Next Article in Journal
Gonad Ontogeny and Sex Differentiation in a Poeciliid, Gambusia holbrooki: Transition from a Bi- to a Mono-Lobed Organ
Previous Article in Journal
Comparison of the Impact between Classical and Novel Strains of Rabbit Haemorrhagic Disease on Wild Rabbit Populations in Spain
Previous Article in Special Issue
Understanding the Wnt Signaling Pathway in Acute Myeloid Leukemia Stem Cells: A Feasible Key against Relapses
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Wnt Signaling in Brain Tumors: A Challenging Therapeutic Target

by
Lorenzo Manfreda
1,2,†,
Elena Rampazzo
1,2,† and
Luca Persano
1,2,*
1
Department of Women and Children’s Health, University of Padova, Via Giustininani, 3, 35128 Padova, Italy
2
Pediatric Research Institute, Corso Stati Uniti, 4, 35127 Padova, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biology 2023, 12(5), 729; https://doi.org/10.3390/biology12050729
Submission received: 31 March 2023 / Revised: 12 May 2023 / Accepted: 15 May 2023 / Published: 16 May 2023
(This article belongs to the Special Issue Molecular Mechanisms behind the Wnt Signalling Pathways)

Abstract

:

Simple Summary

Wnt signaling is one of the main evolutionarily conserved developmental pathways needed to instruct embryo formation and maintenance of organ tissues. Decades of research also clearly recognize the fundamental role of Wnt signaling during carcinogenesis. Indeed, dysregulation of Wnt pathway components has been suggested as a relevant hallmark of several neoplastic malignancies. In this review, we summarize the main molecular mechanism through which the Wnt pathway exerts its intracellular effects, with a specific focus on brain development and brain tumors, and how Wnt interacts with the surrounding brain environment. In this context, we review the latest anti-cancer therapeutic approaches employed to specifically target Wnt signaling in cancer, and their potential application in the brain tumor context. Moreover, we discuss the additional efforts that will be needed to define the real clinical impact of Wnt modulation in different types of brain tumors and even how to overcome the unsolved concerns about the potential systemic effects of such therapeutic approaches.

Abstract

The involvement of Wnt signaling in normal tissue homeostasis and disease has been widely demonstrated over the last 20 years. In particular, dysregulation of Wnt pathway components has been suggested as a relevant hallmark of several neoplastic malignancies, playing a role in cancer onset, progression, and response to treatments. In this review, we summarize the current knowledge on the instructions provided by Wnt signaling during organogenesis and, particularly, brain development. Moreover, we recapitulate the most relevant mechanisms through which aberrant Wnt pathway activation may impact on brain tumorigenesis and brain tumor aggressiveness, with a particular focus on the mutual interdependency existing between Wnt signaling components and the brain tumor microenvironment. Finally, the latest anti-cancer therapeutic approaches employing the specific targeting of Wnt signaling are extensively reviewed and discussed. In conclusion, here we provide evidence that Wnt signaling, due to its pleiotropic involvement in several brain tumor features, may represent a relevant target in this context, although additional efforts will be needed to: (i) demonstrate the real clinical impact of Wnt inhibition in these tumors; (ii) overcome some still unsolved concerns about the potential systemic effects of such approaches; (iii) achieve efficient brain penetration.

1. Introduction

Vertebrate developing embryos are extremely complex entities. Every single embryo develops from one single cell through the instructions provided by peculiar signaling pathways able to guide cellular specification in both space and time. Most importantly, during adulthood, the same pathways are fundamental to maintain the structure and functionality of specific organs and, consequently, control the homeostasis of the entire organism. This means that during the entire life of a vertebrate organism, the disruption, imbalanced dosages, or altered activation patterns of these fundamental pathways can lead to the onset of various diseases, including cancer, which may occur at different life stages. Among the fundamental molecules responsible for the development of several organs and the maintenance of their homeostasis, the Wnt (Wingless-related MMTV integration site) family of secreted lipid-modified glycoproteins and its downstream effectors is key. In 1980, the Wnt cascade was identified as an early segmental patterning signal for the correct development of Drosophila larva [1] and a strong oncogene, able to induce breast cancer in mouse models [2]. Since then, Wnt signaling has emerged as a fundamental regulator of cell fate decisions and migration, organismal development, tissue homeostasis and, in general, cell proliferation and behavior [3,4]. Indeed, Wnt signaling serves as a crucial stimulus for the correct development of several organs and tissues, including the skin and its appendages [5], the heart [6], the liver [7], the intestinal epithelium [8], the kidney [9,10], the bones [11], as well as many others [12]. In particular, Wnt signaling plays a crucial role during different stages of brain development, from early neurogenesis to the differentiation of neural progenitors in the adult hippocampus [9,10]. Hence, abnormal regulation of these mechanisms may severely contribute to several diseases, including cancer.
It is now evident that Wnt signaling is linked with a variety of human diseases [13,14] spanning from brain pathologies [15,16] to bone [17], vascular [18], and genetic disorders [19], not forgetting cancer. As a matter of fact, a large number of previous studies indicate that Wnt signaling must be considered a crucial pathway during oncogenesis [4,20], tumor progression [21], and cancer resistance to treatments [22,23]. Moreover, based on the suggested increased Wnt signaling with aging [24], one may also correlate this observation to the substantial augmented risk of cancer onset in the older population. Familial adenomatous polyposis (FAP) is the typical example of a disease caused by dysregulation of the Wnt pathway. The causative role of Wnt signaling for FAP development was firstly identified in 1991 [25,26]. FAP patients usually develop hundreds of polyps at the level of the colon and rectum, with a high probability to progress to colorectal cancers. Wnt signaling dysregulation is also considered a hallmark of brain tumors [9], here playing a fundamental role in cell proliferation, phenotype, and modulation of their stem cell properties [27,28], as already reported for many other cancer types [21]. Based on this knowledge, the possible targeting of the Wnt/β-catenin signaling pathway has emerged as a promising therapeutic strategy for the treatment of various cancers [21]. Nevertheless, relatively little is known about the potential clinical impact of Wnt signaling inhibition in brain tumors [29].
In this review, we summarize the role played by Wnt signaling in normal brain and brain cancers. In particular, we focus on the determinant role played by Wnt during brain development and specification and in brain cancer physiology, with an important inset in the reciprocal stimulation (Wnt-dependent) occurring between cancer cells and their microenvironment. Moreover, we extensively report on and discuss the most promising pharmacological tools available for the inhibition of Wnt signaling in cancer and, possibly, in brain tumors, considering the key obstacles still present on the way to their clinical translation and administration to patients.

2. The Wnt Signaling Molecular Cascade

2.1. The Wnt Family Canonical Landscape

Wnt ligands comprise a family of 19 secreted hydrophobic glycoproteins that exert their function by binding to Frizzled (FZD) receptors and transmembrane low-density lipoprotein receptor-related protein 5/6 (LRP5/6) co-receptors located on the target cell surface [30] (Figure 1). The ligand–receptor interaction induces the dismantling of an intracellular destruction complex composed of the Adenomatous Polyposis Coli (APC) protein, the Axis Inhibition Protein (Axin), the Glycogen Synthase Kinase-3β (GSK-3β), and the serine/threonine (ser/thr) Casein Kinase 1α (CK1α). In particular, Wnt molecules, upon binding to FZD receptors, recruit the evolutionarily conserved protein Dishevelled (DVL) to the membrane, providing a site for Axin and GSK-3β to bind and phosphorylate LRP5/6, finally preventing β-catenin degradation. The main function of the destruction complex is to phosphorylate the β-catenin amino terminal ser/thr-rich sequence (Ser33 and 37), thus generating an E3-ubiquitin ligase β-TrCP recognition site that targets β-catenin to the proteasome for its degradation [31]. Conversely, concomitant FZD and LRP5/6 activation results in the recruitment of the β-catenin destruction complex to cadherin proteins located at the cell membrane and the formation of a membrane-associated puncta allowing β-catenin accumulation and its nuclear translocation [32] (Figure 1).
The cytoplasmic-nuclear shuttling of β-catenin is the most importantly regulated molecular mechanism which modulates the Wnt/β-catenin pathway activation. Indeed, additional tyrosine phosphorylation (Tyr142/Tyr654) of β-catenin probably dictates peculiar interactions with α-catenin and E-cadherin and results in its dissociation from cadherin-containing adherens junctions, cytoplasmic accumulation, and nuclear translocation to promote gene transcription [33]. Once released from the destruction complex, or adherens junctions, active β-catenin translocates into the nucleus to exert its function as a transcriptional co-activator by interacting with the T Cell Factor/Lymphoid Enhancer Factor (TCF/LEF) family of transcription factors [34,35]. The TCF/LEF family of genes (TCF7, TCF7L2, TCF7L1, and LEF1) encodes for specific transcription factors (TCF1, TCF4, TCF3, and LEF1, respectively) that bind to DNA through a SRY-Box Transcription Factor (SOX)-like high mobility group domain, which recognizes a specific DNA consensus around the core “CTTTG” sequence, known as the Wnt Response Element (WRE) [36] (Figure 1). Recent research has demonstrated that more than 85% of the β-catenin-dependent transcriptional effects directly depend on TCF/LEF transcription factors and that the genome-wide physical association of β-catenin with its specific consensus sequences is severely affected when TCF/LEF are mutated or knocked-down [37]. Despite the consolidated and pivotal role of the β-catenin-TCF/LEF complex, recent findings highlight the existence of other branches within the canonical Wnt/β-catenin signaling pathway that act independently from TCF/LEF transcription factors. In particular, this has been demonstrated in the physical and functional interaction between β-catenin and the Hypoxia-Inducible Factor-1α (HIF-1α) [38], the basic helix-loop-helix (bHLH) transcription factor essential for muscle differentiation MyoD [39], various SOX proteins [40], and many members of the Forkhead Box O (FOXO) family of transcription factors [41]. This intricate, and potentially redundant, set of β-catenin-containing protein complexes, able to interact with DNA and regulate the expression of target genes, confirms the involvement of Wnt signaling in several essential biological processes controlling cell behavior and tissue integrity. In particular, these have been described to heavily affect embryonic development [42], stem-cell maintenance and differentiation [12], bone regeneration [43], but also to support the onset of many diseases such as cancer [44], diabetes [45], and oxidative stress and inflammation [46,47].

2.2. The Non-Canonical Wnt Signaling

In addition to the previously described canonical signaling, the Wnt pathway may also take advantage of alternative non-canonical molecular mechanisms. The term non-canonical pathway refers to the Wnt-dependent but β-catenin-independent signaling pathways. The two main and well-characterized non-canonical Wnt pathways are the planar cell polarity (PCP) and the Wnt-Calcium (Wnt/Ca2+) (Figure 1).
Wnt/PCP signaling is an evolutionarily conserved pathway, both in vertebrates and invertebrates, whose function is to control polarized cell behavior, a process that involves the asymmetric distribution of a set of core proteins within the cell and the subsequent cell polarization across the tissue plane [48,49]. Genetic studies performed across different stages of Drosophila development identified a group of core PCP proteins: Frizzled (Fz), Van Gogh (Vang), Dishevelled (Dsh), Prickle (Pk), Diego (Dgo), and Flamingo (Fmi), which control the orientation of ommatidial clusters in the eye disc and of the bristles and hairs on the fly body [50]. In vertebrates, such as Zebrafish, PCP is fundamental for the convergent extension movement process and serves as a key determinant of the elongation of the anterior–posterior body axis [51]. In mammals, the PCP complex, composed of Frizzled receptors (FZD1-10), Van Gogh-like proteins (VANGL1-2), Dishevelled transducers (DVL), Prickle Planar Cell Polarity Protein (PRICKLE1-3) nuclear receptors, Inversin/Diversin proteins (INVS/ANKRD6), and the Cadherin EGF LAG seven-pass G-type receptors (CELSR1-3), has been shown to control a diverse array of cellular, developmental, and physiological processes whose disruption determines a great variety of developmental defects and prenatal abnormalities [52,53]. The DVL-dependent Wnt/PCP signals are transduced to the Ras Homolog Family Member A (RHOA) signaling cascade through the Formin proteins Dishevelled Associated Activator of Morphogenesis (DAAM1-2) [54,55,56,57] (Figure 1). Furthermore, the altered activation of PCP has been implicated in the progression of various cancers. Indeed, the PCP pathway is extremely sensitive not only to the expression levels of the above-mentioned core proteins, but also to how the core proteins interact both at the intracellular and intercellular level, thus inducing the correct establishment of the PCP cellular asymmetry or a randomized polarity [58]. In this context, a better understanding of how PCP signaling is transduced and finely molecularly regulated is, therefore, essential to harness this signaling pathway for therapeutic purposes.
In Wnt/Ca2+ signaling, the Wnt-FZD binding and, in particular, the binding of the Wnt-5a ligand to the FZD5 receptor and the Receptor Tyrosine Kinase Like Orphan Receptor (ROR1/2) family of co-receptors, leads to the activation of the DVL family of transducers (Figure 1). Through these intracellular effectors, FZD receptors activate the heterotrimeric GTP-Binding Proteins (GTP-BP), leading to the activation of Calcium/Calmodulin-dependent Kinase II (CamK II), Protein Kinase C (PKC), and Phospholipase C (PLC), together with a release of Ca2+ from the endoplasmic reticulum [54,57,59,60]. In particular, by recruiting different DVL transducers, FZD receptors can activate phosphodiesterase (PDE) and the smaller GTP-binding proteins, such as RHO and Cell Division Cycle 42 (CDC42), involved in cell cycle and cell migration [61]. The Wnt/Ca2+ pathway plays a fundamental role in early vertebrate development by regulating blastula cell fate, orchestrating morphogenetic movement during gastrulation [62,63], and finely tuning organogenesis, particularly in the nervous system, the hematopoietic compartment, and the cardiocirculatory system, together with many other ventral organs [64,65]. A pivotal and controversial role of the Wnt/Ca2+ pathway is still under investigation in the cancer context where its activation can act both as a proto-oncogene or a tumor suppressor, depending on the cell type and the specific expression of pathway receptors [66]. As an example, Wnt-5a stimulation is sufficient to induce melanoma cell metastasis by inducing epithelial to mesenchymal transition (EMT) through the PKC/Ca2+ cascade [67,68]. On the contrary, Wnt-5a was reported to act as a tumor suppressor in neuroblastoma [69] and colon cancer [70].

3. Wnt Signaling in Central Nervous System Development

In 2001, Kiecker and Niehrs demonstrated that an activity gradient of Wnt/β-catenin signaling acts as a transforming morphogen to pattern the Central Nervous System (CNS) in Xenopus. In particular, they found that the Wnt signaling gradient is higher in the posterior and lower in the anterior Xenopus body axis, thus properly specifying the anterior–posterior polarity of the neural plate during development [71] (Figure 2A). As soon as the neural plate is specified, the invagination process starts to generate the neural tube. The already mentioned Wnt/PCP non-canonical signaling has been demonstrated to play a key role in neural tube closure. Indeed, mouse-based genetic studies unraveled that specific mutations in the Wnt/PCP core proteins such as FZD4, Wnt-11, CELSRs, LRP6, VANGL2, and PRICKLE-1 result in neural tube defects (NTDs) due to failure of the neural tube closure [72]. In addition, the Wnt/β-catenin canonical pathway and its finely tuned regulations also contribute to neural tube formation. In this context, the LRP6-dependent Wnt/β-catenin signaling allows the posterior neuropore closure and elongation in both mice [73] and humans [73], by means of a β-catenin-mediated activation of the transcription factors Paired Box 3 (PAX3) and Caudal Type Homeobox 2 (CDX2) [74]. Based on this knowledge, the manipulation of both the canonical and non-canonical Wnt signaling activities could provide a relevant therapeutic target for NTDs.
The neural tube is made up of pluripotent precursor cells that proliferate, commit to a specific cell fate, and then migrate to their final destination to arrange the different layers of the CNS, including neuronal ganglia and nuclei, and the cerebral cortex. The Wnt/β-catenin pathway has been associated with both proliferation and specification of neural stem cells (NSCs) during CNS development, in cooperation with other vestigial developmental pathways such as Notch, Sonic Hedgehog (SHH), Bone morphogenetic protein (BMPs) and Fibroblast Growth Factor (FGF) signaling. Indeed, the dorsal–ventral/rostral–caudal gradients of these morphogens and their relative receptors, together with specific cell–cell contacts between NSCs, finely specify the differentiated cell types that compose the nervous system, including mature neurons, astrocytes, and oligodendrocytes [75]. The Wnt/β-catenin pathway maintains the stemness of NSCs by positively or negatively modulating the activity of the BMP pathway during embryonic development, depending on the peculiar microenvironment NSCs are exposed to during specific developmental stages [76,77]. Once the brain is fully developed, the pivotal regulatory role that Wnt signaling still maintains during adult neurogenesis has been demonstrated, by acting as a neuronal pro-differentiation signal. Indeed, several Wnt family members, including Wnt-3, have shown to be expressed by adult hippocampal astrocytes, thus stimulating neuroblast proliferation and instructing adult hippocampal neural progenitors to acquire a neuronal fate [78] (Figure 2B). In particular, Kuwabara and co-workers demonstrated that Neuronal Differentiation 1 (NeuroD1), a pro-neurogenic bHLH transcription factor, is a downstream effector of Wnt signaling, needed to induce an efficient neuronal differentiation [10]. In addition, the β-catenin/TCF complex directly induces the expression of Neurogenin 1 (NGN1), which participates in stimulating cortical neuronal differentiation [79].
Besides substantial evidence for the role of Wnt/β-catenin in controlling neurogenesis through the promotion of neuronal differentiation, some studies have demonstrated that this signaling can also stimulate neural progenitor cell proliferation, since the β-catenin-dependent signal induces the expansion of proliferating precursors in the sub-ventricular zone [80,81]. Therefore, it seems that Wnt/β-catenin may affect both proliferation and differentiation of neural precursors in the CNS, depending on the concomitant activation of other signaling cascades in restricted brain loci and developmental stages [75].
As suggested by the above considerations, it is clear that any dysregulation occurring at the level of both canonical and non-canonical Wnt signaling components may severely affect the fine equilibrium existing in the cellular composition of the CNS, eventually contributing to the onset, progression, and peculiar behavior of different brain tumors. Accordingly, in the following sections, the major known roles played by Wnt signaling in a series of brain cancers are described, thus providing the rationale for considering Wnt signaling as a relevant therapeutic target against brain malignancies.

4. Wnt Signaling in Brain Tumors

4.1. Unraveling the Complexity of Wnt Signaling in Glioblastoma

The involvement of the Wnt signaling pathway in brain tumors has been extensively reported, with its functionality found to be largely contingent upon the tumor subtype [9,82,83]. This pathway has been subjected to intensive investigation in the context of Glioblastoma Multiforme (GBM) [22], recognized as one of the most lethal and aggressive brain tumors [84]. Indeed, despite the implementation of highly aggressive therapeutic approaches, including surgical resection, radiotherapy, and chemotherapy, GBM patients display a median progression-free survival of 12–15 months, with only 3–5% of individuals surviving beyond 3 years [85,86,87]. Currently, recurrent GBM is nearly untreatable, as no targeted therapies have been authorized for its effective eradication. Accordingly, the low survival rate of GBM patients is primarily attributable to disease recurrence, which arises in nearly all patients after completion of the available standard treatments, due to their intrinsic resistance to any additional chemotherapy and radiotherapy cycle [88].
The role of Wnt signaling in the onset and progression of GBM has been extensively studied and characterized, resulting in complex and sometimes contradictory evidence [89,90,91,92,93]. According to recent studies, the contribution of the Wnt pathway to GBM features has been demonstrated to be highly heterogeneous, depending, to different extents, on the microenvironment, the experimental conditions, and the specific experimental models employed. As a result, it remains unclear whether Wnt has a positive or negative impact on GBM development, progression, and aggressiveness [89,94,95]. The clarification of this dichotomy is not obvious and requires careful consideration of multiple microenvironmental factors, starting from the pivotal role played by the intra-tumoral oxygen availability [96].

