Next Article in Journal
Pharmacological Potential of 3-Benzazepines in NMDAR-Linked Pathophysiological Processes
Next Article in Special Issue
Anionic Phospholipids Shift the Conformational Equilibrium of the Selectivity Filter in the KcsA Channel to the Conductive Conformation: Predicted Consequences on Inactivation
Previous Article in Journal
Zinc Finger E-Box Binding Homeobox Family: Non-Coding RNA and Epigenetic Regulation in Gliomas
Previous Article in Special Issue
Structural Basis of the Interaction of the G Proteins, Gαi1, Gβ1γ2 and Gαi1β1γ2, with Membrane Microdomains and Their Relationship to Cell Localization and Activity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Evolving Diagnostic and Treatment Strategies for Pediatric CNS Tumors: The Impact of Lipid Metabolism

by
Paula Fernández-García
1,2,†,
Gema Malet-Engra
1,2,†,
Manuel Torres
1,
Derek Hanson
3,
Catalina A. Rosselló
1,2,
Ramón Román
1,2,
Victoria Lladó
1,2 and
Pablo V. Escribá
1,2,*
1
Laboratory of Molecular Cell Biomedicine, University of the Balearic Islands, 07122 Palma de Mallorca, Spain
2
Laminar Pharmaceuticals, Isaac Newton, 07121 Palma de Mallorca, Spain
3
Hackensack Meridian Health, 343 Thornall Street, Edison, NJ 08837, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2023, 11(5), 1365; https://doi.org/10.3390/biomedicines11051365
Submission received: 10 March 2023 / Revised: 21 April 2023 / Accepted: 27 April 2023 / Published: 5 May 2023

Abstract

:
Pediatric neurological tumors are a heterogeneous group of cancers, many of which carry a poor prognosis and lack a “standard of care” therapy. While they have similar anatomic locations, pediatric neurological tumors harbor specific molecular signatures that distinguish them from adult brain and other neurological cancers. Recent advances through the application of genetics and imaging tools have reshaped the molecular classification and treatment of pediatric neurological tumors, specifically considering the molecular alterations involved. A multidisciplinary effort is ongoing to develop new therapeutic strategies for these tumors, employing innovative and established approaches. Strikingly, there is increasing evidence that lipid metabolism is altered during the development of these types of tumors. Thus, in addition to targeted therapies focusing on classical oncogenes, new treatments are being developed based on a broad spectrum of strategies, ranging from vaccines to viral vectors, and melitherapy. This work reviews the current therapeutic landscape for pediatric brain tumors, considering new emerging treatments and ongoing clinical trials. In addition, the role of lipid metabolism in these neoplasms and its relevance for the development of novel therapies are discussed.

1. Introduction

After hematological cancers, pediatric central nervous system (CNS) or neurological tumors, including pediatric brain tumors (PBTs), are the second most common childhood malignancies. The incidence of pediatric neurological cancers is around 3–6 cases per 100,000 children [1,2], one-third of the rate in adults. PBTs are the main cause of death among all childhood cancers [3,4]. Gliomas account for half of PBTs, followed by neuronal tumors (33%) and embryonal tumors (15%). The molecular basis of PBTs may differ considerably from those in adults, although they share common characteristics [5,6]. The updates of the WHO classification of pediatric CNS tumors in 2016 incorporated histological data and information regarding the molecular signature of specific tumor subtypes. The last update in 2021 [5] included the findings in pediatric tumor genomics, showing the rapid transition from a mainly microscopic to a molecular classification [6,7] (Table 1). This helped to define new tumor subgroups and changed the diagnosis and treatment landscape [8,9,10].
Like adult brain tumors, the treatment for pediatric brain and other CNS tumors includes radiotherapy, surgery, and chemotherapy [34]. The challenge posed by pediatric neurological tumors in the context of basic and clinical research accounts for the multiple clinical trials developing chemotherapies that still rely on cytotoxic agents, but also new drugs in development encompass innovative and multidisciplinary approaches, from cancer vaccines to lipid-based therapies. Despite the hurdles that limit the progress of drug-development for PBTs, rapid regulatory changes and close international cooperation are expected to favor PBT research and improve the outlook for patients [35,36]. Currently, improvements in the diagnosis and treatment of PBTs have led to a cure rate above 50% [37], which may be increased with new therapeutic approaches.
Alterations to lipid metabolism are a hallmark of cancer and of brain tumors in particular, whereby lipogenesis, lipid uptake, and lipid storage are upregulated [38,39,40,41,42,43,44]. However, we are only now beginning to understand how this may affect neurological tumor progression in children. In addition, the plasma membrane composition of brain cancer cells differs from that of healthy cells, which has a direct impact on proliferative signal transduction [44,45,46,47,48]. Targeting pathways that regulate lipid metabolism provides a novel strategy to combat PBTs, which will require a better understanding of lipid metabolism.
As such, this review provides an overview of the potential relevance of lipids in the diagnosis, classification and treatment of PBTs. We will summarize the recent advances in the genetics associated with PBTs and how they could be related to lipids, the weight of lipid metabolism in prognosis, the new advances in diagnostic and imaging tools based on lipid content and the endeavors to develop new treatments for PBTs that have entered clinical trials. In this context, we provide a perspective on how understanding the dysregulation of lipid metabolism associated with PBTs may shed light on future diagnostic and therapeutic outcomes.

2. Cytogenetic Alterations and Lipidomic Landscape of Pediatric Neurological Tumors

The classical nomenclature for classifying gliomas as low-grade (LGG) or high-grade (HGG) has been applied in both adults and children. However, in the last decade, it has become increasingly evident that there are key genetic differences between adult (LGGs/HGGs) and pediatric (pLGGs/pHGGs) gliomas in terms of onset, location, clinical outcome and histopathological features [49,50]. Recent advances in analytical techniques enable tumor genome sequencing and methylation profile, paving the way towards precise diagnosis and personalized therapies. The relevance of these molecular characteristics has led to a new classification based on them rather than on histopathological characteristics [6,51], sorting PBTs beyond their designation as pHGG or pLGG. Indeed, the suitability of including the molecular features of the tumors, as shown in Table 1, is seen in the fact that many of the genes involved in the genetic alterations of CNS tumors encode for proteins that are somehow regulated by lipids or, conversely, that modulate lipid metabolism or composition (Table 1, e.g., Dicer1, CD24, SWI/SNF, P53 or Cyclin D1) [11,52].
Although progress has been made in the identification of PBT driver genes and those with diagnostic value, many targets remain to be discovered, in particular genes related to disease progression, response to treatment, metastasis and relapse [53]. While this classification focuses on the genomic perspective, many of the genetic or proteomic alterations could have an impact on the lipidomic landscape.
Pediatric cancer genomes are characterized by a heterogeneous group of genetic alterations that include germline and somatic mutations, gene fusions, deletions, abnormal gene expression, chromosomal rearrangements, and altered methylation patterns (Table 1) [34,54,55,56,57]. The histone H3 mutation status of the tumors has led to the new classification of the different types of tumors in the pediatric-type diffuse high-grade glioma family, considering the associated genetic alterations and modifications to cell signaling (Table 1) [6,58]. There are two main variants of histone H3 identified in pediatric-type diffuse high-grade gliomas besides the H3 wild-type: the K27 alteration (inducing H3.3K27M expression and promoting genomic instability [59]) and the G34V mutation (inducing H3.3G34V expression and in turn affecting methylation at K36 and K27, and inducing gliomagenesis with a worse prognosis [60,61]). Apart from the characterization of the mutation/methylation status of histone H3 and a low proportion of pediatric-type diffuse high-grade gliomas carrying IDH mutations, there are also a series of genes defining the tumor molecular signature that can be altered as a consequence of a constitutional deficiency for DNA repair which may (1) serve as regulators of the lipid metabolism and participate of the change in the tumor lipid composition, such as TP53 [11,12], (2) act as transmembrane proteins whose location and activity are affected by membrane lipid structure and composition, such as NTRK, or (3) be regulated by lipid modifications and enzymes involved in lipid metabolism, e.g., EGFR and MCYN [13,15]. In view of this, the development of potential anti-tumor treatments aimed at modulating lipid metabolism and/or membrane composition is not surprising. On top of that, the retention of a wild-type IDH genotype supports lipid biosynthesis and preserves the antioxidant level’s rise during the tumor growth [14].
Pediatric-type diffuse low-grade glioma family tumors (formerly pLGGs) are essentially as common as malignant gliomas and embryonal tumors but are expected to have a better prognosis [62]. Considering that, even if the 10-year median survival is higher than 90%, the challenges regarding their treatment consist of avoiding recurrences and long-term sequelae after surgery, radio- and chemotherapy. This family includes different tumor types according to their molecular characteristics with different outcomes and signaling pathways altered, non-related to IDH mutations as opposed to their adult counterparts, and frequently associated with an enhanced MAPK pathway and BRAF gene alterations [6,63].
Interestingly, BRAF regulation is closely related to the lipogenic pathway, polyunsaturated free fatty acid (PUFA) metabolism, and lipid droplet accumulation in the cells [16,17], while the MAPK pathway is tightly controlled by lipid membrane modifications that regulate the activity of growth factor receptors as well as signaling intermediates (such as Ras) [18]. Another family of pediatric gliomas also characterized by BRAF mutations and MAPK enhancement is the circumscribed astrocytic gliomas, which include pilocytic astrocytoma (PA), frequently associated with KIAA1549:BRAF gene fusion [64], and pleomorphic xanthoastrocytoma (PAX), commonly carrying BRAF mutations and homozygous CDKN2A deletion [65].
Moreover, mutations in the fibroblast growth factor receptor (FGFR) family have been identified and characterized in different pediatric brain tumor types via large-scale genetic analysis, such as recurrent FGFR1 somatic mutations (N546K and K656E), FGFR1–TACC1 gene fusions, and duplications of the FGFR1 tyrosine kinase domain in patients with PA and dysembryoplastic neuroepithelial tumors (DNETs) [66]. In addition, their membrane location has been associated with malignancy and tumor grade [66]. Finally, these receptors activate a lipid-anchored Grb2-binding protein (FRS2) that ultimately triggers MAPK pathways as well [67].
In addition, there are other signaling pathways than the MAPK axis (activated by BRAF and FGFR mutations, among others) which are altered in the pediatric-type diffuse low-grade gliomas—for example, MYB/MYBL1 amplifications and rearrangements have been identified in diffuse astrocytomas, becoming a new type of tumor in the last WHO classification update.
On the other hand, unlike most adult gliomas, a notable fraction of PBTs have a hereditary component. For example, subependymal giant cell astrocytomas (SEGAs) are closely associated with germline mutations in the TSC1/TSC2 genes, whose defective phenotype could be reversed by inhibiting lipophagy, as has been shown in mouse models, where this inhibition reduces tumorigenesis [20]. In addition, in the ependymal tumor family, the majority of the tumors belong to the supratentorial ependymomas ZFTA fusion-positive containing a C11ORF95-RELA gene fusion, while the remainder harbor fusions involving the YAP1 oncogene (supratentorial ependymomas, YAP1 fusion-positive) [68]. RELA encodes the p65 subunit of the nuclear factor-κB (NF-κB), one regulator of the inflammatory response that can be activated by saturated fatty acids and lipid peroxidation-derived aldehydes [69,70]. On the other hand, YAP1 plays an important role in the lipid metabolism adaptation during tumorigenesis, acting in different pathways such as its activation by stearoyl-CoA-desaturase-1 (SCD1) and its interaction with beta-catenin, among others [71].
Finally, embryonal tumors are a heterogeneous group of malignant neuroepithelial tumors making up about 15–20% of the pediatric CNS tumors (WHO grade 4). Embryonal tumors include medulloblastoma, atypical teratoid rhaboid tumors (ATRTs) and embryonal tumors with multi-layered rosettes (ETMRs) (Table 1, [72]). Medulloblastoma is the most frequent embryonal tumor and the second most common PBT, with frequent MYC amplifications [55]. Fatty acids act as inhibitors for the DNA-binding c-Myc/Mas dimer, being a potential treatment for this type of PBT [28]. The latest classification divided medulloblastoma into four genetically defined subsets based on the oncogenic profile of the CTNNB1/APC (WNT activated), SMO/GLI2 (SHH activated) with or without and TP53 mutation signaling pathways. All of these pathways are modulated by different lipid species and their metabolism or, conversely, as happens with beta-catenin and p53, can control the lipid catabolism (Table 1).

3. Lipid Metabolism and Pediatric Brain Tumor Prognosis

Lipid metabolic reprogramming is an established hallmark of cancer progression and recent findings suggest that it may influence therapeutical response and resistance [73]. In addition to the above-mentioned relationship between the alterations implicated in CNS tumors and the lipid metabolism or lipid composition, other regulation pathways must be considered. Microdomain localization and activity of transmembrane proteins are likely to be affected by lipid membrane composition (Ntrk), as are those proteins that are regulated by lipidation (EGFR, K-Ras), since their lipid modifications show preference for different types of membrane microdomains, which in turn depend on membrane lipid composition [13]. Altered lipid metabolism or content may affect the general activity of cells, influencing several proteins involved in the pathologic profile of CNS tumors. Alterations to the β-oxidation pathway (e.g., β-catenin malfunction) are implicated in general cell metabolism. For example, those affecting the production of acetyl-CoA are involved in the Krebs cycle, fatty acid metabolism, acetylcholine synthesis, acetylation, etc. Accordingly, the regulation of lipid metabolism or membrane lipid composition offers therapeutic advantages with respect to the possible emergence of resistance and possibly greater guarantees of success. Indeed, the clear relationship between the genetic alterations described in pediatric patients with CNS tumors and the alterations in lipid metabolism highlights the importance of lipids as potential targets for new therapies to manage these conditions.
The role of lipids in cancer cell signaling has been highlighted recently and alterations to the expression of genes that influence lipids are at least as relevant to tumorigenesis as those in conventional oncogenes [74].
In oncological research, gene expression datasets have provided relevant information that helps us to understand the molecular basis of different cancers, identify new therapeutic targets and biomarkers, as well as define prognosis [42,75]. However, there are very few PBT datasets available, which limits the potential advances in this field.
In a study of adult gliomas [74], the expression of eleven genes involved in lipid metabolism (e.g., SGMS, FASN, SPHK, etc.) was significantly altered and their profiles were compared to those of eleven oncogenes that were also significantly affected (e.g., AKT, MYC, RAS, etc.). In these patients from the REMBRANDT database, the probability of developing glioma when the expression of one gene that affects lipids is altered was approximately 21%, whereas the probability of developing glioma with alterations to one conventional oncogene was approximately 12% (Figure 1), suggesting that the genes involved in lipid metabolism may have a strong impact on tumorigenesis. Moreover, the median survival time of patients was more strongly affected by alterations to lipid-related genes rather than conventional oncogenes. These results suggest that the altered expression of genes involved in lipid metabolism could have a similar or stronger influence on the development of brain tumors than oncogenes.
Lipids are major structural elements in the brain, with fatty acids making up about 50% of the total mass of CNS membranes. Deregulation of fatty acid uptake and lipid metabolism has been described in malignant adult gliomas, resulting in marked differences in lipid metabolism between LGGs and HGGs [76]. Membrane lipids define the type and abundance of peripheral proteins that bind to membranes, influencing the propagation of signals driving proliferation or other events [46,77]. Recently, cerebrospinal fluid was explored for potential metabolites and proteome biomarkers of childhood brain tumors, showing that 6% of the proteins altered in the cerebrospinal fluid from extraventricular drainage in pediatric and brain tumor samples, compared to control samples, were associated with membranes [78]. Even more importantly, the metabolic and lipidomic profile of cerebrospinal fluid from patients with medulloblastoma, the most common malignant brain tumor in childhood, are distinctive in terms of their TNF-beta, TNF-alpha, and adipogenesis signatures, allowing the classification of patients with medulloblastoma by analyzing their cerebrospinal fluid and comparing it to that of normal patients [79].
To determine the impact of genes involved in lipid metabolism on the survival of pediatric glioma patients, we analyzed the expression of genes controlling membrane lipids. The raw dataset on the XENA website was used (http://xena.ucsc.edu/ (accessed on 8 September 2019)), available at the Array Express data repository of the European Bioinformatics Institute (http://www.ebi.ac.uk/arrayexpress/ (accessed on 8 September 2019)) under the accession number E-TABM-1107 [80]. RNA and clinical data were considered to analyze the genes related to lipid metabolism, and only those patients with a known overall survival (OS) were included. A total of 66 pediatric patients were studied, of whom 17 were censored for the Kaplan–Meier determination, generating Kaplan–Meier curves using the “survival” and “survminer” packages in the R software. Not all of the selected genes associated with lipid metabolism were included in the array (Table 2). For each of the genes selected, patients were divided into “Low” or “High” groups to reflect expressions above or below the median expression of the whole population. Kaplan–Meier curves were produced for all of these genes and significant differences in median OS were evident for five genes related to lipid metabolism based on their expression when the log-rank test was used (SMS2, FADS1, FABP5, GALC and ACSL4; p-value < 0.05: Figure 2).
The SMS2 gene encodes sphingomyelin synthase 2, one of the two isoforms (SMS1 and SMS2) which mediate sphingomyelin (SM) production [81]. SMS2 expression was previously related to the survival of adult glioma patients, where lower expression is associated with a better prognosis [82]. Interestingly, similar results were obtained in pediatric patients (Figure 2A), although unfortunately, the databases used did not contain data on SMS1, which is associated with better prognosis in adult patients. Nevertheless, this represents further evidence that membrane sphingolipids are critical for brain tumor cell proliferation.
FADS1 (Fatty Acid Desaturase 1) enzyme regulates fatty acid unsaturation and controls the metabolism of inflammatory lipids such as prostaglandin E2. Interestingly, low FADS1 expression in pediatric glioma patients correlates with better prognosis, with a higher 2- and 5-year survival, yet these differences were only evident in patients that survived at least 2 years (Figure 2B). Inhibiting FADS impairs cancer cell proliferation, and it has been proposed as a potential antitumor strategy [83,84].
The FABP5 gene encodes a fatty-acid-binding protein that preferentially transports saturated fatty acids and retinoic acid to the cytoplasm and nucleus [85]. FABP5 is expressed in the mid-term embryonic rat brain, peaking at birth before gradually declining in post-natal life. Contrary to FABP5’s role as tumor promoter in some types of cancer, such as cervical cancer or hepatocellular carcinoma [74], and as a malignancy element in adult LGG via NF-κB pathway activation [86], in the pHGG dataset studied, higher FABP5 expression was associated with a better prognosis (Figure 2C), pointing out the specific molecular pattern followed by pediatric CNS tumors.
The GALC gene encodes a lysosomal galactose-ester-bond hydrolase for galactosylceramide, galactosylsphingosine, lactosylceramide and monogalactosyldiglyceride, and constitutes a marker for mature cells of oligodendroglia [87]. Although this protein has been described as a pro-angiogenic factor that may negatively influence cancer progression [88], a positive association was evident between high GALC expression and median survival in pediatric glioma patients (Figure 2D).
The protein encoded by the ACSL4 gene is an isozyme of the long-chain fatty-acid-coenzyme A ligase family converting free long-chain fatty acids into fatty acyl-CoA esters in what is a key reaction in lipid biosynthesis, fatty acid metabolism and fatty acid uptake. Similarly to what has been described in adult patients, high expression of this gene in pediatric patients is associated with a better prognosis [89]. In particular, high ACSL4 expression is associated with a significantly higher 5-year survival (Figure 2E).
Both SMS2 and GALC are involved in sphingolipid metabolism, while FABP5, FADS1 and ACSL4 are involved in fatty acid biosynthesis, metabolism and transport. Targeting sphingolipids has been proposed as a treatment for GBM patients, including pediatric ones [90]. For sphingolipids, low SMS2 expression and high GALC expression are associated with a better prognosis, and both of these conditions are related to an increase in ceramides [91]. In the case of fatty acids, the better prognostic signature of high ACSL4 and FABP5 and low FADS1 (Figure 2E) might be explained by the increased cellular uptake of arachidonic acid at high ACSL4 expression [92] and the increased pool of saturated fatty acids upon high FABP5 expression and low FADS1 expression [93]. Saturated fatty acids represent a more efficient cellular fuel than unsaturated fatty acids, and the higher energetic demands of cancer cells may partially justify this inverse association with pediatric patient survival. Deregulated fatty acid synthesis may contribute to cancer in many ways, modifying membrane lipid composition or the provision of substrates for ATP production, influencing signaling pathways involved in inflammation and in cell differentiation, proliferation, and survival [92]. Thus, some of the genes involved in lipid metabolism studied here showed similar trends in pediatric and adult brain tumors, while FABP5 and GALC expression was unexpectedly related to OS. Hence, certain molecular features of PBTs might differ from those in adult patients, and further studies will be necessary to reveal the full role of genes involved in lipid metabolism in cancer cells. It is not surprising that molecular differences occur between pediatric and adult brain tumors given the increase in genetic aberrations with age. Therefore, a lower mutation frequency and stronger requirement for lipogenesis in the developing brain is expected in children with cancer. Some of these genetic differences between pediatric and adult brain tumors were highlighted above and they anticipate that significantly different treatments may be necessary for PBTs with respect to their adult counterparts. Indeed, it is noteworthy that such a small patient population (only 66 patients evaluated) could display significant differences in the expression of certain genes that might be useful for diagnosis. Additionally, databases containing information from more patients could reveal more genes involved in lipid metabolism that might be associated with survival. The context within which the prognostic genes are integrated in the lipid metabolism landscape is schematically illustrated in Figure 3.
In light of the results that show the relevance of the expression levels of genes related to lipid metabolism in the prognosis of patient survival, it is logical to suggest that another control point of special interest is the transcription factors that regulate the expression of these genes. Examples of transcription factors that regulate the expression of mediators of lipid metabolism are listed here as potential biomarkers of PBT prognosis and survival (Table 3).

