Next Article in Journal
Location and Movement of the Oxytocin Receptor Differ Between the Normal and Diseased Prostate
Next Article in Special Issue
Clinical Benefit of Pegylated Liposomal Doxorubicin and High Prevalence of Pre-Existing Psychiatric Conditions in Patients with Desmoid-Type Fibromatosis
Previous Article in Journal
Proximity Labeling: Precise Proteomics Technology for Mapping Receptor Protein Neighborhoods at the Cancer Cell Surface
Previous Article in Special Issue
UBR5 in Tumor Biology: Exploring Mechanisms of Immune Regulation and Possible Therapeutic Implications in MPNST
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Sequencing Overview of Malignant Peripheral Nerve Sheath Tumors: Findings and Implications for Treatment

Division of Oncology, Department of Internal Medicine, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(2), 180; https://doi.org/10.3390/cancers17020180
Submission received: 23 November 2024 / Revised: 6 January 2025 / Accepted: 7 January 2025 / Published: 8 January 2025
(This article belongs to the Special Issue Sarcoma: Clinical Trials and Management)

Simple Summary

In recent decades, advancements in high-throughput sequencing technology, coupled with reduced sequencing costs, have led to a significant increase in the genomic profiling of benign and malignant peripheral nerve sheath tumors. This review synthesizes recent sequencing discoveries from multiple sequencing technologies, underscores the critical mutation events involved in tumor pathogenesis, and explores their potential therapeutic implications. By elucidating the molecular basis of these tumors, clinicians and researchers can improve patient outcomes and provide a foundation for more effective treatment strategies.

Abstract

Malignant peripheral nerve sheath tumors (MPNSTs) are rare but aggressive malignancies with a low 5-year survival rate despite current treatments. MPNSTs frequently harbor mutations in key genes such as NF1, CDKN2A, TP53, and PRC2 components (EED or SUZ12) across different disease stages. With the rapid advancement of high-throughput sequencing technologies, the molecular characteristics driving MPNST development are becoming clearer. This review summarizes recent sequencing studies on peripheral nerve sheath tumors, including plexiform neurofibromas (PNs), atypical neurofibromatous neoplasm with uncertain biologic potential (ANNUBP), and MPNSTs, highlighting key mutation events in tumor progression from the perspectives of epigenetics, transcriptomics, genomics, proteomics, and metabolomics. We also discuss the therapeutic implications of these genomic findings, focusing on preclinical and clinical trials targeting these alterations. Finally, we conclude that overcoming tumor resistance through combined targeted therapies and personalized treatments based on the molecular characteristics of MPNSTs will be a key direction for future treatment strategies.

Graphical Abstract

1. Overview of MPNSTs

The earliest MPNST in a human can be traced back to 1952, which was described as an unclassified tumor which resembles a neurofibroma in a Caucasian male [1]. In 1984, Ducatman et al. reported 16 cases of MPNSTs in children under 16, with a mean survival of only 1.8 years [2]. In 1986, a study reviewed 120 cases of MPNSTs and concluded that total resection of the tumor could improve the prognosis of the MPNST. Surgery is still the main treatment for MPNSTs today, but recurrence rates are high [3]. MPNSTs predominant locate along the main nerve bundles [4]. While MPNSTs are traditionally characterized as Schwann cell-derived tumors, this may oversimplify their heterogeneous nature. A recent study highlights the fact that low-grade MPNSTs should be identified as ANNUBP with increased proliferation [5]. The presenting symptom of MPNSTs is typically a gradually enlarging painless mass, which, as the tumor grows to a certain size, compresses adjacent tissues and becomes painful [6]. The MPNST is ranked as the sixth most common soft tissue sarcoma, consisting of 2%~10% of soft tissue sarcomas, annually [7].
The treatment for MPNSTs depends on the stage of the disease. For localized MPNSTs, complete surgical resection with or without chemotherapy and radiation remains the most effective therapy to date [8,9]. A recent retrospective analysis of an MPNST cohort showed that some patients receiving radiation therapy had a better overall survival rate, indicating that radiation therapy may be beneficial for MPNSTs [10]. However, the effect on overall survival with MPNST requires further clinical validation [11]. For those patients who have already developed metastasis, chemotherapy—typically doxorubicin and ifosfamide-based regimen—remain the main treatment, but only extends life expectancy by 1 to 2 years [12,13]. The overall prognosis remains poor, despite surgical resection, adjuvant radiation, or chemotherapy, with the five-year overall survival rate of 47.2% [14].
NF1 is one of the most common tumor predisposition syndromes and is caused by the loss of function of the NF1 tumor suppressor gene. Patients have an increased risk of developing benign tumors, as well as malignancies including soft tissue sarcoma, glioma, breast cancer, and melanoma [15]. Fifty percent of MPNSTs occur in patients with NF1, while 40% occur sporadically, and 10% occur in the setting of prior radiation [16]. The etiology of sporadic MPNSTs is not entirely understood, but the tumors are thought to arise de novo instead of from PNs and ANNUBP [17]. The pathogenesis of NF1-associated MPNST involves dysregulation of multiple genes and progression through several disease stages. In addition to the NF1 gene loss, several other key genes including CDKN2A, TP53 and EED/SUZ12 have been identified as being involved in the pathogenesis of MPNSTs [18,19,20]. In the context of NF1, the loss of the second copy of the NF1 gene along with cues from the heterozygous microenvironment leads to the development of a PN, which is a benign tumor that can cause pain, mobility dysfunction, and organ compromise, depending on the location [21]. The ANNUBP was proposed in 2017 and is an intermediate pre-malignant disease stage between PNs and MPNST [22]. It has been shown that loss of CDKN2A is correlated with ANNUBP [23] and further loss of PRC2 components, TP53, or other alterations are likely to drive the malignant transformation from ANNUBP to MPNST [24,25,26].
In the last several decades, with the rapid development of high-throughput sequencing technology and the reduction of sequencing costs, many MPNST samples have been sent for sequencing, including micro-array, RNA-sequencing (RNA-seq), methylation-sequencing, single-cell RNA sequencing, whole exome sequencing (WES) and whole genome sequencing (WGS). The complex genomic characteristics of MPNSTs are gradually being uncovered at multiple levels. Here we summarize recent MPNST-associated sequencing studies, highlighting potential targeted therapy options.

2. Sequencing Technologies and Its Application in MPNST

Researchers have utilized sequencing studies on MPNSTs with several research focuses. (1) Identification of molecular subtypes and diagnostic tools of MPNST. (2) Comparison of differential gene expression between MPNST and peripheral nerve sheath tumor (PNST). (3) Investigation of the pharmacogenomic changes following various drug treatments. (4) Identification of functional genes in MPNSTs by analyzing genetic mutations and gene-noncoding RNA interactions. The following chapters will introduce in detail the application of various sequencing technologies in MPNSTs. The summary of key findings is shown in Table 1 and Figure 1.