4.1.1. The Role of Microenvironmental Oxygen

As discussed in previous sections, Wnt pathway activation requires the participation of several, and sometimes redundant, molecular transducers, co-activators, repressors, transcription factors, etc., thus making its study very complex and challenging. In GBM, it is well-known that the binding of Wnt ligands to FZD receptors activates the canonical β-catenin-dependent signaling pathway, which then promotes the expression of target genes through the formation of a large molecular complex together with TCF/LEF transcription factors and CREB Binding Protein (CBP)/p300 transcriptional co-factors [3,28,30,97,98,99] (Figure 1). In normoxic conditions (i.e., environmental 20% oxygen), it has been demonstrated that canonical Wnt signaling activation enhances the expression of some EMT activators in GBM cells, including Zinc Finger E-Box Binding Homeobox 1 (ZEB1), Twist Family bHLH Transcription Factor 1 (TWIST1), and Snail Family Transcriptional Repressor 2 (SLUG), thus enhancing their migratory properties in vitro. In the same conditions, it has been reported that increased Wnt activation, dependent on several mechanisms, including both genetic and epigenetic factors, sustains Glioma Stem Cell (GSC) maintenance and function [99,100,101,102,103]. For example, amplification/gain of PLAG1 Like Zinc Finger 2 (PLAGL2) expression has been linked to the upregulation of FZD2-9 receptors, thus promoting Wnt pathway activation and contributing to GSC self-renewal and maintenance [104]. Moreover, Wnt-regulated Forkhead Box M1 (FOXM1) also potentiates GBM cell stemness by directly binding the promoter and thus activating the expression of the NSC transcription factor SOX2 [105].
Further confirming a prominent Wnt signaling activation in GBM cells, through large-scale whole genome approaches it has been demonstrated that they exhibit epigenetic-dependent decreased expression of several Wnt pathway inhibitors, such as Wnt Inhibitory Factor 1 (WIF1), Dickkopf inhibitors (DKKs), and Secreted Frizzled Related Protein 1 (SFRP1) [29,101,106,107,108]. Intriguingly, authors have shown that these tumor suppressors are epigenetically silenced by histone modification and DNA methylation in their promoter region and that histone deacetylase (HDAC) inhibition, but not azacytidine treatment, is sufficient to restore the expression of all three genes, with a clear impact on cell proliferation [100,109,110]. We should also emphasize that activated Wnt/β-catenin signaling has been associated with increased activity of O6-Methylguanine-DNA Methyltransferase (MGMT). Importantly, this enzyme serves as an efficient DNA repair mechanism for GBM cells, shielding them against the mutagenic impact of the alkylating agent temozolomide, the gold standard chemotherapeutic drug administered to patients during adjuvant therapy [23,111,112]. Accordingly, increased levels of the Wnt signaling inhibitor DKK-1 have been demonstrated to enhance the sensitivity of GBM cells to chemotherapy [113]. Finally, from a more translational point of view, Wnt/β-catenin activation has been generally associated with a decreased survival of GBM patients [114], underlining the importance of additional studies aimed at defining the precise contribution of canonical Wnt signaling components to GBM onset, progression, and relapse.
Data presented so far seem to delineate a quite shared consensus on the fundamental role played by the Wnt pathway in GBM, by promoting cell motility and invasion through increased EMT, sustaining the growth and maintenance of GSCs, and contributing to intrinsic chemo- and radio-resistance [115]. On the other hand, multiple studies performed in tightly controlled microenvironmental conditions (i.e., hypoxia) suggest that Wnt signaling activation may exert opposing effects in GBM. Indeed, several normal and pathological tissues, including the brain and GBM, are known to be exposed to reduced oxygen tensions [116,117,118,119,120]. In the case of GBM, these appear to act as a fundamental modulator of Wnt pathway-dependent effects. It has been demonstrated that Wnt pathway activation in the normal brain [121] or GBM may promote different cellular functions based on oxygen availability, with conflicting effects observed upon Wnt signaling stimulation in hypoxic or normoxic environments. In particular, we previously demonstrated that Wnt pathway activation under hypoxic conditions (depending on the presence of a functional HIF-1α protein) promotes a strong differentiation of GSC toward a neuronal phenotype through a NUMB Endocytic Adaptor (NUMB) protein-dependent Notch signaling impairment [122,123]. Based on this evidence, we recently proposed a molecular mechanism that accounts for a potential dual role of Wnt in either inducing differentiation or maintaining GSCs, depending on intra-tumoral hypoxia and TCF proteins availability. Indeed, members of the TCF/LEF family of transcription factors are extremely heterogeneous in structure and function, with their intracellular assortment able to potentially influence the behavior of neural cells, but also GBM cells, during stem cell maintenance and differentiation [124]. Accordingly, we demonstrated that the Wnt signaling-induced formation of a HIF-1α/TCF1/β-catenin complex activates a potent pro-neuronal transcriptional program in GBM cells, which is counteracted, in normoxia, by the accumulation of high-molecular-weight TCF4 isoforms, which act as transcriptional repressors and prevent the complex binding to DNA [125] (Figure 3).
Since we provided examples that Wnt signaling may serve both as a pro-cancerous and a pro-differentiation stimulus in GBM, a better comprehension of the complex molecular interactions contributing to Wnt signaling modulation is a mandatory goal to be achieved soon. This will allow us to increase our molecular knowledge of GBM biology and even identify relevant intracellular signaling nodes with potential therapeutic impact. Nevertheless, despite the reported dichotomous effect of Wnt signaling activation in GBM tumors, several studies still consider Wnt inhibition as a promising therapeutic strategy in these tumors.

4.1.2. Wnt Signaling-Dependent Remodeling of GBM Microenvironment

In the previous section, we described the intimate molecular dependency between oxygen availability and the modulation of Wnt pathway-dependent functions. However, it is important to recognize that this relationship is not one-sided, but rather bidirectional. In this paragraph, we aim to summarize the existing knowledge on how the Wnt pathway can affect tumor microenvironment (TME), with a focus on how GBM tumors, through activation of the Wnt pathway, can manipulate neighboring cell behavior and growth. GBM tumors evolve within an intricate and interdependent microcosm of diverse cellular components, such as immune cells, normal astrocytes, and blood vessels, as well as a dense extracellular matrix [126,127]. The TME composition has been shown to guide the fate and the phenotype of GBM cells, by sustaining proliferation, angiogenesis, invasion, and resistance to treatments [128,129,130,131,132,133,134,135]. Intriguingly, the Wnt pathway may be involved in some of these processes. A recent study elegantly demonstrated that GBM cells can stimulate endothelial cells (ECs) to transdifferentiate into mesenchymal stem-like cells, thus sustaining chemotherapy resistance [136]. This appears to be mediated, at least in part, by a Hepatocyte Growth Factor (HGF)/MET Proto-Oncogene Receptor Tyrosine Kinase (MET) signaling-dependent Wnt pathway activation, nuclear β-catenin accumulation, and Multidrug Resistance-associated Protein-1 (MRP-1) expression, eventually promoting EC stemness and chemoresistance (Figure 4). Accordingly, the pharmacological inhibition of Wnt signaling was shown to decrease MRP-1 expression in ECs and improve mouse survival, when combined with TMZ treatment. Intriguingly, this molecular loop not only influences EC chemoresistance, but also the response of GBM cells to chemotherapy [136]. These findings align with the observation that GBM cell-released Wnt-7a can stimulate vessel co-option, further reducing therapy response [133].
Recently, the Wnt signaling pathway has also emerged as a key player in the regulation of immune cell behavior in GBM tumors. The composition of immune cells infiltrating the GBM TME is highly variable during tumor progression. These include tumor-associated macrophages (TAMs), neutrophils, dendritic cells, plasmacytoid cells, lymphocytes, natural killer cells, mast cells, and a significant amount of microglia [137,138]. In recent years, accumulating evidence suggests that the Wnt pathway strongly influences tumor-associated microglia in GBM. Several studies have demonstrated that co-culture of GBM and microglia, or stimulation of microglial cells by GBM cell-conditioned medium, leads to Wnt pathway activation [139,140]. This enhances microglial cell proliferation and their maturation towards an M2 phenotype, significantly contributing to the onset of a pro-inflammatory and immunosuppressive environment, with a negative impact on patient prognosis. These effects seem to be mediated by the secretion of Wnt-3a [140] or other secreted proteins, such as Wnt-1 Induced Secreted Protein 1 (WISP1), from GBM cells [141] (Figure 4). In this context, Tao et al. have provided a clear explanation of the dual role played by WISP1 in promoting the growth of GBM cells, through both autocrine and paracrine mechanisms. Specifically, WISP1 supports the maintenance of GSCs through autocrine signaling by interacting with Integrin α6β1, which activates the AKT pathway, eventually promoting cell survival. Moreover, the paracrine interaction between WISP1 and tumor-associated macrophages (TAMs) enhances their maintenance and function, which in turn promotes GBM growth by sustaining the pro-inflammatory and immunosuppressive tumor niche. To support their hypothesis, the authors demonstrated that inhibiting the WISP1 signaling pathway, or targeting its upstream regulators, disrupts GSC maintenance, inhibits TAM survival, and effectively suppresses GBM growth. These findings suggest that targeting WISP1 signaling may be a potential therapeutic strategy for GBM treatment. [141,142].

4.2. Medulloblastoma: Focus on the Wnt Subgroup

Cerebellar medulloblastoma (MB) is a highly malignant (grade IV) and invasive brain tumor with a preferential manifestation in the pediatric age. MBs are classified into four molecular groups: Wnt-activated, Shh-activated, group 3, and group 4 MBs [143]. These subgroups were established from cluster analyses performed on genome, transcriptome, methylome, and microRNA profiling of MB tumor samples [144,145,146], consistently displaying a significant correlation with clinical data, histopathological features, and patient survival in both children and adults [144,147,148].
The best characterized is the Wnt subgroup, due to its very good long-term survival of patients, exceeding 90% [149]. Several molecular alterations have been described for this neoplasm, including isochromosome 17q (50% of cases), monosomy of chromosome 6, and Tumor Protein P53 (TP53), Patched 1 (PTCH1), and β-catenin (CTNNB1) gene mutations [150]. It is noteworthy that MB occurs in a considerable proportion (40%) of FAP patients [151], but APC mutations have also been found in a fraction (4.3%) of sporadic MB [152]. Moreover, CTNNB1 mutations, considered the main hallmark of Wnt-driven MB, are found in 86% of patients, clearly suggesting the molecular dependency of Wnt MB onset and progression on the canonical Wnt/β-catenin signaling [153].
In 2012, Gibson et al. generated a mouse model of Wnt-driven MB (brain lipid-binding protein (Blbp)-Cre; Ctnnb1+/lox(Ex3); Trp53flx/flx) in which a conditional stabilized allele of Ctnnb1 is targeted to neural progenitor cells of the lower rhombic lip [154]. These mice, expressing the activated Ctnnb1 transgene in a Trp53-deleted cellular context, develop classical MB tumors [155], clearly correlated with the human Wnt MB counterparts. Moreover, for the first time, they demonstrated that Wnt MB tumors very likely arise from the dorsal brainstem. In 2012, this model was further tuned by the addition of the PI3K catalytic-α polypeptide mutant allele (Pik3caE545K), previously identified in human MB [156], thus generating Blbp-Cre; Ctnnb1+/lox(Ex3); Trp53+/flx; Pik3caE545K mice, which develop Wnt-like MB tumors with 100% penetrance within 3 months [156]. These tools are fundamental to understanding the deep molecular mechanism underneath Wnt MB insurgence and to better specify the Wnt MB cell of origin. Moreover, from a therapeutic point of view, these could be considered invaluable tools for the in vivo study of possible chemotherapy de-escalation protocols, based on the extremely favorable prognosis displayed by these patients.

4.3. Other Brain Tumors

There is limited understanding of the impact of Wnt signaling on the development, growth, and aggressiveness of other brain tumors. However, previous research has explored the involvement of the Wnt pathway in meningiomas and pituitary adenomas.
Meningiomas (MG) are usually considered benign tumors originating at the level of brain meninges, more precisely from meningothelial arachnoid cells [157]. They are the most common CNS tumors with a generally good prognosis, depending on the localization and extension of the mass. To date, there is no approved drug therapy for the treatment of MG [158], even for the 20% of MG cases for which surgery is not curative. Several studies have shown that the Wnt pathway may play a significant role in MG, with genes involved in this signaling being differentially expressed between non-malignant leptomeningeal cells and malignant MGs [159]. In addition, Wrobel et al. reported the overexpression of various Wnt pathway-correlated genes, such as CTNNB1, Cyclin-Dependent Kinase 5 Regulatory Subunit 1 (CDK5R1), Cyclin D1 (CCND1), and Ectodermal-Neural Cortex 1 (ENC1) in atypical and anaplastic MGs, relative to benign tumors [160]. Similarly, the downregulation of specific micro-RNAs, predicted to target Wnt-related genes, has also been demonstrated in anaplastic compared to benign MGs [161]. Supporting these findings, various studies have demonstrated a heterogeneous expression of certain Wnt signaling components such as TCF3, SFRP3, SFRP1, Cadherin 1 (CDH1), and FZD7, when comparing atypical, anaplastic, and benign MGs [162,163]. Finally, recent studies have shown that the activation of the Wnt-FOXM1 axis, at both the genomic and epigenomic level, is associated with poor prognosis, highly mitotic phenotype, and increased aggressiveness in the most severe cases of MG [164,165].
Pituitary adenomas (PA), which are typically benign tumors originating from the pituitary gland [166], are the subject of an ongoing debate regarding the role of Wnt in contributing to their development. On one hand, it has been observed that increased Wnt signaling in pituitary progenitor/stem cells can lead to the formation of PAs in both humans and mice [167]. Additionally, overexpression of WNT4 has been linked to the over-activation of β-catenin-dependent and independent pathways, eventually increasing PA invasiveness [168,169,170]. On the other hand, some studies have not confirmed this difference in the expression of Wnt-related genes. Nonetheless, recent research has shown that decreased expression of SFRP2 is associated with the development of corticotrophic adenomas [171], while overexpression of Solute Carrier Family 20 Member 1 (SLC20A1), which may be linked to the Wnt pathway, has been associated with larger tumor size, invasive behavior, and tumor recurrence in somatotroph adenomas [172].

5. Wnt Signaling as a Therapeutic Target: Achievements and Challenges

In this section, we will try to provide a comprehensive description of the most promising approaches developed in the last years to achieve a therapeutic anti-cancer targeting of the Wnt/β-catenin pathway, and some examples of their application to treat brain tumors. In this context, it is worth investigating the potential employment of Wnt inhibition on certain brain cancers, particularly MB. Indeed, although aberrant Wnt signaling activation is the recognized oncogenic driver of Wnt MBs [145], its overexpression eventually results in a vascular dysfunction-induced weaker blood–brain barrier. This seems not to ease cancer cell invasion but may rather contribute to the relatively good outcome of these patients due to a chemo-sensitization mechanism and even a less invasive disease [173,174,175]. As a consequence, the inhibition of the Wnt pathway in these MB tumors may reduce chemotherapy penetration, with no trials initiated for Wnt targeting, but rather focused on treatment de-escalation [174]. Furthermore, inhibition of Wnt signaling in MB tumors belonging to other subgroups has been only proposed for very rare relapses occurring in good prognosis patients, with restricted verification in preclinical models [176]. Finally, in contrast to the above-described role of Wnt signaling over-activation as a driver of tumorigenesis, some studies have suggested that, in peculiar contexts, β-catenin overexpression may even result in anti-tumoral effects in MBs and GBM, by reducing proliferation and self-renewal of cells and prolonging the survival of MB murine models [122,125,177,178,179]. For all these reasons, the following applications of therapeutic Wnt signaling inhibition against brain tumors are nearly limited to gliomas, particularly GBM.
In general, Wnt signaling inhibitors may be classified into four quite broad groups: (1) non-steroidal anti-inflammatory drugs (NSAIDs), (2) vitamins, natural compounds, and their derivatives, (3) small molecule chemical inhibitors (with a direct or even indirect effect on Wnt signaling), and (4) antibodies against Wnt pathway components (Table 1).

5.1. NSAIDs

NSAIDs are drugs of common use and some of them, including aspirin, indomethacin, sulindac, and celecoxib, have been proposed for the treatment of different types of cancer. Besides their anti-inflammatory properties that may represent by themself an adjuvant strategy during cancer prevention and therapy [106], NSAIDs have been reported for a long time to specifically reduce Wnt/β-catenin activation in human cancer cells and animal models [181]. NSAIDs attenuate the synthesis of prostaglandin by inhibiting cyclooxygenase enzymes (COX-1 and COX-2), demonstrating a prominent anti-inflammatory effect and a relatively good brain distribution [269]. Interestingly, one of their proposed mechanisms of Wnt signaling interference is a COX-dependent modulation of prostaglandin levels, which in turn can affect β-catenin stability [28]. However, NSAIDs have also shown anti-cancer effects in COX activity-lacking cancer cells, thus revealing a COX-independent mechanism of action and a potential direct effect on peculiar Wnt signaling components [270]. In this context, aspirin and indomethacin have been reported to hamper β-catenin/TCF complex transcriptional activity [181], with a suggested onco-preventive action, through inhibiting the Wnt/β-catenin signaling pathway [180,182,184]. Aspirin treatment also reduced proliferation, invasiveness, and Wnt-signaling target genes in GBM cell lines [183]. A similar effect, including the reduction of nuclear β-catenin accumulation, was demonstrated for sulindac, when used to treat colon cancer [185,186]. Moreover, sulindac treatment was shown to induce differentiation of GSCs, although without suggesting a specific mechanism of action [187]. In line with these studies, the selective COX-2 inhibitor celecoxib was shown to inhibit Wnt signaling in colon cancer cell lines, by inducing the degradation of TCFs and hampering the expression of Wnt signaling target genes, independently from COX-2 activity [46,188,190]. In GBM, both diclofenac and celecoxib similarly inhibited cell proliferation and migration [189], supporting a phase II clinical trial for their combination with temozolomide, which unfortunately did not establish any benefit for patients [271].
From a clinical point of view, celecoxib is the only NSAID approved by the Food and Drug Administration (FDA) for the reduction in the number of colorectal polyps in FAP patients. However, despite apparent effectiveness, reports of COX-2 inhibitor-dependent cardiotoxicity now limit their use in FAP patients [272], supporting their more recent market withdrawal by the European Medicines Agency.

5.2. Vitamins, Natural Compounds, and Derivatives

Natural compounds are a major source of drugs with anti-cancer properties. Indeed, at least one-third of drugs FDA-approved for the treatment of cancer are composed of natural products or their direct derivatives [273]. Vitamins and natural compounds can influence very different molecular processes of Wnt signaling, from modulating the abundance of pathway components and their regulators to affecting Wnt-dependent transcriptional activity. In particular, although the mechanism by which Wnt/β-catenin signaling is inhibited by certain vitamins is not completely explained, it has been reported that vitamin-activated nuclear receptors may compete with TCFs for the binding with β-catenin, eventually hampering its transcriptional activity [274,275]. Moreover, vitamin A-derived retinoids, besides their recognized pro-differentiating effects in various cancers, have been suggested to interfere with β-catenin intracellular localization [276] and to induce Wnt signaling inhibitors such as Disabled-2 (Dab2) and Axin [191]. Similarly, vitamin D was shown to reduce Wnt signaling activity through DKK-1 and 4 induction in colon cancer [192]. In this context, morphogens such as Differentiation-Inducing Factors (DIFs) potently inhibit cancer cell proliferation, with DIF-1 and 3 being demonstrated to suppress Cyclin D1 expression by activating GSK-3β [193,194].
In addition to the above-mentioned pro-differentiating compounds, several other drugs of natural origin have shown promising Wnt signaling inhibition-dependent, anti-cancer (including GBM) effects, although their proposed mechanisms of action would benefit from further clarification, due to lack of specificity. Nevertheless, curcumin [195,197] and shikonin [196] displayed inhibitory effects on β-catenin activation. In glioma, trichostatin [199] and diallyl trisulfide [199] modulated the expression of Wnt signaling components LGR5 and LRP6, respectively. Both the extracts of the root Rhodiola crenulata [201] and resveratrol [200] have been shown to affect β-catenin localization. Quercetin disrupted β-catenin/TCF-dependent transcriptional activity in colon cancer [202] and heavily affected Wnt signaling by modulating peculiar miRNAs in GBM [203]. Wnt-dependent transcription was also blocked by the green tea-derived epigallocatechin-3-gallate (EGCG) through induction of peculiar transcriptional repressors [204] or by preventing β-catenin nuclear translocation [205]. Certain indirubin derivatives such as 6-bromoindirubin-oxime (BIO), -acetoxime (BIA), and indirubin-3′-oxime (IO) demonstrated a strong anti-invasive effect in GBM models through inhibition of GSK-3 [206]. Moreover, some derivatives from natural sources, including naringenin (NAR) and phloroglucinol (PGL) have been suggested to antagonize canonical Wnt signaling receptors [207].
However, due to a general lack of specificity and poor knowledge of their molecular mechanisms of action, many of these natural products have failed to proceed to the clinal phases of experimentation, with only a few of them, such as vitamins and resveratrol, being included in clinical trials, essentially based on their pro-differentiation activity on various cancers [28].

5.3. Small Molecule Inhibitors

Several small chemical inhibitors of the Wnt/β-catenin signaling have been identified/developed during recent years through in silico and in vitro high throughput screening (HTS) approaches. Nevertheless, before providing a representative description of these discovery process-derived compounds, at least a few drugs, characterized by a repurposed action against Wnt signaling and a reported anti-cancer effect in multiple GBM models, should be described. The anthelmintic compound niclosamide, in addition to a prominent anti-migratory action on human cells [208,209], displayed a consistent inhibitory effect on nuclear β-catenin accumulation, and was therapeutically effective in in vivo GBM models [208,277]. Similarly, another anthelminthic compound, pyrvinium pamoate, was demonstrated to significantly reduce self-renewal and proliferation of GSCs, in part through inhibition of the Wnt/β-catenin transcriptional activity [210], although this may represent only a secondary output, dependent on the modulation of other pathways [278]. Moreover, the antipsychotic drug quetiapine has been recently reported to induce oligodendroglial differentiation of GSCs through the reduction of GSK-3β phosphorylation [211]. As a final example, pioglitazone, an antidiabetic drug used to lower blood glucose levels in type 2 diabetes patients, was also shown to reduce β-catenin expression in patient-derived GBM cultures [212].
In addition to repurposed drugs, several other compounds with the ability to interfere with various Wnt pathway components have been identified and developed during the last 20 years. A brief description of these compounds, subgrouped according to their suggested mechanism of action, is reported here below.