4. Neuroimaging of Lipids in Pediatric Brain Tumors

As lipids are a major component of brain tissue, the lipid profile of a cell is a molecular signature or its cell type, and its growth and differentiation status. The lipid composition of brain tumors differs from that of healthy brain tissues and reflects the lipid metabolism reprogramming of cancer cells [115,116,117,118]. Transformed cells have higher lipid uptake and storage (often stored in lipid droplets), increased levels of choline-containing compounds, higher lipid biosynthesis and a lipid-dependent catabolism [44]. The rewiring of lipid metabolism through cancer progress has been widely studied in adults and to a lesser extent in children. Nevertheless, the battery of neuroimaging techniques for the analysis of PBTs has been increasing exponentially in recent years, providing powerful non-invasive tools to monitor the biochemical composition of tumors and providing valuable information for diagnosis, surgery planning and for post-surgery follow-up. The analysis of the protein and lipid profile of tumors is able to (1) distinguish between healthy and tumor tissue, defining the tumor contour, (2) identify and diagnose specific tumor types and sub-type, and (3) monitor changes in the biochemical composition of tumors associated with tumor progression and response to treatments.
Raman spectroscopy has been applied to assess the alterations in the protein and lipid content in tumors, as well as defining their morphology, discriminating between medulloblastoma and healthy tissues [119]. Additionally, this technique has proven useful in discriminating medulloblastoma from several types of low- and high-grade tumors such as astrocytoma, glioma, ganglioglioma and medulloblastoma [120]. For this reason, the metabolic profiles of different types of lipid peaks (sphingomyelin, phospholipids, cholesterol, etc.) are a valuable tool to distinguish healthy brain tissue from malignant tissue as well as to distinguish among different types of tumors.
In this regard, in vivo magnetic resonance spectroscopy (MRS) detects lipid peaks that resonate at 1.3 ppm and 0.9 chemical shift in 1H MR spectra. These peaks correspond to the methylene and methyl signals from CH2 and CH3 groups, respectively, in fatty acyl chains of triacylglycerides. Thus, the lipid peaks indicate tissue damage (necrosis) and release of membrane lipids and of cytoplasmic mobile lipids contained in lipid droplets of intact cancer cells. Instead, the choline-containing compounds present in the membranes do not correspond to those peaks but to molecules participating in phospholipid metabolism [121,122]. Magnetic resonance imaging (MRI) helps to predict the tumor type and grade, while high-resolution proton magnetic angle spinning spectroscopy (HRMAS) has been applied to the study of PBT biopsies. Recent studies using HRMAS have examined the metabolic profile of ependymoma, medulloblastoma and pilocytic astrocytoma samples, demonstrating a correlation between choline-containing lipid compounds with tumor grade (glycerophosphocholine, phosphocholine, choline) [123], supporting the results found in different PBTs using the total choline levels determined by MR spectroscopy as a diagnosis tool for neuropathologies in children [124,125]. Choline is required for phospholipid synthesis and is a marker of cell membrane integrity and density, and increased choline levels are thought to reflect increased cell turnover [126]. The levels of phosphocholine may be associated with changes in membrane composition and structure, which may in turn have an impact on cell proliferation. Indeed, medulloblastoma (grade 4) had higher levels of choline-containing compounds than pilocytic astrocytoma (grade 2) and ependymoma (grade 1), while there were fewer fatty acids as the tumor grade worsened.
Lipidic profiles of tumors obtained by MSI (magnetic spectroscopy imaging) have been used to distinguish medulloblastoma and pineoblastoma, with glycerophosphoglycerols and sphingolipids considered the best markers, respectively [127]. Several studies have shown phosphatidylcholine levels to be significantly higher in medulloblastoma and glioblastoma, and in LGG, than in tissue from healthy individuals. Proton NMR analysis demonstrated that the total cholesterol and choline-containing phospholipid levels of medulloblastoma patients (and other adult brain tumor types) were higher than those in blood serum and tumor tissue from healthy individuals [128].
The tumor lipid levels measured by MR spectroscopy are strongly associated with tumor grade, but importantly, they also predict the survival of children with brain tumors [129]. In the study reported by Wilson et al., in a cohort of 115 pediatric patients diagnosed with different types of PBTs (gliomas and embryonic tumors), lipid levels were correlated with survival but also with the glutamine content in the tumor. This correlation suggests a possible link between tumor development and lipogenesis where glutamine serves as a carbon source (Figure 4).
The levels of lipids alone or in combination with choline-containing compounds (signals from choline, phosphocholine and glycerophosphocholine) were also explored using proton magnetic resonance spectroscopic imaging (MRSI) in 76 children with PBTs of diverse types of malignancies, indicating possible alterations to the phospholipid metabolism of tumor cells [130]. In this in vivo analysis, MRSI was used in conjunction with an analysis of tumor grade via standard histopathology. Brain MRSI measurements in vivo show that tumor lipid levels are associated with tumor grade and predict the survival of children with brain tumors.
In another study reported by Bennet et al., HRMAS was used to obtain the ex vivo metabolite profiles of a cohort of 133 pediatric patients diagnosed with a wide variety of different patient brain tumor types (gliomas, ATRT, medulloblastoma among others). In this study, high intratumor lipid levels measured ex vivo by HRMAS correlated with a poor OS in children, confirming the clinical value of lipid metabolite profiling for PBTs as prognostic markers [131].
Moreover, recent data using proton magnetic resonance spectroscopic imaging (MRSI) of child brain tumors in vivo demonstrated a prediction of the outcome of treatments based on the choline-containing compounds, lipids, lactate, and N-acetyl aspartate peaks [132]. The findings of this study showed that patients who responded to chemotherapy or radiation displayed higher total creatine and lower choline, lactate and lipid levels than the patients who did not respond to treatment or were not treated.
Finally, although there is little information as to how lipid metabolites may be differentially associated with PBT metastasis development, recent evidence suggests that lipids are differential components of metastasizing as opposed to non-metastasizing medulloblastomas in mouse models analyzed via 3D-MALDI MS (three-dimensional matrix-assisted laser desorption/ionization mass spectrometry imaging) [133]. This technique allowed the identification of the spatial distribution of lipids within the metastasis sites including phosphatidylserine, phosphoinositides and phosphatidylethanolamines. This and future studies in this line can not only provide relevant information on the mechanistic steps of the metastasis progress of PBTs, but could also define biomarkers and targets that might prove fruitful for the development of novel therapies.
As a result of the abovementioned studies, the application of imaging plus other metabolomic techniques to the field of pediatric oncology is likely to elucidate the lipid metabolic pathways that contribute to the malignant transformation of PBTs, providing complementary information to standard histopathology and genetics that will surely contribute to the classification and treatment of PBTs in the future.

5. Current Trends in the Treatment of Pediatric Neurological Tumors Targeting the Lipid Metabolism

The treatment of pediatric neurological tumors requires a multi-disciplinary approach that may incorporate interventions involving neurosurgery, radiotherapy, and chemotherapy [34]. The specific treatment for an individual PBT depends on the type, size, and location of the tumor, as well as the child’s age and overall health. Several advances in neurosurgery have improved the success rates of tumor surgery, including image guidance, functional mapping, neuroendoscopy, and ultrasonic aspiration [134,135]. Surgical excision is the initial approach recommended in all cases except those in which the tumor is close to vital structures or due to the infiltration of the tumor, as is the case of diffuse midline glioma H3K27M-mutant (formerly classified as diffuse intrinsic pontine glioma [DIPGs]) [136]. Depending on the type of the tumor, its location, and the percentage of resection achieved, the pediatric patient can be administered radiotherapy, with all of the secondary effects associated and variable efficacy outcomes [137,138,139,140,141,142].
While surgery and radiotherapy were initially the two mainstays of PBT therapy, chemotherapy has taken on a more important role in the past 30 years, although it still often involves the use of non-specific cytotoxic conventional chemotherapy. However, there are determined chemotherapy drugs preferred for each of the different kinds of PBTs [34], during or after the radiotherapy treatment, based on the side effects induced, the signaling pathway targeted by the treatment, and the prognosis and sensibility to the drug of the tumor, which do not need to be related to the outcomes observed in adult brain tumors [136,141,142,143]. For instance, regarding the treatment of the CNS embryonal tumor family, adjuvant lomustine, vincristine, cisplatin and cyclophosphamide are used to treat medulloblastomas, while intensive treatment with alkylating agents, high-dose methotrexate and high-dose chemotherapy with stem cell rescue are used to treat ATRTs and ETMRs [84]. On the other hand, chemotherapy plays a palliative role in diffuse midline glioma H3K27M-mutant tumors, yet there is no established role for systemic chemotherapy to treat craniopharyngioma [144].
Considering that the best approach to deal with the pediatric neurological tumors is still tumor resection as much as possible, and that prognosis is worse in patients with local residual or disseminated disease compared to patients with no evidence of disease after surgery [145,146], new therapies that can the pass through the BBB [147,148,149] to reach and reduce CNS tumors are under study [150,151]. The tight junctions of the endothelial cells in cerebral capillaries impair the mobilization of large polar compounds between the blood and brain tissue. By contrast, small non-polar lipid-soluble compounds rapidly traverse the BBB and hence, in addition to their safety and efficacy, small hydrophobic drugs used in membrane lipid therapy (melitherapy) have tremendous potential to treat pediatric CNS tumors [46,49].
Currently, several targeted therapies are under clinical trials to treat PBTs and other pediatric neurological tumors, yet since 2010–2011, only the mTORC1 inhibitor everolimus has been approved by the EMA and FDA for the treatment of SEGAs [152]. To date, most of the drugs have been tested as off-label treatments and no drugs have been specifically designed for children with neurological tumors from scratch, although there has been much recent progress in the generation of PBT cell models, patient-derived orthotopic xenografts and biobanks [153,154]. For example, a combination of temozolomide, which is used in the standard of care treatment of glioma in adults, with O6-benzylguanine has been investigated for the treatment of children with refractory or recurrent brain tumors (NCT00052780). A wide variety of other new therapies are currently being tested in clinical trials, including mutation-specific targeted therapies (Table 4) [155,156].
Currently, conventional cytotoxic agents (such as temozolomide, platinum acetylacetonate, carboplatin, gemcitabine, cytarabine, vincristine and cisplatin), kinase inhibitors (such as erlotinib, ribociclib and other compounds), immunotherapies (including Nivolumab and bevacizumab, among others), and immunomodulators such as CAR T-cells and vaccines are being used alone or in combination with other therapies in various clinical trials to determine their efficacy in the treatment of PBTs and other pediatric CNS tumors (Table 4). Interestingly, there are different clinical trials to identify the appropriate treatment for the different pediatric neurological tumors according to their molecular features; however, most of them were initiated before the establishment of the new classification criteria and maintain the classic nomenclature (pHGG/pLGG and former type descriptions) (Table 4). Indeed, the Pediatric MATCH Screening Trial searches for correlations between drug efficacy and specific mutations in a series of PBTs and other solid tumors (NCT03155620). Another interesting preclinical initiative involves the use of computational tools to reposition already approved drugs targeting PBT cancer stem cells and stemness signaling pathways [157].
Immunotherapy is also being explored for the treatment of cancer [158]. Thus, bevacizumab, which inhibits the VEGFR, has been shown to be effective against several types of cancer, increasing progression-free survival (PFS) in adult patients with glioma but without significantly enhancing median survival [159]. Depatuxizumab mafadotin (ABT-414) is a drug-conjugated antibody that preferentially binds to EGFR-amplified cells (such as GBM cells), and this immunotherapy, alone or in combination with temozolomide, has been under study in pediatric patients with recurrent GBM, but none of the enrolled patients showed a complete or partial response due to tumor progression, without the possibility of completing the treatment (study results published on clinicaltrials.gov: NCT02343406) [160]. Although the efficacy of the antibody therapies has yet to be determined in the treatment of PBTs, different immunomodulators are also under study. Poly-ICLC has been used to treat pLGGs (NCT01188096), either alone or in combination with synthetic peptides such as neoepitope-based personalized vaccines in patients with recurrent brain tumors (NCT03068832) [161]. Moreover, several trials use chimeric antigen receptor (CAR) T cells for PBTs, such as those directed against antigens such as HER2, EGFR806 and B7-H3, whose elevated expression in PBTs supports their potential as therapeutic targets, as acknowledged by the FDA in 2017 [162]. As for immune checkpoint inhibitors, to date no such inhibitors exist for the treatment of PBTs, although promising results were obtained in the first-in-child phase I clinical trial with indoximod plus temozolomide, with improved clinical outcomes in newly diagnosed DIPG patients (median OS and 12-months OS) (NCT02502708, [163]). Indoximod is currently in phase II studies for the treatment of PBTs (Table 4).
Modified viral tools [164] and several PBT cancer vaccines [165,166] are other approaches currently in phase I studies for the treatment of different pediatric neurological tumors, which is a sign of a new trend of innovative therapeutic development.
Lipids have commonly been used as drug delivery vehicles for other molecules [167,168]. However, during recent years, their potential as therapeutic targets has been unveiled by the relevance of lipid metabolism processes in several types of cancer, including lipogenesis, lipolysis, fatty acid oxidation, lipid uptake, and lipid desaturation [169,170]. One example would be CLR131, a radiolabeled molecule that has shown its ability to cross the BBB and provide preliminary activity in children and adolescents with relapsed or refractory cancers, specifically high-grade gliomas (HGGs) and high-risk neuroblastomas, by targeting the preference among malignant cells for phospholipid ethers (NCT03478462). Despite an increasing number of publications pointing to lipid metabolism as a promising therapeutic target in cancer, translation of this potential into actual clinical trials remains scarce. As reviewed by Yan Fu et al. [171], several molecules targeting fatty acid, cholesterol or phospholipid metabolism are under development, mostly at the preclinical stage (Table 5).
Melitherapy is a promising new approach to combat tumors by means of the use of lipids or hydrophobic molecules modulating the composition/structure of lipid membranes as well as lipid metabolism, with potentially high efficacy and safety, suggesting that it could be an appropriate approach to treat PBTs [46,49,170]. Although these are small molecules amenable to production via chemical synthesis or semi-synthetic procedures, their unique mechanisms of action differ from those of more conventional chemotherapeutic agents. The benefits and safety associated with melitherapy recently led to this approach entering the therapeutic arena to treat PBTs and other CNS tumors. Indeed, two different melitherapy agents are currently in phase I clinical studies (LAM561 and BXQ-350) to confirm the safety profile shown in adults and to determine their efficacy in the treatment of the PBTs.
LAM561 (2-hydroxyoleic acid, 2OHOA) has shown clinical activity in adult patients with glioma and other advanced solid tumors and also proved its safety in phase I and II clinical trials, alone and in combination with TMZ, in contrast with most anticancer drugs (clinicaltrials.gov identifier #NCT01792310 and NCT03867123); in addition, its efficacy is being assessed in a phase II/III trial for the treatment of newly diagnosed glioblastoma in adults in combination with the standard of care (radiotherapy and TMZ, clinicaltrials.gov identifier #NCT04250922). The excellent safety profile and promising efficacy of this hydroxylated fatty acid has led to the investigation of its potential use against PBTs in a phase I/II trial (NCT04299191). Significantly, LAM561 was shown to normalize membrane lipid composition, disrupting the association of key peripheral membrane proteins involved in the propagation of messages that drive cancer cell proliferation [190]. LAM561 triggers macroautophagic death of the glioma cells by inducing a relevant reduction in pRb phosphorylation and dihydrofolate reductase expression [190,191], and by dissociating K-Ras from the plasma membrane, all in all inhibiting the MAPK, CDK and PI3K pathways [192].
On the other hand, BXQ-350 (SapC–DOPS) is a nanosome composed of the lysosomal protein saposin C (SapC) and dioleoylphosphatidylserine (DOPS) that targets the surface PS exposed by cancer cells and ceramide-enriched membranes. It is then internalized by endocytosis and induces cell death (necrosis, apoptosis and autophagy) through lysosome activation [193,194]. In addition, the treatment of cancer cells with BXQ-350 sensitizes the cells to irradiation [195]. After showing a good safety profile and promising clinical activity in a phase I clinical trial in adult patients with refractory solid tumors or HGG (NCT02859857), BXQ-350 is currently being investigated to extend these results in a phase I trial in children with newly diagnosed DIPG or diffuse midline glioma (NCT04771897).
Interestingly, valproic acid, a short branched fatty acid that is used as an anticonvulsant, increases the PFS and OS of pediatric DIPG patients [196]. Its hydrophobic nature suggests that, in addition to histone deacetylase inhibition, it may also act through other mechanisms of action related to melitherapy [197]. Furthermore, this study highlights the efficacy of molecules that regulate lipid metabolism alone or in combination with conventional chemotherapy agents, such as temozolomide, for the treatment of brain tumors.
In general, all drugs whose anticancer mechanism of action targets lipid metabolism or is related to cell membrane modulation could be categorized as melitherapy agents. Further examples are farnesyl transferase inhibitors, that prevent the localization of peripheral signaling proteins bearing a farnesyl moiety to the plasma membrane (e.g., Ras).
Ras is a member of the MAPK axis and is regulated by different RTK and growth factor receptors (mutated in different kind of pediatric CNS tumors, see Table 1), as well as by the H3K27M mutation present in the diffuse midline glioma H3 K27-altered, inducing its aberrant activation [198]. Ras localization at the plasma membrane is necessary to propagate growth signals from tyrosine kinase receptors to Raf and other effectors involved in cancer cell proliferation [199]. In this regard, farnesyl transferase inhibitors prevent the localization of peripheral signaling proteins bearing a farnesyl moiety to the plasma membrane, e.g., Ras, and so could act as melitherapeutic drugs. Indeed, the combination of farnesyl transferase inhibitors, tipifarnib and sorafenib, has been studied in adult patients with GBM [200], while lonafarnib has been investigated in children with recurrent or progressive brain tumors (NCT00015899). Despite their limited efficacy, the lack of serious adverse effects suggests that they could be used in combination with other compounds.
Besides the molecules that have been and are already being explored in patients (adult or pediatric) for the treatment of CNS tumors, there are also different compounds that can be endorsed in melitherapy in preclinical research (Table 5). This is the case for ophiobolin A, which has shown promising results against GB in an orthotopic model, by destabilizing the membrane after covalent modification of phosphatidylethanolamine [201].
Many of these molecules directly affect lipid metabolism or biosynthesis, such as fluoxetine, which inhibits sphingomyelin phosphodiesterase 1 (SMPD1) in GBM cells and prevents the conversion of sphingomyelin to ceramide [179] or YTX-7739 and CAY10566, which act as stearoyl CoA desaturase (SCD) inhibitors and trigger lipotoxicity, impairing de novo lipid synthesis [184]. A combination of GSK126 and atorvastatin, a cholesterol biosynthesis inhibitor, showed good results in a murine DIPG model [173], and LXR-623, an agonist of LXR, has been found to be very effective in killing GBM cells in a xenograft model by depleting cholesterol levels [188]. Fatty acid uptake by pediatric ependymoma cells in a 3D spheroid has been also targeted using GW9662 through inhibiting the brain-lipid-binding protein (BLBP or FABP7) gene expression [176]. In addition to molecules targeting lipid-related pathways, other drugs of a lipid nature are being investigated. Ljungblad et al. showed that omega-3 fatty acids can suppress the growth of tumors in pediatric medulloblastoma cells by altering fatty acid composition and decreasing CRYAB expression levels [177]. In addition, a triple-front approach using LAU-0901 (a PAF agonist), avastin (VEGF suppressor) and elovanoids (synthetic dihydroxylated derivatives of PUFAs acting as lipid mediators) has demonstrated therapeutic efficacy suppressing GBM tumors in orthotopic xenograft mice models [181].
Lipid-metabolism-based therapies may have the advantage over single-protein-based therapies in that there are common lipidomic profiles for several tumor types that promote tumor growth, as described above. However, some of the mutated genes are specific to certain types of diseases and therefore targeting metabolic energy sources or the important energy dependence of PBT may represent an opportunity for therapeutic intervention in these conditions. Furthermore, lipids not only represent a source of energy, but also define the membrane structure and its recruitment ability for peripheral signaling proteins, which can activate or inhibit cancer cell proliferation. Thus, monotherapy or combinational therapy with agents targeting lipid metabolic pathways with antiproliferative drugs could be more efficacious against glioma and may show efficacy against PBTs. Moreover, targeting lipid metabolism-based tumor growth could in turn revert the neoplastic proliferative phenotype, while targeting oncogenes could have an impact on the lipid metabolism reprogramming.