2.1. Microarray and RNA-Seq Analysis

Microarrays are based on hybridization of pre-designed labeled probes with target cDNA sequences [27]. To date, microarray sequencing technology is still one of the most common technologies due to its relatively low cost and straightforward pipeline and has been widely applied in MPNST samples. These datasets employ a range of platforms, including Affymetrix, Agilent, and Illumina. However, the accuracy of this technology relies heavily on the pre-designed probes, and the affinity of the probe hybridization [28]. Therefore, microarray technology is not the best fit for samples with low-abundance transcripts, and cannot distinguish isoforms or identify genetic variations. In addition, probes are often accompanied by problems such as cross-hybridization and non-specific hybridization [28].
Table 1. Recent sequencing summary of MPNST-related samples.
Table 1. Recent sequencing summary of MPNST-related samples.
Data TypeOrganismSample SettingsReference
MPNSTPNNFANNUBP
MicroarrayHumanTissue64NA15NAGSE241224 (Høland et al., 2023) [29]
MicroarrayHumanTissue610910GSE239561 (Rhodes, 2023) [30]
MicroarrayHumanTissue10NANANAGSE52390, GSE52391 (Wolf et al., 2013) [31]
DNA methylationHumanTissue10NANANA
MicroarrayHuman Tissue6NA26NAGSE41747 (Jessen et al., 2012) [32]
MicroarrayMouseTissue18NA15NA
MicroarrayHuman Tissue16NANANAGSE17118 (Lafferty-Whyte et al., 2010) [33]
MicroarrayHuman Tissue3NA3NAGSE52252 (Wang et al., 2014) [34]
MicroarrayHuman Tissue30NA8NAGSE66743 (Kolberg et al., 2015) [35]
MicroarrayHuman Tissue4NANANAGSE77203 (Yasuhiro et al., 2019) [36]
MicroarrayHuman Cell21NANANAGSE39764 (Sun et al., 2013) [37]
MicroarrayHuman Tissue3NANANAGSE35852, GSE35851 (Kelly et al., 2012) [38]
micro-RNAHumanTissue3NANANA
MicroarrayHuman Cell22NANANAGSE8717 (Mahller et al., 2007) [39]
MicroarrayHuman Cell20NANANAGSE47476 and GSE47477 (Zhang et al., 2013) [40]
micro-RNAHumanCell12NANANA
MicroarrayHuman Cell9NANANAGSE62500 (De Raedt et al., 2014) [41]
MicroarrayHuman Cell12NANANAGSE84205 (Malone et al., 2017) [42]
MicroarrayHuman PDX/Tissue11NANANAGSE60082 (Castellsagué et al., 2015) [43]
MicroarrayMouseTissue8NANANAGSE57141 (Malone et al., 2014) [44]
Bulk RNA-seq HumanPDX13NANANAsyn11638893 (Hirbe et al., 2022) [45]
WES/WGSHumanPDX16NANANA
scRNA-seq HumanPDX7 MPNST
Bulk RNA-seq HumanTissue73NA12EGAD00001008608 (Genomics of Malignant Peripheral Nerve Sheath Tumor (GeM) Consortium, 2020) [46]
WGSHumanTissue72NA23
Bulk RNA-seq HumanTissue41NANANAGSE206527 and GSE179699 (Chi et al., 2022) [47]
ATAC-seqHumanCell12NANANA
Bulk RNA-seq HumanTissue9NA8NAGSE178989, GSE179033 and GSE179041 (Wu et al., 2022) [48]
scRNA-seq HumanTissue PN vs. MPNST
scRNA-seq MouseTissue1.5-month MPNST vs. 4-months MPNST
DNA methylationHumanCell63NANANAGSE141438, GSE141435 and GSE141437 (Kochat et al., 2021) [49]
Bulk RNA-seq HumanTissue7NA3NA
Bulk RNA-seq HumanCell36NANANA
Bulk RNA-seq HumanTissue2521NANAGSE145064 (Kohlmeyer et al., 2020) [50]
Bulk RNA-seq HumanTissue1212NANAGSE212964 (Vasudevan et al., 2023) [51]
scRNA-seqHumanTissue3 MPNST vs. 3 PN
Bulk RNA-seq HumanTissue14NA34NAGSE207400, PRJNA854920 and GSe207399 (Suppiah et al., 2023) [52]
WESHumanTissue18NA16NA
scRNA-seqHumanTissueANNUBP vs. MPNST
Bulk RNA-seq HumanTissue10NANANATCGA-SARC (https://www.cancer.gov/tcga, accesed on 28 August 2024)
Bulk RNA-seq HumanTissue6NANANAGSE120685 (Woodhoo et al., 2021) [53]
Bulk RNA-seq HumanCell28NANANAGSE183308, GSE183307 (Zhang et al., 2022) [54]
scRNA-seq HumanTissue primary vs. metastasis
Bulk RNA-seq HumanTissue3NA3NAGSE270880 (Zhang et al., 2024) [55]
Bulk RNA-seq HumanCell16NANANAGSE179585, GSe179586 (Patel et al., 2022) [56]
DNA methylationHumanCell16NANANA
Bulk RNA-seq HumanCell6NA10NAGSE118185 (Wassef et al., 2019) [57]
Bulk RNA-seq HumanCell6NANANAGSE213988 (Chung et al., 2022) [58]
Bulk RNA-seq HumanCell4NANANAGSE216792
WGSHuman Tissue (blood)4623NANAsyn23651229 (Shern et al., 2020) [59]
WESHumanTissue51NANANAEGAS0000100452 (Lyskjær et al., 2020) [60]
WESHumanTissue23NANAHirbe et al., 2015 [61]
WESHumanTissue15NANANALee et al., 2014 [19]
WESHumanTissue6NANANAGodec et al., 2020 [62]
WES/WGSHumanTissue11NA2NAKinoshita et al., 2020 [63]
DNA methylationHumanTissue1NA1NAGSE21714 (Feber et al., 2011) [64]
DNA methylationHumanCell16NANANAGSE263127 (Bhunia et al., 2024) [65]
DNA methylationHumanTissue102NANANAE-MTAB-8864 www.ebi.ac.uk/biostudies/arrayexpress/studies/E-MTAB-6961, accesed on 12 December 2024)
DNA methylationHumanTissue8NANANAGSE36982 (Renner et al., 2012) [66]
micro-RNAHumanTissue199NANAGSE140987 (Wiemer, 2019) [67]
ATAC-seqHumanCell6NANANAGSE275047
NA: Not applicable; PDX: Patient-derived xenograft; ATAC-seq: Assay for Transposase-Accessible Chromatin with sequencing.
Given the limitation of microarray sequencing, RNA-seq, one of the most common next-generation sequencing technologies, has been widely applied in the field of biomedicine [68]. RNA-seq has several advantages: first, it can detect low-abundance transcripts. Second, it does not rely on designed probes or primers in advance. Thirdly, new transcripts and splice variants can also be discovered [69].
Molecular classification has become increasingly important in tumor diagnostics and treatment. For instance, in 2016, medulloblastoma was subdivided into multiple molecular subtypes, including the Sonic Hedgehog (SHH) and WNT subtypes, based on transcriptomic data [70]. This subtype-specific classification has since enabled the development of tailored therapeutic strategies, significantly improving patient prognoses [71]. Similarly, several studies have utilized computational algorithms to identify different gene expression patterns to define subtypes of MPNST. For example, Holand et al. utilized the transcriptome of the MPNST to perform clustering analysis and identified two immune-related clusters of MPNSTs which correlated with patient prognosis [29]. They predicted that EGFR, EZH2, KIF11, PLK1 and RRM2 could be potential therapeutic targets for MPNSTs. The largest MPNST sequencing study to date was performed by the GeM consortium, which suggested that genomic expression pattern could predict the prognosis of MPNST better than clinical or pathological evidence alone [72]. In 2023, Suppiah et al. identified two distinct MPNST subtypes, which were associated with activated SHH pathway and WNT pathway, respectively. They found that further targeting of the SHH pathway inhibited growth of MPNST with poor prognosis [52]. Taken together, these analyses highlight the heterogeneity of MPNSTs and revealed different gene expression signatures, which may inform the precision medicine of MPNSTs in the future.
Other studies have focused on comparison between MPNSTs and normal adjacent samples, PN, ANNUBP and benign NF, respectively. For example, Kohlmeyer et al. identified the upregulation of genes such as TWIST1, AURKA, BUB1, and NEK2 in MPNSTs compared to PN, which appears to be important for MPNST growth and proliferation [50]. The comparison of MPNSTs with ANNUBP by Mitchell et al. identified CCMB1, WNT3A, GAS1, and PROM1 as significantly upregulated genes [30]. It is plausible that these could be key genes during the transformation of ANNUBP to MPNST. Wu et al. noted the upregulation of epithelial–mesenchymal transition (EMT) and EZH2 pathways in MPNSTs and upregulation of P53 and interferon signaling in benign neurofibromas (NF) [48]. However, few recurrent DEGs or pathways are identified among these studies, highlighting the heterogeneity throughout MPNST development.
Biomarkers related to Schwann cell development have been identified as potential predictive and diagnostic tools for MPNST. Recently, Holand et al. found CDH19, ERBB3, S100B and other Schwann cell differentiation markers were downregulated in MPNSTs compared to neurofibroma, indicating dysregulation of Schwann cell development in MPNSTs [29]. Vasudevan et al. added to this by showing that compared to neurofibroma cells, Schwann cell differentiation markers S100B and SOX10 were downregulated in PRC2 mutant MPNST cells, suggesting that PRC2 loss is related to the transformation of Schwann cells into MPNSTs [51]. The above sequencing data have shown distinct gene expression signatures between other PNST and MPNSTs and identified a few gene candidates for MPNSTs. However, there is still a lack of RNA-seq-based predictors to clinically assess the risk of patients transforming from PNs to MPNST and then formulate personalized treatment plans. In addition, there is also a lack of a consistent set of genes or pathways implicated across all studies, which highlights the inherent complexity and heterogeneity of MPNSTs. Future research needs to focus on identifying common molecular pathways that can be targeted for therapy, as well as developing reliable predictive models based on RNA-seq data in a prospective manner to guide clinical decisions.
Gene expression analysis can also help assess the drug synergy effect and uncover potential mechanisms to target the MPNST. For example, Borcherding et al. treated MPNST cells with TYK2 inhibitor, and then performed RNA-seq which showed the upregulation of the MAPK pathway after treatment. Following this finding, they combined mirdametinib, a MEK inhibitor, with deucravacitinib, a TYK2 inhibitor, and these two drugs synergistically inhibited the MPNST [73]. In another example, Sun et al. found that the BMP2-SMAD1/5/8 pathway was activated in NF1-associated MPNSTs, and inhibition of this pathway reduced tumor cell proliferation and invasion, hinting at a possible therapeutic target [37]. These studies have uncovered novel therapeutic targets for MPNSTs, and guide more effective treatment strategies.
Overall, microarray and RNA-seq have greatly deepened our understanding of the pathogenesis of MPNSTs at the gene expression level. However, traditional bulk RNA-seq extracts mixed RNA from tissues or a group of cells for sequencing to obtain the average gene expression of all cells, which cannot reveal the expression specificity of individual cells, and cannot understand the interaction between tumor cells and other cells in their microenvironment and the role of each cell type. Future studies should be focused on combination with single-cell and spatial transcriptomic approaches to dissect the cellular heterogeneity within the MPNST.

2.2. Single-Cell RNA-Seq

The single-cell RNA-seq reveals the gene structure and gene expression level of individual cells in the MPNST, reflecting the heterogeneity of cells and allowing for analysis of the contribution of individual cells to the MPNST. Several studies have revealed that MPNSTs are composed predominantly of tumor cells, whereas PNs show a higher proportion of immune-rich environments. For example, Vasudevan et al. showed that MPNSTs were mainly enriched in tumor proliferation cell clusters and growth factor-stimulated tumor cell clusters, while PNs were mainly enriched in T cell and endothelial clusters, which are non-tumor cells [51]. Zhang et al. showed a higher proportion of MPNST cells and fewer immune cell clusters in metastatic MPNSTs, compared to primary MPNSTs, which suggests that metastatic MPNSTs may suppress the immune environment more effectively than even the primary tumors, leading to their aggressiveness and ability to evade immune surveillance [54].
Similar trends are observed in mice models. Wu et al. found that the microenvironment in early-tumor stages of MPNST had more antitumor characteristics, with a higher presence of antitumor macrophages, while the microenvironment in advanced tumor stage became more pro-tumorigenic, characterized by a rise in activated fibroblasts and anti-inflammatory macrophages. They also compared the PNs with MPNSTs and determined unique cell clusters including hypoxic malignant Schwann cell precursors, mesenchymal-like neural crest cells, and mesenchymal-like malignant cells in MPNST, while immature Schwann cells as well as mesenchymal-associated macrophages were enriched in PNs [48].
In another study, Suppiah et al. found that MPNSTs with worse prognosis had a larger proportion of neoplastic cells, while in MPNSTs with better prognosis and atypical NF, the proportion of immune cells are significantly higher. They also found that the known marker of Schwann cells including S100B and ERBB3 were decreased in MPNSTs with worse prognosis, while early neural crest cell markers SOX9 and OTX2 were increased in MPNSTs with better prognosis, indicating that the de-differentiation of Schwann cells is related to the progression to MPNSTs [52].
The above analysis showed that unique cell types, including malignant Schwann cell precursors and mesenchymal-like cells, have been identified in MPNSTs, while Schwann cells and macrophages dominate in PNs. These findings highlight the cellular heterogeneity of MPNSTs and their evolving tumor microenvironment as the disease progresses. However, several limitations of single-cell sequencing technologies cannot be ignored: first, the low-expression transcripts are still likely to escape the detection from sequencing. Second, it requires good practice and relatively complex training to obtain a qualified dataset. Third, it is easy to produce artifacts such as multiplets. Finally, it mainly focuses on the expression of different types of cells and ignores the spatial information and physical interaction of different cells in the tumor microenvironment. Future research should strive to improve the sensitivity of the low-abundance transcripts and simplify the pipeline of single-cell RNA-seq.