5.3.1. Antagonists of β-Catenin/Transcriptional Co-Activators Interaction

A small series of Wnt signaling inhibitors, acting as β-catenin/TCF interaction disrupting agents, was first identified in 2004 by Lepourcelet et al. through a HTS approach of a large library of compounds [279]. However, these compounds were not further developed due to lack of selectivity. Then, the synthetic compound PNU 74654 was identified for its ability to antagonize β-catenin/TCF-4 binding [216], with recently reported biological activity against both colorectal and hepatocellular carcinomas [213,217], but not brain tumors. Through a similar strategy, 2,4-diamino-quinazoline was also identified as a β-catenin/TCF-4 inhibitor, with therapeutic effects in colorectal and gastric cancers [214,215].
ICG-001 exerts its Wnt signaling inhibitory properties by selectively binding the β-catenin transcriptional co-factor CBP, but not its homologous p300, thus only suppressing a CBP-dependent transcriptional program [219], with reported pro-differentiation activity against GSCs [221]. More recently, the ICG-001 derivative PRI-724 was developed as a second-generation β-catenin/CBP antagonist. PRI-724 is endowed with increased potency and selectivity [280], thus entering different phase I/II clinical trials for the treatment of both leukemia and solid tumors, thanks to its verified safety in preclinical studies [222]. Furthermore, PMED-1 was found to weaken the β-catenin/CBP interaction but with poorer activity, despite its highly structural homology with ICG-001 [218]. E7386 is considered the first orally available compound of this class of inhibitors, however little is known about its precise mechanism of action and relative biological activity [222]. Interestingly, ICG-001 was shown to interact with the β-catenin-associating portion of the CBP N-terminus, which also contains retinoic acid and vitamin-D-interacting sites, suggesting that these compounds may share identical mechanisms of action [28,220].
The more recent inhibitors of the β-catenin-containing transcriptional complex [13] are BC2059 and methyl 3-([(4-methylphenyl)sulfonyl]amino)benzoate (MSAB), which attenuate active β-catenin levels, eventually impacting on its transcriptional activity [221,223,224]; CGP049090, CWP232228, and LF3, all inhibiting the β-catenin/TCF4 complex by sabotaging their interaction [225,226,227]; and SAH-BCL9, developed to block the interaction of β-catenin with B cell lymphoma 9 (BCL9), a co-activator of β-catenin-mediated transcription, by directly interacting with β-catenin and dissociating the β-catenin/BCL9 complexes [228].

5.3.2. DVL Inhibitors

This class of compounds is designed to inhibit the capability of DVL to intracellularly transduce Wnt ligand-dependent FZD receptor activation. NSC668036, FJ9, and 3289-8625 have been reported to interact with the PZD protein–protein interaction domain of DVL and to block Wnt signaling activation in vivo in Xenopus embryos and lung cancer mouse models [229,230,231].

5.3.3. Axin Modulators

One of the first examples of compounds affecting Axin protein stability were the Intracellular Wnt Response (IWR) inhibitors. IWRs are targeted at blocking Axin destruction, thus favoring the suppression of Wnt signaling through the increase of β-catenin proteasomal degradation [232,234]. Comparable results, through Axin stabilization, were achieved by using SEN46 and the Tankyrase inhibitor XAV939 in GBM cells [233]. Tankyrase enzymes, belonging to the Poly(ADP-Ribose) Polymerase (PARP) family of transferases, are known to promote ubiquitin-dependent degradation of Axin proteins, thus enhancing Wnt signaling activation. Tankyrase inhibitors stabilize Axins, critically affecting Wnt pathway components [281]. XAV939 has been widely used for experimentally treating GBM models, displaying promising chemo- and radio-sensitizing effects [136,236]. Additional Tankyrase inhibitors, such as AZ1366, G007-LK, and NVP-TNKS656, have been used with similar therapeutic effects in several cancer types, with some of them also displaying efficacy in gliomas [235,237,238,239]. Despite being characterized by a promising Wnt signaling inhibitory action, since Tankyrase targeting may exert additional undesired effects on multiple intracellular pathways, such inhibitors have failed to proceed toward clinical investigation.

5.3.4. Inhibitors of Wnt Ligands Production (PORCN Inhibitors)

The HTS approach implemented by Chen and colleagues in 2009, besides identifying IWRs, uncovered a subset of Inhibitors of Wnt Production (IWPs) [232]. Wnt signaling inhibition mediated by these compounds is based on their ability to strongly reduce the levels of lipidated Wnt ligands by antagonizing O-acyltransferase Porcupine (PORCN) [232,282]. Indeed PORCN-dependent acylation of Wnt ligands eases their lipidation, favoring their secretion, the generation of proper extracellular ligand gradients, and, finally, their biological activity [282,283]. LGK974 was reported to hamper Wnt signaling through PORCN inhibition in several solid cancers, including both mammary and brain tumors, without displaying toxic effects [94,133,242,244]. ETC-159, WNT-C59, and GNF-6231 were identified, through various screening approaches and progressive chemical modifications, as more potent PORCN inhibitors than LGK974, able to dramatically inhibit tumor growth in colorectal, nasopharyngeal, and breast cancer models, respectively [240,241,243]. As for LGK974, no toxic effects of these compounds have been recognized. Interestingly, LGK974 was the first PORCN inhibitor to enter a clinical trial for the treatment of various solid cancers generally characterized by the over-activation of Wnt signaling [284]. Therefore, inhibitors of PORCN can be considered effective therapeutics against several cancers with known Wnt pathway over-activation.

5.3.5. Others

ROR receptors may participate in Wnt signaling by serving as co-receptors for FZDs and enhancing specific Wnt-5a/ROR/FDZ non-canonical intracellular signaling (Figure 1), which is involved in tumor cell proliferation and invasion/metastasis, particularly in bones [285,286,287]. KAN 0439834 is a specific small molecule ROR1 inhibitor which has been reported to retain cytotoxic effects against ROR1-expressing cancer cells [245]. In addition, ROR1 has been used as a promising target for the development of antibodies for Wnt signaling inhibition (see also the following paragraph).
Another promising agent is ONC201, generally indicated as a dopamine receptor D2 antagonist, which has been shown to reduce the expression of several Wnt pathway components, including peculiar Wnt ligands, receptors, and co-factors [247]. In addition, besides its potential effects against the Wnt signal, ONC201 is emerging as a promising TNF-related apoptosis-inducing ligand (TRAIL)-inducing compound in GBM, with reported effectiveness in preliminary small patient cohorts [288,289].
We and others previously suggested that certain HDAC inhibitors of the hydroxamate class, including suberoylanilide hydroxamic acid (SAHA, Vorinostat), could be indicated as Wnt signaling antagonists since they were able to deplete TCF4-dependent Wnt pathway activation in colon carcinoma cells [246] and to cause a general shutdown of the Wnt signaling cascade in GBM cells, finally impairing their proliferation and migration [248]. In the same context, additional epigenetic modulators such as demethylating agents (i.e., azacytidine) have been proposed to hamper Wnt signaling activation by inducing the re-expression of several Wnt pathway inhibitor genes, which are generally turned off due to promoter hypermethylation in gliomas and other tumors, as already stated in previous paragraphs [29,101,106,107,108,249,250].
Finally, small molecule inhibitors such as proteolysis-targeting chimeras (PROTACs) have recently attracted interest for their potential application against several cancers by targeting protein degradation. As an example, a novel PROTAC β-catenin has demonstrated efficacy in inhibiting Wnt signaling in colorectal cancer cell lines and patient-derived organoids [251]. These approaches may represent an interesting option for future drug development, although experimental validation is still required for a proper assessment of their efficacy and safety profiles.

5.4. Antibodies

Several therapeutic antibodies against Wnt signaling pathway components have been developed in recent years. Indeed, based on the relevance of ROR1 expression in human cancers, cirmtuzumab was developed as a humanized antibody inhibiting Wnt-5a-ROR-induced signaling, which then entered a phase I clinical trial for CLL patients [252]. Along this line, ROR1 CAR-T cells have also been developed, demonstrating high efficacy and safety in preclinical animal models [253]. In addition, antibody–drug conjugates (ADC) have also been recently developed for targeting ROR1, including VLS-101, which comprises a cirmtuzumab-linked anti-microtubule toxin such as monomethyl auristatin E [255], and NBE-002, an anti-ROR1 antibody carrying a novel anthracycline payload [254].
Additional antibodies have been mainly developed to trap Wnt ligands or target FZD receptors. Antibodies against Wnt-1, Wnt-2, Wnt-5a (pAb5a-5), and secreted frizzled receptor protein 2 (SFRP2) have all been demonstrated to induce prominent apoptosis and cell death in several solid tumor models [256,257,258,259,260]. Moreover, chimeric proteins composed of the FZD8 peptide fragments fused with the human FC domains (F8CRDhFc and Ipafricept) have been also designed and tested with promising efficacy and adequate tolerability [261,262]. In the group of antibodies targeting FZD receptors, vantictumab (OMP18R5), which targets several FZDs [263,264], the 90Y labeled OTSA101-DPTA-90Y monoclonal antibody (mAb) [267], TT641 polyclonal antibody [266], and MAb 92-13 [265], all designed to target FZD10, have shown promising therapeutic effects against several solid tumors. Moreover, the anti-R-Spondin 3 (RSPO3) mAb OMP-131R10 has demonstrated promising canonical Wnt signaling attenuation, by inducing FZD proteasomal degradation in non-cancerous models [268].
Despite increasingly promising results being reported for the use of these therapeutics against several cancers, we still have no data on the possible implementation of Wnt-targeting antibody-based therapies in the brain tumor context.

6. Additional Considerations and Perspectives

Although we provided evidence that emerging compounds targeting Wnt signaling (or its ancillary modulators), may represent an effective therapeutic strategy against several cancers, inhibition of Wnt signaling in brain tumors still remains a challenging deal, due to its recognized role in brain vascularization and blood–brain barrier (BBB) integrity. Indeed, Wnt-7 ligands produced by neural progenitors activate canonical Wnt signaling through FDZ binding, thus stimulating ECs [290,291]. In addition, the Wnt pathway is known to regulate the expression of pro-angiogenic factors such as Vascular Endothelial Growth Factor (VEGF) which, besides serving as a relevant target of anti-angiogenic therapies in several contexts, including GBM, may dramatically affect the normal physiological functions of brain vasculature [292]. Therefore, further studies will be needed in order to understand the impact of Wnt signaling inhibitors on GBM angiogenesis and normal brain microvascular network, since their modification could provoke undesired BBB alterations, with relevant consequences on its permeability to certain drugs.
It is mandatory to also consider the tight epigenetic regulation that acts upstream of the Wnt pathway function. In particular, it has been reported that hundreds of non-coding RNAs are able to regulate (positively or negatively) several Wnt signaling components. Although these have not been mentioned within the previous sections, an exhaustive summary of the main micro-RNAs and long non-coding (lnc)RNAs able to affect Wnt signaling was recently reported by Daisy Precilla et al. [293].
Collectively, the factors to examine and the problems to overcome are multiple and could be also related to the mechanism of action of the proposed drugs/therapeutics and the knowledge that Wnt signaling is fundamentally involved in the homeostasis of nearly all adult tissues. Indeed, there are few clinical trials verifying the use of Wnt inhibitors in brain tumors (summarized in Table 2; https://clinicaltrials.gov/, accessed on 21 April 2023), with most of them withdrawn early due to bone and gastrointestinal toxicity [294]. As an example, the great promises of vantictumab were not fulfilled when clinical trials stopped due to bone-related safety [295]. Indeed, the expression of multiple secreted Wnt ligands has been associated with the regulation of bone integrity, density, and mineralization [296].
Based on these considerations, the blockade of the Wnt system, which in normal tissues controls vital functions, is thus limited in its clinical applications. Consequently, it is necessary to identify additional pharmacological strategies to restrict Wnt inhibition exclusively, or mainly, at the level of cancer cells.
One proposed approach could be the specific targeting of defined Wnt branches, rather than the whole Wnt system. Indeed, as described above, the non-canonical signaling downstream of the Wnt/ROR cascade is more associated with cell migration and invasion. For this reason, besides potential concerns regarding its impact on the wound healing process, this pathway may be considered less toxic systemically, but nonetheless endowed with therapeutic efficacy [305]. In agreement with this view, preclinical and clinical trials investigating ROR1 as a drug target are emerging as relevant anti-cancer approaches. Adding further complexity, one may also keep in mind that Wnt ligands, through the interaction with multiple receptors, often result in redundant intracellular responses, sustained by non-shared molecular machineries. This should stimulate the research toward the setup of combined target strategies, with the promise to enhance the possibility to achieve an efficient pharmacological suppression of peculiar Wnt-dependent cellular functions. Accordingly, Wnt signaling inhibition may represent only one tile of a more complex multi-target therapeutic puzzle that, unfortunately, is still far from being solved. Nevertheless, although we provided evidence that a full inhibition of the Wnt-dependent signaling cascades should be viewed with caution due to their pleiotropic action, the recent advances made in the preclinical development of multiple potent Wnt signaling inhibitors should not be discarded a priori, but rather considered a fundamental basis for increasing the feasibility of clinically relevant Wnt inhibition to treat patients.

7. Conclusions

The Wnt pathway is a complex, multifaceted, and tightly regulated signaling pathway involved in vertebrate development and tissue homeostasis. The role of Wnt is also pivotal during adulthood, thus impacting on the functionality and structure of several organs. In this review, we focused on the role of Wnt in determining brain tumor onset, aggressiveness, and microenvironment. Moreover, we evidenced that Wnt signaling also depends on, and modulates, several other pathways to control cell differentiation and sensitization to therapies. In this context, the modulation of the Wnt signal achieved through specific compounds or additional epigenetic mechanisms must be finely tuned for its activation level and correct time of intervention during cancer patient treatment. Despite several clinical trials concentrating on Wnt pathway inhibition as a promising anti-cancer therapy, data collected so far suggest that targeting multiple signaling pathways, aberrantly activated in tumor cells, may be the only reliable strategy to include Wnt signaling inhibitors in the future treatment of multiple cancer types, with a clear anticipated benefit for patients.

Author Contributions

Writing—Original Draft Preparation, L.M., E.R. and L.P. Writing—Review and Editing, L.P. Supervision, L.P. All authors have read and agreed to the published version of the manuscript.