6. Concluding Remarks

Currently, therapies to combat adult cancers are also used against pediatric neurological tumors, yet the standard of care for each type of tumor may differ depending on the molecular characteristics and location of the tumor as well as the age of the patient. The latest data concerning the molecular similarities and differences found between child and adult tumors will have important implications for the treatment of PBTs. Beyond toxic chemotherapeutic compounds, radiotherapy and surgery, innovative approaches based on the molecular signature of pediatric cancers may lead to new, safer and more efficacious therapies. Moreover, lipidic molecules, enzymes related to lipid metabolism, and the lipid composition of tumors represent potential biomarkers for the prognosis and characterization of pediatric CNS tumors, as well as therapeutic targets for a new class of compounds with high safety and potential efficacy in their treatment.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biomedicines11051365/s1.

Author Contributions

Conceptualization, P.F.-G., G.M.-E., D.H. and P.V.E.; methodology, P.F.-G. and G.M.-E.; formal analysis, P.F.-G.; writing—original draft preparation, All Authors; writing—review and editing, G.M.-E., C.A.R., R.R. and V.L.; supervision, P.F.-G. and P.V.E.; funding acquisition, P.V.E. and M.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Govern de les Illes Balears i del Fons social Europeu (ES01/TCAI/53_2016, ES01/TCAI/21_2017, ES01/TCAI/24_2018 and PROCOE/5/2017). The European Commission also supported in part this work (H2020 Framework Programme Project CLINGLIO 755179). This research was also funded by the Ministerio de Economía y Competitividad co-financed by FEDER funds (M.T. and P.V.E.: RTC2019-007399-1). G.M.-E. was supported by a Torres-Quevedo Research contract from the Spanish Ministerio de Economía y Competitividad (PTQ-19-010601) and a Marie Sklodowska-Curie grant (agreement No 101033275). R.R. was supported by a postdoctoral contract Felip Bauça (PD/058/2020) by the Govern de les Illes Balears i del Fons social Europeu (inverteix en el teu futur).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Raw dataset from XENA website is publicily available in the website under the number E-TABM-1107 and was analyzed using the Pedriatic_review_v7.Rmd code file available in the supplementary material of this article.

Acknowledgments

We thank David Castillo for his help with the R code.

Conflicts of Interest

P.F.-G., G.M.-E., M.T., C.A.R., V.L. and P.V.E. declare that they are shareholders in the biotech company, Laminar Pharmaceuticals SL. P.V.E. declares that he is also a shareholder in the biotech companies Pharmaconcept, Neurofix and Ability Therapeutics.

Abbreviations

2OHOA2-hydroxyoleic acid
ATRTatypical teratoid/rhabdoid tumor
ATRXα thalassemia/intellectual disability syndrome X–linked gene
BBBblood–brain barrier
BRAFB-Raf proto-oncogene serine/threonineprotein kinase
CARchimeric antigen receptor
CNScentral nervous system
CSFcerebrospinal fluid
DIPGdiffuse intrinsic pontine glioma
DNETsdysembryoplastic neuroepithelial tumors
ETMRembryonal tumor with multilayer rosettes
FABPfatty-acid-binding protein
GBMsglioblastomas
HGGhigh-grade glioma
LGGlow-grade glioma
MAPKmitogen-activated protein kinase
MRSIproton magnetic resonance spectroscopic imaging
MSmass spectroscopy
NMRnuclear magnetic resonance
OSoverall survival
PApilocytic astrocytoma
PBTspediatric brain tumors
PFSprogression-free survival
pLGG/HGGpediatric low-/high-grade glioma
PXApleomorphic xanthoastrocytoma
SCDstearoyl CoA desaturase
SEGAsubependymal giant cell astrocytomas
SHHSonic Hedgehog
SMOsmoothened: frizzled class receptor
SMsphingomyelin
SUFUsuppressor of fused homolog (Drosophila)
TP53tumor protein p53
WHOWorld Health Organization
WTwild-type