2.3. Whole Exome Sequencing (WES) and Whole Genome Sequencing (WGS)

WES and WGS have been widely used to uncover MPNST mutation patterns. NF1 germline mutation and the inactivation of this tumor suppressor gene, is known to predispose individuals to various cancers, including MPNSTs. Additional somatic mutations in NF1 can lead to biallelic inactivation, further increasing the malignancy rate of the disease [74]. In 2015, Hirbe et al. sequenced the same MPNST patient with different stages of diseases including PN, MPNST and metastasis MPNST. They found that the proportion of NF1 mutations increased with the progression of the MPNST and that the tumors became much more copy-number aberrant as they progressed to cancer [61].
PRC2 complex mutations, TP53 loss, and CDKN2A loss are other common events in MPNSTs. In 2021, Dehner et al. performed whole exome sequencing in MPNST PDX lines. They found that SUZ12 loss occurred in 62.5% of PDX lines while proportion of TP53 loss was only 12.5% [45]. Similarly, Lee et al. noticed that the proportion of PRC2 components EED/SUZ12 loss in sporadic MPNST, NF1-MPNST, and radiation-induced MPNST was 92%, 70%, and 90%, respectively. In addition, they found that somatic mutation of CDKN2A co-occurred with PRC2 loss, accounting for 81% of all MPNSTs [19]. Cortes-Ciriano et al. also showed that histone H3 on lysine 27 (H3K27me3) loss occurred in 55% of MPNSTs and biallelic inactivation of SUZ12 and EED occurred in 28% and 17% of MPNSTs, respectively. They also found that CDKN2A biallelic inactivation occurred in 63% of NF1-related MPNST and 55% of sporadic MPNST [72]. In Lorenz’s study, CDKN2A loss existed in 100% of MPNSTs (n = 15) and 100% of MPNST cell lines (n = 8) [75]. These recurrent gene mutation events also suggest that these genes are of great significance in the development of MPNSTs.
Chromosome aberrations have been found in MPNSTs, especially the chromosome 8 gain. Szymanski et al. showed extensive chromosome aberrations, including gain of 1q, 7p, 8q, 9q, and 17q and chromosome loss in 6p and 9p in MPNST [76]. Similarly, Suppiah et al. also showed that gain of SMO, 1q, 8q,13q, 17p and 18q occurred in MPNSTs with poor prognosis [52]. Dehner et al. also found that chromosome 8 gain was exclusively occurring in 87.5% of MPNSTs, while no chromosome 8 gain was observed in PN samples, indicating chromosome 8 gain may be an important genomic event in MPNST development [45].
Together, the above sequencing studies displayed multiple events that occur during the transformation from PNs to MPNSTs, including loss of NF1, CDKN2A, PRC2 components and TP53, which are considered as common genetic mutation events in MPNSTs. Many other gene structural variations are identified such as chromosome gain of 8q, 1q, and 19q. However, it remains unclear at which stage these structural variations occur. Additionally, to understand polyclone evolution during the MPNST transformation, it would require more spatial transcriptome and genome sequencing evidence.

2.4. Epigenetics Sequencing

Epigenetic alterations, including DNA methylation, histone modifications, and non-coding RNA regulation, play a critical role in the pathogenesis of MPNSTs, as well.
Few recurrent gene methylations are reported in MPNSTs. CDKN2A, however, has been identified as one of the few recurrent hypermethylated genes in MPNSTs. In 2011, Feber et al. found hypermethylation of CDKN2A in MPNSTs, which indicated that the repression of the CDKN2A might be due to the epigenetic dysregulation in some cases [64]. In addition, Renner et al. found that CpG sites of CDKN2A could reflect the different types of soft tissue sarcomas [66]. Other distinct DNA methylations in MPNSTs have been also identified. For example, several differentially methylated CpGs located in IL17 were shown to be related to MPNST progression [77]. Danielsen et al. detected hypermethylation of promoter of RASSF1A in 60% of MPNST samples, which is also correlated with poor prognosis of NF1-associated MPNST, indicating RASSF1A might be a prognostic predictor in a subset of MPNSTs [78]. These studies have implications for the epigenetic-based therapies of MPNSTs.
In addition to CDKN2A alterations, mutations in components of the PRC2 complex are common in MPNSTs. PRC2 primarily catalyzes the methylation of H3K27me3 and has been identified as an important modification in MPNSTs. For example, Kochat et al. found that PRC2 loss would reduce the repression of several enhancers, which further promotes MPNST progression [49]. In 2016, Röhrich et al. found that loss or reduction of H3K27me3 was exclusively seen in a subset of MPNSTs based on methylation clustering, which could be used as a biomarker to differentiate the cellular schwannoma and MPNST [79]. Cortes-Ciriano et al. found that global hypomethylation occurred in MPNSTs and hypermethylation of CpG islands and PRC2-component mutations occurred in MPNSTs with poor prognosis compared to MPNSTs with a better prognosis in patients with NF1 [72]. In 2022, Yan et al. performed assay for transposase-accessible chromatin (ATAC-seq) to detect the chromosome accessibility after the knockout of SUZ12. The results showed that the loss of PRC2 due to the knockout of SUZ12 led to decreased chromosome accessibility, which further impaired IFN-gamma response in MPNST cells [47]. However, a previous study showed that was no significant survival difference between MPNSTs with the loss and retaining of PRC2 (n = 100), but they did not stratify patients by NF1 status, which may account for the lack of correlation with survival [60].
Micro-RNA array sequencing has been applied in MPNSTs recently. Amirnasr et al. identified upregulated micro-RNAs miR135b and miR-889 in MPNSTs compared to PN. Subsequent functional inhibition of these two micro-RNAs led to impaired Wnt/β-catenin pathways and inhibited MPNST growth. They also established a micro-RNA expression signature that could differentiate the sporadic and NF1-associated MPNSTs, indicating that micro-RNA could potentially be used as a promising diagnostic tool [67]. Zhang et al. observed an increased expression of miR-30d after the knockdown of EZH2, which further induced the proliferation of MPNST cells and revealed that altered micro-RNA expression could be related to tumor aggressiveness [40]. Overall, these epigenetic studies suggest that PRC2 loss and CDKN2A hypermethylation are common pathways that could serve as prognostic indicators or therapeutic targets for MPNSTs. Future research should leverage a combination of high-resolution methylation profiling, ATAC-seq for chromatin accessibility, and ChIP-seq for histone modifications—to map out the full landscape of epigenetic alterations in MPNSTs.

2.5. Emerging Sequencing Technologies: Proteomics and Metabolomic Sequencing

Recently, proteomics and metabolomics sequencing are beginning to be applied in MPNST. Tsuchiya et al. performed proteomics sequencing for 23 MPNSTs and found that the MET pathway was upregulated in the recurrent or metastatic MPNST group. Further drug screens also identified the fact that one of the MET inhibitors, crizotinib, could effectively inhibit MPNST growth, highlighting the potential to use this technology for therapeutic drug discovery [80]. Jia et al. performed proteomics sequencing of MPNST samples with different prognosis (less than 2 years’ survival vs. more than 5 years’ survival). They observed that expression of decorin, a protein maintaining the stability of extracellular matrix, was significantly lower in the poor-prognosis MPNST group, highlighting the use of this technology to identify prognostic biomarkers [81].
No large metabolomic screens have been performed. However, Lemberg et al. evaluated the effect of an inhibitor of glutamine, JHU395, on MPNST growth. JHU395 could inhibit the MPNST growth in vivo and in vitro with mild toxicity. They further analyzed the metabolomic changes following JHU395 inhibition in mice and identified formylglycinamide ribonucleotide as the most significantly altered metabolite. This metabolite plays a key role in purine synthesis, suggesting that the inhibition of purine synthesis could be the mechanism by which JHU395 impedes MPNST progression [82]. More recently, they evaluated a combination therapy of pro905 and JHU395 in MPNSTs, and detected the metabolites that change after the treatment in a mouse model. Similarly, purine and pyrimidine metabolism were still the top differentially altered pathways [83]. These studies highlight the possibility of identifying combination therapies through metabolomic analyses. Future studies should focus on cell type-specific metabolic and proteomic signatures, and within the MPNST.

3. From Sequencing to Implications: Targeted Therapy

3.1. MEK Inhibitor

MEK pathway activation has been observed in MPNSTs [52,84]. Although several studies showed promising results utilizing MEK inhibitors in benign PN, single-agent MEK inhibition has not been nearly as effective in MPNST models [32,85], likely due to adaptive signaling leading to drug resistance [86]. However, a MEK inhibitor is a potentially promising partner in combination therapies. Borcherding et al. showed the synergistic anti-tumor effect of a TYK2 inhibitor and MEK inhibitor in NF1-associated MPNST in vivo and in vitro [73]. Other promising combinations have included CDK4/6 inhibition with MEK inhibition in MPNST models. Additionally, that combination treatment increased the MPNST response rate to anti-PD-L1 immunotherapy in the mice model [87]. Wang et al. identified SHP2 as a central node in MPNST pathogenesis and the combination of an SHP2 inhibitor and MEK inhibitor looked promising in vivo, in MPNST models [88]. The above studies highlight the importance of combination therapy in MPNST to overcome drug resistance. Several clinical trials are evaluating MEK inhibitor combinations in MPNSTs (NCT05107037, NCT05253131 and NCT03433183) and other trials based on the pre-clinical studies described here are in development.

3.2. Histone Deacetylase (HDAC) Inhibitors

Much evidence from RNA-seq and DNA methylation sequencing has shown that PRC2 loss is a key molecular mutation event during the MPNST transformation [19,49]. PRC2 is crucial for the methylation of H3k27me3, which is the rationale for attempting to treat MPNSTs using HDAC inhibitors. Several HDAC inhibitors have been evaluated, showing promising results in vitro and in vivo in MPNST models [89,90,91]. To date, there are several HDAC inhibitors that have been approved to treat other cancers, including romidepsin, vorinostat, belinostat and panobinostat, which are currently used for lymphoma and multiple myeloma [92]. However, there is only one completed phase II clinical trial testing the effect of HDAC inhibitor romidepsin on various sarcomas (NCT00112463), and no studies to date have focused solely on MPNSTs. These findings underscore the need for further research and clinical trials to explore the potential of HDAC inhibitors in the treatment of MPNSTs, potentially in combination with other therapies.