Funding

The Rally Foundation for Childhood Cancer Research (#20IN28 and #21IC16) and Cassa di Risparmio di Padova e Rovigo (CARIPARO) Foundation (Project no. 20/16 FCR). Funding sources were not involved in any process of manuscript writing and submission.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We are grateful to Valentina Serafin from University of Padova for sharing Premium access to BioRender.com.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Nusslein-Volhard, C.; Wieschaus, E. Mutations affecting segment number and polarity in Drosophila. Nature 1980, 287, 795–801. [Google Scholar] [CrossRef]
  2. Nusse, R.; Varmus, H.E. Many tumors induced by the mouse mammary tumor virus contain a provirus integrated in the same region of the host genome. Cell 1982, 31, 99–109. [Google Scholar] [CrossRef] [PubMed]
  3. Nusse, R. Wnt signaling. Cold Spring Harb. Perspect. Biol. 2012, 4, a011163. [Google Scholar] [CrossRef] [PubMed]
  4. Nusse, R.; Varmus, H. Three decades of Wnts: A personal perspective on how a scientific field developed. EMBO J. 2012, 31, 2670–2684. [Google Scholar] [CrossRef] [PubMed]
  5. Lim, X.; Nusse, R. Wnt signaling in skin development, homeostasis, and disease. Cold Spring Harb. Perspect. Biol. 2013, 5, a008029. [Google Scholar] [CrossRef]
  6. Li, D.; Sun, J.; Zhong, T.P. Wnt Signaling in Heart Development and Regeneration. Curr. Cardiol. Rep. 2022, 24, 1425–1438. [Google Scholar] [CrossRef]
  7. Russell, J.O.; Monga, S.P. Wnt/beta-Catenin Signaling in Liver Development, Homeostasis, and Pathobiology. Annu. Rev. Pathol. 2018, 13, 351–378. [Google Scholar] [CrossRef]
  8. Cui, C.; Wang, F.; Zheng, Y.; Wei, H.; Peng, J. From birth to death: The hardworking life of Paneth cell in the small intestine. Front. Immunol. 2023, 14, 1122258. [Google Scholar] [CrossRef]
  9. Alkailani, M.I.; Aittaleb, M.; Tissir, F. WNT signaling at the intersection between neurogenesis and brain tumorigenesis. Front. Mol. Neurosci. 2022, 15, 1017568. [Google Scholar] [CrossRef]
  10. Kuwabara, T.; Hsieh, J.; Muotri, A.; Yeo, G.; Warashina, M.; Lie, D.C.; Moore, L.; Nakashima, K.; Asashima, M.; Gage, F.H. Wnt-mediated activation of NeuroD1 and retro-elements during adult neurogenesis. Nat. Neurosci. 2009, 12, 1097–1105. [Google Scholar] [CrossRef]
  11. Craig, S.E.L.; Michalski, M.N.; Williams, B.O. Got WNTS? Insight into bone health from a WNT perspective. Curr. Top. Dev. Biol. 2023, 153, 327–346. [Google Scholar] [CrossRef]
  12. Chidiac, R.; Angers, S. Wnt signaling in stem cells during development and cell lineage specification. Curr. Top. Dev. Biol. 2023, 153, 121–143. [Google Scholar] [CrossRef]
  13. Katoh, M.; Katoh, M. Molecular genetics and targeted therapy of WNT-related human diseases (Review). Int. J. Mol. Med. 2017, 40, 587–606. [Google Scholar] [CrossRef]
  14. Clevers, H.; Nusse, R. Wnt/beta-catenin signaling and disease. Cell 2012, 149, 1192–1205. [Google Scholar] [CrossRef]
  15. Kuechler, A.; Willemsen, M.H.; Albrecht, B.; Bacino, C.A.; Bartholomew, D.W.; van Bokhoven, H.; van den Boogaard, M.J.; Bramswig, N.; Buttner, C.; Cremer, K.; et al. De novo mutations in beta-catenin (CTNNB1) appear to be a frequent cause of intellectual disability: Expanding the mutational and clinical spectrum. Hum. Genet. 2015, 134, 97–109. [Google Scholar] [CrossRef]
  16. Marzo, A.; Galli, S.; Lopes, D.; McLeod, F.; Podpolny, M.; Segovia-Roldan, M.; Ciani, L.; Purro, S.; Cacucci, F.; Gibb, A.; et al. Reversal of Synapse Degeneration by Restoring Wnt Signaling in the Adult Hippocampus. Curr. Biol. 2016, 26, 2551–2561. [Google Scholar] [CrossRef]
  17. Huybrechts, Y.; Mortier, G.; Boudin, E.; Van Hul, W. WNT Signaling and Bone: Lessons From Skeletal Dysplasias and Disorders. Front. Endocrinol. 2020, 11, 165. [Google Scholar] [CrossRef]
  18. Wang, Z.; Liu, C.H.; Huang, S.; Chen, J. Wnt Signaling in vascular eye diseases. Prog. Retin. Eye Res. 2019, 70, 110–133. [Google Scholar] [CrossRef]
  19. Mandel, H.; Shemer, R.; Borochowitz, Z.U.; Okopnik, M.; Knopf, C.; Indelman, M.; Drugan, A.; Tiosano, D.; Gershoni-Baruch, R.; Choder, M.; et al. SERKAL syndrome: An autosomal-recessive disorder caused by a loss-of-function mutation in WNT4. Am. J. Hum. Genet. 2008, 82, 39–47. [Google Scholar] [CrossRef]
  20. Nusse, R.; Clevers, H. Wnt/beta-Catenin Signaling, Disease, and Emerging Therapeutic Modalities. Cell 2017, 169, 985–999. [Google Scholar] [CrossRef]
  21. Zhan, T.; Rindtorff, N.; Boutros, M. Wnt signaling in cancer. Oncogene 2017, 36, 1461–1473. [Google Scholar] [CrossRef] [PubMed]
  22. Latour, M.; Her, N.G.; Kesari, S.; Nurmemmedov, E. WNT Signaling as a Therapeutic Target for Glioblastoma. Int. J. Mol. Sci. 2021, 22, 8428. [Google Scholar] [CrossRef]
  23. Wickstrom, M.; Dyberg, C.; Milosevic, J.; Einvik, C.; Calero, R.; Sveinbjornsson, B.; Sanden, E.; Darabi, A.; Siesjo, P.; Kool, M.; et al. Wnt/beta-catenin pathway regulates MGMT gene expression in cancer and inhibition of Wnt signalling prevents chemoresistance. Nat. Commun. 2015, 6, 8904. [Google Scholar] [CrossRef]
  24. Liu, H.; Fergusson, M.M.; Castilho, R.M.; Liu, J.; Cao, L.; Chen, J.; Malide, D.; Rovira, I.I.; Schimel, D.; Kuo, C.J.; et al. Augmented Wnt signaling in a mammalian model of accelerated aging. Science 2007, 317, 803–806. [Google Scholar] [CrossRef] [PubMed]
  25. Kinzler, K.W.; Nilbert, M.C.; Su, L.K.; Vogelstein, B.; Bryan, T.M.; Levy, D.B.; Smith, K.J.; Preisinger, A.C.; Hedge, P.; McKechnie, D.; et al. Identification of FAP locus genes from chromosome 5q21. Science 1991, 253, 661–665. [Google Scholar] [CrossRef]
  26. Nishisho, I.; Nakamura, Y.; Miyoshi, Y.; Miki, Y.; Ando, H.; Horii, A.; Koyama, K.; Utsunomiya, J.; Baba, S.; Hedge, P. Mutations of chromosome 5q21 genes in FAP and colorectal cancer patients. Science 1991, 253, 665–669. [Google Scholar] [CrossRef]
  27. Bisht, S.; Nigam, M.; Kunjwal, S.S.; Sergey, P.; Mishra, A.P.; Sharifi-Rad, J. Cancer Stem Cells: From an Insight into the Basics to Recent Advances and Therapeutic Targeting. Stem Cells Int. 2022, 2022, 9653244. [Google Scholar] [CrossRef]
  28. Takahashi-Yanaga, F.; Kahn, M. Targeting Wnt signaling: Can we safely eradicate cancer stem cells? Clin. Cancer Res. 2010, 16, 3153–3162. [Google Scholar] [CrossRef] [PubMed]
  29. Lee, Y.; Lee, J.K.; Ahn, S.H.; Lee, J.; Nam, D.H. WNT signaling in glioblastoma and therapeutic opportunities. Lab. Investig. 2016, 96, 137–150. [Google Scholar] [CrossRef]
  30. Mikels, A.J.; Nusse, R. Wnts as ligands: Processing, secretion and reception. Oncogene 2006, 25, 7461–7468. [Google Scholar] [CrossRef]
  31. Stamos, J.L.; Weis, W.I. The beta-catenin destruction complex. Cold Spring Harb. Perspect. Biol. 2013, 5, a007898. [Google Scholar] [CrossRef]
  32. Cliffe, A.; Hamada, F.; Bienz, M. A role of Dishevelled in relocating Axin to the plasma membrane during wingless signaling. Curr. Biol. 2003, 13, 960–966. [Google Scholar] [CrossRef]
  33. Brembeck, F.H.; Rosario, M.; Birchmeier, W. Balancing cell adhesion and Wnt signaling, the key role of beta-catenin. Curr. Opin. Genet. Dev. 2006, 16, 51–59. [Google Scholar] [CrossRef]
  34. Munoz-Castaneda, J.R.; Rodelo-Haad, C.; Pendon-Ruiz de Mier, M.V.; Martin-Malo, A.; Santamaria, R.; Rodriguez, M. Klotho/FGF23 and Wnt Signaling as Important Players in the Comorbidities Associated with Chronic Kidney Disease. Toxins 2020, 12, 185. [Google Scholar] [CrossRef]
  35. Reyes, M.; Flores, T.; Betancur, D.; Pena-Oyarzun, D.; Torres, V.A. Wnt/beta-Catenin Signaling in Oral Carcinogenesis. Int. J. Mol. Sci. 2020, 21, 4682. [Google Scholar] [CrossRef]
  36. Cadigan, K.M.; Waterman, M.L. TCF/LEFs and Wnt signaling in the nucleus. Cold Spring Harb. Perspect. Biol. 2012, 4, a007906. [Google Scholar] [CrossRef] [PubMed]
  37. Doumpas, N.; Lampart, F.; Robinson, M.D.; Lentini, A.; Nestor, C.E.; Cantu, C.; Basler, K. TCF/LEF dependent and independent transcriptional regulation of Wnt/beta-catenin target genes. EMBO J. 2019, 38, e98873. [Google Scholar] [CrossRef]
  38. Kaidi, A.; Williams, A.C.; Paraskeva, C. Interaction between beta-catenin and HIF-1 promotes cellular adaptation to hypoxia. Nat. Cell Biol. 2007, 9, 210–217. [Google Scholar] [CrossRef]
  39. Kim, C.H.; Neiswender, H.; Baik, E.J.; Xiong, W.C.; Mei, L. Beta-catenin interacts with MyoD and regulates its transcription activity. Mol. Cell. Biol. 2008, 28, 2941–2951. [Google Scholar] [CrossRef]
  40. Kormish, J.D.; Sinner, D.; Zorn, A.M. Interactions between SOX factors and Wnt/beta-catenin signaling in development and disease. Dev. Dyn. 2010, 239, 56–68. [Google Scholar] [CrossRef]
  41. Essers, M.A.; de Vries-Smits, L.M.; Barker, N.; Polderman, P.E.; Burgering, B.M.; Korswagen, H.C. Functional interaction between beta-catenin and FOXO in oxidative stress signaling. Science 2005, 308, 1181–1184. [Google Scholar] [CrossRef]
  42. Sokol, S.Y. Maintaining embryonic stem cell pluripotency with Wnt signaling. Development 2011, 138, 4341–4350. [Google Scholar] [CrossRef] [PubMed]
  43. Chen, Y.; Alman, B.A. Wnt pathway, an essential role in bone regeneration. J. Cell. Biochem. 2009, 106, 353–362. [Google Scholar] [CrossRef] [PubMed]
  44. Clevers, H. Wnt/beta-catenin signaling in development and disease. Cell 2006, 127, 469–480. [Google Scholar] [CrossRef]
  45. Jin, T. The WNT signalling pathway and diabetes mellitus. Diabetologia 2008, 51, 1771–1780. [Google Scholar] [CrossRef]
  46. Sakoguchi-Okada, N.; Takahashi-Yanaga, F.; Fukada, K.; Shiraishi, F.; Taba, Y.; Miwa, Y.; Morimoto, S.; Iida, M.; Sasaguri, T. Celecoxib inhibits the expression of survivin via the suppression of promoter activity in human colon cancer cells. Biochem. Pharmacol. 2007, 73, 1318–1329. [Google Scholar] [CrossRef]
  47. Vallee, A.; Lecarpentier, Y. Crosstalk between Peroxisome Proliferator-Activated Receptor Gamma and the Canonical WNT/beta-Catenin Pathway in Chronic Inflammation and Oxidative Stress during Carcinogenesis. Front. Immunol. 2018, 9, 745. [Google Scholar] [CrossRef]
  48. Veeman, M.T.; Axelrod, J.D.; Moon, R.T. A second canon. Functions and mechanisms of beta-catenin-independent Wnt signaling. Dev. Cell 2003, 5, 367–377. [Google Scholar] [CrossRef]
  49. Yang, Y.; Mlodzik, M. Wnt-Frizzled/planar cell polarity signaling: Cellular orientation by facing the wind (Wnt). Annu. Rev. Cell Dev. Biol. 2015, 31, 623–646. [Google Scholar] [CrossRef]
  50. Goodrich, L.V.; Strutt, D. Principles of planar polarity in animal development. Development 2011, 138, 1877–1892. [Google Scholar] [CrossRef]
  51. Roszko, I.; Sawada, A.; Solnica-Krezel, L. Regulation of convergence and extension movements during vertebrate gastrulation by the Wnt/PCP pathway. Semin. Cell Dev. Biol. 2009, 20, 986–997. [Google Scholar] [CrossRef] [PubMed]
  52. Butler, M.T.; Wallingford, J.B. Planar cell polarity in development and disease. Nat. Rev. Mol. Cell Biol. 2017, 18, 375–388. [Google Scholar] [CrossRef]
  53. Devenport, D. The cell biology of planar cell polarity. J. Cell Biol. 2014, 207, 171–179. [Google Scholar] [CrossRef]
  54. Barzegar Behrooz, A.; Talaie, Z.; Jusheghani, F.; Los, M.J.; Klonisch, T.; Ghavami, S. Wnt and PI3K/Akt/mTOR Survival Pathways as Therapeutic Targets in Glioblastoma. Int. J. Mol. Sci. 2022, 23, 1353. [Google Scholar] [CrossRef]
  55. Garcia, A.L.; Udeh, A.; Kalahasty, K.; Hackam, A.S. A growing field: The regulation of axonal regeneration by Wnt signaling. Neural Regen. Res. 2018, 13, 43–52. [Google Scholar] [CrossRef]
  56. Liu, W.; Sato, A.; Khadka, D.; Bharti, R.; Diaz, H.; Runnels, L.W.; Habas, R. Mechanism of activation of the Formin protein Daam1. Proc. Natl. Acad. Sci. USA 2008, 105, 210–215. [Google Scholar] [CrossRef]
  57. Seifert, J.R.; Mlodzik, M. Frizzled/PCP signalling: A conserved mechanism regulating cell polarity and directed motility. Nat. Rev. Genet. 2007, 8, 126–138. [Google Scholar] [CrossRef]
  58. Humphries, A.C.; Mlodzik, M. From instruction to output: Wnt/PCP signaling in development and cancer. Curr. Opin. Cell Biol. 2018, 51, 110–116. [Google Scholar] [CrossRef]
  59. Kohn, A.D.; Moon, R.T. Wnt and calcium signaling: Beta-catenin-independent pathways. Cell Calcium 2005, 38, 439–446. [Google Scholar] [CrossRef]
  60. Gomez-Orte, E.; Saenz-Narciso, B.; Moreno, S.; Cabello, J. Multiple functions of the noncanonical Wnt pathway. Trends Genet. 2013, 29, 545–553. [Google Scholar] [CrossRef]
  61. Sharma, M.; Castro-Piedras, I.; Simmons, G.E., Jr.; Pruitt, K. Dishevelled: A masterful conductor of complex Wnt signals. Cell. Signal. 2018, 47, 52–64. [Google Scholar] [CrossRef] [PubMed]
  62. Freisinger, C.M.; Fisher, R.A.; Slusarski, D.C. Regulator of g protein signaling 3 modulates wnt5b calcium dynamics and somite patterning. PLoS Genet. 2010, 6, e1001020. [Google Scholar] [CrossRef]
  63. He, X.; Saint-Jeannet, J.P.; Wang, Y.; Nathans, J.; Dawid, I.; Varmus, H. A member of the Frizzled protein family mediating axis induction by Wnt-5A. Science 1997, 275, 1652–1654. [Google Scholar] [CrossRef] [PubMed]
  64. Davis, E.K.; Zou, Y.; Ghosh, A. Wnts acting through canonical and noncanonical signaling pathways exert opposite effects on hippocampal synapse formation. Neural Dev. 2008, 3, 32. [Google Scholar] [CrossRef]
  65. Yoshida, Y.; Kim, S.; Chiba, K.; Kawai, S.; Tachikawa, H.; Takahashi, N. Calcineurin inhibitors block dorsal-side signaling that affect late-stage development of the heart, kidney, liver, gut and somitic tissue during Xenopus embryogenesis. Dev. Growth Differ. 2004, 46, 139–152. [Google Scholar] [CrossRef] [PubMed]
  66. McDonald, S.L.; Silver, A. The opposing roles of Wnt-5a in cancer. Br. J. Cancer 2009, 101, 209–214. [Google Scholar] [CrossRef]
  67. Mohapatra, P.; Yadav, V.; Toftdahl, M.; Andersson, T. WNT5A-Induced Activation of the Protein Kinase C Substrate MARCKS Is Required for Melanoma Cell Invasion. Cancers 2020, 12, 346. [Google Scholar] [CrossRef]
  68. Weeraratna, A.T.; Jiang, Y.; Hostetter, G.; Rosenblatt, K.; Duray, P.; Bittner, M.; Trent, J.M. Wnt5a signaling directly affects cell motility and invasion of metastatic melanoma. Cancer Cell 2002, 1, 279–288. [Google Scholar] [CrossRef]
  69. Illendula, A.; Fultang, N.; Peethambaran, B. Retinoic acid induces differentiation in neuroblastoma via ROR1 by modulating retinoic acid response elements. Oncol. Rep. 2020, 44, 1013–1024. [Google Scholar] [CrossRef]
  70. MacLeod, R.J.; Hayes, M.; Pacheco, I. Wnt5a secretion stimulated by the extracellular calcium-sensing receptor inhibits defective Wnt signaling in colon cancer cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2007, 293, G403–G411. [Google Scholar] [CrossRef]
  71. Kiecker, C.; Niehrs, C. A morphogen gradient of Wnt/beta-catenin signalling regulates anteroposterior neural patterning in Xenopus. Development 2001, 128, 4189–4201. [Google Scholar] [CrossRef] [PubMed]
  72. Chen, Z.; Lei, Y.; Cao, X.; Zheng, Y.; Wang, F.; Bao, Y.; Peng, R.; Finnell, R.H.; Zhang, T.; Wang, H. Genetic analysis of Wnt/PCP genes in neural tube defects. BMC Med. Genom. 2018, 11, 38. [Google Scholar] [CrossRef] [PubMed]
  73. Shi, Z.; Yang, X.; Li, B.B.; Chen, S.; Yang, L.; Cheng, L.; Zhang, T.; Wang, H.; Zheng, Y. Novel Mutation of LRP6 Identified in Chinese Han Population Links Canonical WNT Signaling to Neural Tube Defects. Birth Defects Res. 2018, 110, 63–71. [Google Scholar] [CrossRef]
  74. Zhao, T.; Gan, Q.; Stokes, A.; Lassiter, R.N.; Wang, Y.; Chan, J.; Han, J.X.; Pleasure, D.E.; Epstein, J.A.; Zhou, C.J. beta-catenin regulates Pax3 and Cdx2 for caudal neural tube closure and elongation. Development 2014, 141, 148–157. [Google Scholar] [CrossRef]
  75. Manzari-Tavakoli, A.; Babajani, A.; Farjoo, M.H.; Hajinasrollah, M.; Bahrami, S.; Niknejad, H. The Cross-Talks among Bone Morphogenetic Protein (BMP) Signaling and Other Prominent Pathways Involved in Neural Differentiation. Front. Mol. Neurosci. 2022, 15, 827275. [Google Scholar] [CrossRef]
  76. Feigenson, K.; Reid, M.; See, J.; Crenshaw, I.E.; Grinspan, J.B. Canonical Wnt signalling requires the BMP pathway to inhibit oligodendrocyte maturation. ASN Neuro 2011, 3, e00061. [Google Scholar] [CrossRef] [PubMed]
  77. Haegele, L.; Ingold, B.; Naumann, H.; Tabatabai, G.; Ledermann, B.; Brandner, S. Wnt signalling inhibits neural differentiation of embryonic stem cells by controlling bone morphogenetic protein expression. Mol. Cell. Neurosci. 2003, 24, 696–708. [Google Scholar] [CrossRef]
  78. Lie, D.C.; Colamarino, S.A.; Song, H.J.; Desire, L.; Mira, H.; Consiglio, A.; Lein, E.S.; Jessberger, S.; Lansford, H.; Dearie, A.R.; et al. Wnt signalling regulates adult hippocampal neurogenesis. Nature 2005, 437, 1370–1375. [Google Scholar] [CrossRef]
  79. Hirabayashi, Y.; Itoh, Y.; Tabata, H.; Nakajima, K.; Akiyama, T.; Masuyama, N.; Gotoh, Y. The Wnt/beta-catenin pathway directs neuronal differentiation of cortical neural precursor cells. Development 2004, 131, 2791–2801. [Google Scholar] [CrossRef]
  80. Adachi, K.; Mirzadeh, Z.; Sakaguchi, M.; Yamashita, T.; Nikolcheva, T.; Gotoh, Y.; Peltz, G.; Gong, L.; Kawase, T.; Alvarez-Buylla, A.; et al. Beta-catenin signaling promotes proliferation of progenitor cells in the adult mouse subventricular zone. Stem Cells 2007, 25, 2827–2836. [Google Scholar] [CrossRef]
  81. Faigle, R.; Song, H. Signaling mechanisms regulating adult neural stem cells and neurogenesis. Biochim. Biophys. Acta 2013, 1830, 2435–2448. [Google Scholar] [CrossRef]
  82. Kalani, M.Y.; Cheshier, S.H.; Cord, B.J.; Bababeygy, S.R.; Vogel, H.; Weissman, I.L.; Palmer, T.D.; Nusse, R. Wnt-mediated self-renewal of neural stem/progenitor cells. Proc. Natl. Acad. Sci. USA 2008, 105, 16970–16975. [Google Scholar] [CrossRef] [PubMed]