References

  1. Udaka, Y.T.; Packer, R.J. Pediatric Brain Tumors. Neurol Clin. 2018, 36, 533–556. [Google Scholar] [CrossRef]
  2. Ostrom, Q.T.; Gittleman, H.; Xu, J.; Kromer, C.; Wolinsky, Y.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2009–2013. Neuro. Oncol. 2016, 18, v1–v75. [Google Scholar] [CrossRef]
  3. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA. Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef]
  4. Dang, M.; Phillips, P.C. Pediatric Brain Tumors. Continuum (Minneap. Minn.) 2017, 23, 1727–1757. [Google Scholar] [CrossRef]
  5. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A summary. Neuro. Oncol. 2021, 23, 1231–1251. [Google Scholar] [CrossRef]
  6. Pfister, S.M.; Reyes-Múgica, M.; Chan, J.K.C.; Hasle, H.; Lazar, A.J.; Rossi, S.; Ferrari, A.; Jarzembowski, J.A.; Pritchard-Jones, K.; Hill, D.A.; et al. A Summary of the Inaugural WHO Classification of Pediatric Tumors: Transitioning from the Optical into the Molecular Era. Cancer Discov. 2022, 12, 331–355. [Google Scholar] [CrossRef]
  7. Komori, T. The 2021 WHO classification of tumors, 5th edition, central nervous system tumors: The 10 basic principles. Brain Tumor Pathol. 2022, 39, 47–50. [Google Scholar] [CrossRef]
  8. Louis, D.N.; Perry, A.; Reifenberger, G.; von Deimling, A.; Figarella-Branger, D.; Cavenee, W.K.; Ohgaki, H.; Wiestler, O.D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A summary. Acta Neuropathol. 2016, 131, 803–820. [Google Scholar] [CrossRef] [PubMed]
  9. Louis, D.N.; Wesseling, P.; Brandner, S.; Brat, D.J.; Ellison, D.W.; Giangaspero, F.; Hattab, E.M.; Hawkins, C.; Judge, M.J.; Kleinschmidt-DeMasters, B.; et al. Data sets for the reporting of tumors of the central nervous system recommendations from the international collaboration on cancer reporting. Arch. Pathol. Lab. Med. 2020, 144, 196–206. [Google Scholar] [CrossRef] [PubMed]
  10. Fangusaro, J.; Bandopadhayay, P. Advances in the classification and treatment of pediatric brain tumors. Curr. Opin. Pediatr. 2021, 33, 26–32. [Google Scholar] [CrossRef] [PubMed]
  11. Parrales, A.; Iwakuma, T. p53 as a regulator of lipid metabolism in cancer. Int. J. Mol. Sci. 2016, 17, 2074. [Google Scholar] [CrossRef]
  12. Kang, J.G.; Lago, C.U.; Lee, J.E.; Park, J.H.; Donnelly, M.P.; Starost, M.F.; Liu, C.; Kwon, J.; Noguchi, A.C.; Ge, K.; et al. A Mouse Homolog of a Human TP53 Germline Mutation Reveals a Lipolytic Activity of p53. Cell Rep. 2020, 30, 783–792.e5. [Google Scholar] [CrossRef]
  13. Runkle, K.B.; Kharbanda, A.; Stypulkowski, E.; Cao, X.J.; Wang, W.; Garcia, B.A.; Witze, E.S. Inhibition of DHHC20-Mediated EGFR Palmitoylation Creates a Dependence on EGFR Signaling. Mol. Cell 2016, 62, 385–396. [Google Scholar] [CrossRef]
  14. Calvert, A.E.; Chalastanis, A.; Wu, Y.; Hurley, L.A.; Kouri, F.M.; Bi, Y.; Kachman, M.; May, J.L.; Bartom, E.; Hua, Y.; et al. Cancer-Associated IDH1 Promotes Growth and Resistance to Targeted Therapies in the Absence of Mutation. Cell Rep. 2017, 19, 1858–1873. [Google Scholar] [CrossRef]
  15. Qin, X.Y.; Su, T.; Yu, W.; Kojima, S. Lipid desaturation-associated endoplasmic reticulum stress regulates MYCN gene expression in hepatocellular carcinoma cells. Cell Death Dis. 2020, 11, 1–13. [Google Scholar] [CrossRef] [PubMed]
  16. Turner, J.A.; Paton, E.L.; Gulick, R.V.; Stefanoni, D.; Cendali, F.; Reisz, J.; Tobin, R.P.; McCarter, M.; D’Alessandro, A.; Torres, R.M.; et al. BRAF Modulates Lipid Use and Accumulation. Cancers 2022, 14, 2110. [Google Scholar] [CrossRef] [PubMed]
  17. Talebi, A.; Dehairs, J.; Rambow, F.; Rogiers, A.; Nittner, D.; Derua, R.; Vanderhoydonc, F.; Duarte, J.A.G.; Bosisio, F.; Van Den Eynde, K.; et al. Sustained SREBP-1-dependent lipogenesis as a key mediator of resistance to BRAF-targeted therapy. Nat. Commun. 2018, 9, 2500. [Google Scholar] [CrossRef]
  18. Anderson, D.H. Role of lipids in the MAPK signaling pathway. Prog. Lipid Res. 2006, 45, 102–119. [Google Scholar] [CrossRef] [PubMed]
  19. D’Angelo, I.; Welti, S.; Bonneau, F.; Scheffzek, K. A novel bipartite phospholipid-binding module in the neurofibromatosis type 1 protein. EMBO Rep. 2006, 7, 174–179. [Google Scholar] [CrossRef]
  20. Wang, C.; Haas, M.A.; Yang, F.; Yeo, S.; Okamoto, T.; Chen, S.; Wen, J.; Sarma, P.; Plas, D.R.; Guan, J.L. Autophagic lipid metabolism sustains mTORC1 activity in TSC-deficient neural stem cells. Nat. Metab. 2019, 1, 1127–1140. [Google Scholar] [CrossRef]
  21. Priolo, C.; Ricoult, S.J.H.; Khabibullin, D.; Filippakis, H.; Yu, J.; Manning, B.D.; Clish, C.; Henske, E.P. Tuberous sclerosis complex 2 loss increases lysophosphatidylcholine synthesis in lymphangioleiomyomatosis. Am. J. Respir. Cell Mol. Biol. 2015, 53, 33–41. [Google Scholar] [CrossRef]
  22. Lee, H.C.; Ou, C.H.; Huang, Y.C.; Hou, P.C.; Creighton, C.J.; Lin, Y.S.; Hu, C.Y.; Lin, S.C. YAP1 overexpression contributes to the development of enzalutamide resistance by induction of cancer stemness and lipid metabolism in prostate cancer. Oncogene 2021, 40, 2407–2421. [Google Scholar] [CrossRef] [PubMed]
  23. Chinthalapudi, K.; Mandati, V.; Zheng, J.; Sharff, A.J.; Bricogne, G.; Griffin, P.R.; Kissil, J.; Izard, T. Lipid binding promotes the open conformation and tumor-suppressive activity of neurofibromin 2. Nat. Commun. 2018, 9, 1–10. [Google Scholar] [CrossRef] [PubMed]
  24. Gorbenko, O.; Panayotou, G.; Zhyvoloup, A.; Volkova, D.; Gout, I.; Filonenko, V. Identification of novel PTEN-binding partners: PTEN interaction with fatty acid binding protein FABP4. Mol. Cell. Biochem. 2010, 337, 299–305. [Google Scholar] [CrossRef]
  25. Senni, N.; Savall, M.; Cabrerizo Granados, D.; Alves-Guerra, M.C.; Sartor, C.; Lagoutte, I.; Gougelet, A.; Terris, B.; Gilgenkrantz, H.; Perret, C.; et al. β-catenin-activated hepatocellular carcinomas are addicted to fatty acids. Gut 2019, 68, 322–334. [Google Scholar] [CrossRef] [PubMed]
  26. Blassberg, R.; Jacob, J. Lipid metabolism fattens up hedgehog signaling. BMC Biol. 2017, 15, 95. [Google Scholar] [CrossRef]
  27. Xiao, X.; Tang, J.J.; Peng, C.; Wang, Y.; Fu, L.; Qiu, Z.P.; Xiong, Y.; Yang, L.F.; Cui, H.W.; He, X.L.; et al. Cholesterol Modification of Smoothened Is Required for Hedgehog Signaling. Mol. Cell 2017, 66, 154–162.e10. [Google Scholar] [CrossRef]
  28. Casciano, J.C.; Perry, C.; Cohen-Nowak, A.J.; Miller, K.D.; Vande Voorde, J.; Zhang, Q.; Chalmers, S.; Sandison, M.E.; Liu, Q.; Hedley, A.; et al. MYC regulates fatty acid metabolism through a multigenic program in claudin-low triple negative breast cancer. Br. J. Cancer 2020, 122, 868–884. [Google Scholar] [CrossRef]
  29. Ricci, F.; Brunelli, L.; Talapatra, J.; Reddy, M.M. Lipid Metabolic Reprogramming in Embryonal Neoplasms with MYCN Amplification. Cancers 2023, 15, 2144. [Google Scholar] [CrossRef]
  30. Zhang, P.; Li, L.; Bao, Z.; Huang, F. Role of BAF60a/BAF60c in chromatin remodeling and hepatic lipid metabolism. Nutr. Metab. 2016, 13, 30. [Google Scholar] [CrossRef]
  31. Liu, M.X.; Gao, M.; Li, C.Z.; Yu, C.Z.; Yan, H.; Peng, C.; Li, Y.; Li, C.G.; Ma, Z.L.; Zhao, Y.; et al. Dicer1/miR-29/HMGCR axis contributes to hepatic free cholesterol accumulation in mouse non-alcoholic steatohepatitis. Acta Pharmacol. Sin. 2017, 38, 660–671. [Google Scholar] [CrossRef]
  32. Huang, T.C.; Sahasrabuddhe, N.A.; Kim, M.S.; Getnet, D.; Yang, Y.; Peterson, J.M.; Ghosh, B.; Chaerkady, R.; Leach, S.D.; Marchionni, L.; et al. Regulation of lipid metabolism by dicer revealed through SILAC mice. J. Proteome Res. 2012, 11, 2193–2205. [Google Scholar] [CrossRef]
  33. Li, Z.; Martin, M.; Zhang, J.; Huang, H.Y.; Bai, L.; Zhang, J.; Kang, J.; He, M.; Li, J.; Maurya, M.R.; et al. Krüppel-Like Factor 4 Regulation of Cholesterol-25-Hydroxylase and Liver X Receptor Mitigates Atherosclerosis Susceptibility. Circulation 2017, 136, 1315–1320. [Google Scholar] [CrossRef] [PubMed]
  34. Lutz, K.; Jünger, S.T.; Messing-Jünger, M. Essential Management of Pediatric Brain Tumors. Children 2022, 9, 498. [Google Scholar] [CrossRef]
  35. Laetsch, T.W.; Dubois, S.G.; Bender, J.G.; Macy, M.E.; Moreno, L. Opportunities and challenges in drug development for pediatric cancers. Cancer Discov. 2021, 11, 545–559. [Google Scholar] [CrossRef] [PubMed]
  36. Aldape, K.; Brindle, K.M.; Chesler, L.; Chopra, R.; Gajjar, A.; Gilbert, M.R.; Gottardo, N.; Gutmann, D.H.; Hargrave, D.; Holland, E.C.; et al. Challenges to curing primary brain tumours. Nat. Rev. Clin. Oncol. 2019, 16, 509–520. [Google Scholar] [CrossRef]
  37. Girardi, F.; Allemani, C.; Coleman, M.P. Worldwide Trends in Survival From Common Childhood Brain Tumors: A Systematic Review. J. Glob. Oncol. 2019. [Google Scholar] [CrossRef]
  38. Cheng, C.; Geng, F.; Cheng, X.; Guo, D. Lipid metabolism reprogramming and its potential targets in cancer. Cancer Commun. (Lond. Engl.) 2018, 38, 27. [Google Scholar] [CrossRef]
  39. Koundouros, N.; Poulogiannis, G. Reprogramming of fatty acid metabolism in cancer. Br. J. Cancer 2019, 122, 4–22. [Google Scholar] [CrossRef] [PubMed]
  40. Wang, W.; Bai, L.; Li, W.; Cui, J. The Lipid Metabolic Landscape of Cancers and New Therapeutic Perspectives. Front. Oncol. 2020, 10, 2686. [Google Scholar] [CrossRef]
  41. Snaebjornsson, M.T.; Janaki-Raman, S.; Schulze, A. Greasing the Wheels of the Cancer Machine: The Role of Lipid Metabolism in Cancer. Cell Metab. 2020, 31, 62–76. [Google Scholar] [CrossRef]
  42. Santos, C.R.; Schulze, A. Lipid metabolism in cancer. FEBS J. 2012, 279, 2610–2623. [Google Scholar] [CrossRef]
  43. Wang, B.; Tontonoz, P. Phospholipid Remodeling in Physiology and Disease. Annu. Rev. Physiol. 2019, 81, 165–188. [Google Scholar] [CrossRef] [PubMed]
  44. Bian, X.; Liu, R.; Meng, Y.; Xing, D.; Xu, D.; Lu, Z. Lipid metabolism and cancer. J. Exp. Med. 2021, 218, e20201606. [Google Scholar] [CrossRef] [PubMed]
  45. Casares, D.; Escribá, P.V.; Rosselló, C.A. Membrane lipid composition: Effect on membrane and organelle structure, function and compartmentalization and therapeutic avenues. Int. J. Mol. Sci. 2019, 20, 2167. [Google Scholar] [CrossRef]
  46. Escribá, P.V.; Busquets, X.; Inokuchi, J.I.; Balogh, G.; Török, Z.; Horváth, I.; Harwood, J.L.; Vígh, L. Membrane lipid therapy: Modulation of the cell membrane composition and structure as a molecular base for drug discovery and new disease treatment. Prog. Lipid Res. 2015, 59, 38–53. [Google Scholar] [CrossRef]
  47. Agarwala, P.K.; Aneja, R.; Kapoor, S. Lipidomic landscape in cancer: Actionable insights for membrane-based therapy and diagnoses. Med. Res. Rev. 2022, 42, 983–1018. [Google Scholar] [CrossRef]
  48. Preta, G. New Insights Into Targeting Membrane Lipids for Cancer Therapy. Front. Cell Dev. Biol. 2020, 8, 876. [Google Scholar] [CrossRef] [PubMed]
  49. Gröbner, S.N.; Worst, B.C.; Weischenfeldt, J.; Buchhalter, I.; Kleinheinz, K.; Rudneva, V.A.; Johann, P.D.; Balasubramanian, G.P.; Segura-Wang, M.; Brabetz, S.; et al. The landscape of genomic alterations across childhood cancers. Nature 2018, 555, 321–327. [Google Scholar] [CrossRef]
  50. Ma, X.; Liu, Y.; Liu, Y.; Alexandrov, L.B.; Edmonson, M.N.; Gawad, C.; Zhou, X.; Li, Y.; Rusch, M.C.; John, E.; et al. Pan-cancer genome and transcriptome analyses of 1,699 paediatric leukaemias and solid tumours. Nature 2018, 555, 371–376. [Google Scholar] [CrossRef]
  51. Śledzińska, P.; Bebyn, M.G.; Furtak, J.; Kowalewski, J.; Lewandowska, M.A. Prognostic and Predictive Biomarkers in Gliomas. Int. J. Mol. Sci. 2021, 22, 10373. [Google Scholar] [CrossRef] [PubMed]
  52. Fairbridge, N.A.; Southall, T.M.; Ayre, D.C.; Komatsu, Y.; Raquet, P.I.; Brown, R.J.; Randell, E.; Kovacs, C.S.; Christian, S.L. Loss of CD24 in mice leads to metabolic dysfunctions and a reduction in white adipocyte tissue. PLoS ONE 2015, 10, e0141966. [Google Scholar] [CrossRef] [PubMed]
  53. Sweet-Cordero, E.A.; Biegel, J.A. The genomic landscape of pediatric cancers: Implications for diagnosis and treatment. Science 2019, 363, 1170–1175. [Google Scholar] [CrossRef]
  54. Mackay, A.; Burford, A.; Carvalho, D.; Izquierdo, E.; Fazal-Salom, J.; Taylor, K.R.; Bjerke, L.; Clarke, M.; Vinci, M.; Nandhabalan, M.; et al. Integrated Molecular Meta-Analysis of 1,000 Pediatric High-Grade and Diffuse Intrinsic Pontine Glioma. Cancer Cell 2017, 32, 520–537.e5. [Google Scholar] [CrossRef]
  55. Jones, C.; Baker, S.J. Unique genetic and epigenetic mechanisms driving paediatric diffuse high-grade glioma. Nat. Rev. Cancer 2014, 14, 651–661. [Google Scholar] [CrossRef] [PubMed]
  56. Kumar, R.; Liu, A.P.Y.; Orr, B.A.; Northcott, P.A.; Robinson, G.W. Advances in the classification of pediatric brain tumors through DNA methylation profiling: From research tool to frontline diagnostic. Cancer 2018, 124, 4168–4180. [Google Scholar] [CrossRef]
  57. Capper, D.; Jones, D.T.W.; Sill, M.; Hovestadt, V.; Schrimpf, D.; Sturm, D.; Koelsche, C.; Sahm, F.; Chavez, L.; Reuss, D.E.; et al. DNA methylation-based classification of central nervous system tumours. Nature 2018, 555, 469–474. [Google Scholar] [CrossRef]
  58. Pasqualetti, F.; Bertero, L.; Buccoliero, A.M.; Giunti, L.; Moscardi, S.; Castiglione, F.; Provenzano, A.; Sardi, I.; Scagnet, M.; Genitori, L.; et al. Pediatric High Grade Glioma Classification Criteria and Molecular Features of a Case Series. Genes 2022, 13, 624. [Google Scholar] [CrossRef]
  59. Bočkaj, I.; Martini, T.E.I.; De Camargo Magalhães, E.S.; Bakker, P.L.; Meeuwsen-De Boer, T.G.J.; Armandari, I.; Meuleman, S.L.; Mondria, M.T.; Stok, C.; Kok, Y.P.; et al. The H3.3K27M oncohistone affects replication stress outcome and provokes genomic instability in pediatric glioma. PLOS Genet. 2021, 17, e1009868. [Google Scholar] [CrossRef]
  60. Huang, T.Y.T.; Piunti, A.; Qi, J.; Morgan, M.; Bartom, E.; Shilatifard, A.; Saratsis, A.M. Effects of H3.3G34V mutation on genomic H3K36 and H3K27 methylation patterns in isogenic pediatric glioma cells. Acta Neuropathol. Commun. 2020, 8, 1–13. [Google Scholar] [CrossRef]
  61. Liu, X.; McEachron, T.A.; Schwartzentruber, J.; Wu, G. Histone H3 mutations in pediatric brain tumors. Cold Spring Harb. Perspect. Biol. 2014, 6. [Google Scholar] [CrossRef] [PubMed]
  62. Bale, T.A.; Rosenblum, M.K. The 2021 WHO Classification of Tumors of the Central Nervous System: An update on pediatric low-grade gliomas and glioneuronal tumors. Brain Pathol. 2022, 32, e13060. [Google Scholar] [CrossRef]
  63. Ryall, S.; Tabori, U.; Hawkins, C. Pediatric low-grade glioma in the era of molecular diagnostics. Acta Neuropathol. Commun. 2020, 8, 1–22. [Google Scholar] [CrossRef] [PubMed]
  64. Jones, D.T.W.; Kocialkowski, S.; Liu, L.; Pearson, D.M.; Bäcklund, L.M.; Ichimura, K.; Collins, V.P. Tandem duplication producing a novel oncogenic BRAF fusion gene defines the majority of pilocytic astrocytomas. Cancer Res. 2008, 68, 8673–8677. [Google Scholar] [CrossRef] [PubMed]
  65. Lassaletta, A.; Zapotocky, M.; Mistry, M.; Ramaswamy, V.; Honnorat, M.; Krishnatry, R.; Stucklin, A.G.; Zhukova, N.; Arnoldo, A.; Ryall, S.; et al. JOURNAL OF CLINICAL ONCOLOGY Therapeutic and Prognostic Implications of BRAF V600E in Pediatric Low-Grade Gliomas. J. Clin. Oncol. 2017, 35, 2934–2941. [Google Scholar] [CrossRef] [PubMed]
  66. Egbivwie, N.; Cockle, J.V.; Humphries, M.; Ismail, A.; Esteves, F.; Taylor, C.; Karakoula, K.; Morton, R.; Warr, T.; Short, S.C.; et al. FGFR1 expression and role in migration in low and high grade pediatric gliomas. Front. Oncol. 2019, 9, 103. [Google Scholar] [CrossRef]
  67. Kouhara, H.; Hadari, Y.R.; Spivak-Kroizman, T.; Schilling, J.; Bar-Sagi, D.; Lax, I.; Schlessinger, J. A Lipid-Anchored Grb2-Binding Protein That Links FGF-Receptor Activation to the Ras/MAPK Signaling Pathway. Cell 1997, 89, 693–702. [Google Scholar] [CrossRef]
