentiation and migration
FGFFibroblast growth factorGrowth factorAngiogenesis, wound healing, embryonic development, and various endocrine signaling pathways, proliferation, and differentiation of various cell types
G-CSFGranulocyte-colony stimulating factorCytokineSurvival, proliferation, differentiation, and function of neutrophil precursors and mature neutrophils
HGFHepatocyte growth factorGrowth, motility and morphogenic factorEmbryonic organ development, specifically in myogenesis, in adult organ regeneration, and in wound healing, mediates pro-tumorigenic roles in growing tumors
HIF-1αHypoxia-inducible factor 1 αTranscription factorReleased in response to hypoxia, neovascularization, energy metabolism, cell survival, and tumor invasion
IGFInsulin-like growth factorGrowth factorPromotes growth and survival of tumor cells
MCP-1/CCL2Monocyte chemoattractant protein 1ChemokineRecruitment of several inflammatory monocytes, memory T cells, and dendritic cells to the tumor
MGMTO-6-methylguanine-DNA methyltransferaseEnzyme/proteinDNA Repair promotes resistance of tumor cells to chemotherapy (esp. Temozolomide (TMZ))
MMP-2 and 9Matrix Metalloproteinases-2 and 9Proteinase enzymesDegradation of extra-cellular matrix (ECM) proteins, promotes angiogenesis by ECM remodeling
NG2Neuron-glial antigen 2/Chondroitin sulfate proteoglycan 4Chondroitin sulfate proteoglycanTumor cell metastasis and invasion
PDGFPlatelet-derived growth factorGrowth factorPro-angiogenic molecule
PLGFPlacental growth factorGrowth factorPro-angiogenic molecule
SDF-1αStromal-derived factor 1 αChemokineChemotactic protein to facilitate recruitment of bone marrow-derived cells and endothelial progenitor cells
VEGFVascular endothelial growth factorGrowth factorPromotes neovascularisation by facilitating survival and development of endothelial cells and proliferation of endothelial progenitor cells
VEGFRVascular endothelial growth factor receptorReceptor tyrosine kinaseReceptor for VEGF to promote neovascularization
TN-CTenascin CGlycoproteinTumor cell proliferation and migration
20-HETE20-Hydroxyeicosatetraenoic acidEicosanoid metabolite of Arachidonic acidNeovascularization, tumor cell growth, proliferation, migration, and recruitment of angiogenic myeloid cells to tumor microenvironment

Share and Cite

MDPI and ACS Style

Arbab, A.S.; Rashid, M.H.; Angara, K.; Borin, T.F.; Lin, P.-C.; Jain, M.; Achyut, B.R. Major Challenges and Potential Microenvironment-Targeted Therapies in Glioblastoma. Int. J. Mol. Sci. 2017, 18, 2732. https://doi.org/10.3390/ijms18122732

AMA Style

Arbab AS, Rashid MH, Angara K, Borin TF, Lin P-C, Jain M, Achyut BR. Major Challenges and Potential Microenvironment-Targeted Therapies in Glioblastoma. International Journal of Molecular Sciences. 2017; 18(12):2732. https://doi.org/10.3390/ijms18122732

Chicago/Turabian Style

Arbab, Ali S., Mohammad H. Rashid, Kartik Angara, Thaiz F. Borin, Ping-Chang Lin, Meenu Jain, and Bhagelu R. Achyut. 2017. "Major Challenges and Potential Microenvironment-Targeted Therapies in Glioblastoma" International Journal of Molecular Sciences 18, no. 12: 2732. https://doi.org/10.3390/ijms18122732

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