  83. Manoranjan, B.; Venugopal, C.; Bakhshinyan, D.; Adile, A.A.; Richards, L.; Kameda-Smith, M.M.; Whitley, O.; Dvorkin-Gheva, A.; Subapanditha, M.; Savage, N.; et al. Wnt activation as a therapeutic strategy in medulloblastoma. Nat. Commun. 2020, 11, 4323. [Google Scholar] [CrossRef] [PubMed]
  84. Omuro, A.; DeAngelis, L.M. Glioblastoma and other malignant gliomas: A clinical review. JAMA 2013, 310, 1842–1850. [Google Scholar] [CrossRef]
  85. Krex, D.; Klink, B.; Hartmann, C.; von Deimling, A.; Pietsch, T.; Simon, M.; Sabel, M.; Steinbach, J.P.; Heese, O.; Reifenberger, G.; et al. Long-term survival with glioblastoma multiforme. Brain 2007, 130, 2596–2606. [Google Scholar] [CrossRef]
  86. Marenco-Hillembrand, L.; Wijesekera, O.; Suarez-Meade, P.; Mampre, D.; Jackson, C.; Peterson, J.; Trifiletti, D.; Hammack, J.; Ortiz, K.; Lesser, E.; et al. Trends in glioblastoma: Outcomes over time and type of intervention: A systematic evidence based analysis. J. Neurooncol. 2020, 147, 297–307. [Google Scholar] [CrossRef]
  87. 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]
  88. Campos, B.; Olsen, L.R.; Urup, T.; Poulsen, H.S. A comprehensive profile of recurrent glioblastoma. Oncogene 2016, 35, 5819–5825. [Google Scholar] [CrossRef]
  89. Nowicki, M.O.; Dmitrieva, N.; Stein, A.M.; Cutter, J.L.; Godlewski, J.; Saeki, Y.; Nita, M.; Berens, M.E.; Sander, L.M.; Newton, H.B.; et al. Lithium inhibits invasion of glioma cells; possible involvement of glycogen synthase kinase-3. Neuro Oncol. 2008, 10, 690–699. [Google Scholar] [CrossRef]
  90. Kotliarova, S.; Pastorino, S.; Kovell, L.C.; Kotliarov, Y.; Song, H.; Zhang, W.; Bailey, R.; Maric, D.; Zenklusen, J.C.; Lee, J.; et al. Glycogen synthase kinase-3 inhibition induces glioma cell death through c-MYC, nuclear factor-kappaB, and glucose regulation. Cancer Res. 2008, 68, 6643–6651. [Google Scholar] [CrossRef] [PubMed]
  91. Korur, S.; Huber, R.M.; Sivasankaran, B.; Petrich, M.; Morin, P., Jr.; Hemmings, B.A.; Merlo, A.; Lino, M.M. GSK3beta regulates differentiation and growth arrest in glioblastoma. PLoS ONE 2009, 4, e7443. [Google Scholar] [CrossRef]
  92. Liu, X.; Wang, L.; Zhao, S.; Ji, X.; Luo, Y.; Ling, F. beta-Catenin overexpression in malignant glioma and its role in proliferation and apoptosis in glioblastma cells. Med. Oncol. 2011, 28, 608–614. [Google Scholar] [CrossRef]
  93. Pu, P.; Zhang, Z.; Kang, C.; Jiang, R.; Jia, Z.; Wang, G.; Jiang, H. Downregulation of Wnt2 and beta-catenin by siRNA suppresses malignant glioma cell growth. Cancer Gene Ther. 2009, 16, 351–361. [Google Scholar] [CrossRef]
  94. Kahlert, U.D.; Suwala, A.K.; Koch, K.; Natsumeda, M.; Orr, B.A.; Hayashi, M.; Maciaczyk, J.; Eberhart, C.G. Pharmacologic Wnt Inhibition Reduces Proliferation, Survival, and Clonogenicity of Glioblastoma Cells. J. Neuropathol. Exp. Neurol. 2015, 74, 889–900. [Google Scholar] [CrossRef]
  95. Zuccarini, M.; Giuliani, P.; Ziberi, S.; Carluccio, M.; Iorio, P.D.; Caciagli, F.; Ciccarelli, R. The Role of Wnt Signal in Glioblastoma Development and Progression: A Possible New Pharmacological Target for the Therapy of This Tumor. Genes 2018, 9, 105. [Google Scholar] [CrossRef]
  96. Persano, L.; Rampazzo, E.; Basso, G.; Viola, G. Glioblastoma cancer stem cells: Role of the microenvironment and therapeutic targeting. Biochem. Pharmacol. 2013, 85, 612–622. [Google Scholar] [CrossRef]
  97. Hecht, A.; Vleminckx, K.; Stemmler, M.P.; van Roy, F.; Kemler, R. The p300/CBP acetyltransferases function as transcriptional coactivators of beta-catenin in vertebrates. EMBO J. 2000, 19, 1839–1850. [Google Scholar] [CrossRef] [PubMed]
  98. Takemaru, K.I.; Moon, R.T. The transcriptional coactivator CBP interacts with beta-catenin to activate gene expression. J. Cell Biol. 2000, 149, 249–254. [Google Scholar] [CrossRef]
  99. Tompa, M.; Kalovits, F.; Nagy, A.; Kalman, B. Contribution of the Wnt Pathway to Defining Biology of Glioblastoma. Neuromol. Med. 2018, 20, 437–451. [Google Scholar] [CrossRef]
  100. Foltz, G.; Ryu, G.Y.; Yoon, J.G.; Nelson, T.; Fahey, J.; Frakes, A.; Lee, H.; Field, L.; Zander, K.; Sibenaller, Z.; et al. Genome-wide analysis of epigenetic silencing identifies BEX1 and BEX2 as candidate tumor suppressor genes in malignant glioma. Cancer Res. 2006, 66, 6665–6674. [Google Scholar] [CrossRef]
  101. Foltz, G.; Yoon, J.G.; Lee, H.; Ma, L.; Tian, Q.; Hood, L.; Madan, A. Epigenetic regulation of wnt pathway antagonists in human glioblastoma multiforme. Genes Cancer 2010, 1, 81–90. [Google Scholar] [CrossRef] [PubMed]
  102. Guan, R.; Zhang, X.; Guo, M. Glioblastoma stem cells and Wnt signaling pathway: Molecular mechanisms and therapeutic targets. Chin. Neurosurg. J. 2020, 6, 25. [Google Scholar] [CrossRef]
  103. Hersh, A.M.; Gaitsch, H.; Alomari, S.; Lubelski, D.; Tyler, B.M. Molecular Pathways and Genomic Landscape of Glioblastoma Stem Cells: Opportunities for Targeted Therapy. Cancers 2022, 14, 3743. [Google Scholar] [CrossRef] [PubMed]
  104. Zheng, H.; Ying, H.; Wiedemeyer, R.; Yan, H.; Quayle, S.N.; Ivanova, E.V.; Paik, J.H.; Zhang, H.; Xiao, Y.; Perry, S.R.; et al. PLAGL2 regulates Wnt signaling to impede differentiation in neural stem cells and gliomas. Cancer Cell 2010, 17, 497–509. [Google Scholar] [CrossRef]
  105. Zhang, N.; Wei, P.; Gong, A.; Chiu, W.T.; Lee, H.T.; Colman, H.; Huang, H.; Xue, J.; Liu, M.; Wang, Y.; et al. FoxM1 promotes beta-catenin nuclear localization and controls Wnt target-gene expression and glioma tumorigenesis. Cancer Cell 2011, 20, 427–442. [Google Scholar] [CrossRef] [PubMed]
  106. Arnes, M.; Casas Tinto, S. Aberrant Wnt signaling: A special focus in CNS diseases. J. Neurogenet. 2017, 31, 216–222. [Google Scholar] [CrossRef]
  107. Kim, S.A.; Kwak, J.; Nam, H.Y.; Chun, S.M.; Lee, B.W.; Lee, H.J.; Khang, S.K.; Kim, S.W. Promoter methylation of WNT inhibitory factor-1 and expression pattern of WNT/beta-catenin pathway in human astrocytoma: Pathologic and prognostic correlations. Mod. Pathol. 2013, 26, 626–639. [Google Scholar] [CrossRef]
  108. Kierulf-Vieira, K.S.; Sandberg, C.J.; Grieg, Z.; Gunther, C.C.; Langmoen, I.A.; Vik-Mo, E.O. Wnt inhibition is dysregulated in gliomas and its re-establishment inhibits proliferation and tumor sphere formation. Exp. Cell Res. 2016, 340, 53–61. [Google Scholar] [CrossRef]
  109. Allen, B.K.; Stathias, V.; Maloof, M.E.; Vidovic, D.; Winterbottom, E.F.; Capobianco, A.J.; Clarke, J.; Schurer, S.; Robbins, D.J.; Ayad, N.G. Epigenetic pathways and glioblastoma treatment: Insights from signaling cascades. J. Cell. Biochem. 2015, 116, 351–363. [Google Scholar] [CrossRef]
  110. Schiefer, L.; Visweswaran, M.; Perumal, V.; Arfuso, F.; Groth, D.; Newsholme, P.; Warrier, S.; Dharmarajan, A. Epigenetic regulation of the secreted frizzled-related protein family in human glioblastoma multiforme. Cancer Gene Ther. 2014, 21, 297–303. [Google Scholar] [CrossRef]
  111. Cao, V.T.; Jung, T.Y.; Jung, S.; Jin, S.G.; Moon, K.S.; Kim, I.Y.; Kang, S.S.; Park, C.S.; Lee, K.H.; Chae, H.J. The correlation and prognostic significance of MGMT promoter methylation and MGMT protein in glioblastomas. Neurosurgery 2009, 65, 866–875; discussion 875. [Google Scholar] [CrossRef] [PubMed]
  112. Hau, P.; Stupp, R.; Hegi, M.E. MGMT methylation status: The advent of stratified therapy in glioblastoma? Dis. Markers 2007, 23, 97–104. [Google Scholar] [CrossRef]
  113. Shou, J.; Ali-Osman, F.; Multani, A.S.; Pathak, S.; Fedi, P.; Srivenugopal, K.S. Human Dkk-1, a gene encoding a Wnt antagonist, responds to DNA damage and its overexpression sensitizes brain tumor cells to apoptosis following alkylation damage of DNA. Oncogene 2002, 21, 878–889. [Google Scholar] [CrossRef] [PubMed]
  114. Park, A.K.; Kim, P.; Ballester, L.Y.; Esquenazi, Y.; Zhao, Z. Subtype-specific signaling pathways and genomic aberrations associated with prognosis of glioblastoma. NeuroOncol. 2019, 21, 59–70. [Google Scholar] [CrossRef] [PubMed]
  115. Liu, Y.; Shen, Y.; Sun, T.; Yang, W. Mechanisms regulating radiosensitivity of glioma stem cells. Neoplasma 2017, 64, 655–665. [Google Scholar] [CrossRef] [PubMed]
  116. Bhandari, V.; Hoey, C.; Liu, L.Y.; Lalonde, E.; Ray, J.; Livingstone, J.; Lesurf, R.; Shiah, Y.J.; Vujcic, T.; Huang, X.; et al. Molecular landmarks of tumor hypoxia across cancer types. Nat. Genet. 2019, 51, 308–318. [Google Scholar] [CrossRef]
  117. Evans, S.M.; Jenkins, K.W.; Jenkins, W.T.; Dilling, T.; Judy, K.D.; Schrlau, A.; Judkins, A.; Hahn, S.M.; Koch, C.J. Imaging and analytical methods as applied to the evaluation of vasculature and hypoxia in human brain tumors. Radiat. Res. 2008, 170, 677–690. [Google Scholar] [CrossRef]
  118. Evans, S.M.; Judy, K.D.; Dunphy, I.; Jenkins, W.T.; Nelson, P.T.; Collins, R.; Wileyto, E.P.; Jenkins, K.; Hahn, S.M.; Stevens, C.W.; et al. Comparative measurements of hypoxia in human brain tumors using needle electrodes and EF5 binding. Cancer Res. 2004, 64, 1886–1892. [Google Scholar] [CrossRef]
  119. Pistollato, F.; Abbadi, S.; Rampazzo, E.; Persano, L.; Della Puppa, A.; Frasson, C.; Sarto, E.; Scienza, R.; D’Avella, D.; Basso, G. Intratumoral hypoxic gradient drives stem cells distribution and MGMT expression in glioblastoma. Stem Cells 2010, 28, 851–862. [Google Scholar] [CrossRef]
  120. Domenech, M.; Hernandez, A.; Plaja, A.; Martinez-Balibrea, E.; Balana, C. Hypoxia: The Cornerstone of Glioblastoma. Int. J. Mol. Sci. 2021, 22, 12608. [Google Scholar] [CrossRef]
  121. Mazumdar, J.; O’Brien, W.T.; Johnson, R.S.; LaManna, J.C.; Chavez, J.C.; Klein, P.S.; Simon, M.C. O2 regulates stem cells through Wnt/beta-catenin signalling. Nat. Cell Biol. 2010, 12, 1007–1013. [Google Scholar] [CrossRef]
  122. Rampazzo, E.; Persano, L.; Pistollato, F.; Moro, E.; Frasson, C.; Porazzi, P.; Della Puppa, A.; Bresolin, S.; Battilana, G.; Indraccolo, S.; et al. Wnt activation promotes neuronal differentiation of glioblastoma. Cell Death Dis. 2013, 4, e500. [Google Scholar] [CrossRef] [PubMed]
  123. Katoh, M.; Katoh, M. NUMB is a break of WNT-Notch signaling cycle. Int. J. Mol. Med. 2006, 18, 517–521. [Google Scholar] [CrossRef] [PubMed]
  124. Arce, L.; Yokoyama, N.N.; Waterman, M.L. Diversity of LEF/TCF action in development and disease. Oncogene 2006, 25, 7492–7504. [Google Scholar] [CrossRef]
  125. Boso, D.; Rampazzo, E.; Zanon, C.; Bresolin, S.; Maule, F.; Porcu, E.; Cani, A.; Della Puppa, A.; Trentin, L.; Basso, G.; et al. HIF-1alpha/Wnt signaling-dependent control of gene transcription regulates neuronal differentiation of glioblastoma stem cells. Theranostics 2019, 9, 4860–4877. [Google Scholar] [CrossRef] [PubMed]
  126. Bikfalvi, A.; da Costa, C.A.; Avril, T.; Barnier, J.V.; Bauchet, L.; Brisson, L.; Cartron, P.F.; Castel, H.; Chevet, E.; Chneiweiss, H.; et al. Challenges in glioblastoma research: Focus on the tumor microenvironment. Trends Cancer 2023, 9, 9–27. [Google Scholar] [CrossRef]
  127. Perus, L.J.M.; Walsh, L.A. Microenvironmental Heterogeneity in Brain Malignancies. Front. Immunol. 2019, 10, 2294. [Google Scholar] [CrossRef]
  128. Kowal, J.; Kornete, M.; Joyce, J.A. Re-education of macrophages as a therapeutic strategy in cancer. Immunotherapy 2019, 11, 677–689. [Google Scholar] [CrossRef]
  129. Venkataramani, V.; Tanev, D.I.; Strahle, C.; Studier-Fischer, A.; Fankhauser, L.; Kessler, T.; Korber, C.; Kardorff, M.; Ratliff, M.; Xie, R.; et al. Glutamatergic synaptic input to glioma cells drives brain tumour progression. Nature 2019, 573, 532–538. [Google Scholar] [CrossRef]
  130. Seano, G. Targeting the perivascular niche in brain tumors. Curr. Opin. Oncol. 2018, 30, 54–60. [Google Scholar] [CrossRef]
  131. Guelfi, S.; Duffau, H.; Bauchet, L.; Rothhut, B.; Hugnot, J.P. Vascular Transdifferentiation in the CNS: A Focus on Neural and Glioblastoma Stem-Like Cells. Stem Cells Int. 2016, 2016, 2759403. [Google Scholar] [CrossRef]
  132. Deshors, P.; Toulas, C.; Arnauduc, F.; Malric, L.; Siegfried, A.; Nicaise, Y.; Lemarie, A.; Larrieu, D.; Tosolini, M.; Cohen-Jonathan Moyal, E.; et al. Ionizing radiation induces endothelial transdifferentiation of glioblastoma stem-like cells through the Tie2 signaling pathway. Cell Death Dis. 2019, 10, 816. [Google Scholar] [CrossRef] [PubMed]
  133. Griveau, A.; Seano, G.; Shelton, S.J.; Kupp, R.; Jahangiri, A.; Obernier, K.; Krishnan, S.; Lindberg, O.R.; Yuen, T.J.; Tien, A.C.; et al. A Glial Signature and Wnt7 Signaling Regulate Glioma-Vascular Interactions and Tumor Microenvironment. Cancer Cell 2018, 33, 874–889.e7. [Google Scholar] [CrossRef] [PubMed]
  134. Clavreul, A.; Menei, P. Mesenchymal Stromal-Like Cells in the Glioma Microenvironment: What Are These Cells? Cancers 2020, 12, 2628. [Google Scholar] [CrossRef] [PubMed]
  135. Miroshnikova, Y.A.; Mouw, J.K.; Barnes, J.M.; Pickup, M.W.; Lakins, J.N.; Kim, Y.; Lobo, K.; Persson, A.I.; Reis, G.F.; McKnight, T.R.; et al. Tissue mechanics promote IDH1-dependent HIF1alpha-tenascin C feedback to regulate glioblastoma aggression. Nat. Cell Biol. 2016, 18, 1336–1345. [Google Scholar] [CrossRef]
  136. Huang, M.; Zhang, D.; Wu, J.Y.; Xing, K.; Yeo, E.; Li, C.; Zhang, L.; Holland, E.; Yao, L.; Qin, L.; et al. Wnt-mediated endothelial transformation into mesenchymal stem cell-like cells induces chemoresistance in glioblastoma. Sci. Transl. Med. 2020, 12, eaay7522. [Google Scholar] [CrossRef]
  137. Broekman, M.L.; Maas, S.L.N.; Abels, E.R.; Mempel, T.R.; Krichevsky, A.M.; Breakefield, X.O. Multidimensional communication in the microenvirons of glioblastoma. Nat. Rev. Neurol. 2018, 14, 482–495. [Google Scholar] [CrossRef]
  138. Yeo, A.T.; Rawal, S.; Delcuze, B.; Christofides, A.; Atayde, A.; Strauss, L.; Balaj, L.; Rogers, V.A.; Uhlmann, E.J.; Varma, H.; et al. Single-cell RNA sequencing reveals evolution of immune landscape during glioblastoma progression. Nat. Immunol. 2022, 23, 971–984. [Google Scholar] [CrossRef]
  139. Fan, D.; Yue, Q.; Chen, J.; Wang, C.; Yu, R.; Jin, Z.; Yin, S.; Wang, Q.; Chen, L.; Liao, X.; et al. Reprogramming the immunosuppressive microenvironment of IDH1 wild-type glioblastoma by blocking Wnt signaling between microglia and cancer cells. Oncoimmunology 2021, 10, 1932061. [Google Scholar] [CrossRef]
  140. Matias, D.; Dubois, L.G.; Pontes, B.; Rosario, L.; Ferrer, V.P.; Balca-Silva, J.; Fonseca, A.C.C.; Macharia, L.W.; Romao, L.; TCLS, E.S.; et al. GBM-Derived Wnt3a Induces M2-Like Phenotype in Microglial Cells Through Wnt/beta-Catenin Signaling. Mol. Neurobiol. 2019, 56, 1517–1530. [Google Scholar] [CrossRef]
  141. Tao, W.; Chu, C.; Zhou, W.; Huang, Z.; Zhai, K.; Fang, X.; Huang, Q.; Zhang, A.; Wang, X.; Yu, X.; et al. Dual Role of WISP1 in maintaining glioma stem cells and tumor-supportive macrophages in glioblastoma. Nat. Commun. 2020, 11, 3015. [Google Scholar] [CrossRef]
  142. Zheng, Y.; Graeber, M.B. Microglia and Brain Macrophages as Drivers of Glioma Progression. Int. J. Mol. Sci. 2022, 23, 15612. [Google Scholar] [CrossRef]
  143. 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]
  144. Coltin, H.; Sundaresan, L.; Smith, K.S.; Skowron, P.; Massimi, L.; Eberhart, C.G.; Schreck, K.C.; Gupta, N.; Weiss, W.A.; Tirapelli, D.; et al. Subgroup and subtype-specific outcomes in adult medulloblastoma. Acta Neuropathol. 2021, 142, 859–871. [Google Scholar] [CrossRef] [PubMed]
  145. Northcott, P.A.; Korshunov, A.; Witt, H.; Hielscher, T.; Eberhart, C.G.; Mack, S.; Bouffet, E.; Clifford, S.C.; Hawkins, C.E.; French, P.; et al. Medulloblastoma comprises four distinct molecular variants. J. Clin. Oncol. 2011, 29, 1408–1414. [Google Scholar] [CrossRef] [PubMed]
  146. Parsons, D.W.; Li, M.; Zhang, X.; Jones, S.; Leary, R.J.; Lin, J.C.; Boca, S.M.; Carter, H.; Samayoa, J.; Bettegowda, C.; et al. The genetic landscape of the childhood cancer medulloblastoma. Science 2011, 331, 435–439. [Google Scholar] [CrossRef] [PubMed]
  147. Cho, Y.J.; Tsherniak, A.; Tamayo, P.; Santagata, S.; Ligon, A.; Greulich, H.; Berhoukim, R.; Amani, V.; Goumnerova, L.; Eberhart, C.G.; et al. Integrative genomic analysis of medulloblastoma identifies a molecular subgroup that drives poor clinical outcome. J. Clin. Oncol. 2011, 29, 1424–1430. [Google Scholar] [CrossRef] [PubMed]
  148. Kool, M.; Korshunov, A.; Remke, M.; Jones, D.T.; Schlanstein, M.; Northcott, P.A.; Cho, Y.J.; Koster, J.; Schouten-van Meeteren, A.; van Vuurden, D.; et al. Molecular subgroups of medulloblastoma: An international meta-analysis of transcriptome, genetic aberrations, and clinical data of WNT, SHH, Group 3, and Group 4 medulloblastomas. Acta Neuropathol. 2012, 123, 473–484. [Google Scholar] [CrossRef]
  149. Taylor, M.D.; Northcott, P.A.; Korshunov, A.; Remke, M.; Cho, Y.J.; Clifford, S.C.; Eberhart, C.G.; Parsons, D.W.; Rutkowski, S.; Gajjar, A.; et al. Molecular subgroups of medulloblastoma: The current consensus. Acta Neuropathol. 2012, 123, 465–472. [Google Scholar] [CrossRef]
  150. Northcott, P.A.; Buchhalter, I.; Morrissy, A.S.; Hovestadt, V.; Weischenfeldt, J.; Ehrenberger, T.; Grobner, S.; Segura-Wang, M.; Zichner, T.; Rudneva, V.A.; et al. The whole-genome landscape of medulloblastoma subtypes. Nature 2017, 547, 311–317. [Google Scholar] [CrossRef]
  151. Khattab, A.; Monga, D.K. Turcot Syndrome. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  152. Huang, H.; Mahler-Araujo, B.M.; Sankila, A.; Chimelli, L.; Yonekawa, Y.; Kleihues, P.; Ohgaki, H. APC mutations in sporadic medulloblastomas. Am. J. Pathol. 2000, 156, 433–437. [Google Scholar] [CrossRef] [PubMed]
  153. Zurawel, R.H.; Chiappa, S.A.; Allen, C.; Raffel, C. Sporadic medulloblastomas contain oncogenic beta-catenin mutations. Cancer Res. 1998, 58, 896–899. [Google Scholar]
  154. Gibson, P.; Tong, Y.; Robinson, G.; Thompson, M.C.; Currle, D.S.; Eden, C.; Kranenburg, T.A.; Hogg, T.; Poppleton, H.; Martin, J.; et al. Subtypes of medulloblastoma have distinct developmental origins. Nature 2010, 468, 1095–1099. [Google Scholar] [CrossRef]