  68. Zaytseva, M.; Papusha, L.; Novichkova, G.; Druy, A. Molecular Stratification of Childhood Ependymomas as a Basis for Personalized Diagnostics and Treatment. Cancers 2021, 13, 4954. [Google Scholar] [CrossRef]
  69. Hommelberg, P.P.H.; Plat, J.; Langen, R.C.J.; Schols, A.M.W.J.; Mensink, R.P. Fatty acid-induced NF-κB activation and insulin resistance in skeletal muscle are chain length dependent. Am. J. Physiol.-Endocrinol. Metab. 2009, 296, 114–120. [Google Scholar] [CrossRef]
  70. Yadav, U.C.S.; Ramana, K.V. Regulation of NF-κB-induced inflammatory signaling by lipid peroxidation-derived aldehydes. Oxid. Med. Cell. Longev. 2013, 2013. [Google Scholar] [CrossRef]
  71. Yamaguchi, H.; Taouk, G.M. A Potential Role of YAP/TAZ in the Interplay Between Metastasis and Metabolic Alterations. Front. Oncol. 2020, 10, 928. [Google Scholar] [CrossRef]
  72. Stucklin, A.S.G.; Ramaswamy, V.; Daniels, C.; Taylor, M.D. Review of molecular classification and treatment implications of pediatric brain tumors. Curr. Opin. Pediatr. 2018, 30, 3–9. [Google Scholar] [CrossRef] [PubMed]
  73. Bacci, M.; Lorito, N.; Smiriglia, A.; Morandi, A. Fat and Furious: Lipid Metabolism in Antitumoral Therapy Response and Resistance. Trends Cancer 2021, 7, 198–213. [Google Scholar] [CrossRef] [PubMed]
  74. Lladó, V.; López, D.J.; Ibarguren, M.; Alonso, M.; Soriano, J.B.; Escribá, P.V.; Busquets, X. Regulation of the cancer cell membrane lipid composition by NaCHOleate: Effects on cell signaling and therapeutical relevance in glioma. Biochim. Biophys. Acta-Biomembr. 2014, 1838, 1619–1627. [Google Scholar] [CrossRef]
  75. Hanauer, D.; Rhodes, D.; Sinha-Kumar, C.; Chinnaiyan, A. Bioinformatics Approaches in the Study of Cancer. Curr. Mol. Med. 2007, 7, 133–141. [Google Scholar] [CrossRef] [PubMed]
  76. Owada, Y. Fatty acid binding protein: Localization and functional significance in the brain. Tohoku J. Exp. Med. 2008, 214, 213–220. [Google Scholar] [CrossRef] [PubMed]
  77. Torres, M.; Rosselló, C.A.; Fernández-García, P.; Lladó, V.; Kakhlon, O.; Escribá, P.V. The Implications for Cells of the Lipid Switches Driven by Protein–Membrane Interactions and the Development of Membrane Lipid Therapy. Int. J. Mol. Sci. 2020, 21, 2322. [Google Scholar] [CrossRef]
  78. Bruschi, M.; Petretto, A.; Cama, A.; Pavanello, M.; Bartolucci, M.; Morana, G.; Ramenghi, L.A.; Garré, M.L.; Ghiggeri, G.M.; Panfoli, I.; et al. Potential biomarkers of childhood brain tumor identified by proteomics of cerebrospinal fluid from extraventricular drainage (EVD). Sci. Rep. 2021, 11, 1–13. [Google Scholar] [CrossRef]
  79. Lee, B.; Mohamad, I.; Pokhrel, R.; Murad, R.; Yuan, M.; Stapleton, S.; Bettegowda, C.; Jallo, G.; Eberhart, C.G.; Garrett, T.; et al. Medulloblastoma cerebrospinal fluid reveals metabolites and lipids indicative of hypoxia and cancer-specific RNAs. Acta Neuropathol. Commun. 2022, 10, 1–14. [Google Scholar] [CrossRef]
  80. Puget, S.; Philippe, C.; Bax, D.A.; Job, B.; Varlet, P.; Junier, M.P.; Andreiuolo, F.; Carvalho, D.; Reis, R.; Guerrini-Rousseau, L.; et al. Mesenchymal transition and pdgfra amplification/mutation are key distinct oncogenic events in pediatric diffuse intrinsic pontine gliomas. PLoS ONE 2012, 7, e3031. [Google Scholar] [CrossRef]
  81. Ullman, M.D.; Radin, N.S. The enzymatic formation of sphingomyelin from ceramide and lecithin in mouse liver. J. Biol. Chem. 1974, 249, 1506–1512. [Google Scholar] [CrossRef] [PubMed]
  82. Fernández-García, P.; Rosselló, C.A.; Rodríguez-Lorca, R.; Beteta-Göbel, R.; Fernández-Díaz, J.; Lladó, V.; Busquets, X.; Escribá, P.V. The opposing contribution of SMS1 and SMS2 to glioma progression and their value in the therapeutic response to 2OHOA. Cancers 2019, 11, 88. [Google Scholar] [CrossRef] [PubMed]
  83. Liu, G.; Kuang, S.; Cao, R.; Wang, J.; Peng, Q.; Sun, C. Sorafenib kills liver cancer cells by disrupting SCD1-mediated synthesis of monounsaturated fatty acids via the ATP-AMPK-mTOR-SREBP1 signaling pathway. FASEB J. 2019, 33, 10089–10103. [Google Scholar] [CrossRef]
  84. Martin, M.L.; Barceló-Coblijn, G.; De Almeida, R.F.M.; Noguera-Salvà, M.A.; Terés, S.; Higuera, M.; Liebisch, G.; Schmitz, G.; Busquets, X.; Escribá, P.V. The role of membrane fatty acid remodeling in the antitumor mechanism of action of 2-hydroxyoleic acid. Biochim. Biophys. Acta-Biomembr. 2013, 1828, 1405–1413. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, W.; Chu, H.J.; Liang, Y.C.; Huang, J.M.; Shang, C.L.; Tan, H.; Liu, D.; Zhao, Y.H.; Liu, T.Y.; Yao, S.Z. FABP5 correlates with poor prognosis and promotes tumor cell growth and metastasis in cervical cancer. Tumour Biol. 2016, 37, 14873–14883. [Google Scholar] [CrossRef]
  86. Wang, Y.; Wahafu, A.; Wu, W.; Xiang, J.; Huo, L.; Ma, X.; Wang, N.; Liu, H.; Bai, X.; Xu, D.; et al. FABP5 enhances malignancies of lower-grade gliomas via canonical activation of NF-κB signaling. J. Cell. Mol. Med. 2021, 25, 4487–4500. [Google Scholar] [CrossRef]
  87. Dyer, C.A.; Benjamins, J.A. Organization of oligodendroglial membrane sheets: II. Galactocerebroside: Antibody interactions signal changes in cytoskeleton and myelin basic protein. J. Neurosci. Res. 1989, 24, 212–221. [Google Scholar] [CrossRef]
  88. Liu, D.G.; Xue, L.; Li, J.; Yang, Q.; Peng, J.Z. Epithelial-mesenchymal transition and GALC expression of circulating tumor cells indicate metastasis and poor prognosis in non-small cell lung cancer. Cancer Biomark. 2018, 22, 417–426. [Google Scholar] [CrossRef]
  89. Chen, W.C.; Wang, C.Y.; Hung, Y.H.; Weng, T.Y.; Yen, M.C.; Lai, M.D. Systematic Analysis of Gene Expression Alterations and Clinical Outcomes for Long-Chain Acyl-Coenzyme A Synthetase Family in Cancer. PLoS ONE 2016, 11, e0155660. [Google Scholar] [CrossRef]
  90. Tea, M.N.; Poonnoose, S.I.; Pitson, S.M. Targeting the Sphingolipid System as a Therapeutic Direction for Glioblastoma. Cancers 2020, 12, 111. [Google Scholar] [CrossRef]
  91. Sheridan, M.; Ogretmen, B. The Role of Ceramide Metabolism and Signaling in the Regulation of Mitophagy and Cancer Therapy. Cancers 2021, 13, 2475. [Google Scholar] [CrossRef] [PubMed]
  92. Kuwata, H.; Hara, S. Role of acyl-CoA synthetase ACSL4 in arachidonic acid metabolism. Prostaglandins Other Lipid Mediat. 2019, 144, 106363. [Google Scholar] [CrossRef]
  93. Röhrig, F.; Schulze, A. The multifaceted roles of fatty acid synthesis in cancer. Nat. Rev. Cancer 2016, 16, 732–749. [Google Scholar] [CrossRef] [PubMed]
  94. Gabriely, G.; Wheeler, M.A.; Takenaka, M.C.; Quintana, F.J. Role of AHR and HIF-1$α$ in Glioblastoma Metabolism. Trends Endocrinol. Metab. 2017, 28, 428–436. [Google Scholar] [CrossRef] [PubMed]
  95. Parveen, F.; Bender, D.; Law, S.H.; Mishra, V.K.; Chen, C.C.; Ke, L.Y. Role of Ceramidases in Sphingolipid Metabolism and Human Diseases. Cells 2019, 8, 1573. [Google Scholar] [CrossRef]
  96. Bonica, J.; Mao, C.; Obeid, L.M.; Hannun, Y.A. Transcriptional Regulation of Sphingosine Kinase 1. Cells 2020, 9, 2437. [Google Scholar] [CrossRef]
  97. Dyer, M.A.; Qadeer, Z.A.; Valle-Garcia, D.; Bernstein, E.; Armstrong, S.A.; Henikoff, S.; Vakoc, C.R. ATRX and DAXX: Mechanisms and Mutations. Cold Spring Harb. Perspect. Med. 2017, 7, a026567. [Google Scholar] [CrossRef] [PubMed]
  98. Pyne, N.J.; Pyne, S. Recent advances in the role of sphingosine 1-phosphate in cancer. FEBS Lett. 2020, 594, 3583–3601. [Google Scholar] [CrossRef] [PubMed]
  99. Chen, C.; Zhang, Z.; Liu, C.; Wang, B.; Liu, P.; Fang, S.; Yang, F.; You, Y.; Li, X. ATF4-dependent fructolysis fuels growth of glioblastoma forme n.d. Nat. Commun. 2022, 13, 6108. [Google Scholar] [CrossRef] [PubMed]
  100. Arlotta, P.; Molyneaux, B.J.; Jabaudon, D.; Yoshida, Y.; Macklis, J.D. Ctip2 Controls the Differentiation of Medium Spiny Neurons and the Establishment of the Cellular Architecture of the Striatum. J. Neurosci. 2008, 28, 622–632. [Google Scholar] [CrossRef]
  101. Yoon, J.; Grinchuk, O.V.; Tirado-Magallanes, R.; Ngian, Z.K.; Tay, E.X.Y.; Chuah, Y.H.; Lee, B.W.L.; Feng, J.; Crasta, K.C.; Ong, C.T.; et al. E2F and STAT3 provide transcriptional synergy for histone variant H2AZ activation to sustain glioblastoma chromatin accessibility and tumorigenicity. Cell Death Differ. 2022, 29, 1379–1394. [Google Scholar] [CrossRef]
  102. Stoffel, W.; Jenke, B.; Schmidt-Soltau, I.; Binczek, E.; Brodesser, S.; Hammels, I. SMPD3 deficiency perturbs neuronal proteostasis and causes progressive cognitive impairment. Cell Death Dis. 2018, 9, 1–14. [Google Scholar] [CrossRef] [PubMed]
  103. Pan, J.; Sheng, S.; Ye, L.; Xu, X.; Ma, Y.; Feng, X.; Qiu, L.; Fan, Z.; Wang, Y.; Xia, X.; et al. Extracellular vesicles derived from glioblastoma promote proliferation and migration of neural progenitor cells via PI3K-Akt pathway. Cell Commun. Signal. 2022, 20, 1–16. [Google Scholar] [CrossRef]
  104. Marques, C.; Unterkircher, T.; Kroon, P.; Oldrini, B.; Izzo, A.; Dramaretska, Y.; Ferrarese, R.; Kling, E.; Schnell, O.; Nelander, S.; et al. Nf1 regulates mesenchymal glioblastoma plasticity and aggressiveness through the ap-1 transcription factor fosl1. Elife 2021, 10, e64846. [Google Scholar] [CrossRef]
  105. Prucca, C.G.; Racca, A.C.; Velazquez, F.N.; Gizzi, A.M.C.; Berdini, L.R.; Caputto, B.L. Impairing activation of phospholipid synthesis by c-Fos interferes with glioblastoma cell proliferation. Biochem. J. 2020, 477, 4675–4688. [Google Scholar] [CrossRef] [PubMed]
  106. Giacopelli, F.; Cappato, S.; Tonachini, L.; Mura, M.; Di Lascio, S.; Fornasari, D.; Ravazzolo, R.; Bocciardi, R. Identification and characterization of regulatory elements in the promoter of ACVR1, the gene mutated in Fibrodysplasia Ossificans Progressiva. Orphanet J. Rare Dis. 2013, 8, 1–16. [Google Scholar] [CrossRef]
  107. Peng, G.; Wang, Y.; Ge, P.; Bailey, C.; Zhang, P.; Zhang, D.; Meng, Z.; Qi, C.; Chen, Q.; Chen, J.; et al. The HIF1$α$-PDGFD-PDGFR$α$ axis controls glioblastoma growth at normoxia/mild-hypoxia and confers sensitivity to targeted therapy by echinomycin. J. Exp. Clin. Cancer Res. 2021, 40, 1–16. [Google Scholar] [CrossRef]
  108. Renfrow, J.J.; Soike, M.H.; West, J.L.; Ramkissoon, S.H.; Metheny-Barlow, L.; Mott, R.T.; Kittel, C.A.; D’Agostino, R.B.; Tatter, S.B.; Laxton, A.W.; et al. Attenuating hypoxia driven malignant behavior in glioblastoma with a novel hypoxia-inducible factor 2 alpha inhibitor. Sci. Rep. 2020, 10, 1–11. [Google Scholar] [CrossRef]
  109. Gan, B. ACSL4, PUFA, and Ferroptosis: New Arsenal in Anti-Tumor Immunity. Signal. Transduct. Target. Ther. 2022, 7, 1–3. [Google Scholar] [CrossRef]
  110. Liang, J.; Piao, Y.; Henry, V.; Tiao, N.; de Groot, J.F.; Liang, J.; Piao, Y.; Henry, V.; Tiao, N.; de Groot, J.F. Interferon-Regulatory Factor-1 (IRF1) Regulates Bevacizumab Induced Autophagy. Oncotarget 2015, 6, 31479–31492. [Google Scholar] [CrossRef]
  111. Kim, S.H.; Kim, E.J.; Hitomi, M.; Oh, S.Y.; Jin, X.; Jeon, H.M.; Beck, S.; Jin, X.; Kim, J.K.; Park, C.G.; et al. The LIM-only transcription factor LMO2 determines tumorigenic and angiogenic traits in glioma stem cells. Cell Death Differ. 2015, 22, 1517–1525. [Google Scholar] [CrossRef]
  112. Nishi-Tatsumi, M.; Yahagi, N.; Takeuchi, Y.; Toya, N.; Takarada, A.; Murayama, Y.; Aita, Y.; Sawada, Y.; Piao, X.; Oya, Y.; et al. A key role of nuclear factor Y in the refeeding response of fatty acid synthase in adipocytes. FEBS Lett. 2017, 591, 965–978. [Google Scholar] [CrossRef] [PubMed]
  113. Cui, H.; Zhang, M.; Wang, Y.; Wang, Y. NF-YC in glioma cell proliferation and tumor growth and its role as an independent predictor of patient survival. Neurosci. Lett. 2016, 631, 40–49. [Google Scholar] [CrossRef]
  114. Wegner, M.S.; Gruber, L.; Mattjus, P.; Geisslinger, G.; Grösch, S. The UDP-glucose ceramide glycosyltransferase (UGCG) and the link to multidrug resistance protein 1 (MDR1). BMC Cancer 2018, 18, 1–10. [Google Scholar] [CrossRef]
  115. Gopal, K.; Grossi, E.; Paoletti, P.; Usardi, M. Lipid composition of human intracranial tumors: A biochemical study. Acta Neurochir. 1963, 11, 333–347. [Google Scholar] [CrossRef] [PubMed]
  116. Campenella, R. Membrane lipids modifications in human gliomas of different degree of malignancy. J. Neurosurg. Sci. 1992, 36, 11–25. [Google Scholar]
  117. Martin, D.D.; Robbins, M.E.C.; Spector, A.A.; Wen, B.C.; Hussey, D.H. The fatty acid composition of human gliomas differs from that found in nonmalignant brain tissue. Lipids 1996, 31, 1283–1288. [Google Scholar] [CrossRef]
  118. Aramesh, M.; Stoycheva, D.; Sandu, I.; Ihle, S.J.; Zünd, T.; Shiu, J.Y.; Forró, C.; Asghari, M.; Bernero, M.; Lickert, S.; et al. Nanoconfinement of microvilli alters gene expression and boosts T cell activation. Proc. Natl. Acad. Sci. USA 2021, 118, e2107535118. [Google Scholar] [CrossRef]
  119. Polis, B.; Imiela, A.; Polis, L.; Abramczyk, H. Raman spectroscopy for medulloblastoma. Child’s Nerv. Syst. 2018, 34, 2425–2430. [Google Scholar] [CrossRef]
  120. Leslie, D.G.; Kast, R.E.; Poulik, J.M.; Rabah, R.; Sood, S.; Auner, G.W.; Klein, M.D. Identification of pediatric brain neoplasms using Raman spectroscopy. Pediatr. Neurosurg. 2012, 48, 109–117. [Google Scholar] [CrossRef] [PubMed]
  121. Glunde, K.; Bhujwalla, Z.M. Metabolic tumor imaging using magnetic resonance spectroscopy. Semin. Oncol. 2011, 38, 26–41. [Google Scholar] [CrossRef] [PubMed]
  122. Gillies, R.J.; Morse, D.L. In Vivo Magnetic Resonance Spectroscopy in Cancer. Annu. Rev. Biomed. Eng. 2005, 7, 287–326. [Google Scholar] [CrossRef]
  123. Cuellar-Baena, S.; Morales, J.M.; Martinetto, H.; Calvar, J.; Sevlever, G.; Castellano, G.; Cerdá-Nicolás, M.; Celda, B.; Monle-on, D. Comparative metabolic profiling of paediatric ependymoma, medulloblastoma and pilocytic astrocytoma. Int. J. Mol. Med. 2010, 26, 941–948. [Google Scholar] [CrossRef]
  124. Liserre, R.; Pinelli, L.; Gasparotti, R. MR spectroscopy in pediatric neuroradiology. Transl. Pediatr. 2021, 10, 1169. [Google Scholar] [CrossRef] [PubMed]
  125. Orphanidou-Vlachou, E.; Kohe, S.E.; Brundler, M.A.; MacPherson, L.; Sun, Y.; Davies, N.; Wilson, M.; Pan, X.; Arvanitis, T.N.; Grundy, R.G.; et al. Metabolite Levels in Paediatric Brain Tumours Correlate with Histological Features. Pathobiology 2018, 85, 157–168. [Google Scholar] [CrossRef] [PubMed]
  126. Galanaud, D.; Nicoli, F.; Confort-Gouny, S.; Le Fur, Y.; Dormont, D.; Girard, N.; Ranjeva, J.P.; Cozzone, P.J. Spectroscopie par résonance magnétique cérébrale. J. Radiol. 2007, 88, 483–496. [Google Scholar] [CrossRef]
  127. Clark, A.R.; Calligaris, D.; Regan, M.S.; Pomeranz Krummel, D.; Agar, J.N.; Kallay, L.; MacDonald, T.; Schniederjan, M.; Santagata, S.; Pomeroy, S.L.; et al. Rapid discrimination of pediatric brain tumors by mass spectrometry imaging. J. Neurooncol. 2018, 140, 269–279. [Google Scholar] [CrossRef]
  128. Srivastava, N.K.; Pradhan, S.; Gowda, G.A.N.; Kumar, R. In vitro, high-resolution 1H and 31P NMR based analysis of the lipid components in the tissue, serum, and CSF of the patients with primary brain tumors: One possible diagnostic view. NMR Biomed. 2010, 23, 113–122. [Google Scholar] [CrossRef]
  129. Wilson, M.; Cummins, C.L.; MacPherson, L.; Sun, Y.; Natarajan, K.; Grundy, R.G.; Arvanitis, T.N.; Kauppinen, R.A.; Peet, A.C. Magnetic resonance spectroscopy metabolite profiles predict survival in paediatric brain tumours. Eur. J. Cancer 2013, 49, 457–464. [Google Scholar] [CrossRef]