3.3. Other Emerging Targeted Therapies

Several DNMT inhibitors have been evaluated in MPNSTs. Patel et al. found that a DNMT1 inhibitor selectively inhibited PRC2-loss and MPNST growth and activated transcription of a portion of PRC2 target genes [56]. Similarly, decitabine, a DNA methyltransferase inhibitor, has been approved to treat myelodysplastic syndrome [93] and an ongoing phase II clinical trial is testing the effect of ASTX727, a combination of decitabine and cedazurine, in MPNSTs (NCT04872543). These studies underscore the therapeutic potential of DNMT inhibitors in MPNST, particularly in targeting MPNSTs with PRC2 loss.
The mTOR signaling pathway has also been found to be activated in MPNSTs, and increased expression of p-mTOR is related to the poor prognosis of MPNSTs [94]. Preclinical studies have implicated mTOR signaling in MPNSTs. Johasson et al. found that the single mTOR inhibitor everolimus could inhibit the sporadic and NF1-associated MPNST growth in vivo [95]. They also showed that a combination of everolimus and erlotinib, an EGFR inhibitor, could achieve an additive effect on inhibition of tumor growth. However, clinical trial results with mTOR inhibitors have not shown benefit [96,97,98]. This may be due to poor selection of partners, specificity of mTOR inhibitors, or perhaps the fact that these combinations were studied in a single preclinical model which may not be representative of MPNSTs.
In 2013, Peacock et al. found up-regulated DNA repair mechanisms in MPNSTs, which indicates that DNA repair inhibitors might be a potential treatment for MPNSTs. Poly (ADP-ribose) polymerase (PARP) is a protein involved in DNA repair, and has been studied in MPNSTs recently. Several PARP inhibitors (e.g., olaparib, rucaparib, niraparib and talazoparib) have been approved by FDA and applied in many cancers. Larsson et al. tested a combination of trabectidin and olaparib in MPNSTs and found that this combination therapy reduced tumor growth in vivo [99]. Kivlin et al. also tested olaparib in MPNST and mouse models and found that olaparib could help decrease tumor growth and metastasis [100]. Unfortunately, no clinical trials report the effects of PARP inhibition on MPNST, and we look forward to further investigation of this target in MPNST patients.

3.4. Implications for Immunotherapy

Although there is currently no approved immunotherapy regimen for MPNSTs, several preclinical studies have shown that this could be a promising treatment strategy. Holand et al. identified two subtypes of MPNSTs based on transcriptome data, named immune deficient and immune active. They found that MPNSTs from an immune-deficient group had a poor prognosis and upregulated LGR5, IGF2BP1, PROM1, which could be used as potential druggable targets [29].
PRC2 status is also correlated with immune infiltration in MPNSTs. Yan et al. compared gene expression of MPNSTs with PRC2 retaining or loss [47]. They found that adaptive immune response pathway and T-cell receptor pathway are enriched in the PRC2-retained group while spinal cord development and other differentiation pathways are enriched in the PRC2-loss group. Further, they found that immunogenic virus injection into the PRC2-loss group could enhance the tumor immune infiltration and sensitize the tumor to immune checkpoint therapy. In addition, Cortes-Ciriano et al. identified two transcriptomic subtypes of MPNSTs, which were correlated with the H3Kme27 status, and the subtype with H3K27me3 loss had decreased immune infiltration and immune checkpoint expression inferred from RNAseq data [72].
The alteration of the immune microenvironment is being explored as a therapeutic strategy. Patwardhan et al. depleted macrophages in an MPNST xenograft model using pexidartinib, which led to reduced tumor growth [101]. In addition, Somatilaka et al. were able to cause a shift in MPNSTs from immune cold- to immune hot-tumor, using stimulator of IFN genes (STING) signaling, which sensitized the MPNST to the PD-1/PD-L1 blockade treatment [102]. Future studies could be aimed at therapeutic strategies to shift this immune cold state to an immune-rich environment, to harness immunotherapies.

4. Conclusions and Implications for Clinical Practice

As sequencing technologies have advanced, there has been an explosion in the volume and variety of data from MPNST, derived at multiple biological levels. These data have led to the identification of MPNST driver genes including NF1, CDKN2A, PRC2 components and TP53, as well as numerous potential targeted therapies including MEK inhibition combinations, HDAC inhibitors, SHP2 inhibitors, TYK2 inhibitors, and CDK4/6 inhibitors, etc. Additionally, these data have begun to uncover the potential of targeting the immune system, as well.
Given the relatively low cost of clinical sequencing platforms and potential information that can be gained to guide clinical practice, obtaining molecular information on MPNST patients is potentially beneficial. For example, MTAP loss, which occurs with CDKN2A loss in at least 25% of MPNSTs, confers sensitivity to PRMT5 inhibitors which are currently in clinical trials [103]. Having this information would allow for patients to be enrolled in such a clinical trial. Additionally, MDM2 inhibitors are being utilized in clinical trials for MPNSTs. However, 25% of MPNSTs have TP53 mutations, and as such, an MDM2 inhibitor would not be beneficial in this patient population [104]. Thus, molecular data can again guide decisions for clinical trial enrollment.
Despite these advancements, significant challenges remain. First, we are only starting to understand molecular subtypes of MPNSTs. For example, 25% of MPNSTs have TP53 mutations, and approximately 65% have mutations in the PRC2 complex. However, for at least 10% of MPNSTs, the driver is unclear. Additionally, we have not had validated studies to understand whether or not there are clear transcriptomic subtypes that should guide therapy. Second, once a therapy begins, drug resistance driven by adaptive changes is bound to occur, which will certainly impact therapeutic decisions. In the coming years, development of larger cohorts in clinical trials and the integration of multi-omics data, combined with targeted therapies to overcome tumor resistance and personalized treatments, based on the molecular characteristics of MPNST, will shape the therapeutic trends of the future.

Author Contributions

Conceptualization, K.X. and A.C.H.; supervision, A.C.H.; writing—original draft, K.X., K.Y. and A.C.H.; writing—review and editing, K.X., K.Y. and A.C.H. All authors have read and agreed to the published version of the manuscript.

Funding

K.X. is funded by R01NS134815-02 awarded to ACH through NINDS. K.Y. is partially funded by a grant from the St. Louis Men’s Group Against Cancer.

Acknowledgments

The authors thank Yang Lyu and Diana Akinyi Odhiambo for their suggestions and help with the review paper.

Conflicts of Interest

A.C.H. has the following COI: Serving on the Advisory Board of Aadi Subsidiary, Inc., engaging in licensing activities with Boehringer Ingelheim GmbH, providing consulting services to Springworks Therapeutic Operating Company, PBC, receiving an honorarium from the American Physician Institute for Advanced Professional Studies, and receiving research funding for Tango. The other authors declare no conflicts of interest.