  155. Bartlett, F.; Kortmann, R.; Saran, F. Medulloblastoma. Clin. Oncol. 2013, 25, 36–45. [Google Scholar] [CrossRef]
  156. Robinson, G.; Parker, M.; Kranenburg, T.A.; Lu, C.; Chen, X.; Ding, L.; Phoenix, T.N.; Hedlund, E.; Wei, L.; Zhu, X.; et al. Novel mutations target distinct subgroups of medulloblastoma. Nature 2012, 488, 43–48. [Google Scholar] [CrossRef] [PubMed]
  157. Preusser, M.; Brastianos, P.K.; Mawrin, C. Advances in meningioma genetics: Novel therapeutic opportunities. Nat. Rev. Neurol. 2018, 14, 106–115. [Google Scholar] [CrossRef] [PubMed]
  158. Wen, P.Y.; Quant, E.; Drappatz, J.; Beroukhim, R.; Norden, A.D. Medical therapies for meningiomas. J. Neurooncol. 2010, 99, 365–378. [Google Scholar] [CrossRef]
  159. Watson, M.A.; Gutmann, D.H.; Peterson, K.; Chicoine, M.R.; Kleinschmidt-DeMasters, B.K.; Brown, H.G.; Perry, A. Molecular characterization of human meningiomas by gene expression profiling using high-density oligonucleotide microarrays. Am. J. Pathol. 2002, 161, 665–672. [Google Scholar] [CrossRef]
  160. Wrobel, G.; Roerig, P.; Kokocinski, F.; Neben, K.; Hahn, M.; Reifenberger, G.; Lichter, P. Microarray-based gene expression profiling of benign, atypical and anaplastic meningiomas identifies novel genes associated with meningioma progression. Int. J. Cancer 2005, 114, 249–256. [Google Scholar] [CrossRef]
  161. Ludwig, N.; Kim, Y.J.; Mueller, S.C.; Backes, C.; Werner, T.V.; Galata, V.; Sartorius, E.; Bohle, R.M.; Keller, A.; Meese, E. Posttranscriptional deregulation of signaling pathways in meningioma subtypes by differential expression of miRNAs. Neuro-Oncol. 2015, 17, 1250–1260. [Google Scholar] [CrossRef]
  162. Chang, X.; Shi, L.; Gao, F.; Russin, J.; Zeng, L.; He, S.; Chen, T.C.; Giannotta, S.L.; Weisenberger, D.J.; Zada, G.; et al. Genomic and transcriptome analysis revealing an oncogenic functional module in meningiomas. Neurosurg. Focus 2013, 35, E3. [Google Scholar] [CrossRef] [PubMed]
  163. He, S.; Pham, M.H.; Pease, M.; Zada, G.; Giannotta, S.L.; Wang, K.; Mack, W.J. A review of epigenetic and gene expression alterations associated with intracranial meningiomas. Neurosurg. Focus 2013, 35, E5. [Google Scholar] [CrossRef] [PubMed]
  164. Laurendeau, I.; Ferrer, M.; Garrido, D.; D’Haene, N.; Ciavarelli, P.; Basso, A.; Vidaud, M.; Bieche, I.; Salmon, I.; Szijan, I. Gene expression profiling of the hedgehog signaling pathway in human meningiomas. Mol. Med. 2010, 16, 262–270. [Google Scholar] [CrossRef]
  165. Vasudevan, H.N.; Braunstein, S.E.; Phillips, J.J.; Pekmezci, M.; Tomlin, B.A.; Wu, A.; Reis, G.F.; Magill, S.T.; Zhang, J.; Feng, F.Y.; et al. Comprehensive Molecular Profiling Identifies FOXM1 as a Key Transcription Factor for Meningioma Proliferation. Cell Rep. 2018, 22, 3672–3683. [Google Scholar] [CrossRef] [PubMed]
  166. Ezzat, S.; Asa, S.L.; Couldwell, W.T.; Barr, C.E.; Dodge, W.E.; Vance, M.L.; McCutcheon, I.E. The prevalence of pituitary adenomas: A systematic review. Cancer 2004, 101, 613–619. [Google Scholar] [CrossRef]
  167. Gaston-Massuet, C.; Andoniadou, C.L.; Signore, M.; Jayakody, S.A.; Charolidi, N.; Kyeyune, R.; Vernay, B.; Jacques, T.S.; Taketo, M.M.; Le Tissier, P.; et al. Increased Wingless (Wnt) signaling in pituitary progenitor/stem cells gives rise to pituitary tumors in mice and humans. Proc. Natl. Acad. Sci. USA 2011, 108, 11482–11487. [Google Scholar] [CrossRef]
  168. Li, W.; Zhang, Y.; Zhang, M.; Huang, G.; Zhang, Q. Wnt4 is overexpressed in human pituitary adenomas and is associated with tumor invasion. J. Clin. Neurosci. 2014, 21, 137–141. [Google Scholar] [CrossRef]
  169. Elston, M.S.; Gill, A.J.; Conaglen, J.V.; Clarkson, A.; Shaw, J.M.; Law, A.J.; Cook, R.J.; Little, N.S.; Clifton-Bligh, R.J.; Robinson, B.G.; et al. Wnt pathway inhibitors are strongly down-regulated in pituitary tumors. Endocrinology 2008, 149, 1235–1242. [Google Scholar] [CrossRef]
  170. Miyakoshi, T.; Takei, M.; Kajiya, H.; Egashira, N.; Takekoshi, S.; Teramoto, A.; Osamura, R.Y. Expression of Wnt4 in human pituitary adenomas regulates activation of the beta-catenin-independent pathway. Endocr. Pathol. 2008, 19, 261–273. [Google Scholar] [CrossRef]
  171. Ren, J.; Jian, F.; Jiang, H.; Sun, Y.; Pan, S.; Gu, C.; Chen, X.; Wang, W.; Ning, G.; Bian, L.; et al. Decreased expression of SFRP2 promotes development of the pituitary corticotroph adenoma by upregulating Wnt signaling. Int. J. Oncol. 2018, 52, 1934–1946. [Google Scholar] [CrossRef]
  172. Li, J.; Dong, W.; Li, Z.; Wang, H.; Gao, H.; Zhang, Y. Impact of SLC20A1 on the Wnt/beta-catenin signaling pathway in somatotroph adenomas. Mol. Med. Rep. 2019, 20, 3276–3284. [Google Scholar] [CrossRef] [PubMed]
  173. Phoenix, T.N.; Patmore, D.M.; Boop, S.; Boulos, N.; Jacus, M.O.; Patel, Y.T.; Roussel, M.F.; Finkelstein, D.; Goumnerova, L.; Perreault, S.; et al. Medulloblastoma Genotype Dictates Blood Brain Barrier Phenotype. Cancer Cell 2016, 29, 508–522. [Google Scholar] [CrossRef]
  174. Thomas, A.; Noel, G. Medulloblastoma: Optimizing care with a multidisciplinary approach. J. Multidiscip. Healthc. 2019, 12, 335–347. [Google Scholar] [CrossRef]
  175. Lhermitte, B.; Blandin, A.F.; Coca, A.; Guerin, E.; Durand, A.; Entz-Werle, N. Signaling pathway deregulation and molecular alterations across pediatric medulloblastomas. Neurochirurgie 2021, 67, 39–45. [Google Scholar] [CrossRef] [PubMed]
  176. Rodriguez-Blanco, J.; Pednekar, L.; Penas, C.; Li, B.; Martin, V.; Long, J.; Lee, E.; Weiss, W.A.; Rodriguez, C.; Mehrdad, N.; et al. Inhibition of WNT signaling attenuates self-renewal of SHH-subgroup medulloblastoma. Oncogene 2017, 36, 6306–6314. [Google Scholar] [CrossRef] [PubMed]
  177. Geron, L.; Salomao, K.B.; Borges, K.S.; Andrade, A.F.; Correa, C.A.P.; Scrideli, C.A.; Tone, L.G. Molecular characterization of Wnt pathway and function of beta-catenin overexpression in medulloblastoma cell lines. Cytotechnology 2018, 70, 1713–1722. [Google Scholar] [CrossRef]
  178. Poschl, J.; Bartels, M.; Ohli, J.; Bianchi, E.; Kuteykin-Teplyakov, K.; Grammel, D.; Ahlfeld, J.; Schuller, U. Wnt/beta-catenin signaling inhibits the Shh pathway and impairs tumor growth in Shh-dependent medulloblastoma. Acta Neuropathol. 2014, 127, 605–607. [Google Scholar] [CrossRef]
  179. Ray, S.; Chaturvedi, N.K.; Bhakat, K.K.; Rizzino, A.; Mahapatra, S. Subgroup-Specific Diagnostic, Prognostic, and Predictive Markers Influencing Pediatric Medulloblastoma Treatment. Diagnostics 2021, 12, 61. [Google Scholar] [CrossRef]
  180. Chan, T.A. Nonsteroidal anti-inflammatory drugs, apoptosis, and colon-cancer chemoprevention. Lancet Oncol. 2002, 3, 166–174. [Google Scholar] [CrossRef]
  181. Dihlmann, S.; Siermann, A.; von Knebel Doeberitz, M. The nonsteroidal anti-inflammatory drugs aspirin and indomethacin attenuate beta-catenin/TCF-4 signaling. Oncogene 2001, 20, 645–653. [Google Scholar] [CrossRef]
  182. Friis, S.; Riis, A.H.; Erichsen, R.; Baron, J.A.; Sorensen, H.T. Low-Dose Aspirin or Nonsteroidal Anti-inflammatory Drug Use and Colorectal Cancer Risk: A Population-Based, Case-Control Study. Ann. Intern. Med. 2015, 163, 347–355. [Google Scholar] [CrossRef] [PubMed]
  183. Lan, F.; Yue, X.; Han, L.; Yuan, X.; Shi, Z.; Huang, K.; Yang, Y.; Zou, J.; Zhang, J.; Jiang, T.; et al. Antitumor effect of aspirin in glioblastoma cells by modulation of beta-catenin/T-cell factor-mediated transcriptional activity. J. Neurosurg. 2011, 115, 780–788. [Google Scholar] [CrossRef]
  184. Thun, M.J.; Namboodiri, M.M.; Heath, C.W., Jr. Aspirin use and reduced risk of fatal colon cancer. N. Engl. J. Med. 1991, 325, 1593–1596. [Google Scholar] [CrossRef]
  185. Boon, E.M.; Keller, J.J.; Wormhoudt, T.A.; Giardiello, F.M.; Offerhaus, G.J.; van der Neut, R.; Pals, S.T. Sulindac targets nuclear beta-catenin accumulation and Wnt signalling in adenomas of patients with familial adenomatous polyposis and in human colorectal cancer cell lines. Br. J. Cancer 2004, 90, 224–229. [Google Scholar] [CrossRef]
  186. Giardiello, F.M.; Hamilton, S.R.; Krush, A.J.; Piantadosi, S.; Hylind, L.M.; Celano, P.; Booker, S.V.; Robinson, C.R.; Offerhaus, G.J. Treatment of colonic and rectal adenomas with sulindac in familial adenomatous polyposis. N. Engl. J. Med. 1993, 328, 1313–1316. [Google Scholar] [CrossRef] [PubMed]
  187. Allani, S.K.; Weissbach, H.; Lopez Toledano, M.A. Sulindac induces differentiation of glioblastoma stem cells making them more sensitive to oxidative stress. Neoplasma 2018, 65, 376–388. [Google Scholar] [CrossRef]
  188. Grosch, S.; Tegeder, I.; Niederberger, E.; Brautigam, L.; Geisslinger, G. COX-2 independent induction of cell cycle arrest and apoptosis in colon cancer cells by the selective COX-2 inhibitor celecoxib. FASEB J. 2001, 15, 2742–2744. [Google Scholar] [CrossRef] [PubMed]
  189. Sareddy, G.R.; Kesanakurti, D.; Kirti, P.B.; Babu, P.P. Nonsteroidal anti-inflammatory drugs diclofenac and celecoxib attenuates Wnt/beta-catenin/Tcf signaling pathway in human glioblastoma cells. Neurochem. Res. 2013, 38, 2313–2322. [Google Scholar] [CrossRef]
  190. Takahashi-Yanaga, F.; Yoshihara, T.; Jingushi, K.; Miwa, Y.; Morimoto, S.; Hirata, M.; Sasaguri, T. Celecoxib-induced degradation of T-cell factors-1 and -4 in human colon cancer cells. Biochem. Biophys. Res. Commun. 2008, 377, 1185–1190. [Google Scholar] [CrossRef]
  191. Jiang, Y.; Prunier, C.; Howe, P.H. The inhibitory effects of Disabled-2 (Dab2) on Wnt signaling are mediated through Axin. Oncogene 2008, 27, 1865–1875. [Google Scholar] [CrossRef]
  192. Pendas-Franco, N.; Aguilera, O.; Pereira, F.; Gonzalez-Sancho, J.M.; Munoz, A. Vitamin D and Wnt/beta-catenin pathway in colon cancer: Role and regulation of DICKKOPF genes. Anticancer Res. 2008, 28, 2613–2623. [Google Scholar] [PubMed]
  193. Mori, J.; Takahashi-Yanaga, F.; Miwa, Y.; Watanabe, Y.; Hirata, M.; Morimoto, S.; Shirasuna, K.; Sasaguri, T. Differentiation-inducing factor-1 induces cyclin D1 degradation through the phosphorylation of Thr286 in squamous cell carcinoma. Exp. Cell Res. 2005, 310, 426–433. [Google Scholar] [CrossRef]
  194. Takahashi-Yanaga, F.; Taba, Y.; Miwa, Y.; Kubohara, Y.; Watanabe, Y.; Hirata, M.; Morimoto, S.; Sasaguri, T. Dictyostelium differentiation-inducing factor-3 activates glycogen synthase kinase-3beta and degrades cyclin D1 in mammalian cells. J. Biol. Chem. 2003, 278, 9663–9670. [Google Scholar] [CrossRef]
  195. Jaiswal, A.S.; Marlow, B.P.; Gupta, N.; Narayan, S. Beta-catenin-mediated transactivation and cell-cell adhesion pathways are important in curcumin (diferuylmethane)-induced growth arrest and apoptosis in colon cancer cells. Oncogene 2002, 21, 8414–8427. [Google Scholar] [CrossRef]
  196. Zhang, F.Y.; Hu, Y.; Que, Z.Y.; Wang, P.; Liu, Y.H.; Wang, Z.H.; Xue, Y.X. Shikonin Inhibits the Migration and Invasion of Human Glioblastoma Cells by Targeting Phosphorylated beta-Catenin and Phosphorylated PI3K/Akt: A Potential Mechanism for the Anti-Glioma Efficacy of a Traditional Chinese Herbal Medicine. Int. J. Mol. Sci. 2015, 16, 23823–23848. [Google Scholar] [CrossRef]
  197. Hesari, A.; Rezaei, M.; Rezaei, M.; Dashtiahangar, M.; Fathi, M.; Rad, J.G.; Momeni, F.; Avan, A.; Ghasemi, F. Effect of curcumin on glioblastoma cells. J. Cell. Physiol. 2019, 234, 10281–10288. [Google Scholar] [CrossRef] [PubMed]
  198. Miao, J.; Jiang, Y.; Wang, D.; Zhou, J.; Fan, C.; Jiao, F.; Liu, B.; Zhang, J.; Wang, Y.; Zhang, Q. Trichosanthin suppresses the proliferation of glioma cells by inhibiting LGR5 expression and the Wnt/beta-catenin signaling pathway. Oncol. Rep. 2015, 34, 2845–2852. [Google Scholar] [CrossRef] [PubMed]
  199. Tao, Q.; Wu, C.; Xu, R.; Niu, L.; Qin, J.; Liu, N.; Zhang, P.; Wang, C. Diallyl trisulfide inhibits proliferation, invasion and angiogenesis of glioma cells by inactivating Wnt/beta-catenin signaling. Cell Tissue Res. 2017, 370, 379–390. [Google Scholar] [CrossRef]
  200. Cilibrasi, C.; Riva, G.; Romano, G.; Cadamuro, M.; Bazzoni, R.; Butta, V.; Paoletta, L.; Dalpra, L.; Strazzabosco, M.; Lavitrano, M.; et al. Resveratrol Impairs Glioma Stem Cells Proliferation and Motility by Modulating the Wnt Signaling Pathway. PLoS ONE 2017, 12, e0169854. [Google Scholar] [CrossRef]
  201. Mora, M.C.; Bassa, L.M.; Wong, K.E.; Tirabassi, M.V.; Arenas, R.B.; Schneider, S.S. Rhodiola crenulata inhibits Wnt/beta-catenin signaling in glioblastoma. J. Surg. Res. 2015, 197, 247–255. [Google Scholar] [CrossRef]
  202. Park, C.H.; Chang, J.Y.; Hahm, E.R.; Park, S.; Kim, H.K.; Yang, C.H. Quercetin, a potent inhibitor against beta-catenin/Tcf signaling in SW480 colon cancer cells. Biochem. Biophys. Res. Commun. 2005, 328, 227–234. [Google Scholar] [CrossRef]
  203. Yilmaz, U.C.; Bagca, B.G.; Karaca, E.; Durmaz, A.; Durmaz, B.; Aykut, A.; Kayalar, H.; Avci, C.B.; Susluer, S.Y.; Pariltay, E.; et al. Propolis Extract Regulates microRNA Expression in Glioblastoma and Brain Cancer Stem Cells. Anticancer Agents Med. Chem. 2022, 22, 378–389. [Google Scholar] [CrossRef] [PubMed]
  204. Kim, J.; Zhang, X.; Rieger-Christ, K.M.; Summerhayes, I.C.; Wazer, D.E.; Paulson, K.E.; Yee, A.S. Suppression of Wnt signaling by the green tea compound (-)-epigallocatechin 3-gallate (EGCG) in invasive breast cancer cells. Requirement of the transcriptional repressor HBP1. J. Biol. Chem. 2006, 281, 10865–10875. [Google Scholar] [CrossRef] [PubMed]
  205. Xie, C.R.; You, C.G.; Zhang, N.; Sheng, H.S.; Zheng, X.S. Epigallocatechin Gallate Preferentially Inhibits O(6)-Methylguanine DNA-Methyltransferase Expression in Glioblastoma Cells Rather than in Nontumor Glial Cells. Nutr. Cancer 2018, 70, 1339–1347. [Google Scholar] [CrossRef] [PubMed]
  206. Williams, S.P.; Nowicki, M.O.; Liu, F.; Press, R.; Godlewski, J.; Abdel-Rasoul, M.; Kaur, B.; Fernandez, S.A.; Chiocca, E.A.; Lawler, S.E. Indirubins decrease glioma invasion by blocking migratory phenotypes in both the tumor and stromal endothelial cell compartments. Cancer Res. 2011, 71, 5374–5380. [Google Scholar] [CrossRef]
  207. Precilla, D.S.; Kuduvalli, S.S.; Purushothaman, M.; Marimuthu, P.; Muralidharan, A.R.; Anitha, T.S. Wnt/beta-catenin Antagonists: Exploring New Avenues to Trigger Old Drugs in Alleviating Glioblastoma Multiforme. Curr. Mol. Pharmacol. 2022, 15, 338–360. [Google Scholar] [CrossRef]
  208. Oh, H.C.; Shim, J.K.; Park, J.; Lee, J.H.; Choi, R.J.; Kim, N.H.; Kim, H.S.; Moon, J.H.; Kim, E.H.; Chang, J.H.; et al. Combined effects of niclosamide and temozolomide against human glioblastoma tumorspheres. J. Cancer Res. Clin. Oncol. 2020, 146, 2817–2828. [Google Scholar] [CrossRef]
  209. Porcu, E.; Maule, F.; Manfreda, L.; Mariotto, E.; Bresolin, S.; Cani, A.; Bortolozzi, R.; Puppa, A.D.; Corallo, D.; Viola, G.; et al. Identification of Homoharringtonine as a potent inhibitor of glioblastoma cell proliferation and migration. Transl. Res. 2023, 251, 41–53. [Google Scholar] [CrossRef]
  210. Venugopal, C.; Hallett, R.; Vora, P.; Manoranjan, B.; Mahendram, S.; Qazi, M.A.; McFarlane, N.; Subapanditha, M.; Nolte, S.M.; Singh, M.; et al. Pyrvinium Targets CD133 in Human Glioblastoma Brain Tumor-Initiating Cells. Clin. Cancer Res. 2015, 21, 5324–5337. [Google Scholar] [CrossRef]
  211. Wang, Y.; Huang, N.; Li, H.; Liu, S.; Chen, X.; Yu, S.; Wu, N.; Bian, X.W.; Shen, H.Y.; Li, C.; et al. Promoting oligodendroglial-oriented differentiation of glioma stem cell: A repurposing of quetiapine for the treatment of malignant glioma. Oncotarget 2017, 8, 37511–37524. [Google Scholar] [CrossRef]
  212. Wan, Z.; Shi, W.; Shao, B.; Shi, J.; Shen, A.; Ma, Y.; Chen, J.; Lan, Q. Peroxisome proliferator-activated receptor gamma agonist pioglitazone inhibits beta-catenin-mediated glioma cell growth and invasion. Mol. Cell. Biochem. 2011, 349, 1–10. [Google Scholar] [CrossRef]
  213. Amerizadeh, F.; Rahmani, F.; Maftooh, M.; Nasiri, S.N.; Hassanian, S.M.; Giovannetti, E.; Moradi-Marjaneh, R.; Sabbaghzadeh, R.; Shahidsales, S.; Joudi-Mashhad, M.; et al. Inhibition of the Wnt/b-catenin pathway using PNU-74654 reduces tumor growth in in vitro and in vivo models of colorectal cancer. Tissue Cell 2022, 77, 101853. [Google Scholar] [CrossRef] [PubMed]
  214. Chang, T.S.; Lu, C.K.; Hsieh, Y.Y.; Wei, K.L.; Chen, W.M.; Tung, S.Y.; Wu, C.S.; Chan, M.W.Y.; Chiang, M.K. 2,4-Diamino-Quinazoline, a Wnt Signaling Inhibitor, Suppresses Gastric Cancer Progression and Metastasis. Int. J. Mol. Sci. 2020, 21, 5901. [Google Scholar] [CrossRef]
  215. Chen, Z.; Venkatesan, A.M.; Dehnhardt, C.M.; Dos Santos, O.; Delos Santos, E.; Ayral-Kaloustian, S.; Chen, L.; Geng, Y.; Arndt, K.T.; Lucas, J.; et al. 2,4-Diamino-quinazolines as inhibitors of beta-catenin/Tcf-4 pathway: Potential treatment for colorectal cancer. Bioorg. Med. Chem. Lett. 2009, 19, 4980–4983. [Google Scholar] [CrossRef]
  216. Trosset, J.Y.; Dalvit, C.; Knapp, S.; Fasolini, M.; Veronesi, M.; Mantegani, S.; Gianellini, L.M.; Catana, C.; Sundstrom, M.; Stouten, P.F.; et al. Inhibition of protein-protein interactions: The discovery of druglike beta-catenin inhibitors by combining virtual and biophysical screening. Proteins 2006, 64, 60–67. [Google Scholar] [CrossRef] [PubMed]
  217. Wu, M.Y.; Wang, C.C.; Chang, Y.C.; Yu, C.Y.; Sung, W.W.; Chen, C.J.; Tsai, M.C. The Therapeutic Role of PNU-74654 in Hepatocellular Carcinoma May Involve Suppression of NF-kappaB Signaling. Medicina 2022, 58, 798. [Google Scholar] [CrossRef]
  218. Delgado, E.R.; Yang, J.; So, J.; Leimgruber, S.; Kahn, M.; Ishitani, T.; Shin, D.; Mustata Wilson, G.; Monga, S.P. Identification and characterization of a novel small-molecule inhibitor of beta-catenin signaling. Am. J. Pathol. 2014, 184, 2111–2122. [Google Scholar] [CrossRef]