  130. Marcus, K.J.; Astrakas, L.G.; Zurakowski, D.; Zarifi, M.K.; Mintzopoulos, D.; Poussaint, T.Y.; Anthony, D.C.; De Girolami, U.; Black, P.M.L.; Tarbell, N.J.; et al. Predicting survival of children with CNS tumors using proton magnetic resonance spectroscopic imaging biomarkers. Int. J. Oncol. 2007, 30, 651–657. [Google Scholar] [CrossRef]
  131. Bennett, C.D.; Gill, S.K.; Kohe, S.E.; Wilson, M.P.; Davies, N.P.; Arvanitis, T.N.; Tennant, D.A.; Peet, A.C. Ex vivo metabolite profiling of paediatric central nervous system tumours reveals prognostic markers. Sci. Rep. 2019, 9, 10473. [Google Scholar] [CrossRef] [PubMed]
  132. Tzika, A.A.; Zurakowski, D.; Poussaint, T.Y.; Goumnerova, L.; Astrakas, L.G.; Barnes, P.D.; Anthony, D.C.; Billett, A.L.; Tarbell, N.J.; Scott, R.M.; et al. Proton magnetic spectroscopic imaging of the child’s brain: The response of tumors to treatment. Neuroradiology 2001, 43, 169–177. [Google Scholar] [CrossRef]
  133. Paine, M.R.L.; Liu, J.; Huang, D.; Ellis, S.R.; Trede, D.; Kobarg, J.H.; Heeren, R.M.A.; Fernández, F.M.; MacDonald, T.J. Three-Dimensional Mass Spectrometry Imaging Identifies Lipid Markers of Medulloblastoma Metastasis. Sci. Rep. 2019, 9, 2205. [Google Scholar] [CrossRef] [PubMed]
  134. Silva, A.H.D.; Aquilina, K. Surgical approaches in pediatric neuro-oncology. Cancer Metastasis Rev. 2019, 38, 723–747. [Google Scholar] [CrossRef]
  135. Zebian, B.; Vergani, F.; Lavrador, J.P.; Mukherjee, S.; Kitchen, W.J.; Stagno, V.; Chamilos, C.; Pettorini, B.; Mallucci, C. Recent technological advances in pediatric brain tumor surgery. CNS Oncol. 2017, 6, 71. [Google Scholar] [CrossRef] [PubMed]
  136. Funakoshi, Y.; Hata, N.; Kuga, D.; Hatae, R.; Sangatsuda, Y.; Fujioka, Y.; Takigawa, K.; Mizoguchi, M. Pediatric Glioma: An Update of Diagnosis, Biology, and Treatment. Cancers 2021, 13, 758. [Google Scholar] [CrossRef]
  137. Laprie, A.; Hu, Y.; Alapetite, C.; Carrie, C.; Habrand, J.L.; Bolle, S.; Bondiau, P.Y.; Ducassou, A.; Huchet, A.; Bertozzi, A.I.; et al. Paediatric brain tumours: A review of radiotherapy, state of the art and challenges for the future regarding protontherapy and carbontherapy. Cancer Radiother. 2015, 19, 775–789. [Google Scholar] [CrossRef]
  138. Baliga, S.; Yock, T.I. Proton beam therapy in pediatric oncology. Curr. Opin. Pediatr. 2019, 31, 28–34. [Google Scholar] [CrossRef]
  139. Kortmann, R.D.; Seidel, C.; Müller, K.; Hirsch, F.W. Irradiation of Intracranial Gliomas in Children. Prog. Neurol. Surg. 2018, 31, 87–101. [Google Scholar] [CrossRef]
  140. Lobón, M.J.; Bautista, F.; Riet, F.; Dhermain, F.; Canale, S.; Dufour, C.; Blauwblomme, T.; Zerah, M.; Beccaria, K.; Saint-Rose, C.; et al. Re-irradiation of recurrent pediatric ependymoma: Modalities and outcomes: A twenty-year survey. Springerplus 2016, 5, 879. [Google Scholar] [CrossRef]
  141. Cohen, K.J.; Pollack, I.F.; Zhou, T.; Buxton, A.; Holmes, E.J.; Burger, P.C.; Brat, D.J.; Rosenblum, M.K.; Hamilton, R.L.; Lavey, R.S.; et al. Temozolomide in the treatment of high-grade gliomas in children: A report from the Children’s Oncology Group. Neuro. Oncol. 2011, 13, 317–323. [Google Scholar] [CrossRef]
  142. Chatwin, H.V.; Cruz, J.C.; Green, A.L.; Chatwin, H.V.; Cruz, J.C.; Green, A.L. Pediatric high-grade glioma: Moving toward subtype-specific multimodal therapy. FEBS J. 2021, 288, 6127–6141. [Google Scholar] [CrossRef] [PubMed]
  143. Hwang, E.I.; Jakacki, R.I.; Fisher, M.J.; Kilburn, L.B.; Horn, M.; Vezina, G.; Rood, B.R.; Packer, R.J. Long-term efficacy and toxicity of bevacizumab-based therapy in children with recurrent low-grade gliomas. Pediatr. Blood Cancer 2013, 60, 776–782. [Google Scholar] [CrossRef] [PubMed]
  144. Rashed, W.M.; Maher, E.; Adel, M.; Saber, O.; Zaghloul, M.S. Pediatric diffuse intrinsic pontine glioma: Where do we stand? Cancer Metastasis Rev. 2019, 38, 759–770. [Google Scholar] [CrossRef]
  145. Stanić, D.; Grujičić, D.; Pekmezović, T.; Bokun, J.; Popović-Vuković, M.; Janić, D.; Paripović, L.; Ilić, V.; Slović, M.P.; Ilić, R.; et al. Clinical profile, treatment and outcome of pediatric brain tumors in Serbia in a 10-year period: A national referral institution experience. PLoS ONE 2021, 16, e0259095. [Google Scholar] [CrossRef] [PubMed]
  146. Mathew, R.K.; O’kane, R.; Parslow, R.; Stiller, C.; Kenny, T.; Picton, S.; Chumas, P.D. Comparison of survival between the UK and US after surgery for most common pediatric CNS tumors. Neuro. Oncol. 2014, 16, 1137–1145. [Google Scholar] [CrossRef] [PubMed]
  147. Arvanitis, C.D.; Ferraro, G.B.; Jain, R.K. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat. Rev. Cancer 2020, 20, 26–41. [Google Scholar] [CrossRef]
  148. Patel, J.P.; Spiller, S.E.; Barker, E.D. Drug penetration in pediatric brain tumors: Challenges and opportunities. Pediatr. Blood Cancer 2021, 68, e28983. [Google Scholar] [CrossRef]
  149. Power, E.A.; Rechberger, J.S.; Gupta, S.; Schwartz, J.D.; Daniels, D.J.; Khatua, S. Drug delivery across the blood-brain barrier for the treatment of pediatric brain tumors—An update. Adv. Drug Deliv. Rev. 2022, 185, 114303. [Google Scholar] [CrossRef]
  150. De Blank, P.; Fouladi, M.; Huse, J.T. Molecular markers and targeted therapy in pediatric low-grade glioma. J. Neurooncol. 2020, 150, 5–15. [Google Scholar] [CrossRef]
  151. Smith, A.; Onar-Thomas, A.; Ellison, D.; Owens-Pickle, E.; Wu, S.; Leary, S.E.S.; Fouladi, M.; Merchant, T.; Gajjar, A.; Foreman, N. EPEN-54. ACNS0831, phase III randomized trial of post-radiation chemotherapy in patients with newly diagnosed ependymoma ages 1 to 21 years. Neuro. Oncol. 2020, 22, iii318. [Google Scholar] [CrossRef]
  152. Franz, D.N.; Belousova, E.; Sparagana, S.; Bebin, E.M.; Frost, M.; Kuperman, R.; Witt, O.; Kohrman, M.H.; Flamini, J.R.; Wu, J.Y.; et al. Efficacy and safety of everolimus for subependymal giant cell astrocytomas associated with tuberous sclerosis complex (EXIST-1): A multicentre, randomised, placebo-controlled phase 3 trial. Lancet (Lond. Engl.) 2013, 381, 125–132. [Google Scholar] [CrossRef]
  153. Brabetz, S.; Leary, S.E.S.; Gröbner, S.N.; Nakamoto, M.W.; Şeker-Cin, H.; Girard, E.J.; Cole, B.; Strand, A.D.; Bloom, K.L.; Hovestadt, V.; et al. A biobank of patient-derived pediatric brain tumor models. Nat. Med. 2018, 24, 1752–1761. [Google Scholar] [CrossRef]
  154. Xu, J.; Margol, A.; Asgharzadeh, S.; Erdreich-Epstein, A. Pediatric Brain Tumor Cell Lines. J. Cell. Biochem. 2015, 116, 218–224. [Google Scholar] [CrossRef] [PubMed]
  155. Hanz, S.Z.; Adeuyan, O.; Lieberman, G.; Hennika, T. Clinical trials using molecular stratification of pediatric brain tumors. Transl. Pediatr. 2020, 9, 144. [Google Scholar] [CrossRef]
  156. Findlay, I.J.; De Iuliis, G.N.; Duchatel, R.J.; Jackson, E.R.; Vitanza, N.A.; Cain, J.E.; Waszak, S.M.; Dun, M.D. Pharmaco-proteogenomic profiling of pediatric diffuse midline glioma to inform future treatment strategies. Oncogene 2021, 41, 461–475. [Google Scholar] [CrossRef]
  157. Bahmad, H.F.; Elajami, M.K.; El Zarif, T.; Bou-Gharios, J.; Abou-Antoun, T.; Abou-Kheir, W. Drug repurposing towards targeting cancer stem cells in pediatric brain tumors. Cancer Metastasis Rev. 2020, 39, 127–148. [Google Scholar] [CrossRef]
  158. Hwang, E.I.; Sayour, E.J.; Flores, C.T.; Grant, G.; Wechsler-Reya, R.; Hoang-Minh, L.B.; Kieran, M.W.; Salcido, J.; Prins, R.M.; Figg, J.W.; et al. The current landscape of immunotherapy for pediatric brain tumors. Nat. Cancer 2022, 3, 11–24. [Google Scholar] [CrossRef]
  159. Wick, W.; Osswald, M.; Wick, A.; Winkler, F. Treatment of glioblastoma in adults. Ther. Adv. Neurol. Disord. 2018, 11. [Google Scholar] [CrossRef] [PubMed]
  160. Van den Bent, M.; Gan, H.K.; Lassman, A.B.; Kumthekar, P.; Merrell, R.; Butowski, N.; Lwin, Z.; Mikkelsen, T.; Nabors, L.B.; Papadopoulos, K.P.; et al. Efficacy of depatuxizumab mafodotin (ABT-414) monotherapy in patients with EGFR-amplified, recurrent glioblastoma: Results from a multi-center, international study. Cancer Chemother. Pharmacol. 2017, 80, 1209. [Google Scholar] [CrossRef] [PubMed]
  161. Hartman, L.L.R.; Crawford, J.R.; Makale, M.T.; Milburn, M.; Joshi, S.; Salazar, A.M.; Hasenauer, B.; Vandenberg, S.R.; Macdonald, T.J.; Durden, D.L. Pediatric Phase II Trials of Poly-ICLC in the Management of Newly Diagnosed and Recurrent Brain Tumors. J. Pediatr. Hematol. Oncol. 2014, 36, 451. [Google Scholar] [CrossRef] [PubMed]
  162. Patterson, J.D.; Henson, J.C.; Breese, R.O.; Bielamowicz, K.J.; Rodriguez, A. CAR T Cell Therapy for Pediatric Brain Tumors. Front. Oncol. 2020, 10, 1582. [Google Scholar] [CrossRef]
  163. Abdel-Khaleq, S.; Alim, L.; Johnston, A.; Adam, K.; Galvin, R.; Maeser, D.; Gruener, R.; Huang, S.; Johnson, T.S.; Pacholczyk, R.; et al. Immu-04. First-in-children phase 1b study using the ido pathway inhibitor indoximod in combination with radiation and chemotherapy for children with newly diagnosed DIPG (NCT02502708, NLG2105). Neuro. Oncol. 2021, 23, i27. [Google Scholar] [CrossRef]
  164. Varela-Guruceaga, M.; Tejada-Solís, S.; García-Moure, M.; Fueyo, J.; Gomez-Manzano, C.; Patiño-García, A.; Alonso, M.M. Oncolytic Viruses as Therapeutic Tools for Pediatric Brain Tumors. Cancers 2018, 10, 226. [Google Scholar] [CrossRef]
  165. Olsen, H.E.; Lynn, G.M.; Valdes, P.A.; Cerecedo Lopez, C.D.; Ishizuka, A.S.; Arnaout, O.; Bi, W.L.; Peruzzi, P.P.; Chiocca, E.A.; Friedman, G.K.; et al. Therapeutic cancer vaccines for pediatric malignancies: Advances, challenges, and emerging technologies. Neuro-Oncology Adv. 2021, 3, 1–14. [Google Scholar] [CrossRef]
  166. Banerjee, K.; Núñez, F.J.; Haase, S.; McClellan, B.L.; Faisal, S.M.; Carney, S.V.; Yu, J.; Alghamri, M.S.; Asad, A.S.; Candia, A.J.N.; et al. Current Approaches for Glioma Gene Therapy and Virotherapy. Front. Mol. Neurosci. 2021, 14, 30. [Google Scholar] [CrossRef] [PubMed]
  167. El Moukhtari, S.H.; Rodríguez-Nogales, C.; Blanco-Prieto, M.J. Oral lipid nanomedicines: Current status and future perspectives in cancer treatment. Adv. Drug Deliv. Rev. 2021, 173, 238–251. [Google Scholar] [CrossRef]
  168. Iturrioz-Rodríguez, N.; Bertorelli, R.; Ciofani, G. Lipid-Based Nanocarriers for The Treatment of Glioblastoma. Adv. NanoBiomed Res. 2021, 1, 2000054. [Google Scholar] [CrossRef] [PubMed]
  169. Hu, J.; Zhang, L.; Chen, W.; Shen, L.; Jiang, J.; Sun, S.; Chen, Z. Role of Intra- and Extracellular Lipid Signals in Cancer Stemness and Potential Therapeutic Strategy. Front. Pharmacol. 2021, 12, 2504. [Google Scholar] [CrossRef] [PubMed]
  170. Torres, M.; Parets, S.; Fernández-Díaz, J.; Beteta-Göbel, R.; Rodríguez-Lorca, R.; Román, R.; Lladó, V.; Rosselló, C.A.; Fernández-García, P.; Escribá, P.V. Lipids in Pathophysiology and Development of the Membrane Lipid Therapy: New Bioactive Lipids. Membranes 2021, 11, 919. [Google Scholar] [CrossRef]
  171. Fu, Y.; Zou, T.; Shen, X.; Nelson, P.J.; Li, J.; Wu, C.; Yang, J.; Zheng, Y.; Bruns, C.; Zhao, Y.; et al. Lipid metabolism in cancer progression and therapeutic strategies. MedComm 2020, 2, 27–59. [Google Scholar] [CrossRef] [PubMed]
  172. Matsui, Y. Mbrs-23. Significance of mi-r33 in generation and progression of medulloblastoma. Neuro. Oncol. 2020, 22, iii402. [Google Scholar] [CrossRef]
  173. Rahal, F.; Capdevielle, C.; Rousseau, B.; Izotte, J.; Dupuy, J.-W.; Cappellen, D.; Chotard, G.; Ménard, M.; Charpentier, J.; Jecko, V.; et al. An EZH2 blocker sensitizes histone mutated diffuse midline glioma to cholesterol metabolism inhibitors through an off-target effect. Neuro-Oncology Adv. 2022, 4, 1–13. [Google Scholar] [CrossRef] [PubMed]
  174. Dai, L.; Chen, J.; Lin, Z.; Wang, Z.; Mu, S.; Qin, Z. Targeting sphingosine kinase by ABC294640 against diffuse intrinsic pontine glioma (DIPG). J. Cancer 2020, 11, 4683–4691. [Google Scholar] [CrossRef] [PubMed]
  175. Daggubati, V.; Hochstelter, J.; Bommireddy, A.; Choudhury, A.; Krup, A.L.; Kaur, P.; Tong, P.; Li, A.; Xu, L.; Reiter, J.F.; et al. Smoothened-activating lipids drive resistance to CDK4/6 inhibition in Hedgehog-associated medulloblastoma cells and preclinical models. J. Clin. Invest. 2021, 131, e141171. [Google Scholar] [CrossRef] [PubMed]
  176. Sabnis, D.H.; Liu, J.-F.; Simmonds, L.; Blackburn, S.; Grundy, R.G.; Kerr, I.D.; Coyle, B. BLBP Is Both a Marker for Poor Prognosis and a Potential Therapeutic Target in Paediatric Ependymoma. Cancers 2021, 13, 2100. [Google Scholar] [CrossRef]
  177. Ljungblad, L.; Bergqvist, F.; Tümmler, C.; Madawala, S.; Olsen, T.K.; Andonova, T.; Jakobsson, P.J.; Johnsen, J.I.; Pickova, J.; Strandvik, B.; et al. Omega-3 fatty acids decrease CRYAB, production of oncogenic prostaglandin E2 and suppress tumor growth in medulloblastoma. Life Sci. 2022, 295, 120394. [Google Scholar] [CrossRef]
  178. Jendrossek, V. Erucylphosphocholine, a novel antineoplastic ether lipid, blocks growth and induces apoptosis in brain tumor cell lines in vitro. Int. J. Oncol. 1999, 14, 15–22. [Google Scholar] [CrossRef]
  179. Bi, J.; Khan, A.; Tang, J.; Armando, A.M.; Wu, S.; Zhang, W.; Gimple, R.C.; Reed, A.; Jing, H.; Koga, T.; et al. Targeting glioblastoma signaling and metabolism with a re-purposed brain-penetrant drug. Cell Rep. 2021, 37, 109957. [Google Scholar] [CrossRef]
  180. Xu, C.; Zhao, J.; Song, J.; Xiao, M.; Cui, X.; Xin, L.; Xu, J.; Zhang, Y.; Yi, K.; Hong, B.; et al. lncRNA PRADX is a Mesenchymal Glioblastoma Biomarker for Cellular Metabolism Targeted Therapy. Front. Oncol. 2022, 12, 1865. [Google Scholar] [CrossRef]
  181. Cruz Flores, V.A.; Menghani, H.; Mukherjee, P.K.; Marrero, L.; Obenaus, A.; Dang, Q.; Khoutorova, L.; Reid, M.M.; Belayev, L.; Bazan, N.G. Combined Therapy With Avastin, a PAF Receptor Antagonist and a Lipid Mediator Inhibited Glioblastoma Tumor Growth. Front. Pharmacol. 2021, 12, 2554. [Google Scholar] [CrossRef] [PubMed]
  182. Yi, K.; Zhan, Q.; Wang, Q.; Tan, Y.; Fang, C.; Wang, Y.; Zhou, J.; Yang, C.; Li, Y.; Kang, C. PTRF/cavin-1 remodels phospholipid metabolism to promote tumor proliferation and suppress immune responses in glioblastoma by stabilizing cPLA2. Neuro. Oncol. 2021, 23, 387–399. [Google Scholar] [CrossRef]
  183. Oatman, N.; Dasgupta, N.; Arora, P.; Choi, K.; Gawali, M.V.; Gupta, N.; Parameswaran, S.; Salomone, J.; Reisz, J.A.; Lawler, S.; et al. Mechanisms of stearoyl CoA desaturase inhibitor sensitivity and acquired resistance in cancer. Sci. Adv. 2021, 7, 7459. [Google Scholar] [CrossRef] [PubMed]
  184. Eyme, K.M.; Sammarco, A.; Jha, R.; Mnatsakanyan, H.; Pechdimaljian, C.; Carvalho, L.; Neustadt, R.; Moses, C.; Alnasser, A.; Tardiff, D.F.; et al. Targeting de novo lipid synthesis induces lipotoxicity and impairs DNA damage repair in glioblastoma mouse models. Sci. Transl. Med. 2023, 15, eabq6288. [Google Scholar] [CrossRef] [PubMed]
  185. Lin, H.; Patel, S.; Affeck, V.S.; Wilson, I.; Turnbull, D.M.; Joshi, A.R.; Maxwell, R.; Stoll, E.A. Fatty acid oxidation is required for the respiration and proliferation of malignant glioma cells. Neuro. Oncol. 2017, 19, 43–54. [Google Scholar] [CrossRef] [PubMed]
  186. Nam, H.J.; Kim, Y.E.; Moon, B.S.; Kim, H.Y.; Jung, D.; Choi, S.; Jang, J.W.; Nam, D.H.; Cho, H. Azathioprine antagonizes aberrantly elevated lipid metabolism and induces apoptosis in glioblastoma. iScience 2021, 24, 102238. [Google Scholar] [CrossRef] [PubMed]
  187. Dasari, R.; Masi, M.; Lisy, R.; Ferdérin, M.; English, L.R.; Cimmino, A.; Mathieu, V.; Brenner, A.J.; Kuhn, J.G.; Whitten, S.T.; et al. Fungal metabolite ophiobolin A as a promising anti-glioma agent: In vivo evaluation, structure-activity relationship and unique pyrrolylation of primary amines. Bioorg. Med. Chem. Lett. 2015, 25, 4544–4548. [Google Scholar] [CrossRef] [PubMed]