References

  1. Cuneo, H.M.; Rand, C.W. Tumors of the Gasserian Ganglion: Tumor of the Left Gasserian Ganglion Associated with Enlargement of the Mandibular Nerve a Review of the Literature and Case Report. J. Neurosurg. 1952, 9, 423–431. [Google Scholar] [CrossRef] [PubMed]
  2. Ducatman, B.; Scheithauer, B.; Piepgras, D.; Reiman, H. Malignant Peripheral Nerve Sheath Tumors in Childhood. J. Neuro-Oncology 1984, 2, 241–248. [Google Scholar] [CrossRef] [PubMed]
  3. Ducatman, B.S.; Scheithauer, B.W.; Piepgras, D.G.; Reiman, H.M.; Ilstrup, D.M. Malignant Peripheral Nerve Sheath Tumors. A Clinicopathologic Study of 120 Cases. Cancer 1986, 57, 2006–2021. [Google Scholar] [CrossRef]
  4. Anghileri, M.; Miceli, R.; Fiore, M.; Mariani, L.; Ferrari, A.; Mussi, C.; Lozza, L.; Collini, P.; Olmi, P.; Casali, P.G.; et al. Malignant Peripheral Nerve Sheath Tumors: Prognostic Factors and Survival in a Series of Patients Treated at a Single Institution. Cancer 2006, 107, 1065–1074. [Google Scholar] [CrossRef] [PubMed]
  5. Lucas, C.-H.G.; Gross, A.M.; Romo, C.G.; Dehner, C.A.; Lazar, A.J.; Miettinen, M.; Pekmezci, M.; Quezado, M.; Rodriguez, F.J.; Stemmer-Rachamimov, A.; et al. Consensus Recommendations for an Integrated Diagnostic Approach to Peripheral Nerve Sheath Tumors Arising in the Setting of Neurofibromatosis Type 1 (NF1). Neuro-Oncology 2024, noae235. [Google Scholar] [CrossRef] [PubMed]
  6. Hirbe, A.C.; Dehner, C.A.; Dombi, E.; Eulo, V.; Gross, A.M.; Sundby, T.; Lazar, A.J.; Widemann, B.C. Contemporary Approach to Neurofibromatosis Type 1–Associated Malignant Peripheral Nerve Sheath Tumors. Am. Soc. Clin. Oncol. Educ. Book. 2024, 44, e432242. [Google Scholar] [CrossRef]
  7. Siegel, R.L.; Giaquinto, A.N.; Jemal, A. Cancer Statistics, 2024. CA Cancer J Clin. 2024, 74, 12–49. [Google Scholar] [CrossRef]
  8. Clark, M.A.; Fisher, C.; Judson, I.; Thomas, J.M. Soft-Tissue Sarcomas in Adults. N. Engl. J. Med. 2005, 353, 701–711. [Google Scholar] [CrossRef]
  9. Dunn, G.P.; Spiliopoulos, K.; Plotkin, S.R.; Hornicek, F.J.; Harmon, D.C.; Delaney, T.F.; Williams, Z. Role of Resection of Malignant Peripheral Nerve Sheath Tumors in Patients with Neurofibromatosis Type 1: Clinical Article. J. Neurosurg. 2013, 118, 142–148. [Google Scholar] [CrossRef]
  10. Lucas, C.-H.G.; Vasudevan, H.N.; Chen, W.C.; Magill, S.T.; Braunstein, S.E.; Jacques, L.; Dahiya, S.; Rodriguez, F.J.; Horvai, A.E.; Perry, A.; et al. Histopathologic Findings in Malignant Peripheral Nerve Sheath Tumor Predict Response to Radiotherapy and Overall Survival. Neuro-Oncol. Adv. 2020, 2, vdaa131. [Google Scholar] [CrossRef] [PubMed]
  11. Sloan, L.; Terezakis, S.A.; Blakeley, J.O.; Slobogean, B.; Kleinberg, L.R. Long-Term Outcomes of Radiation Therapy (RT) in the Management of Malignant Peripheral Nerve Sheath Tumors (MPNST) in Patients with Neurofibromatosis Type 1 (NF1). Int. J. Radiat. Oncol. Biol. Phys. 2018, 102, e474–e475. [Google Scholar] [CrossRef]
  12. Van Noesel, M.M.; Orbach, D.; Brennan, B.; Kelsey, A.; Zanetti, I.; De Salvo, G.L.; Gaze, M.N.; Craigie, R.J.; McHugh, K.; Francotte, N.; et al. Outcome and Prognostic Factors in Pediatric Malignant Peripheral Nerve Sheath Tumors: An Analysis of the European Pediatric Soft Tissue Sarcoma Group (EpSSG) NRSTS-2005 Prospective Study. Pediatr. Blood Cancer 2019, 66, e27833. [Google Scholar] [CrossRef]
  13. Hruban, R.H.; Shiu, M.H.; Senie, R.T.; Woodruff, J.M. Malignant Peripheral Nerve Sheath Tumors of the Buttock and Lower Extremity. A Study of 43 Cases. Cancer 1990, 66, 1253–1265. [Google Scholar] [CrossRef] [PubMed]
  14. Sharma, M.; Puj, K.; Salunke, A.; Pandya, S.; Gandhi, J.; Parikh, A. Malignant Peripheral Nerve Sheath Tumor with Analysis of Various Prognostic Factors: A Single-Institutional Experience. J. Can. Res. Ther. 2021, 17, 106. [Google Scholar] [CrossRef] [PubMed]
  15. Landry, J.P.; Schertz, K.L.; Chiang, Y.-J.; Bhalla, A.D.; Yi, M.; Keung, E.Z.; Scally, C.P.; Feig, B.W.; Hunt, K.K.; Roland, C.L.; et al. Comparison of Cancer Prevalence in Patients With Neurofibromatosis Type 1 at an Academic Cancer Center vs in the General Population From 1985 to 2020. JAMA Netw. Open 2021, 4, e210945. [Google Scholar] [CrossRef] [PubMed]
  16. Stucky, C.-C.H.; Johnson, K.N.; Gray, R.J.; Pockaj, B.A.; Ocal, I.T.; Rose, P.S.; Wasif, N. Malignant Peripheral Nerve Sheath Tumors (MPNST): The Mayo Clinic Experience. Ann. Surg. Oncol. 2012, 19, 878–885. [Google Scholar] [CrossRef] [PubMed]
  17. Pemov, A.; Li, H.; Presley, W.; Wallace, M.R.; Miller, D.T. Genetics of Human Malignant Peripheral Nerve Sheath Tumors. Neuro-Oncol. Adv. 2020, 2, i50–i61. [Google Scholar] [CrossRef]
  18. Spurlock, G.; Knight, S.J.L.; Thomas, N.; Kiehl, T.-R.; Guha, A.; Upadhyaya, M. Molecular Evolution of a Neurofibroma to Malignant Peripheral Nerve Sheath Tumor (MPNST) in an NF1 Patient: Correlation between Histopathological, Clinical and Molecular Findings. J. Cancer Res. Clin. Oncol. 2010, 136, 1869–1880. [Google Scholar] [CrossRef]
  19. Lee, W.; Teckie, S.; Wiesner, T.; Ran, L.; Prieto Granada, C.N.; Lin, M.; Zhu, S.; Cao, Z.; Liang, Y.; Sboner, A.; et al. PRC2 Is Recurrently Inactivated through EED or SUZ12 Loss in Malignant Peripheral Nerve Sheath Tumors. Nat. Genet. 2014, 46, 1227–1232. [Google Scholar] [CrossRef]
  20. Yao, C.; Zhou, H.; Dong, Y.; Alhaskawi, A.; Hasan Abdullah Ezzi, S.; Wang, Z.; Lai, J.; Goutham Kota, V.; Hasan Abdulla Hasan Abdulla, M.; Lu, H. Malignant Peripheral Nerve Sheath Tumors: Latest Concepts in Disease Pathogenesis and Clinical Management. Cancers 2023, 15, 1077. [Google Scholar] [CrossRef] [PubMed]
  21. Packer, R.J.; Gutmann, D.H.; Rubenstein, A.; Viskochil, D.; Zimmerman, R.A.; Vezina, G.; Small, J.; Korf, B. Plexiform Neurofibromas in NF1: Toward Biologic-Based Therapy. Neurology 2002, 58, 1461–1470. [Google Scholar] [CrossRef]
  22. Miettinen, M.M.; Antonescu, C.R.; Fletcher, C.D.M.; Kim, A.; Lazar, A.J.; Quezado, M.M.; Reilly, K.M.; Stemmer-Rachamimov, A.; Stewart, D.R.; Viskochil, D.; et al. Histopathologic Evaluation of Atypical Neurofibromatous Tumors and Their Transformation into Malignant Peripheral Nerve Sheath Tumor in Patients with Neurofibromatosis 1—A Consensus Overview. Hum. Pathol. 2017, 67, 1–10. [Google Scholar] [CrossRef] [PubMed]
  23. Rhodes, S.D.; He, Y.; Smith, A.; Jiang, L.; Lu, Q.; Mund, J.; Li, X.; Bessler, W.; Qian, S.; Dyer, W.; et al. Cdkn2a (Arf) Loss Drives NF1-Associated Atypical Neurofibroma and Malignant Transformation. Hum. Mol. Genet. 2019, 28, 2752–2762. [Google Scholar] [CrossRef] [PubMed]
  24. Brockman, Q.R.; Scherer, A.; McGivney, G.R.; Gutierrez, W.R.; Voigt, A.P.; Isaacson, A.L.; Laverty, E.A.; Roughton, G.; Knepper-Adrian, V.; Darbro, B.; et al. PRC2 Loss Drives MPNST Metastasis and Matrix Remodeling. JCI Insight 2022, 7, e157502. [Google Scholar] [CrossRef]
  25. Kohlmeyer, J.L.; Kaemmer, C.A.; Lingo, J.J.; Voigt, E.; Leidinger, M.R.; McGivney, G.R.; Scherer, A.; Koppenhafer, S.L.; Gordon, D.J.; Breheny, P.; et al. Oncogenic RABL6A Promotes NF1-Associated MPNST Progression in Vivo. Neuro-Oncol. Adv. 2022, 4, vdac047. [Google Scholar] [CrossRef]
  26. Rahrmann, E.P.; Watson, A.L.; Keng, V.W.; Choi, K.; Moriarity, B.S.; Beckmann, D.A.; Wolf, N.K.; Sarver, A.; Collins, M.H.; Moertel, C.L.; et al. Forward Genetic Screen for Malignant Peripheral Nerve Sheath Tumor Formation Identifies New Genes and Pathways Driving Tumorigenesis. Nat. Genet. 2013, 45, 756–766. [Google Scholar] [CrossRef] [PubMed]
  27. Heller, M.J. DNA Microarray Technology: Devices, Systems, and Applications. Annu. Rev. Biomed. Eng. 2002, 4, 129–153. [Google Scholar] [CrossRef]
  28. Reilly, C.; Raghavan, A.; Bohjanen, P. Global Assessment of Cross-Hybridization for Oligonucleotide Arrays. J. Biomol. Tech. 2006, 17, 163–172. [Google Scholar]