  219. Emami, K.H.; Nguyen, C.; Ma, H.; Kim, D.H.; Jeong, K.W.; Eguchi, M.; Moon, R.T.; Teo, J.L.; Kim, H.Y.; Moon, S.H.; et al. A small molecule inhibitor of beta-catenin/CREB-binding protein transcription. Proc. Natl. Acad. Sci. USA 2004, 101, 12682–12687. [Google Scholar] [CrossRef]
  220. McMillan, M.; Kahn, M. Investigating Wnt signaling: A chemogenomic safari. Drug Discov. Today 2005, 10, 1467–1474. [Google Scholar] [CrossRef]
  221. Zhang, Y.; Wen, Y.L.; Ma, J.W.; Ye, J.C.; Wang, X.; Huang, J.X.; Meng, C.Y.; Xu, X.Z.; Wang, S.X.; Zhong, X.Y. Tetrandrine inhibits glioma stem-like cells by repressing beta-catenin expression. Int. J. Oncol. 2017, 50, 101–110. [Google Scholar] [CrossRef] [PubMed]
  222. Yang, P.; Zhu, Y.; Zheng, Q.; Meng, S.; Wu, Y.; Shuai, W.; Sun, Q.; Wang, G. Recent advances of beta-catenin small molecule inhibitors for cancer therapy: Current development and future perspectives. Eur. J. Med. Chem. 2022, 243, 114789. [Google Scholar] [CrossRef]
  223. Fiskus, W.; Sharma, S.; Saha, S.; Shah, B.; Devaraj, S.G.; Sun, B.; Horrigan, S.; Leveque, C.; Zu, Y.; Iyer, S.; et al. Pre-clinical efficacy of combined therapy with novel beta-catenin antagonist BC2059 and histone deacetylase inhibitor against AML cells. Leukemia 2015, 29, 1267–1278. [Google Scholar] [CrossRef] [PubMed]
  224. Hwang, S.Y.; Deng, X.; Byun, S.; Lee, C.; Lee, S.J.; Suh, H.; Zhang, J.; Kang, Q.; Zhang, T.; Westover, K.D.; et al. Direct Targeting of beta-Catenin by a Small Molecule Stimulates Proteasomal Degradation and Suppresses Oncogenic Wnt/beta-Catenin Signaling. Cell Rep. 2016, 16, 28–36. [Google Scholar] [CrossRef] [PubMed]
  225. Fang, L.; Zhu, Q.; Neuenschwander, M.; Specker, E.; Wulf-Goldenberg, A.; Weis, W.I.; von Kries, J.P.; Birchmeier, W. A Small-Molecule Antagonist of the beta-Catenin/TCF4 Interaction Blocks the Self-Renewal of Cancer Stem Cells and Suppresses Tumorigenesis. Cancer Res. 2016, 76, 891–901. [Google Scholar] [CrossRef]
  226. Jang, G.B.; Hong, I.S.; Kim, R.J.; Lee, S.Y.; Park, S.J.; Lee, E.S.; Park, J.H.; Yun, C.H.; Chung, J.U.; Lee, K.J.; et al. Wnt/beta-Catenin Small-Molecule Inhibitor CWP232228 Preferentially Inhibits the Growth of Breast Cancer Stem-like Cells. Cancer Res. 2015, 75, 1691–1702. [Google Scholar] [CrossRef]
  227. Trautmann, M.; Sievers, E.; Aretz, S.; Kindler, D.; Michels, S.; Friedrichs, N.; Renner, M.; Kirfel, J.; Steiner, S.; Huss, S.; et al. SS18-SSX fusion protein-induced Wnt/beta-catenin signaling is a therapeutic target in synovial sarcoma. Oncogene 2014, 33, 5006–5016. [Google Scholar] [CrossRef] [PubMed]
  228. Takada, K.; Zhu, D.; Bird, G.H.; Sukhdeo, K.; Zhao, J.J.; Mani, M.; Lemieux, M.; Carrasco, D.E.; Ryan, J.; Horst, D.; et al. Targeted disruption of the BCL9/beta-catenin complex inhibits oncogenic Wnt signaling. Sci. Transl. Med. 2012, 4, 148ra117. [Google Scholar] [CrossRef] [PubMed]
  229. Fujii, N.; You, L.; Xu, Z.; Uematsu, K.; Shan, J.; He, B.; Mikami, I.; Edmondson, L.R.; Neale, G.; Zheng, J.; et al. An antagonist of dishevelled protein-protein interaction suppresses beta-catenin-dependent tumor cell growth. Cancer Res. 2007, 67, 573–579. [Google Scholar] [CrossRef]
  230. Grandy, D.; Shan, J.; Zhang, X.; Rao, S.; Akunuru, S.; Li, H.; Zhang, Y.; Alpatov, I.; Zhang, X.A.; Lang, R.A.; et al. Discovery and characterization of a small molecule inhibitor of the PDZ domain of dishevelled. J. Biol. Chem. 2009, 284, 16256–16263. [Google Scholar] [CrossRef] [PubMed]
  231. Shan, J.; Shi, D.L.; Wang, J.; Zheng, J. Identification of a specific inhibitor of the dishevelled PDZ domain. Biochemistry 2005, 44, 15495–15503. [Google Scholar] [CrossRef] [PubMed]
  232. Chen, B.; Dodge, M.E.; Tang, W.; Lu, J.; Ma, Z.; Fan, C.W.; Wei, S.; Hao, W.; Kilgore, J.; Williams, N.S.; et al. Small molecule-mediated disruption of Wnt-dependent signaling in tissue regeneration and cancer. Nat. Chem. Biol. 2009, 5, 100–107. [Google Scholar] [CrossRef]
  233. De Robertis, A.; Valensin, S.; Rossi, M.; Tunici, P.; Verani, M.; De Rosa, A.; Giordano, C.; Varrone, M.; Nencini, A.; Pratelli, C.; et al. Identification and characterization of a small-molecule inhibitor of Wnt signaling in glioblastoma cells. Mol. Cancer Ther. 2013, 12, 1180–1189. [Google Scholar] [CrossRef]
  234. Suwala, A.K.; Hanaford, A.; Kahlert, U.D.; Maciaczyk, J. Clipping the Wings of Glioblastoma: Modulation of WNT as a Novel Therapeutic Strategy. J. Neuropathol. Exp. Neurol. 2016, 75, 388–396. [Google Scholar] [CrossRef] [PubMed]
  235. Arques, O.; Chicote, I.; Puig, I.; Tenbaum, S.P.; Argiles, G.; Dienstmann, R.; Fernandez, N.; Caratu, G.; Matito, J.; Silberschmidt, D.; et al. Tankyrase Inhibition Blocks Wnt/beta-Catenin Pathway and Reverts Resistance to PI3K and AKT Inhibitors in the Treatment of Colorectal Cancer. Clin. Cancer Res. 2016, 22, 644–656. [Google Scholar] [CrossRef] [PubMed]
  236. Kim, Y.; Kim, K.H.; Lee, J.; Lee, Y.A.; Kim, M.; Lee, S.J.; Park, K.; Yang, H.; Jin, J.; Joo, K.M.; et al. Wnt activation is implicated in glioblastoma radioresistance. Lab. Investig. 2012, 92, 466–473. [Google Scholar] [CrossRef]
  237. Lau, T.; Chan, E.; Callow, M.; Waaler, J.; Boggs, J.; Blake, R.A.; Magnuson, S.; Sambrone, A.; Schutten, M.; Firestein, R.; et al. A novel tankyrase small-molecule inhibitor suppresses APC mutation-driven colorectal tumor growth. Cancer Res. 2013, 73, 3132–3144. [Google Scholar] [CrossRef] [PubMed]
  238. Scarborough, H.A.; Helfrich, B.A.; Casas-Selves, M.; Schuller, A.G.; Grosskurth, S.E.; Kim, J.; Tan, A.C.; Chan, D.C.; Zhang, Z.; Zaberezhnyy, V.; et al. AZ1366: An Inhibitor of Tankyrase and the Canonical Wnt Pathway that Limits the Persistence of Non-Small Cell Lung Cancer Cells Following EGFR Inhibition. Clin. Cancer Res. 2017, 23, 1531–1541. [Google Scholar] [CrossRef]
  239. Kierulf-Vieira, K.S.; Sandberg, C.J.; Waaler, J.; Lund, K.; Skaga, E.; Saberniak, B.M.; Panagopoulos, I.; Brandal, P.; Krauss, S.; Langmoen, I.A.; et al. A Small-Molecule Tankyrase Inhibitor Reduces Glioma Stem Cell Proliferation and Sphere Formation. Cancers 2020, 12, 1630. [Google Scholar] [CrossRef]
  240. Cheng, D.; Liu, J.; Han, D.; Zhang, G.; Gao, W.; Hsieh, M.H.; Ng, N.; Kasibhatla, S.; Tompkins, C.; Li, J.; et al. Discovery of Pyridinyl Acetamide Derivatives as Potent, Selective, and Orally Bioavailable Porcupine Inhibitors. ACS Med. Chem. Lett. 2016, 7, 676–680. [Google Scholar] [CrossRef] [PubMed]
  241. Cheng, Y.; Phoon, Y.P.; Jin, X.; Chong, S.Y.; Ip, J.C.; Wong, B.W.; Lung, M.L. Wnt-C59 arrests stemness and suppresses growth of nasopharyngeal carcinoma in mice by inhibiting the Wnt pathway in the tumor microenvironment. Oncotarget 2015, 6, 14428–14439. [Google Scholar] [CrossRef]
  242. Liu, J.; Pan, S.; Hsieh, M.H.; Ng, N.; Sun, F.; Wang, T.; Kasibhatla, S.; Schuller, A.G.; Li, A.G.; Cheng, D.; et al. Targeting Wnt-driven cancer through the inhibition of Porcupine by LGK974. Proc. Natl. Acad. Sci. USA 2013, 110, 20224–20229. [Google Scholar] [CrossRef] [PubMed]
  243. Madan, B.; Ke, Z.; Harmston, N.; Ho, S.Y.; Frois, A.O.; Alam, J.; Jeyaraj, D.A.; Pendharkar, V.; Ghosh, K.; Virshup, I.H.; et al. Wnt addiction of genetically defined cancers reversed by PORCN inhibition. Oncogene 2016, 35, 2197–2207. [Google Scholar] [CrossRef] [PubMed]
  244. Suwala, A.K.; Koch, K.; Rios, D.H.; Aretz, P.; Uhlmann, C.; Ogorek, I.; Felsberg, J.; Reifenberger, G.; Kohrer, K.; Deenen, R.; et al. Inhibition of Wnt/beta-catenin signaling downregulates expression of aldehyde dehydrogenase isoform 3A1 (ALDH3A1) to reduce resistance against temozolomide in glioblastoma in vitro. Oncotarget 2018, 9, 22703–22716. [Google Scholar] [CrossRef] [PubMed]
  245. Hojjat-Farsangi, M. Targeting non-receptor tyrosine kinases using small molecule inhibitors: An overview of recent advances. J. Drug Target. 2016, 24, 192–211. [Google Scholar] [CrossRef]
  246. Gotze, S.; Coersmeyer, M.; Muller, O.; Sievers, S. Histone deacetylase inhibitors induce attenuation of Wnt signaling and TCF7L2 depletion in colorectal carcinoma cells. Int. J. Oncol. 2014, 45, 1715–1723. [Google Scholar] [CrossRef]
  247. Prabhu, V.V.; Lulla, A.R.; Madhukar, N.S.; Ralff, M.D.; Zhao, D.; Kline, C.L.B.; Van den Heuvel, A.P.J.; Lev, A.; Garnett, M.J.; McDermott, U.; et al. Cancer stem cell-related gene expression as a potential biomarker of response for first-in-class imipridone ONC201 in solid tumors. PLoS ONE 2017, 12, e0180541. [Google Scholar] [CrossRef]
  248. Rampazzo, E.; Manfreda, L.; Bresolin, S.; Cani, A.; Mariotto, E.; Bortolozzi, R.; Della Puppa, A.; Viola, G.; Persano, L. Histone Deacetylase Inhibitors Impair Glioblastoma Cell Motility and Proliferation. Cancers 2022, 14, 1897. [Google Scholar] [CrossRef]
  249. Gotze, S.; Wolter, M.; Reifenberger, G.; Muller, O.; Sievers, S. Frequent promoter hypermethylation of Wnt pathway inhibitor genes in malignant astrocytic gliomas. Int. J. Cancer 2010, 126, 2584–2593. [Google Scholar] [CrossRef]
  250. Mazieres, J.; He, B.; You, L.; Xu, Z.; Lee, A.Y.; Mikami, I.; Reguart, N.; Rosell, R.; McCormick, F.; Jablons, D.M. Wnt inhibitory factor-1 is silenced by promoter hypermethylation in human lung cancer. Cancer Res. 2004, 64, 4717–4720. [Google Scholar] [CrossRef]
  251. Liao, H.; Li, X.; Zhao, L.; Wang, Y.; Wang, X.; Wu, Y.; Zhou, X.; Fu, W.; Liu, L.; Hu, H.G.; et al. A PROTAC peptide induces durable beta-catenin degradation and suppresses Wnt-dependent intestinal cancer. Cell Discov. 2020, 6, 35. [Google Scholar] [CrossRef]
  252. Yu, J.; Chen, L.; Cui, B.; Widhopf, G.F., 2nd; Shen, Z.; Wu, R.; Zhang, L.; Zhang, S.; Briggs, S.P.; Kipps, T.J. Wnt5a induces ROR1/ROR2 heterooligomerization to enhance leukemia chemotaxis and proliferation. J. Clin. Investig. 2016, 126, 585–598. [Google Scholar] [CrossRef]
  253. Berger, C.; Sommermeyer, D.; Hudecek, M.; Berger, M.; Balakrishnan, A.; Paszkiewicz, P.J.; Kosasih, P.L.; Rader, C.; Riddell, S.R. Safety of targeting ROR1 in primates with chimeric antigen receptor-modified T cells. Cancer Immunol. Res. 2015, 3, 206–216. [Google Scholar] [CrossRef]
  254. Antibody-drug conjugates for cancer score with ROR1. Nat. Biotechnol. 2021, 39, 10. [CrossRef] [PubMed]
  255. Vaisitti, T.; Arruga, F.; Vitale, N.; Lee, T.T.; Ko, M.; Chadburn, A.; Braggio, E.; Di Napoli, A.; Iannello, A.; Allan, J.N.; et al. ROR1 targeting with the antibody-drug conjugate VLS-101 is effective in Richter syndrome patient-derived xenograft mouse models. Blood 2021, 137, 3365–3377. [Google Scholar] [CrossRef] [PubMed]
  256. He, B.; Reguart, N.; You, L.; Mazieres, J.; Xu, Z.; Lee, A.Y.; Mikami, I.; McCormick, F.; Jablons, D.M. Blockade of Wnt-1 signaling induces apoptosis in human colorectal cancer cells containing downstream mutations. Oncogene 2005, 24, 3054–3058. [Google Scholar] [CrossRef]
  257. He, B.; You, L.; Uematsu, K.; Xu, Z.; Lee, A.Y.; Matsangou, M.; McCormick, F.; Jablons, D.M. A monoclonal antibody against Wnt-1 induces apoptosis in human cancer cells. Neoplasia 2004, 6, 7–14. [Google Scholar] [CrossRef] [PubMed]
  258. You, L.; He, B.; Xu, Z.; Uematsu, K.; Mazieres, J.; Fujii, N.; Mikami, I.; Reguart, N.; McIntosh, J.K.; Kashani-Sabet, M.; et al. An anti-Wnt-2 monoclonal antibody induces apoptosis in malignant melanoma cells and inhibits tumor growth. Cancer Res. 2004, 64, 5385–5389. [Google Scholar] [CrossRef] [PubMed]
  259. Hanaki, H.; Yamamoto, H.; Sakane, H.; Matsumoto, S.; Ohdan, H.; Sato, A.; Kikuchi, A. An anti-Wnt5a antibody suppresses metastasis of gastric cancer cells in vivo by inhibiting receptor-mediated endocytosis. Mol. Cancer Ther. 2012, 11, 298–307. [Google Scholar] [CrossRef] [PubMed]
  260. Fontenot, E.; Rossi, E.; Mumper, R.; Snyder, S.; Siamakpour-Reihani, S.; Ma, P.; Hilliard, E.; Bone, B.; Ketelsen, D.; Santos, C.; et al. A novel monoclonal antibody to secreted frizzled-related protein 2 inhibits tumor growth. Mol. Cancer Ther. 2013, 12, 685–695. [Google Scholar] [CrossRef] [PubMed]
  261. DeAlmeida, V.I.; Miao, L.; Ernst, J.A.; Koeppen, H.; Polakis, P.; Rubinfeld, B. The soluble wnt receptor Frizzled8CRD-hFc inhibits the growth of teratocarcinomas in vivo. Cancer Res. 2007, 67, 5371–5379. [Google Scholar] [CrossRef]
  262. Jimeno, A.; Gordon, M.; Chugh, R.; Messersmith, W.; Mendelson, D.; Dupont, J.; Stagg, R.; Kapoun, A.M.; Xu, L.; Uttamsingh, S.; et al. A First-in-Human Phase I Study of the Anticancer Stem Cell Agent Ipafricept (OMP-54F28), a Decoy Receptor for Wnt Ligands, in Patients with Advanced Solid Tumors. Clin. Cancer Res. 2017, 23, 7490–7497. [Google Scholar] [CrossRef]
  263. Diamond, J.R.; Becerra, C.; Richards, D.; Mita, A.; Osborne, C.; O’Shaughnessy, J.; Zhang, C.; Henner, R.; Kapoun, A.M.; Xu, L.; et al. Phase Ib clinical trial of the anti-frizzled antibody vantictumab (OMP-18R5) plus paclitaxel in patients with locally advanced or metastatic HER2-negative breast cancer. Breast Cancer Res. Treat. 2020, 184, 53–62. [Google Scholar] [CrossRef]
  264. Gurney, A.; Axelrod, F.; Bond, C.J.; Cain, J.; Chartier, C.; Donigan, L.; Fischer, M.; Chaudhari, A.; Ji, M.; Kapoun, A.M.; et al. Wnt pathway inhibition via the targeting of Frizzled receptors results in decreased growth and tumorigenicity of human tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 11717–11722. [Google Scholar] [CrossRef]
  265. Fukukawa, C.; Hanaoka, H.; Nagayama, S.; Tsunoda, T.; Toguchida, J.; Endo, K.; Nakamura, Y.; Katagiri, T. Radioimmunotherapy of human synovial sarcoma using a monoclonal antibody against FZD10. Cancer Sci. 2008, 99, 432–440. [Google Scholar] [CrossRef]
  266. Nagayama, S.; Fukukawa, C.; Katagiri, T.; Okamoto, T.; Aoyama, T.; Oyaizu, N.; Imamura, M.; Toguchida, J.; Nakamura, Y. Therapeutic potential of antibodies against FZD 10, a cell-surface protein, for synovial sarcomas. Oncogene 2005, 24, 6201–6212. [Google Scholar] [CrossRef] [PubMed]
  267. Nielsen, T.O.; Poulin, N.M.; Ladanyi, M. Synovial sarcoma: Recent discoveries as a roadmap to new avenues for therapy. Cancer Discov. 2015, 5, 124–134. [Google Scholar] [CrossRef] [PubMed]
  268. Zhang, M.; Haughey, M.; Wang, N.Y.; Blease, K.; Kapoun, A.M.; Couto, S.; Belka, I.; Hoey, T.; Groza, M.; Hartke, J.; et al. Targeting the Wnt signaling pathway through R-spondin 3 identifies an anti-fibrosis treatment strategy for multiple organs. PLoS ONE 2020, 15, e0229445. [Google Scholar] [CrossRef] [PubMed]
  269. Parepally, J.M.; Mandula, H.; Smith, Q.R. Brain uptake of nonsteroidal anti-inflammatory drugs: Ibuprofen, flurbiprofen, and indomethacin. Pharm. Res. 2006, 23, 873–881. [Google Scholar] [CrossRef]
  270. Zhang, X.; Morham, S.G.; Langenbach, R.; Young, D.A. Malignant transformation and antineoplastic actions of nonsteroidal antiinflammatory drugs (NSAIDs) on cyclooxygenase-null embryo fibroblasts. J. Exp. Med. 1999, 190, 451–459. [Google Scholar] [CrossRef] [PubMed]
  271. 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 Oncol. 2015, 17, 266–273. [Google Scholar] [CrossRef]
  272. Solomon, S.D.; McMurray, J.J.; Pfeffer, M.A.; Wittes, J.; Fowler, R.; Finn, P.; Anderson, W.F.; Zauber, A.; Hawk, E.; Bertagnolli, M.; et al. Cardiovascular risk associated with celecoxib in a clinical trial for colorectal adenoma prevention. N. Engl. J. Med. 2005, 352, 1071–1080. [Google Scholar] [CrossRef] [PubMed]
  273. Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803. [Google Scholar] [CrossRef] [PubMed]
  274. Palmer, H.G.; Gonzalez-Sancho, J.M.; Espada, J.; Berciano, M.T.; Puig, I.; Baulida, J.; Quintanilla, M.; Cano, A.; de Herreros, A.G.; Lafarga, M.; et al. Vitamin D(3) promotes the differentiation of colon carcinoma cells by the induction of E-cadherin and the inhibition of beta-catenin signaling. J. Cell Biol. 2001, 154, 369–387. [Google Scholar] [CrossRef]
  275. Shah, S.; Hecht, A.; Pestell, R.; Byers, S.W. Trans-repression of beta-catenin activity by nuclear receptors. J. Biol. Chem. 2003, 278, 48137–48145. [Google Scholar] [CrossRef]
  276. Shah, S.; Pishvaian, M.J.; Easwaran, V.; Brown, P.H.; Byers, S.W. The role of cadherin, beta-catenin, and AP-1 in retinoid-regulated carcinoma cell differentiation and proliferation. J. Biol. Chem. 2002, 277, 25313–25322. [Google Scholar] [CrossRef] [PubMed]
  277. Wieland, A.; Trageser, D.; Gogolok, S.; Reinartz, R.; Hofer, H.; Keller, M.; Leinhaas, A.; Schelle, R.; Normann, S.; Klaas, L.; et al. Anticancer effects of niclosamide in human glioblastoma. Clin. Cancer Res. 2013, 19, 4124–4136. [Google Scholar] [CrossRef]
  278. Venerando, A.; Girardi, C.; Ruzzene, M.; Pinna, L.A. Pyrvinium pamoate does not activate protein kinase CK1, but promotes Akt/PKB down-regulation and GSK3 activation. Biochem. J. 2013, 452, 131–137. [Google Scholar] [CrossRef] [PubMed]
  279. Lepourcelet, M.; Chen, Y.N.; France, D.S.; Wang, H.; Crews, P.; Petersen, F.; Bruseo, C.; Wood, A.W.; Shivdasani, R.A. Small-molecule antagonists of the oncogenic Tcf/beta-catenin protein complex. Cancer Cell 2004, 5, 91–102. [Google Scholar] [CrossRef]
  280. Kim, Y.M.; Kahn, M. The role of the Wnt signaling pathway in cancer stem cells: Prospects for drug development. Res. Rep. Biochem. 2014, 4, 1–12. [Google Scholar] [CrossRef] [PubMed]
  281. Huang, S.M.; Mishina, Y.M.; Liu, S.; Cheung, A.; Stegmeier, F.; Michaud, G.A.; Charlat, O.; Wiellette, E.; Zhang, Y.; Wiessner, S.; et al. Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling. Nature 2009, 461, 614–620. [Google Scholar] [CrossRef]
  282. Torres, V.I.; Godoy, J.A.; Inestrosa, N.C. Modulating Wnt signaling at the root: Porcupine and Wnt acylation. Pharmacol. Ther. 2019, 198, 34–45. [Google Scholar] [CrossRef] [PubMed]
  283. Megason, S.G.; McMahon, A.P. A mitogen gradient of dorsal midline Wnts organizes growth in the CNS. Development 2002, 129, 2087–2098. [Google Scholar] [CrossRef]