  188. Villa, G.R.; Hulce, J.J.; Zanca, C.; Bi, J.; Ikegami, S.; Cahill, G.L.; Gu, Y.; Lum, K.M.; Masui, K.; Yang, H.; et al. An LXR-Cholesterol Axis Creates a Metabolic Co-Dependency for Brain Cancers. Cancer Cell 2016, 30, 683–693. [Google Scholar] [CrossRef] [PubMed]
  189. Seyfried, T.N.; Mukherjee, P. Ganglioside GM3 is antiangiogenic in malignant brain cancer. J. Oncol. 2010, 2010, 961243. [Google Scholar] [CrossRef]
  190. Barceló-Coblijn, G.; Martin, M.L.; De Almeida, R.F.M.; Noguera-Salvà, M.A.; Marcilla-Etxenike, A.; Guardiola-Serrano, F.; Lüth, A.; Kleuser, B.; Halver, J.E.; Escribá, P.V. Sphingomyelin and sphingomyelin synthase (SMS) in the malignant transformation of glioma cells and in 2-hydroxyoleic acid therapy. Proc. Natl. Acad. Sci. USA 2011, 108, 19569–19574. [Google Scholar] [CrossRef] [PubMed]
  191. Lladó, V.; Terés, S.; Higuera, M.; Álvarez, R.; Noguera-Salva, M.A.; Halver, J.E.; Escribá, P.V.; Busquetsa, X. Pivotal role of dihydrofolate reductase knockdown in the anticancer activity of 2-hydroxyoleic acid. Proc. Natl. Acad. Sci. USA 2009, 106, 13754. [Google Scholar] [CrossRef]
  192. Terés, S.; Lladó, V.; Higuera, M.; Barceló-Coblijn, G.; Martin, M.L.; Noguera-Salvà, M.A.; Marcilla-Etxenike, A.; García-Verdugo, J.M.; Soriano-Navarro, M.; Saus, C.; et al. 2-Hydroxyoleate, anontoxic membrane binding anticancer drug, induces glioma cell differentiation and autophagy. Proc. Natl. Acad. Sci. USA 2012, 109, 8489–8494. [Google Scholar] [CrossRef]
  193. Kaynak, A.; Davis, H.W.; Vallabhapurapu, S.D.; Pak, K.Y.; Gray, B.D.; Qi, X. SapC&ndash;DOPS as a Novel Therapeutic and Diagnostic Agent for Glioblastoma Therapy and Detection: Alternative to Old Drugs and Agents. Pharmaceuticals 2021, 14, 1193. [Google Scholar] [CrossRef]
  194. Davis, H.W.; Kaynak, A.; Vallabhapurapu, S.D.; Qi, X. Targeting of elevated cell surface phosphatidylserine with saposin C-dioleoylphosphatidylserine nanodrug as individual or combination therapy for pancreatic cancer. World J. Gastrointest. Oncol. 2021, 13, 550–559. [Google Scholar] [CrossRef] [PubMed]
  195. Davis, H.W.; Vallabhapurapu, S.D.; Chu, Z.; Vallabhapurapu, S.L.; Franco, R.S.; Mierzwa, M.; Kassing, W.; Barrett, W.L.; Qi, X. Enhanced phosphatidylserine-selective cancer therapy with irradiation and SapC-DOPS nanovesicles. Oncotarget 2019, 10, 856–868. [Google Scholar] [CrossRef] [PubMed]
  196. Felix, F.H.C.; De Araujo, O.L.; Da Trindade, K.M.; Trompieri, N.M.; Fontenele, J.B. Retrospective evaluation of the outcomes of children with diffuse intrinsic pontine glioma treated with radiochemotherapy and valproic acid in a single center. J. Neurooncol. 2014, 116, 261–266. [Google Scholar] [CrossRef] [PubMed]
  197. Chen, X.; Wong, J.Y.C.; Wong, P.; Radany, E.H. Low Dose Valproic Acid Enhances Radiosensitivity of Prostate Cancer through Acetylated p53-Dependent Modulation of Mitochondrial Membrane Potential and Apoptosis. Mol. Cancer Res. 2011, 9, 448. [Google Scholar] [CrossRef]
  198. Pajovic, S.; Siddaway, R.; Bridge, T.; Sheth, J.; Rakopoulos, P.; Kim, B.; Ryall, S.; Agnihotri, S.; Phillips, L.; Yu, M.; et al. Epigenetic activation of a RAS/MYC axis in H3.3K27M-driven cancer. Nat. Commun. 2020, 11, 1–16. [Google Scholar] [CrossRef]
  199. Chavan, T.S.; Muratcioglu, S.; Marszalek, R.; Jang, H.; Keskin, O.; Gursoy, A.; Nussinov, R.; Gaponenko, V. Plasma membrane regulates Ras signaling networks. Cell. Logist. 2016, 5, e1136374. [Google Scholar] [CrossRef]
  200. Bagchi, S.; Rathee, P.; Jayaprakash, V.; Banerjee, S. Farnesyl Transferase Inhibitors as Potential Anticancer Agents. Mini Rev. Med. Chem. 2018, 18, 1611–1623. [Google Scholar] [CrossRef]
  201. Chidley, C.; Trauger, S.A.; Birsoy, K.; O’Shea, E.K. The anticancer natural product ophiobolin A induces cytotoxicity by covalent modification of phosphatidylethanolamine. Elife 2016, 5, e14601. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Frequency distribution of alterations to gene expression in glioma patients. Eleven lipid metabolism genes (ASAH2, FASN, SMPD2, SMPD3, PHYH, SPTLC3, SGMS1, SGMS2, SPHK1, SPHK2, UGCG) and eleven conventional oncogenes (AKT1, AKT2, AKT3, MYC, MAPK13, PIK3CB, PI3KCG, PI3KCD, NRAS, KRAS, TNF) with significantly altered expression were analyzed. The frequency of the alterations in expression (either higher or lower) was analyzed in patients with glioma in the REMBRANDT database. The graph shows data from 0 to 8 alterations in lipid metabolism (closed bars) or conventional oncogene (open bars) transcripts. Reprinted/adapted with permission from Ref. [74] Copyright 2014, Elsevier.
Figure 1. Frequency distribution of alterations to gene expression in glioma patients. Eleven lipid metabolism genes (ASAH2, FASN, SMPD2, SMPD3, PHYH, SPTLC3, SGMS1, SGMS2, SPHK1, SPHK2, UGCG) and eleven conventional oncogenes (AKT1, AKT2, AKT3, MYC, MAPK13, PIK3CB, PI3KCG, PI3KCD, NRAS, KRAS, TNF) with significantly altered expression were analyzed. The frequency of the alterations in expression (either higher or lower) was analyzed in patients with glioma in the REMBRANDT database. The graph shows data from 0 to 8 alterations in lipid metabolism (closed bars) or conventional oncogene (open bars) transcripts. Reprinted/adapted with permission from Ref. [74] Copyright 2014, Elsevier.
Biomedicines 11 01365 g001
Figure 2. Overall survival and gene expression in pediatric gliomas. Kaplan–Meier plots showing the OS in pediatric patients with gliomas with high (red) or low (green) expression of SMS2 (A), FADS1 (B), FABP5 (C), GALC (D) and ASCL4 (E). Survival probability was considered significantly different at p < 0.05 (log rank test). See text for further details.
Figure 2. Overall survival and gene expression in pediatric gliomas. Kaplan–Meier plots showing the OS in pediatric patients with gliomas with high (red) or low (green) expression of SMS2 (A), FADS1 (B), FABP5 (C), GALC (D) and ASCL4 (E). Survival probability was considered significantly different at p < 0.05 (log rank test). See text for further details.
Biomedicines 11 01365 g002
Figure 3. Metabolic pathways regulating lipid metabolism in PBTs, and specific molecular signatures related to plasma membrane receptors, nuclear receptors, and the metabolism of specific lipid subtypes. Arrows indicate good (↑) or bad (↓) prognosis associated with the high expression of ACSL4, GALC, FABP5, FADS1, and SMS2. CER: ceramide FFA: free fatty acids, LDs: lipid droplets, LPA: lysophosphatidic acid, PI3K: phosphatidyl inositol kinases, PLs: phospholipids, PLC: phospholipase C, SM sphingomyelin, S1P: sphingosine phosphate, UPR: unfolded protein response.
Figure 3. Metabolic pathways regulating lipid metabolism in PBTs, and specific molecular signatures related to plasma membrane receptors, nuclear receptors, and the metabolism of specific lipid subtypes. Arrows indicate good (↑) or bad (↓) prognosis associated with the high expression of ACSL4, GALC, FABP5, FADS1, and SMS2. CER: ceramide FFA: free fatty acids, LDs: lipid droplets, LPA: lysophosphatidic acid, PI3K: phosphatidyl inositol kinases, PLs: phospholipids, PLC: phospholipase C, SM sphingomyelin, S1P: sphingosine phosphate, UPR: unfolded protein response.
Biomedicines 11 01365 g003
Figure 4. Survival of PBT patients in relation to the metabolite profiles measured by magnetic resonance spectroscopy in vivo for a range of tumor types. Kaplan–Meier survival plots for (A) lipids at 1.3 PPM, (B) glutamine of 115 pediatric patients study followed up for a median of 35 months. Significance values assessed using a chi squared test for equality. Reproduced with permission of Martin Wilson [129].
Figure 4. Survival of PBT patients in relation to the metabolite profiles measured by magnetic resonance spectroscopy in vivo for a range of tumor types. Kaplan–Meier survival plots for (A) lipids at 1.3 PPM, (B) glutamine of 115 pediatric patients study followed up for a median of 35 months. Significance values assessed using a chi squared test for equality. Reproduced with permission of Martin Wilson [129].
Biomedicines 11 01365 g004
Table 1. Molecular alterations in pediatric CNS tumors and their association with the lipid metabolism and composition.
Table 1. Molecular alterations in pediatric CNS tumors and their association with the lipid metabolism and composition.
Type of TumorWHO GradeMain Molecular AlterationsCodified Proteins Affected by Lipid Metabolism or Lipid Composition RegulationPercentage of Cases
GLIOMAS, GLIONEURONAL TUMORS AND NEURONAL TUMORS (excluding adult-type diffuse glioma)
Pediatric-type diffuse high-grade glioma 11.1
Diffuse midline glioma H3 K27-altered3H3 K27, TP53, ACVR1, PDGFRA, EGFR, EZHIP P53—regulator of lipid metabolism in cancer [11]. Mutations on TP53 provide lipolytic activity to P53 [12].
EGFR is regulated by palmytoilation at Cys1049 and Cys1146 [13].
Diffuse hemispheric glioma, H3 G34-mutant 4H3 G34, TP53, ATRX P53—regulator of lipid metabolism in cancer [11]. Mutations on TP53 provide lipolytic activity to P53 [12].
Atrx—transcriptional factor targeting lipid metabolism mediators.
Diffuse High-grade glioma H3-wild-type and IDH-wild-type 4IDH-wildtype, H3-wildtype, PDGFRA,
MYCN,
EGFR (methylome)
IDH-wild type—IDH1 activity is critical for lipid biosynthesis and its inactivation compromises tumor growth [14]
MCYN—Lipid desaturation-associated endoplasmic reticulum stress regulates MYCN gene expression [15].
EGFR is regulated by palmytoilation at Cys1049 and Cys1146 [13].
Infant-type Hemispheric glioma 4NTRK family, ALK, ROS, MET NTRK, Alk, Ros and MET are transmembrane proteins.
Pediatric-type diffuse Low-grade gliomas25–30
Diffuse astrocytoma, MYB or MYBL1 altered 1–2MYB, MYBL1
Angiocentric glioma1MYB, BRAF V600E mutBRAF V600E mut—induction of lipid droplet accumulation [16]
Polymorphous low-grade neuroepithelial tumor of the young 1BRAF, FGFR family BRAF—the lipogenic pathway is a key mediator of oncogenic BRAF. Inhibition of oncogenic BRAF caused an increase in the proportion of poly-unsaturated membrane phospholipid species at the expense of saturated and mono-unsaturated phospholipids [17].
FGFR—transmembrane protein
Diffuse low-grade glioma MAPK pathway-altered 1FGFR1, BRAFBRAF—the lipogenic pathway is a key mediator of oncogenic BRAF. Inhibition of oncogenic BRAF caused an increase in the proportion of poly-unsaturated membrane phospholipid species at the expense of saturated and mono-unsaturated phospholipids [17].
FGFR1—transmembrane protein.
The altered lipid structure allows one to factor in the protein–lipid interactions and the biophysical properties of the resulting membranes into the regulation of signal transduction pathways such as the MAPK pathway [18].
Circumscribed astrocytic gliomas17.6
Pilocytic astrocytoma1KIAA1549-BRAF, BRAF, NF1BRAF—the lipogenic pathway is a key mediator of oncogenic BRAF. Inhibition of oncogenic BRAF caused an increase in the proportion of poly-unsaturated membrane phospholipid species at the expense of saturated and mono-unsaturated phospholipids [17].
NF1—phospholipid binding protein [19].
High-grade astrocytoma with piloid features 3–4BRAF, NF1, ATRX,
CDKN2A/B
(methylome)
BRAF—the lipogenic pathway is a key mediator of oncogenic BRAF. Inhibition of oncogenic BRAF caused an increase in the proportion of poly-unsaturated membrane phospholipid species at the expense of saturated and mono-unsaturated phospholipids [17].
NF1—phospholipid binding protein [19].
Atrx—transcriptional factor targeting lipid metabolism mediators.
Pleomorphic xanthoastrocytoma2BRAF, CDKN2A/BBRAF—the lipogenic pathway is a key mediator of oncogenic BRAF. Inhibition of oncogenic BRAF caused an increase in the proportion of poly-unsaturated membrane phospholipid species at the expense of saturated and mono-unsaturated phospholipids [17].
Subependymal giant cell astrocytomas (SEGA)1TSC1, TSC2TSC1—inhibition of lipophagy or its downstream catabolic pathway reverses defective phenotypes caused by Tsc1-null NSCs and reduces tumorigenesis in mouse models [20].
TSC2—TSC2-deficient cells have enhanced choline phospholipid metabolism [21]
Astroblastoma, MN1-altered3–4MN1
Ependymal tumors10
Subependymoma1–2
Supratentorial ependymomas ZFTA fusion-positive2ZFTA, RELA
Supratentorial ependymomas, YAP1 fusion positive 2–3YAP1, MAML2YAP1 positively regulates numerous genes related to cancer stemness and lipid metabolism [22]
Posterior fossa ependymomas, group PFA (EZHIP mutation) 2–3H3 K27me3,
EZHIP
(methylome)
Posterior fossa ependymomas, group PFB 2
Spinal ependymomas, MYCN-amplified 3NF2, MYCNNF2—lipid binding results in the open conformation of neurofibromin 2 [23]
MYCN—lipid desaturation-associated endoplasmic reticulum stress regulates MYCN gene expression [15].
Myxopapillary ependymoma2
Neuronal and glioneuronal tumors4.4
Dysembryoplastic neuroepithelial tumors (DNET)2FGFR1FGFR1—transmembrane protein
Gangliogliomas1–2
Diffuse glioneuronal tumor with oligodendroglioma-like features and nuclear clusters (DGONC) 2Chromosome 14, (methylome)
Myxoid glioneuronal tumor (MGT) 2PDFGRA
Multinodular and vacuolating tumor (MVNT) 1MAPK pathway An altered lipid structure allows one to factor in the protein–lipid interactions and the biophysical properties of the resulting membranes into the regulation of signal transduction pathways such as the MAPK pathway [18]
Rosette-forming glioneuronal tumor1FGFR1,
PIK3CA, NF1
FGFR1—transmembrane protein.
PIK3CA—phospholipid binding protein.
NF1—phospholipid binding protein [19].
Myxoid glioneuronal tumor 1PDFGRA
Diffuse leptomeningeal glioneuronal tumor1–3KIAA1549-BRAF fusion, 1p (methylome) BRAF—the lipogenic pathway is a key mediator of oncogenic BRAF. Inhibition of oncogenic BRAF caused an increase in the proportion of poly-unsaturated membrane phospholipid species at the expense of saturated and mono-unsaturated phospholipids [17].
Gangliocytoma1
Dysplastic cerebellar gangliocytoma (Lhermitte-Duclos disease)1PTENPTEN—phospholipid binding protein which also interacts with FABP4 [24]
Central neurocytoma2
Extraventricular neurocytoma2FGFR (FGFR1-TACC1 fusion), IDH-wild typeFGFR—transmembrane protein
Cerebellar liponeurocytoma2
CNS EMBRYONAL TUMORS
Medulloblastoma20.0
Medulloblastoma, molecularly defined4
Medulloblastoma, WNT-activated4CTNNB1, APCCTNNB1—ß-catenin strongly promotes ß-oxidation [25]
Medulloblastoma, SHH-activated and TP53-wild-type4PTCH1, SUFU, SMO, MYCN,
GLI2 (methylome)
PTCH1, GLI2—lipid metabolism has a profound influence on both hedgehog signal transduction and the properties of the ligands themselves [26]
SMO—Hh signaling transduces to SMO through modulating its cholesterylation [27].
Medulloblastoma, SHH-activated and TP53-mutant4TP53, PTCH1, SUFU, SMO, MYCN, GLI2 (methylome) TP53—mutations on TP53 provide lipolytic activity to P53 [12].
PTCH1, GLI2—Lipid metabolism has a profound influence on both hedgehog signal transduction and the properties of the ligands themselves [26]
SMO—Hh signaling transduces to SMO through modulating its cholesterylation [27].
Medulloblastoma, non-WNT/non-SHH3–4MYC, MYCN, PRDM6, KDM6A (methylome) MYC—fatty acids are inhibitors of the DNA binding of c-Myc/Max dimer [28]
MCYN—lipid desaturation-associated endoplasmic reticulum stress regulates MYCN gene expression [15,29].
Medulloblastoma, histologically defined3–4
Other CNS embryonal tumors
Atypical teratoid/rhabdoid tumor (ATRT)4SMARCB1,
SMARCA4
SMARCB1—also known as SWI/SNF-related matrix-associated protein, related also to SMARCA4—BAF60a and BAF60c, two subunits of the SWI/SNF chromatin-remodeling complexes, are important for maintaining hepatic lipid metabolism. SWI/SNF complex might be targeted to develop drugs aimed at regulation of lipid homeostasis in hepatic steatosis [30].