  29. Høland, M.; Berg, K.C.G.; Eilertsen, I.A.; Bjerkehagen, B.; Kolberg, M.; Boye, K.; Lingjærde, O.C.; Guren, T.K.; Mandahl, N.; Van Den Berg, E.; et al. Transcriptomic Subtyping of Malignant Peripheral Nerve Sheath Tumours Highlights Immune Signatures, Genomic Profiles, Patient Survival and Therapeutic Targets. eBioMedicine 2023, 97, 104829. [Google Scholar] [CrossRef] [PubMed]
  30. Mitchell, D.K.; Burgess, B.; White, E.E.; Smith, A.E.; Sierra Potchanant, E.A.; Mang, H.; Hickey, B.E.; Lu, Q.; Qian, S.; Bessler, W.; et al. Spatial Gene-Expression Profiling Unveils Immuno-Oncogenic Programs of NF1-Associated Peripheral Nerve Sheath Tumor Progression. Clin. Cancer Res. 2024, OF1–OF16. [Google Scholar] [CrossRef] [PubMed]
  31. Chudasama, P.; Renner, M.; Straub, M.; Mughal, S.S.; Hutter, B.; Kosaloglu, Z.; Schweßinger, R.; Scheffler, M.; Alldinger, I.; Schimmack, S.; et al. Targeting Fibroblast Growth Factor Receptor 1 for Treatment of Soft-Tissue Sarcoma. Clin. Cancer Res. 2017, 23, 962–973. [Google Scholar] [CrossRef]
  32. Jessen, W.J.; Miller, S.J.; Jousma, E.; Wu, J.; Rizvi, T.A.; Brundage, M.E.; Eaves, D.; Widemann, B.; Kim, M.-O.; Dombi, E.; et al. MEK Inhibition Exhibits Efficacy in Human and Mouse Neurofibromatosis Tumors. J. Clin. Investig. 2013, 123, 340–347. [Google Scholar] [CrossRef] [PubMed]
  33. Lafferty-Whyte, K.; Bilsland, A.; Hoare, S.F.; Burns, S.; Zaffaroni, N.; Cairney, C.J.; Keith, W.N. TCEAL7 Inhibition of C-Myc Activity in Alternative Lengthening of Telomeres Regulates hTERT Expression. Neoplasia 2010, 12, 405–414. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, X.; Bledsoe, K.L.; Graham, R.P.; Asmann, Y.W.; Viswanatha, D.S.; Lewis, J.E.; Lewis, J.T.; Chou, M.M.; Yaszemski, M.J.; Jen, J.; et al. Recurrent PAX3-MAML3 Fusion in Biphenotypic Sinonasal Sarcoma. Nat. Genet. 2014, 46, 666–668. [Google Scholar] [CrossRef]
  35. Kolberg, M.; Høland, M.; Lind, G.E.; Ågesen, T.H.; Skotheim, R.I.; Sundby Hall, K.; Mandahl, N.; Smeland, S.; Mertens, F.; Davidson, B.; et al. Protein Expression of BIRC5, TK1, and TOP2A in Malignant Peripheral Nerve Sheath Tumours—A Prognostic Test after Surgical Resection. Mol. Oncol. 2015, 9, 1129–1139. [Google Scholar] [CrossRef] [PubMed]
  36. Komura, S.; Ito, K.; Ohta, S.; Ukai, T.; Kabata, M.; Itakura, F.; Semi, K.; Matsuda, Y.; Hashimoto, K.; Shibata, H.; et al. Cell-Type Dependent Enhancer Binding of the EWS/ATF1 Fusion Gene in Clear Cell Sarcomas. Nat. Commun. 2019, 10, 3999. [Google Scholar] [CrossRef]
  37. Sun, D.; Haddad, R.; Kraniak, J.M.; Horne, S.D.; Tainsky, M.A. RAS/MEK–Independent Gene Expression Reveals BMP2-Related Malignant Phenotypes in the Nf1 -Deficient MPNST. Mol. Cancer Res. 2013, 11, 616–627. [Google Scholar] [CrossRef]
  38. Fountzilas, E.; Kelly, A.D.; Perez-Atayde, A.R.; Goldsmith, J.; Konstantinopoulos, P.A.; Francoeur, N.; Correll, M.; Rubio, R.; Hu, L.; Gebhardt, M.C.; et al. A microRNA Activity Map of Human Mesenchymal Tumors: Connections to Oncogenic Pathways; an Integrative Transcriptomic Study. BMC Genom. 2012, 13, 332. [Google Scholar] [CrossRef]
  39. Mahller, Y.Y.; Sakthivel, B.; Baird, W.H.; Aronow, B.J.; Hsu, Y.-H.; Cripe, T.P.; Mehrian-Shai, R. Molecular Analysis of Human Cancer Cells Infected by an Oncolytic HSV-1 Reveals Multiple Upregulated Cellular Genes and a Role for SOCS1 in Virus Replication. Cancer Gene Ther. 2008, 15, 733–741. [Google Scholar] [CrossRef] [PubMed]
  40. Zhang, P.; Garnett, J.; Creighton, C.J.; Al Sannaa, G.A.; Igram, D.R.; Lazar, A.; Liu, X.; Liu, C.; Pollock, R.E. EZH2-miR-30d-KPNB1 Pathway Regulates Malignant Peripheral Nerve Sheath Tumour Cell Survival and Tumourigenesis. J. Pathol. 2014, 232, 308–318. [Google Scholar] [CrossRef]
  41. De Raedt, T.; Beert, E.; Pasmant, E.; Luscan, A.; Brems, H.; Ortonne, N.; Helin, K.; Hornick, J.L.; Mautner, V.; Kehrer-Sawatzki, H.; et al. PRC2 Loss Amplifies Ras-Driven Transcription and Confers Sensitivity to BRD4-Based Therapies. Nature 2014, 514, 247–251. [Google Scholar] [CrossRef]
  42. Malone, C.F.; Emerson, C.; Ingraham, R.; Barbosa, W.; Guerra, S.; Yoon, H.; Liu, L.L.; Michor, F.; Haigis, M.; Macleod, K.F.; et al. mTOR and HDAC Inhibitors Converge on the TXNIP/Thioredoxin Pathway to Cause Catastrophic Oxidative Stress and Regression of RAS-Driven Tumors. Cancer Discov. 2017, 7, 1450–1463. [Google Scholar] [CrossRef] [PubMed]
  43. Castellsagué, J.; Gel, B.; Fernández-Rodríguez, J.; Llatjós, R.; Blanco, I.; Benavente, Y.; Pérez-Sidelnikova, D.; García-Del Muro, J.; Viñals, J.M.; Vidal, A.; et al. Comprehensive Establishment and Characterization of Orthoxenograft Mouse Models of Malignant Peripheral Nerve Sheath Tumors for Personalized Medicine. EMBO Mol. Med. 2015, 7, 608–627. [Google Scholar] [CrossRef] [PubMed]
  44. Malone, C.F.; Fromm, J.A.; Maertens, O.; DeRaedt, T.; Ingraham, R.; Cichowski, K. Defining Key Signaling Nodes and Therapeutic Biomarkers in NF1-Mutant Cancers. Cancer Discov. 2014, 4, 1062–1073. [Google Scholar] [CrossRef] [PubMed]
  45. Dehner, C.; Moon, C.I.; Zhang, X.; Zhou, Z.; Miller, C.; Xu, H.; Wan, X.; Yang, K.; Mashl, J.; Gosline, S.J.C.; et al. Chromosome 8 Gain Is Associated with High-Grade Transformation in MPNST. JCI Insight 2021, 6, e146351. [Google Scholar] [CrossRef]
  46. Miller, D.T.; Cortés-Ciriano, I.; Pillay, N.; Hirbe, A.C.; Snuderl, M.; Bui, M.M.; Piculell, K.; Al-Ibraheemi, A.; Dickson, B.C.; Hart, J.; et al. Genomics of MPNST (GeM) Consortium: Rationale and Study Design for Multi-Omic Characterization of NF1-Associated and Sporadic MPNSTs. Genes 2020, 11, 387. [Google Scholar] [CrossRef]
  47. Yan, J.; Chen, Y.; Patel, A.J.; Warda, S.; Lee, C.J.; Nixon, B.G.; Wong, E.W.P.; Miranda-Román, M.A.; Yang, N.; Wang, Y.; et al. Tumor-Intrinsic PRC2 Inactivation Drives a Context-Dependent Immune-Desert Microenvironment and Is Sensitized by Immunogenic Viruses. J. Clin. Investig. 2022, 132, e153437. [Google Scholar] [CrossRef]
  48. Wu, L.M.N.; Zhang, F.; Rao, R.; Adam, M.; Pollard, K.; Szabo, S.; Liu, X.; Belcher, K.A.; Luo, Z.; Ogurek, S.; et al. Single-Cell Multiomics Identifies Clinically Relevant Mesenchymal Stem-like Cells and Key Regulators for MPNST Malignancy. Sci. Adv. 2022, 8, eabo5442. [Google Scholar] [CrossRef] [PubMed]
  49. Kochat, V.; Raman, A.T.; Landers, S.M.; Tang, M.; Schulz, J.; Terranova, C.; Landry, J.P.; Bhalla, A.D.; Beird, H.C.; Wu, C.-C.; et al. Enhancer Reprogramming in PRC2-Deficient Malignant Peripheral Nerve Sheath Tumors Induces a Targetable de-Differentiated State. Acta Neuropathol. 2021, 142, 565–590. [Google Scholar] [CrossRef]
  50. Kohlmeyer, J.L.; Kaemmer, C.A.; Pulliam, C.; Maharjan, C.K.; Samayoa, A.M.; Major, H.J.; Cornick, K.E.; Knepper-Adrian, V.; Khanna, R.; Sieren, J.C.; et al. RABL6A Is an Essential Driver of MPNSTs That Negatively Regulates the RB1 Pathway and Sensitizes Tumor Cells to CDK4/6 Inhibitors. Clin. Cancer Res. 2020, 26, 2997–3011. [Google Scholar] [CrossRef]
  51. Vasudevan, H.N.; Payne, E.; Delley, C.L.; John Liu, S.; Mirchia, K.; Sale, M.J.; Lastella, S.; Nunez, M.S.; Lucas, C.-H.G.; Eaton, C.D.; et al. Functional Interactions between Neurofibromatosis Tumor Suppressors Underlie Schwann Cell Tumor De-Differentiation and Treatment Resistance. Nat. Commun. 2024, 15, 477. [Google Scholar] [CrossRef] [PubMed]
  52. Suppiah, S.; Mansouri, S.; Mamatjan, Y.; Liu, J.C.; Bhunia, M.M.; Patil, V.; Rath, P.; Mehani, B.; Heir, P.; Bunda, S.; et al. Multiplatform Molecular Profiling Uncovers Two Subgroups of Malignant Peripheral Nerve Sheath Tumors with Distinct Therapeutic Vulnerabilities. Nat. Commun. 2023, 14, 2696. [Google Scholar] [CrossRef] [PubMed]
  53. Palomo-Irigoyen, M.; Pérez-Andrés, E.; Iruarrizaga-Lejarreta, M.; Barreira-Manrique, A.; Tamayo-Caro, M.; Vila-Vecilla, L.; Moreno-Cugnon, L.; Beitia, N.; Medrano, D.; Fernández-Ramos, D.; et al. HuR/ELAVL1 Drives Malignant Peripheral Nerve Sheath Tumor Growth and Metastasis. J. Clin. Investig. 2020, 130, 3848–3864. [Google Scholar] [CrossRef] [PubMed]