  284. Zhang, X.; Hao, J. Development of anticancer agents targeting the Wnt/beta-catenin signaling. Am. J. Cancer Res. 2015, 5, 2344–2360. [Google Scholar]
  285. Ford, C.E.; Qian Ma, S.S.; Quadir, A.; Ward, R.L. The dual role of the novel Wnt receptor tyrosine kinase, ROR2, in human carcinogenesis. Int. J. Cancer 2013, 133, 779–787. [Google Scholar] [CrossRef]
  286. Fukuda, T.; Chen, L.; Endo, T.; Tang, L.; Lu, D.; Castro, J.E.; Widhopf, G.F., 2nd; Rassenti, L.Z.; Cantwell, M.J.; Prussak, C.E.; et al. Antisera induced by infusions of autologous Ad-CD154-leukemia B cells identify ROR1 as an oncofetal antigen and receptor for Wnt5a. Proc. Natl. Acad. Sci. USA 2008, 105, 3047–3052. [Google Scholar] [CrossRef] [PubMed]
  287. Kumawat, K.; Gosens, R. WNT-5A: Signaling and functions in health and disease. Cell. Mol. Life Sci. 2016, 73, 567–587. [Google Scholar] [CrossRef] [PubMed]
  288. 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]
  289. 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] [PubMed]
  290. Cho, C.; Smallwood, P.M.; Nathans, J. Reck and Gpr124 Are Essential Receptor Cofactors for Wnt7a/Wnt7b-Specific Signaling in Mammalian CNS Angiogenesis and Blood-Brain Barrier Regulation. Neuron 2017, 95, 1056–1073.e5. [Google Scholar] [CrossRef]
  291. Whelan, R.; Hargaden, G.C.; Knox, A.J.S. Modulating the Blood-Brain Barrier: A Comprehensive Review. Pharmaceutics 2021, 13, 1980. [Google Scholar] [CrossRef]
  292. Reardon, D.A.; Wen, P.Y.; Desjardins, A.; Batchelor, T.T.; Vredenburgh, J.J. Glioblastoma multiforme: An emerging paradigm of anti-VEGF therapy. Expert Opin. Biol. Ther. 2008, 8, 541–553. [Google Scholar] [CrossRef]
  293. Daisy Precilla, S.; Biswas, I.; Kuduvalli, S.S.; Anitha, T.S. Crosstalk between PI3K/AKT/mTOR and WNT/beta-Catenin signaling in GBM—Could combination therapy checkmate the collusion? Cell. Signal. 2022, 95, 110350. [Google Scholar] [CrossRef] [PubMed]
  294. Tai, D.; Wells, K.; Arcaroli, J.; Vanderbilt, C.; Aisner, D.L.; Messersmith, W.A.; Lieu, C.H. Targeting the WNT Signaling Pathway in Cancer Therapeutics. Oncologist 2015, 20, 1189–1198. [Google Scholar] [CrossRef]
  295. Davis, S.L.; Cardin, D.B.; Shahda, S.; Lenz, H.J.; Dotan, E.; O’Neil, B.H.; Kapoun, A.M.; Stagg, R.J.; Berlin, J.; Messersmith, W.A.; et al. A phase 1b dose escalation study of Wnt pathway inhibitor vantictumab in combination with nab-paclitaxel and gemcitabine in patients with previously untreated metastatic pancreatic cancer. Investig. New Drugs 2020, 38, 821–830. [Google Scholar] [CrossRef]
  296. Colaianni, G.; Brunetti, G.; Faienza, M.F.; Colucci, S.; Grano, M. Osteoporosis and obesity: Role of Wnt pathway in human and murine models. World J. Orthop. 2014, 5, 242–246. [Google Scholar] [CrossRef]
  297. Gilbert, M.R.; Gonzalez, J.; Hunter, K.; Hess, K.; Giglio, P.; Chang, E.; Puduvalli, V.; Groves, M.D.; Colman, H.; Conrad, C.; et al. A phase I factorial design study of dose-dense temozolomide alone and in combination with thalidomide, isotretinoin, and/or celecoxib as postchemoradiation adjuvant therapy for newly diagnosed glioblastoma. Neuro Oncol. 2010, 12, 1167–1172. [Google Scholar] [CrossRef] [PubMed]
  298. Halatsch, M.E.; Dwucet, A.; Schmidt, C.J.; Muhlnickel, J.; Heiland, T.; Zeiler, K.; Siegelin, M.D.; Kast, R.E.; Karpel-Massler, G. In Vitro and Clinical Compassionate Use Experiences with the Drug-Repurposing Approach CUSP9v3 in Glioblastoma. Pharmaceuticals 2021, 14, 1241. [Google Scholar] [CrossRef]
  299. Romo-Perez, A.; Dominguez-Gomez, G.; Chavez-Blanco, A.; Taja-Chayeb, L.; Gonzalez-Fierro, A.; Garcia-Martinez, E.; Correa-Basurto, J.; Duenas-Gonzalez, A. BAPST. A Combo of Common Use Drugs as Metabolic Therapy for Cancer: A Theoretical Proposal. Curr. Mol. Pharmacol. 2022, 15, 815–831. [Google Scholar] [CrossRef]
  300. Slavc, I.; Mayr, L.; Stepien, N.; Gojo, J.; Aliotti Lippolis, M.; Azizi, A.A.; Chocholous, M.; Baumgartner, A.; Hedrich, C.S.; Holm, S.; et al. Improved Long-Term Survival of Patients with Recurrent Medulloblastoma Treated with a "MEMMAT-like" Metronomic Antiangiogenic Approach. Cancers 2022, 14, 5128. [Google Scholar] [CrossRef]
  301. Przystal, J.M.; Cianciolo Cosentino, C.; Yadavilli, S.; Zhang, J.; Laternser, S.; Bonner, E.R.; Prasad, R.; Dawood, A.A.; Lobeto, N.; Chin Chong, W.; et al. Imipridones affect tumor bioenergetics and promote cell lineage differentiation in diffuse midline gliomas. Neuro Oncol. 2022, 24, 1438–1451. [Google Scholar] [CrossRef] [PubMed]
  302. Cantor, E.; Wierzbicki, K.; Tarapore, R.S.; Ravi, K.; Thomas, C.; Cartaxo, R.; Nand Yadav, V.; Ravindran, R.; Bruzek, A.K.; Wadden, J.; et al. Serial H3K27M cell-free tumor DNA (cf-tDNA) tracking predicts ONC201 treatment response and progression in diffuse midline glioma. Neuro Oncol. 2022, 24, 1366–1374. [Google Scholar] [CrossRef]
  303. 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]
  304. Stein, M.N.; Malhotra, J.; Tarapore, R.S.; Malhotra, U.; Silk, A.W.; Chan, N.; Rodriguez, L.; Aisner, J.; Aiken, R.D.; Mayer, T.; et al. Safety and enhanced immunostimulatory activity of the DRD2 antagonist ONC201 in advanced solid tumor patients with weekly oral administration. J. Immunother. Cancer 2019, 7, 136. [Google Scholar] [CrossRef] [PubMed]
  305. Henry, C.E.; Llamosas, E.; Djordjevic, A.; Hacker, N.F.; Ford, C.E. Migration and invasion is inhibited by silencing ROR1 and ROR2 in chemoresistant ovarian cancer. Oncogenesis 2016, 5, e226. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Graphic summarizing the main molecular players involved in the transduction and activation of the canonical (left panels) and non-canonical Wnt signaling cascades (right panels). In the canonical Wnt signaling, the absence of Wnt ligands allows the combination of Axin and APC to recruit GSK-3β and CK1α, which then phosphorylate β-catenin, targeting it to proteasomal degradation through β-TrCP. Upon FZD stimulation induced by Wnt ligands, DVL is recruited to the cell membrane, thus providing a site for Axin and GSK-3β to bind and phosphorylate LRP5/6, finally preventing the formation of the destruction complex. This allows the accumulation of nuclear β-catenin, which activates gene transcription through the binding with a series of transcription factors (TCF/LEF) and transcriptional co-activators (i.e., CBP and p300). The activation of downstream non-canonical Wnt pathways is almost independent on β-catenin. In the Wnt/Ca2+ signaling, Wnt ligands stimulate the concomitant activation of FZD receptors and RYK/ROR co-receptors leading to the activation of a PLC-dependent molecular cascade involving the activation of PDE, Inositol 1,4,5-Trisphosphate Receptor (IP3), Ca2+ release from the Endoplasmic Reticulum and the eventual stimulation of CamK II and Calcineurin. This signal is then transduced into a specific gene transcription pattern through the Nuclear Factor Of Activated T Cells (NFAT) transcription factor. On the other hand, in the Wnt/PCP signaling, recruited DVL proteins form a complex with DAAM which concomitantly stimulates the RHOA cascade and a RAK-dependent JNK activation, finally resulting in the recruitment of the JUN transcription factor. Created with Adobe Illustrator.
Figure 1. Graphic summarizing the main molecular players involved in the transduction and activation of the canonical (left panels) and non-canonical Wnt signaling cascades (right panels). In the canonical Wnt signaling, the absence of Wnt ligands allows the combination of Axin and APC to recruit GSK-3β and CK1α, which then phosphorylate β-catenin, targeting it to proteasomal degradation through β-TrCP. Upon FZD stimulation induced by Wnt ligands, DVL is recruited to the cell membrane, thus providing a site for Axin and GSK-3β to bind and phosphorylate LRP5/6, finally preventing the formation of the destruction complex. This allows the accumulation of nuclear β-catenin, which activates gene transcription through the binding with a series of transcription factors (TCF/LEF) and transcriptional co-activators (i.e., CBP and p300). The activation of downstream non-canonical Wnt pathways is almost independent on β-catenin. In the Wnt/Ca2+ signaling, Wnt ligands stimulate the concomitant activation of FZD receptors and RYK/ROR co-receptors leading to the activation of a PLC-dependent molecular cascade involving the activation of PDE, Inositol 1,4,5-Trisphosphate Receptor (IP3), Ca2+ release from the Endoplasmic Reticulum and the eventual stimulation of CamK II and Calcineurin. This signal is then transduced into a specific gene transcription pattern through the Nuclear Factor Of Activated T Cells (NFAT) transcription factor. On the other hand, in the Wnt/PCP signaling, recruited DVL proteins form a complex with DAAM which concomitantly stimulates the RHOA cascade and a RAK-dependent JNK activation, finally resulting in the recruitment of the JUN transcription factor. Created with Adobe Illustrator.
Biology 12 00729 g001
Figure 2. (A) Graphic representing the developing neural tube whose anteroposterior patterning is dictated by a Wnt signaling activation gradient. (B) Schematic representation of the NSC niche in which Wnt ligands, released by astrocytes, induce their neuronal specification. Created with Adobe Illustrator. A few of the icons were sourced from BioRender.com (accessed on 26 April 2023).
Figure 2. (A) Graphic representing the developing neural tube whose anteroposterior patterning is dictated by a Wnt signaling activation gradient. (B) Schematic representation of the NSC niche in which Wnt ligands, released by astrocytes, induce their neuronal specification. Created with Adobe Illustrator. A few of the icons were sourced from BioRender.com (accessed on 26 April 2023).
Biology 12 00729 g002
Figure 3. Graphic summarizing the differential response of Wnt signaling activation in GBM cells upon modulation of oxygen (O2) tension. In normoxic conditions, Wnt signaling activation promotes the translocation of β-catenin into the nucleus, where it associates with TCF/LEF transcription factors to promote gene transcription, resulting in the maintenance and function of GSCs, EMT, and resistance to chemotherapy and radiation. In hypoxia, however, HIF-1α translocates into the nucleus and interacts with TCF1 and β-catenin, promoting neuronal differentiation and neurogenesis. Created with BioRender.com (accessed on 3 May 2023).
Figure 3. Graphic summarizing the differential response of Wnt signaling activation in GBM cells upon modulation of oxygen (O2) tension. In normoxic conditions, Wnt signaling activation promotes the translocation of β-catenin into the nucleus, where it associates with TCF/LEF transcription factors to promote gene transcription, resulting in the maintenance and function of GSCs, EMT, and resistance to chemotherapy and radiation. In hypoxia, however, HIF-1α translocates into the nucleus and interacts with TCF1 and β-catenin, promoting neuronal differentiation and neurogenesis. Created with BioRender.com (accessed on 3 May 2023).
Biology 12 00729 g003
Figure 4. Graphic displaying the referenced Wnt-dependent remodeling of the microenvironment in GBM. GBM cells secrete HGF, which induces EC transformation into MSC-like cells by accumulating nuclear β-catenin. This transformation enhances the survival of both GBM cells and ECs upon chemotherapy. In addition, tumor cells release WISP1, which promotes the survival of M2 TAMs by activating AKT signaling. Furthermore, Wnt-3a induces the maturation of microglia into M2-activated microglia, which contributes to a pro-inflammatory and immunosuppressive environment that supports the growth of GBM cells. Together, these mechanisms allow GBM cells to shape their microenvironment in a way that sustains their growth and survival. Created with BioRender.com (accessed on 3 May 2023).
Figure 4. Graphic displaying the referenced Wnt-dependent remodeling of the microenvironment in GBM. GBM cells secrete HGF, which induces EC transformation into MSC-like cells by accumulating nuclear β-catenin. This transformation enhances the survival of both GBM cells and ECs upon chemotherapy. In addition, tumor cells release WISP1, which promotes the survival of M2 TAMs by activating AKT signaling. Furthermore, Wnt-3a induces the maturation of microglia into M2-activated microglia, which contributes to a pro-inflammatory and immunosuppressive environment that supports the growth of GBM cells. Together, these mechanisms allow GBM cells to shape their microenvironment in a way that sustains their growth and survival. Created with BioRender.com (accessed on 3 May 2023).
Biology 12 00729 g004
Table 1. Summary of Wnt signaling inhibitors mentioned within this review.
Table 1. Summary of Wnt signaling inhibitors mentioned within this review.
ClassAgentProposed Mechanism/TargetReferences
NSAIDsaspirin and indomethacinreduction of β-catenin/TCFs transcriptional activity[180,181,182,183,184]
sulindacreduction of β-catenin nuclear localization[185,186,187]
celecoxib and diclofenacdegradation of TCFs[46,188,189,190]
Natural compoundsretinoidsincrease of Dab-2 and Axin[191]
vitamin Dincrease of DKK-1 and 4[192]
DIF-1 and 3GSK3-β and cyclin D1[193,194]
curcumin and shikoninβ-catenin activation[195,196,197]
trichosantinmodulation of LGR5[198]
diallyl trisulfidemodulation of LRP6[199]
Rhodiola crenulata and resveratrolreduction of β-catenin nuclear localization[200,201]
quercetindestruction of β-catenin/TCFs binding[202,203]
EGCGincrease of Wnt transcriptional repressors[204,205]
BIO, BIA, and IOinhibition of GSK-3[206]
NAR and PGLantagonism on Wnt receptors[207]
Small
molecules
niclosamidereduction of β-catenin nuclear localization[208,209]
pyrvinium pamoatereduction of β-catenin transcriptional activity[210]
quetiapinereduction of GSK-3β phosphorylation[211]
pioglitazonereduction of β-catenin expression[212]
PNU 74654 and 2,4-diamino-quinazolineinhibition of β-catenin/TCF4 binding[213,214,215,216,217]
ICG-001, PRI-724 and PMED-1inhibition of β-catenin/CBP binding[218,219,220,221]
E7386unknown[222]
BC2059 and MSABreduction of active β-catenin [221,223,224]
CGP049090, CWP232228, and LF3inhibition of β-catenin/TCF4 binding[225,226,227]
SAH-BCL9inhibition of β-catenin/BCL9 binding[228]
NSC668036, FJ9, and 3289-8625inhibition of DVL[229,230,231]
IWRs and SEN46inhibition of Axin destruction[232,233,234]
XAV939, AZ1366, G007-LK, and NVP-TNKS656inhibition of Tankyrase[136,235,236,237,238,239]
IWPs, LGK974, ETC-159, WNT-C59, and GNF-62inhibition of PORCN[94,133,232,240,241,242,243,244]
KAN 0439834inhibition of ROR1[245]
ONC201 and SAHAreduction of multiple Wnt signaling components[246,247,248]
azacytidineincrease of Wnt inhibitor expression[249,250]
PROTAC β-catenininduction of β-catenin degradation[251]
AntibodiesCirmtuzumabreduction of Wnt-5a/ROR signaling[252]
CAR-T cellstargeting of ROR1[253]
VLS-101 and NBE-002ADCs targeting ROR1[254,255]
anti-Wnt-1 mAbtargeting of Wnt-1[256,257]
anti-Wnt-2 mAbtargeting of Wnt-2[258]
pAb5a-5targeting of Wnt-5a[259]
SFRP2 mAbtargeting of SFRP2[260]
F8CRDhFc and Ipafricepttargeting of FZD8[261,262]
Vantictumabtargeting multiple FZDs[263,264]
OTSA101-DPTA-90Y, TT641 and MAb 92-13targeting of FZD10[265,266,267]
OMP-131R10targeting of RSPO3[268]
Table 2. Summary of ongoing and recently completed (within 5 years) clinical trials involving the use of previously described Wnt signaling inhibitors in the context of brain tumors. “Active” means a clinical trial that is formally active, but has not yet recruited patients. mut: mutated.
Table 2. Summary of ongoing and recently completed (within 5 years) clinical trials involving the use of previously described Wnt signaling inhibitors in the context of brain tumors. “Active” means a clinical trial that is formally active, but has not yet recruited patients. mut: mutated.
DrugBrain TumorRecruitment StatusPhaseReferences
CelecoxibLow and high-grade gliomasCompletedINCT02115074
GlioblastomaCompletedIINCT00112502[271,297]
Recurrent glioblastomaCompletedI–IINCT02770378[298,299]
Recurrent MB, EPD and ATRTRecruitingIINCT01356290[300]
CurcuminHigh-grade gliomasRecruitingI–IINCT05768919
ETC-159Unresectable refractory solid tumorsRecruitingINCT02521844
ONC201Diffuse gliomasRecruitingIINCT05009992[301]
IIINCT05580562
IIINCT05476939
Recurrent H3K27M-mut gliomaActiveIINCT03295396
H3K27M-mut gliomasActiveINCT03416530[302]
IINCT02525692[303]
Advanced solid tumorsCompletedINCT02250781[304]
SAHADiffuse intrinsic pontine gliomaActiveCompletedINCT02420613
I–IINCT01189266
High-grade gliomaActiveII–IIINCT01236560
INCT00268385
Recurrent glioblastomaCompletedActiveI–IINCT01266031
I–IINCT00555399
GlioblastomaCompletedActiveI–IINCT00731731
INCT03426891
Embryonal tumors of the CNSCompletedINCT00867178
AzacytidineRecurrent gliomas (IDH1/2-mut)RecruitingIINCT03666559
Gliomas (IDH1-mut)CompletedI–IINCT03684811
GlioblastomaCompletedINCT02223052
Recurrent posterior fossa EPDRecruitingINCT03572530
Early INCT04958486
Recurrent/refractory pediatric brain tumorsActiveINCT03206021
OMP-131R10Refractory solid tumorsCompletedINCT02482441
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Manfreda, L.; Rampazzo, E.; Persano, L. Wnt Signaling in Brain Tumors: A Challenging Therapeutic Target. Biology 2023, 12, 729. https://doi.org/10.3390/biology12050729

AMA Style

Manfreda L, Rampazzo E, Persano L. Wnt Signaling in Brain Tumors: A Challenging Therapeutic Target. Biology. 2023; 12(5):729. https://doi.org/10.3390/biology12050729

Chicago/Turabian Style

Manfreda, Lorenzo, Elena Rampazzo, and Luca Persano. 2023. "Wnt Signaling in Brain Tumors: A Challenging Therapeutic Target" Biology 12, no. 5: 729. https://doi.org/10.3390/biology12050729

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