Cribriform neuroepithelial tumor (provisional type)3–4
Embryonal Tumor with Multilayer Rosettes (ETMR)4C19MC, DICER1 DICER1—the loss of miRNAs resulting from Dicer1 deficiency greatly contributes to the progression of many diseases, including lipid dysregulation [31].
Neuroblastoma, FOXR2-activated 4FOXR2
CNS tumor with BCOR internal tandem duplication 4BCOR
Embryonal tumor NEC/NOS4
TUMORS OF THE SELLAR REGION
Craniopharyngioma4.0
Adamantinomatous craniopharyngioma1CTNNB1CTNNB1—ß-catenin strongly promotes ß-oxidation [25]
Papillary craniopharyngioma1BRAFBRAF—the lipogenic pathway is a key mediator of oncogenic BRAF. Inhibition of oncogenic BRAF caused an increase in the proportion of poly-unsaturated membrane phospholipid species at the expense of saturated and mono-unsaturated phospholipids [17].
Pituitary endocrine tumors3.9
Pituitary blastoma 1–4DICER1Dicer—Dicer disruption caused a marked decrease in microsomal triglyceride transfer protein, long-chain fatty acyl-CoA ligase 5, fatty acid binding protein, and very-long-chain fatty acyl-CoA dehydrogenase [32].
MELANOCYTIC TUMORS
Meningeal melanocytosis and melanomatosis1–3 2.5
GERM CELL TUMORS
1 3.7
MENINGIOMAS
Meningioma1–3NF2, AKT1, TRAF7, SMO, PIK3CA; KLF4, SMARCE1,
BAP1 in subtypes; H3K27me3; TERT promoter, CDKN2A/B in CNS WHO grade 3
NF2—lipid binding results in the open conformation of neurofibromin 2 [23].
SMO—Hh signaling transduces to SMO through modulating its cholesterylation [27].
PIK3CA—phospholipid binding protein.
KLF4—regulates cholesterol metabolism by endothelial cells [33].
2.9
CHOROID PLEXUS TUMORS
2.3
Plexus papilloma1
Atypical plexus papilloma2
Plexus carcinoma3
Plexus papilloma1
PINEAL TUMORS
3–11
Pineocytoma1
Pineoblastoma4
Papillary tumor of pineal region2–3
OTHER/UNCLASSIFIED TUMORS
4.9
Table 2. Genes involved in lipid metabolism referred to herein.
Table 2. Genes involved in lipid metabolism referred to herein.
Official SymbolEnzyme Name
ACER1ASAH1; Alkaline Ceramidase 1
ACER3ASAH3; Alkaline Ceramidase 3
ACSL1Acyl-CoA Synthetase Long-Chain Family Member 1
ACSL3Acyl-coA synthetase Long Chain Family member 3
ACSL4Acyl-coA synthetase Long Chain Family member 4
ACSL5Acyl-coA synthetase Long Chain Family member 5
AHRAryl Hydrocarbon Receptor
ALDH3A2Fatty Aldehyde dehydrogenase
ASAH2Ceramidase, non-lysosomal
CD36CD36 Molecule
CEPT1Choline/Ethanolamine Phosphotransferase 1
CERS1LASS1, Ceramide Synthase 1
DEGS1Delta 4-Desaturase, Sphingolipid 1
FA2HFatty Acid 2-hydroxylase
FABP5Fatty Acid Binding Protein 5
FABP7Fatty Acid Binding Protein 7
FADS1Fatty Acid Desaturase 1
FADS2Fatty Acid Desaturase 2
FASNFatty Acid synthase
FFAR1Free Fatty Acid Receptor 1; GPR40
FFAR2Free Fatty Acid Receptor 2; GPR43
FFAR3Free Fatty Acid Receptor 3; GPR41
FFAR4Free Fatty Acid Receptor 4; GPR120; O3FAR1
GALCGalactosylceramidase
GPR42G Protein-Coupled Receptor 42 (Gene/Pseudogene); FFAR1L
HACL12-hydroxypythanoyl-coA-lyase, 2-hydroxyacyl-CoA lyase 1
HSPA5BiP, GRP78; Heat Shock Protein Family A (Hsp70) Member 5
LPAR1LPA1; Lysophosphatidic Acid Receptor 1
LPAR2LPA2; Lysophosphatidic Acid Receptor 2
LPAR3LPA3; Lysophosphatidic Acid Receptor 3
LPAR4LPA4; Lysophosphatidic Acid Receptor 4
LPAR5LPA1; Lysophosphatidic Acid Receptor 5
LPAR6LPA1; Lysophosphatidic Acid Receptor 6
NR1H3LXRA; Liver X Nuclear Receptor Alpha Variant 1
NSMAFN-Smase; Neutral Sphingomyelinase Activation Associated Factor
PEMTPhosphatidylethanolamine N-Methyltransferase
PHYHPhytanoyl-CoA 2-hydroxylase
PPARaPeroxisome Proliferator Activated Receptor Alpha
PPARbPeroxisome Proliferator Activated Receptor Beta
PPARdPeroxisome Proliferator Activated Receptor Delta
PPARgPeroxisome Proliferator Activated Receptor Gamma
S1P1Sphingosine-1-Phosphate Receptor 1
S1P2Sphingosine-1-Phosphate Receptor 2
S1P3Sphingosine-1-Phosphate Receptor 3
S1P4Sphingosine-1-Phosphate Receptor 4
S1P5Sphingosine-1-Phosphate Receptor 5
SAMD8SMSr; CEP Synthase; Sterile Alpha Motif Domain Containing 8
SCDStearoyl CoA desaturase
SMS1Sphingomyelin synthase 1
SMS2Sphingomyelin synthase 2
SGPL1Sphingosine-1-Phosphate Lyase 1
SMPD1Acid Sphingomyelinase
SMPD2Neutral sphingomyelinase 1
SMPD3Neutral sphingomyelinase 2
SMPD4Neutral sphingomyelinase 3
SMPDL3AAcid Sphingomyelinase-Like Phosphodiesterase 3a
SMPDL3BAcid Sphingomyelinase-Like Phosphodiesterase 3b+C1:C63
SPHK1Sphingosine kinase 1
SPHK2Sphingosine kinase 2
SPTLC3Serine palmitoyl Transferase, long chain subunit 3
TLR2Toll-Like Receptor 2
UGCGUDP-Glucose Cer Glucosyltransferase (GluCer synthase)
Genes that were not present in the array from the database used are indicated in red.
Table 3. Cases of transcription factors controlling lipid metabolism mediator expression.
Table 3. Cases of transcription factors controlling lipid metabolism mediator expression.
Official Symbol Transcription Factor Name Target Lipid Metabolism Mediator
AHR Aryl Hydrocarbon Receptor To be determined [94]
AP-1 Activator protein 1 ASAH2 [95]
SPHK1 [96]
AP-2 Transcription Factor AP-2 Alpha ASAH2 [95]
Atrx Alpha Thalassemia/Mental Retardation Syndrome X-Linked Several complexes along the chromosome maintain different states of chromatin [97]
Atf-4 Activating Transcription Factor 4 SPHK2 [98,99]
BCL11B B-Cell Lymphoma/Leukaemia 11B SMPD2 [100]
CREB CAMP Responsive Element Binding Protein 1 SPHK2 [98]
E2F E2F Transcription Factor 1 SPHK1 [101]
Fos FBJ Murine Osteosarcoma Viral Oncogene Homolog SMPD3 [102,103,104,105]
GATA GATA Transcription Factor ASAH2 [95]
Hey1 Hes Related Family BHLH Transcription Factor With YRPW Motif 1 ACVR1 [106]
HIF1α Hypoxia-inducible factor 1-alpha PDGFRA [107]
HIF2α Hypoxia-inducible factor 2-alpha SPHK1 [108]
IRF1Interferon-regulatory factor-1ASCL4 [109,110]
LMO2 LIM domain only 2 rhombotin-like 1 SPHK1 [111]
NF-Y Nuclear factor Y ASAH2 [95]
FASN [112,113]
Oct-1 POU Class 2 Homeobox 1 ASAH2 [95]
SP1 Specificity Protein 1 ASAH2 [95]
UGCG [95,114]
ZBTB7A/LRF Zinc Finger And BTB Domain Containing 7A/Lymphoma Related Factor ACVR1 [106]
Table 4. Selection of current clinical trials in pediatric patients with brain tumors.
Table 4. Selection of current clinical trials in pediatric patients with brain tumors.
Drug TypeExample AgentsTargetDiseasePediatric Clinical Trial
ImmunomodulatorsAPX005MCD40 agonistGBM, A, CNST, E, DIPG, MBNCT03389802
PomalidomideTNFaCNSTSNCT03257631
IndoximodIDO, mTORE, MB, GBM, DIPGNCT05106296
NCT04049669
NKTR-214CD122 IL2 pathway agonistE, HGG, MB, PBTsNCT04730349
AntibodiesMagrolimabCD47PBTsNCT05169944
AvelumabPD-L1CNSTsNCT05081180
NivolumabPD-1 receptorCNSTsNCT03838042
NCT04500548
IpilimumabCTLA-4CNSTsNCT04500548
BevacizumabVEGF-APBTsNCT02698254
CAR T Cells and other cellular immunotherapiesHER2-specific CAR T cell locoregional ImmunotherapyHER2G, E, MB, GCT, ATRT, PBNCT03500991
EGFR806-specific CAR T cell locoregional ImmunotherapyEGFRG, E, MB, GCT, ATRT, PNET, CPC, PBNCT03638167
B7-H3-specific CAR T Cell locoregional ImmunotherapyB7H3DIPG, DMG, E, MB, GCT, ATRT, CPC, PB, GNCT04185038
GD2-CART01 (iC9-GD2-CAR T-cells)Disialoganglioside GD2MB, PBTsNCT05298995
IL13Ralpha2-specific hinge-optimized 41BB-co-stimulatory CAR truncated CD19IL13Ralpha2PBTsNCT04510051
Haploidentical transplant and donor NK cell infusion CNSTsNCT02100891
Bone marrow-derived allogenic mesenchymal stem cells infected with an oncolytic adenovirus, ICOVIR-5pRB pathwayDIPG, MBNCT04758533
VaccinesPEP-CMVCMV antigenHGG, DIPG, MBNCT03299309 NCT05096481
Personalized neoantigen DNA vaccine DMG, DIPGNCT03988283
rHSC-DIPGVax (neo-antigen heat schock protein vaccine) DMG, DIPGNCT04943848
Dendritic cell vaccination: WT1 mRNA-loaded autologous monocyte-derived DCs HGG DIPGNCT04911621
Immunomodulatory DC vaccine DIPG, GBMNCT03914768
SurVaxMSurvivinMB, GBM, AA, A, NOS, AO, AE, E, DIPGNCT04978727
K27M peptide DIPG, DMGNCT02960230
Viral TherapyHSV G207 oncolytic herpes simplex virus-1 (HSV) CNSTsNCT03911388 NCT02457845
Wild-type reovirus (reolysin) HGGsNCT02444546
Polio/rhinovirus recombinant (PVSRIPO)CD155 nectin-like molecule-5CNSTsNCT03043391
DNX-2401 oncolytic adenovirusIntegrinsBSG, DIPGNCT03178032
Conventional chemotherapeuticsMebendazole:TubulinMB, A, GB, AA, Brain Stem Neoplasms, O, AO, GNCT02644291
PTC596TubulinDIPG, HGGNCT03605550
AntimetabolitesPemetrexedFolate analogMBNCT01878617
HydroxyureaRRM2G, GBMNCT03463733
New chemotherapeuticsMarizomibProteasomeDIPG, BSG, PBTsNCT04341311
ALRN-6924MDM2/MDMXPBTsNCT03654716
Curaxin CBL0137FACTDMG, DIPG, CNSTsNCT04870944
Kinase InhibitorsCX-4945 silmitasertibCK2MBNCT03904862
PrexasertibChk1MBNCT04023669
9-ING-41GSK 3βPBTsNCT04239092
TrametinibMEK1, MEK2PBTsNCT03434262
IbrutinibBruton’s tyrosine KinaseE, MB, GBMNCT05106296
LenvatinibVEGFR1, 2 and 3, FGFR1, 2, 3 and 4, PDGFR alpha, c-Kit, RET proto-oncogeneCNSTsNCT05081180
NCT03245151
AlectinibALKCNSTsNCT04774718
LarotrectinibTropomyosin receptor kinasesCNSTsNCT03213704
NCT03834961
NCT03155620
Repotrectinib (TPX-0005)ALK, ROSCNSTsNCT04094610
Downstream signaling pathway inhibitorsVemurafenibB-Raf. BRAFV600GNCT01748149 NCT03220035
EntrectinibTRKA, TRKB, TRKC, ROS1, ALKCNSTsNCT02650401
ONC206Stress response, DRD2/ClpPDMG, CNSTsNCT04732065
Everolimus immunosuprmTor, FKBP-12HGG, PNETNCT03245151
Sirolimus immunosuprmTor, FKBP-12CNSTsNCT02574728
GDC-0084PI3K/mTorCNSTsNCT03696355
WP1066JAK/STAT3PBTsNCT04334863
IndoximodIDO, mTORE, MB, GBMNCT05106296
MEK162Ras/Raf/MEKLGGNCT02285439
TrametinibMEK1/2PBTsNCT04485559
NCT03363217
NCT05180825
NCT02684058
NCT04201457
Developmental pathway inhibitorsVismodegibSMOMBNCT01878617
Cell Death Pathway inducersONC201TRAIL, ISRDIPG, DMG, HGGNCT05009992
NCT05580562
Angiogenesis inhibitorRecombinant human endostatin (rh-ES)Ras, Raf, VEGF, VEGFR2LGGNCT04659421
Epigenetic therapyBMS-986158 and BMS-986378Bromodomain (BRD) and extra-terminal domain (BET)PBTsNCT03936465
RRx-001DNMT and global methylationPBTsNCT04525014
PanobinostatHDACDIPG, BSG, PBTsNCT02717455
NCT04341311
MRT/ATRTNCT04897880
EntinostatClass I and IV HDACCNSTsNCT03838042
TazemetostatEZH2CNSTsNCT03213665
VorinostatHDACBSG, A, CAA, CSCNNCT01236560
BMS-986158Bromodomain and extra-terminal (BET) proteinsPBTsNCT03936465
Melitherapy2-hydroxyoleic acidPlasma membrane compositionPBTsNCT04299191
BXQ-350Plasma membrane sphingolipid modulationDIPG, DMG PBTsNCT04771897
NCT04404569
Radiolabeled drugsRadiolabeled phospholipid drug conjugate: CLR 131 radioiodinated phospholipid ethers (PLEs) Lipid rafts of cancer cell membranesPBTs,NCT03478462
Peptide receptor radionuclide: lutathera (177Lu-DOTATATE) Somatostatin receptorsCNSTsNCT05278208
Radiolabelled monoclonal antibody: iodine I 131 MOAB 8H9 4Ig-B7-H3CNSTsNCT00089245
Abbreviations: A: astrocytoma, AA: anaplastic astrocytoma, AE: anaplastic ependymoma, ATRT: atypical teratoid rhabdoid tumor, AO: anaplastic oligodendroglioma, BSG: brain stem glioma, CAA: cerebellar anaplastic astrocytoma, Chk1: checkpoint kinase 1, CK2: protein casein kinase II, ClpP: human mitochondrial caseinolytic protease P, CNSTs: central nervous system tumors, CPC: choroid plexus carcinoma, CSCN: childhood spinal cord neoplasm, DIPG: diffuse intrinsic pontine glioma, DMG: diffuse midline gliomas, DNMT: DNA methyltransferase, DRD2: dopamine receptor D2, E: ependymoma, G: glioma, FACT: facilitates chromatin transcription complex, G: glioma, GBM: glioblastoma multiforme, GCT: germ cell tumor, HDAC: histone deacetylase, HGG: high-grade glioma, IDO: indoleamine 2,3-dioxygenase, ISR: TRAIL, integrated stress response LGG: low-grade glioma, MB: medulloblastoma, MRT: malignant rhabdoid tumor, O: oligodendroglioma, PB: pineoblastoma, PBTs: pediatric brain tumors, PD-L1: programmed death-ligand 1, pRB: retinoblastoma tumor suppressor protein, RRM2: M2 protein subunit of ribonucleotide reductase.
Table 5. Selection of pre-clinical stage therapeutic drugs of lipid nature (Italic) or targeting lipid-related pathways in brain tumors.
Table 5. Selection of pre-clinical stage therapeutic drugs of lipid nature (Italic) or targeting lipid-related pathways in brain tumors.
CategoryDrug AgentFamilyTargetAffected PathwaysDiseaseModelReference
PediatricCordycepinNucleoside derivativemiR-33Lipid metabolismMBOrthotropic xenograft[172]
GSK126+ AtorvastatinSmall molecule inhibitorsEZH2Cholesterol synthesisDIPGMurine orthotopic model[173]
ABC294640Small molecule inhibitorSphK2Sphingolipid metabolismDIPGSF8628 and SF7761 Soft agar[174]
Carbenoxolone + palbociclibSmall molecule inhibitorsHSD11β2- CDK4/6Oxysterol biosynthesisMBTransgenic[175]
GW9662Small molecule agonistBLBPFatty acid uptakeE3D spheroid[176]
ω3-LCPUFAFatty acidsCRYABProtein foldingMBXenograft[177]
ErucylphosphocholineEther lipidMembraneApoptosisMBD283 Med[178]
GeneralFluoxetineSmall molecule inhibitorSMDP-1Sphingolipid metabolismGBMOrthotropic xenograft[179]
Triacsin C + EtoximirSmall molecule inhibitorsACSL1- ACSL3-CPT1Lipid biosynthesis and fatty acid oxidationMesenchymal GBMXenograft[180]
LAU-0901 + Avastin + ElovanoidsSmall molecule + synthetic lipidsPAFR Tumor cell proliferationGBMOrthotropic xenograft[181]
Arachidonyl trifluoromethyl ketoneSmall molecule inhibitorPTRF(cavin-1)Phospholipid metabolismGBMIntracranial Patient-Derived Xenograft Model[182]
CAY10566Small molecule inhibitorSCD1LipogenesisGBMXenograft[183]
YTX-7739Small molecule inhibitorSCDLipogenesisGBMOrthotropic xenograft[184]
EtomoxirSmall molecule inhibitorCPT1Fatty acid oxidationGBMSyngeneic[185]
AzathioprinePurine analogueEGFR-AKTLipid metabolismGBMOrthotropic xenograft[186]
Ophiobolin ATerpenoid antagonistPEMembrane DestabilizationGBMorthotopic U251-LUC xenograft[187]
LXR-623Small molecule agonistLXRCholesterol metabolismGBMOrthotropic xenograft[188]
GM3GangliosideVEGFTumor angiogenesis.ACT-2A Matrigel[189]
Abbreviations: A: astrocytoma, ACSL: adipose acyl-CoA synthetase, AKT: protein kinase B, BLBP: brain lipid-binding protein, CDK: cyclin-dependent kinase, CPT1: carnitine palmitoyltransferase I, CRYAB: alpha-crystallin B chain, DIPG: diffuse intrinsic pontine glioma, E: ependymoma, EGFR: epidermal growth factor receptor, EZH2: enhancer of zeste homolog 2, GBM: glioblastoma, HSD11b2: 11β-hydroxysteroid dehydrogenase, type 2, LXR: Liver X receptor, MB: medulloblastoma, PE: phosphatidylethanolamine, PTRF: polymerase I and transcript release factor, SCD1: stearoyl-CoA desaturase-1, SMDP1: surfactant metabolism dysfunction, pulmonary, type 1, SPHK2: sphingosine kinase 2, VEGF: vascular endothelial growth factor.
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

Fernández-García, P.; Malet-Engra, G.; Torres, M.; Hanson, D.; Rosselló, C.A.; Román, R.; Lladó, V.; Escribá, P.V. Evolving Diagnostic and Treatment Strategies for Pediatric CNS Tumors: The Impact of Lipid Metabolism. Biomedicines 2023, 11, 1365. https://doi.org/10.3390/biomedicines11051365

AMA Style

Fernández-García P, Malet-Engra G, Torres M, Hanson D, Rosselló CA, Román R, Lladó V, Escribá PV. Evolving Diagnostic and Treatment Strategies for Pediatric CNS Tumors: The Impact of Lipid Metabolism. Biomedicines. 2023; 11(5):1365. https://doi.org/10.3390/biomedicines11051365

Chicago/Turabian Style

Fernández-García, Paula, Gema Malet-Engra, Manuel Torres, Derek Hanson, Catalina A. Rosselló, Ramón Román, Victoria Lladó, and Pablo V. Escribá. 2023. "Evolving Diagnostic and Treatment Strategies for Pediatric CNS Tumors: The Impact of Lipid Metabolism" Biomedicines 11, no. 5: 1365. https://doi.org/10.3390/biomedicines11051365

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