  54. Zhang, X.; Lou, H.E.; Gopalan, V.; Liu, Z.; Jafarah, H.M.; Lei, H.; Jones, P.; Sayers, C.M.; Yohe, M.E.; Chittiboina, P.; et al. Single-Cell Sequencing Reveals Activation of Core Transcription Factors in PRC2-Deficient Malignant Peripheral Nerve Sheath Tumor. Cell Rep. 2022, 40, 111363. [Google Scholar] [CrossRef] [PubMed]
  55. Zhang, X.; Hu, C.; Sun, S.; Guo, C.; Bu, Y.; Wang, Z.; Liu, Z.; Zhang, X.; Li, D.; Liu, S. TSPO Deficiency Promotes the Progression of Malignant Peripheral Sheath Tumors by Regulating the G2/M Phase of the Cell Cycle via CDK1. Sci. Rep. 2024, 14, 26235. [Google Scholar] [CrossRef]
  56. Patel, A.J.; Warda, S.; Maag, J.L.V.; Misra, R.; Miranda-Román, M.A.; Pachai, M.R.; Lee, C.J.; Li, D.; Wang, N.; Bayshtok, G.; et al. PRC2-Inactivating Mutations in Cancer Enhance Cytotoxic Response to DNMT1-Targeted Therapy via Enhanced Viral Mimicry. Cancer Discov. 2022, 12, 2120–2139. [Google Scholar] [CrossRef] [PubMed]
  57. Wassef, M.; Luscan, A.; Aflaki, S.; Zielinski, D.; Jansen, P.W.T.C.; Baymaz, H.I.; Battistella, A.; Kersouani, C.; Servant, N.; Wallace, M.R.; et al. EZH1/2 Function Mostly within Canonical PRC2 and Exhibit Proliferation-Dependent Redundancy That Shapes Mutational Signatures in Cancer. Proc. Natl. Acad. Sci. USA 2019, 116, 6075–6080. [Google Scholar] [CrossRef]
  58. Chung, M.-H.; Aimaier, R.; Yu, Q.; Li, H.; Li, Y.; Wei, C.; Gu, Y.; Wang, W.; Guo, Z.; Long, M.; et al. RRM2 as a Novel Prognostic and Therapeutic Target of NF1-Associated MPNST. Cell Oncol. 2023, 46, 1399–1413. [Google Scholar] [CrossRef]
  59. Szymanski, J.J.; Sundby, R.T.; Jones, P.A.; Srihari, D.; Earland, N.; Harris, P.K.; Feng, W.; Qaium, F.; Lei, H.; Roberts, D.; et al. Cell-Free DNA Ultra-Low-Pass Whole Genome Sequencing to Distinguish Malignant Peripheral Nerve Sheath Tumor (MPNST) from Its Benign Precursor Lesion: A Cross-Sectional Study. PLoS Med. 2021, 18, e1003734. [Google Scholar] [CrossRef]
  60. Lyskjær, I.; Lindsay, D.; Tirabosco, R.; Steele, C.D.; Lombard, P.; Strobl, A.; Rocha, A.M.; Davies, C.; Ye, H.; Bekers, E.; et al. H3K27me3 Expression and Methylation Status in Histological Variants of Malignant Peripheral Nerve Sheath Tumours. J. Pathol. 2020, 252, 151–164. [Google Scholar] [CrossRef] [PubMed]
  61. Hirbe, A.C.; Dahiya, S.; Miller, C.A.; Li, T.; Fulton, R.S.; Zhang, X.; McDonald, S.; DeSchryver, K.; Duncavage, E.J.; Walrath, J.; et al. Whole Exome Sequencing Reveals the Order of Genetic Changes during Malignant Transformation and Metastasis in a Single Patient with NF1-Plexiform Neurofibroma. Clin. Cancer Res. 2015, 21, 4201–4211. [Google Scholar] [CrossRef] [PubMed]
  62. Godec, A.; Jayasinghe, R.; Chrisinger, J.S.A.; Prudner, B.; Ball, T.; Wang, Y.; Srihari, D.; Kaushal, M.; Dietz, H.; Zhang, X.; et al. Whole Exome Sequencing Reveals the Maintained Polyclonal Nature from Primary to Metastatic Malignant Peripheral Nerve Sheath Tumor in Two Patients with NF1. Neuro-Oncol. Adv. 2020, 2, i75–i84. [Google Scholar] [CrossRef] [PubMed]
  63. Kinoshita, I.; Yamada, Y.; Kohashi, K.; Yamamoto, H.; Iwasaki, T.; Ishihara, S.; Toda, Y.; Ito, Y.; Susuki, Y.; Kawaguchi, K.; et al. Frequent MN1 Gene Mutations in Malignant Peripheral Nerve Sheath Tumor. Anticancer Res. 2020, 40, 6221–6228. [Google Scholar] [CrossRef]
  64. Feber, A.; Wilson, G.A.; Zhang, L.; Presneau, N.; Idowu, B.; Down, T.A.; Rakyan, V.K.; Noon, L.A.; Lloyd, A.C.; Stupka, E.; et al. Comparative Methylome Analysis of Benign and Malignant Peripheral Nerve Sheath Tumors. Genome Res. 2011, 21, 515–524. [Google Scholar] [CrossRef]
  65. Bhunia, M.M.; Stehn, C.M.; Jubenville, T.A.; Novacek, E.L.; Larsson, A.T.; Madala, M.; Suppiah, S.; Velez-Reyes, G.L.; Williams, K.B.; Sokolowski, M.; et al. Multiomic Analyses Reveal New Targets of Polycomb Repressor Complex 2 in Schwann Lineage Cells and Malignant Peripheral Nerve Sheath Tumors. Neuro-Oncol. Adv. 2024, 6, vdae188. [Google Scholar] [CrossRef] [PubMed]
  66. Renner, M.; Czwan, E.; Hartmann, W.; Penzel, R.; Brors, B.; Eils, R.; Wardelmann, E.; Büttner, R.; Lichter, P.; Schirmacher, P.; et al. MicroRNA Profiling of Primary High-grade Soft Tissue Sarcomas. Genes Chromosomes Cancer 2012, 51, 982–996. [Google Scholar] [CrossRef]
  67. Amirnasr, A.; Verdijk, R.M.; Van Kuijk, P.F.; Kartal, P.; Vriends, A.L.M.; French, P.J.; Van Royen, M.E.; Taal, W.; Sleijfer, S.; Wiemer, E.A.C. Deregulated microRNAs in Neurofibromatosis Type 1 Derived Malignant Peripheral Nerve Sheath Tumors. Sci. Rep. 2020, 10, 2927. [Google Scholar] [CrossRef]
  68. Wang, Z.; Gerstein, M.; Snyder, M. RNA-Seq: A Revolutionary Tool for Transcriptomics. Nat. Rev. Genet. 2009, 10, 57–63. [Google Scholar] [CrossRef] [PubMed]
  69. Rai, M.F.; Tycksen, E.D.; Sandell, L.J.; Brophy, R.H. Advantages of RNA-seq Compared to RNA Microarrays for Transcriptome Profiling of Anterior Cruciate Ligament Tears. J. Orthop. Res. 2018, 36, 484–497. [Google Scholar] [CrossRef]
  70. 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]
  71. Bagchi, A.; Dhanda, S.K.; Dunphy, P.; Sioson, E.; Robinson, G.W. Molecular Classification Improves Therapeutic Options for Infants and Young Children With Medulloblastoma. J. Natl. Compr. Cancer Netw. 2023, 21, 1097–1105. [Google Scholar] [CrossRef]
  72. Cortes-Ciriano, I.; Steele, C.D.; Piculell, K.; Al-Ibraheemi, A.; Eulo, V.; Bui, M.M.; Chatzipli, A.; Dickson, B.C.; Borcherding, D.C.; Feber, A.; et al. Genomic Patterns of Malignant Peripheral Nerve Sheath Tumor (MPNST) Evolution Correlate with Clinical Outcome and Are Detectable in Cell-Free DNA. Cancer Discov. 2023, 13, 654–671. [Google Scholar] [CrossRef] [PubMed]
  73. Borcherding, D.C.; Amin, N.V.; He, K.; Zhang, X.; Lyu, Y.; Dehner, C.; Bhatia, H.; Gothra, A.; Daud, L.; Ruminski, P.; et al. MEK Inhibition Synergizes with TYK2 Inhibitors in NF1-Associated Malignant Peripheral Nerve Sheath Tumors. Clin. Cancer Res. 2023, 29, 1592–1604. [Google Scholar] [CrossRef]
  74. Bottillo, I.; Ahlquist, T.; Brekke, H.; Danielsen, S.A.; Van Den Berg, E.; Mertens, F.; Lothe, R.A.; Dallapiccola, B. Germline and Somatic NF1 Mutations in Sporadic and NF1-associated Malignant Peripheral Nerve Sheath Tumours. J. Pathol. 2009, 217, 693–701. [Google Scholar] [CrossRef] [PubMed]
  75. Magallón-Lorenz, M.; Fernández-Rodríguez, J.; Terribas, E.; Creus-Batchiller, E.; Romagosa, C.; Estival, A.; Perez Sidelnikova, D.; Salvador, H.; Villanueva, A.; Blanco, I.; et al. Chromosomal Translocations Inactivating CDKN2A Support a Single Path for Malignant Peripheral Nerve Sheath Tumor Initiation. Hum. Genet. 2021, 140, 1241–1252. [Google Scholar] [CrossRef]
  76. Banerjee, J.; Allaway, R.J.; Taroni, J.N.; Baker, A.; Zhang, X.; Moon, C.I.; Pratilas, C.A.; Blakeley, J.O.; Guinney, J.; Hirbe, A.; et al. Integrative Analysis Identifies Candidate Tumor Microenvironment and Intracellular Signaling Pathways That Define Tumor Heterogeneity in NF1. Genes 2020, 11, 226. [Google Scholar] [CrossRef] [PubMed]
  77. Brockman, Q.R.; Rytlewski, J.D.; Milhem, M.; Monga, V.; Dodd, R.D. Integrated Epigenetic and Transcriptomic Analysis Identifies Interleukin 17 DNA Methylation Signature of Malignant Peripheral Nerve Sheath Tumor Progression and Metastasis. JCO Precis. Oncol. 2024, 8, e2300325. [Google Scholar] [CrossRef] [PubMed]
  78. Danielsen, S.A.; Lind, G.E.; Kolberg, M.; Høland, M.; Bjerkehagen, B.; Sundby Hall, K.; Van Den Berg, E.; Mertens, F.; Smeland, S.; Picci, P.; et al. Methylated RASSF1A in Malignant Peripheral Nerve Sheath Tumors Identifies Neurofibromatosis Type 1 Patients with Inferior Prognosis. Neuro-Oncology 2015, 17, 63–69. [Google Scholar] [CrossRef]
  79. Röhrich, M.; Koelsche, C.; Schrimpf, D.; Capper, D.; Sahm, F.; Kratz, A.; Reuss, J.; Hovestadt, V.; Jones, D.T.W.; Bewerunge-Hudler, M.; et al. Methylation-Based Classification of Benign and Malignant Peripheral Nerve Sheath Tumors. Acta Neuropathol. 2016, 131, 877–887. [Google Scholar] [CrossRef]
  80. Tsuchiya, R.; Yoshimatsu, Y.; Noguchi, R.; Sin, Y.; Ono, T.; Akiyama, T.; Kosako, H.; Yoshida, A.; Ohtori, S.; Kawai, A.; et al. Integrating Analysis of Proteome Profile and Drug Screening Identifies Therapeutic Potential of MET Pathway for the Treatment of Malignant Peripheral Nerve Sheath Tumor. Expert Rev. Proteom. 2023, 20, 109–119. [Google Scholar] [CrossRef]
  81. Jia, X.; Chen, C.; Chen, L.; Yu, C.; Kondo, T. Decorin as a Prognostic Biomarker in Patients with Malignant Peripheral Nerve Sheath Tumors. Oncol. Lett. 2019. [Google Scholar] [CrossRef]
  82. Lemberg, K.M.; Zhao, L.; Wu, Y.; Veeravalli, V.; Alt, J.; Aguilar, J.M.H.; Dash, R.P.; Lam, J.; Tenora, L.; Rodriguez, C.; et al. The Novel Glutamine Antagonist Prodrug JHU395 Has Antitumor Activity in Malignant Peripheral Nerve Sheath Tumor. Mol. Cancer Ther. 2020, 19, 397–408. [Google Scholar] [CrossRef]
  83. Lemberg, K.M.; Ali, E.S.; Krecmerova, M.; Aguilar, J.M.H.; Alt, J.; Peters, D.E.; Zhao, L.; Wu, Y.; Nuha, N.; Asara, J.M.; et al. Pro-905, a Novel Purine Antimetabolite, Combines with Glutamine Amidotransferase Inhibition to Suppress Growth of Malignant Peripheral Nerve Sheath Tumor. Mol. Cancer Ther. 2023, 22, 1390–1403. [Google Scholar] [CrossRef] [PubMed]
  84. Brohl, A.S.; Kahen, E.; Yoder, S.J.; Teer, J.K.; Reed, D.R. The Genomic Landscape of Malignant Peripheral Nerve Sheath Tumors: Diverse Drivers of Ras Pathway Activation. Sci. Rep. 2017, 7, 14992. [Google Scholar] [CrossRef] [PubMed]
  85. Gu, Y.; Wang, W.; Li, Y.; Li, H.; Guo, Z.; Wei, C.; Long, M.; Chung, M.; Aimaier, R.; Li, Q.; et al. Preclinical Assessment of MEK Inhibitors for Malignant Peripheral Nerve Sheath Tumors Reveals Differences in Efficacy and Adaptive Response. Front. Oncol. 2022, 12, 903177. [Google Scholar] [CrossRef]
  86. Wang, J.; Pollard, K.; Calizo, A.; Pratilas, C.A. Activation of Receptor Tyrosine Kinases Mediates Acquired Resistance to MEK Inhibition in Malignant Peripheral Nerve Sheath Tumors. Cancer Res. 2021, 81, 747–762. [Google Scholar] [CrossRef]
  87. Kohlmeyer, J.L.; Lingo, J.J.; Kaemmer, C.A.; Scherer, A.; Warrier, A.; Voigt, E.; Raygoza Garay, J.A.; McGivney, G.R.; Brockman, Q.R.; Tang, A.; et al. CDK4/6-MEK Inhibition in MPNSTs Causes Plasma Cell Infiltration, Sensitization to PD-L1 Blockade, and Tumor Regression. Clin. Cancer Res. 2023, 29, 3484–3497. [Google Scholar] [CrossRef] [PubMed]
  88. Wang, J.; Pollard, K.; Allen, A.N.; Tomar, T.; Pijnenburg, D.; Yao, Z.; Rodriguez, F.J.; Pratilas, C.A. Combined Inhibition of SHP2 and MEK Is Effective in Models of NF1-Deficient Malignant Peripheral Nerve Sheath Tumors. Cancer Res. 2020, 80, 5367–5379. [Google Scholar] [CrossRef]
  89. Huang, P.-Y.; Shih, I.-A.; Liao, Y.-C.; You, H.-L.; Lee, M.-J. A Novel HDAC11 Inhibitor Potentiates the Tumoricidal Effects of Cordycepin against Malignant Peripheral Nerve Sheath Tumor through the Hippo Signaling Pathway. Am. J. Cancer Res. 2022, 12, 873–892. [Google Scholar]
  90. Lopez, G.; Torres, K.; Liu, J.; Hernandez, B.; Young, E.; Belousov, R.; Bolshakov, S.; Lazar, A.J.; Slopis, J.M.; McCutcheon, I.E.; et al. Autophagic Survival in Resistance to Histone Deacetylase Inhibitors: Novel Strategies to Treat Malignant Peripheral Nerve Sheath Tumors. Cancer Res. 2011, 71, 185–196. [Google Scholar] [CrossRef] [PubMed]
  91. Lopez, G.; Bill, K.L.J.; Bid, H.K.; Braggio, D.; Constantino, D.; Prudner, B.; Zewdu, A.; Batte, K.; Lev, D.; Pollock, R.E. HDAC8, A Potential Therapeutic Target for the Treatment of Malignant Peripheral Nerve Sheath Tumors (MPNST). PLoS ONE 2015, 10, e0133302. [Google Scholar] [CrossRef]
  92. Yoon, S.; Eom, G.H. HDAC and HDAC Inhibitor: From Cancer to Cardiovascular Diseases. Chonnam Med. J. 2016, 52, 1. [Google Scholar] [CrossRef]
  93. Cacabelos, R.; Torrellas, C. Pharmacoepigenomics. In Medical Epigenetics; Elsevier: Amsterdam, The Netherlands, 2016; pp. 585–617. ISBN 978-0-12-803239-8. [Google Scholar]
  94. Endo, M.; Yamamoto, H.; Setsu, N.; Kohashi, K.; Takahashi, Y.; Ishii, T.; Iida, K.; Matsumoto, Y.; Hakozaki, M.; Aoki, M.; et al. Prognostic Significance of AKT/mTOR and MAPK Pathways and Antitumor Effect of mTOR Inhibitor in NF1-Related and Sporadic Malignant Peripheral Nerve Sheath Tumors. Clin. Cancer Res. 2013, 19, 450–461. [Google Scholar] [CrossRef] [PubMed]
  95. Johansson, G.; Mahller, Y.Y.; Collins, M.H.; Kim, M.-O.; Nobukuni, T.; Perentesis, J.; Cripe, T.P.; Lane, H.A.; Kozma, S.C.; Thomas, G.; et al. Effective in Vivo Targeting of the Mammalian Target of Rapamycin Pathway in Malignant Peripheral Nerve Sheath Tumors. Mol. Cancer Ther. 2008, 7, 1237–1245. [Google Scholar] [CrossRef] [PubMed]
  96. Manji, G.A.; Stanton, L.J.; Ge, L.; Sta Ana, S.; Chrisinger, J.; Titus, S.; Labadie, B.W.; May, M.S.; Lyu, Y.; Sender, N.; et al. A Phase II Study of the Combination of Pexidartinib and Sirolimus to Target Tumor-Associated Macrophages in Unresectable Malignant Peripheral Nerve Sheath Tumors. JCO 2024, 42, 11565. [Google Scholar] [CrossRef]
  97. Widemann, B.C.; Lu, Y.; Reinke, D.; Okuno, S.H.; Meyer, C.F.; Cote, G.M.; Chugh, R.; Milhem, M.M.; Hirbe, A.C.; Kim, A.; et al. Targeting Sporadic and Neurofibromatosis Type 1 (NF1) Related Refractory Malignant Peripheral Nerve Sheath Tumors (MPNST) in a Phase II Study of Everolimus in Combination with Bevacizumab (SARC016). Sarcoma 2019, 2019, 7656747. [Google Scholar] [CrossRef] [PubMed]
  98. Kim, A.; Lu, Y.; Okuno, S.H.; Reinke, D.; Maertens, O.; Perentesis, J.; Basu, M.; Wolters, P.L.; De Raedt, T.; Chawla, S.; et al. Targeting Refractory Sarcomas and Malignant Peripheral Nerve Sheath Tumors in a Phase I/II Study of Sirolimus in Combination with Ganetespib (SARC023). Sarcoma 2020, 2020, 5784876. [Google Scholar] [CrossRef]
  99. Larsson, A.T.; Bhatia, H.; Calizo, A.; Pollard, K.; Zhang, X.; Conniff, E.; Tibbitts, J.F.; Rono, E.; Cummins, K.; Osum, S.H.; et al. Ex Vivo to in Vivo Model of Malignant Peripheral Nerve Sheath Tumors for Precision Oncology. Neuro-Oncology 2023, 25, 2044–2057. [Google Scholar] [CrossRef] [PubMed]
  100. Kivlin, C.M.; Watson, K.L.; Al Sannaa, G.A.; Belousov, R.; Ingram, D.R.; Huang, K.-L.; May, C.D.; Bolshakov, S.; Landers, S.M.; Kalam, A.A.; et al. Poly (ADP) Ribose Polymerase Inhibition: A Potential Treatment of Malignant Peripheral Nerve Sheath Tumor. Cancer Biol. Ther. 2016, 17, 129–138. [Google Scholar] [CrossRef]
  101. Patwardhan, P.P.; Surriga, O.; Beckman, M.J.; De Stanchina, E.; Dematteo, R.P.; Tap, W.D.; Schwartz, G.K. Sustained Inhibition of Receptor Tyrosine Kinases and Macrophage Depletion by PLX3397 and Rapamycin as a Potential New Approach for the Treatment of MPNSTs. Clin. Cancer Res. 2014, 20, 3146–3158. [Google Scholar] [CrossRef]
  102. Somatilaka, B.N.; Madana, L.; Sadek, A.; Chen, Z.; Chandrasekaran, S.; McKay, R.M.; Le, L.Q. STING Activation Reprograms the Microenvironment to Sensitize NF1-Related Malignant Peripheral Nerve Sheath Tumors for Immunotherapy. J. Clin. Investig. 2024, 134, e176748. [Google Scholar] [CrossRef]
  103. Borcherding, D.; Zhang, X.; Zhang, M.; Bhatia, H.; Lyu, Y.; He, K.; Yang, L.; Yang, K.; DiBenedetto, H.; Tsai, A.; et al. EXTH-63. MTA-COOPERATIVE PRMT5 INHIBITORS ARE EFFICACIOUS IN MTAP-DELETED MALIGNANT PERIPHERAL NERVE SHEATH TUMOR MODELS. Neuro-Oncology 2023, 25, v238. [Google Scholar] [CrossRef]
  104. Somaiah, N.; Van Tine, B.A.; Chmielowski, B.; Drabick, J.J.; Chawla, S.P.; McKean, M.; Spira, A.I.; O’Byrne, K.J.; Foresto, S.A.; Movva, S.; et al. A Phase 2 Study of Alrizomadlin, a Novel MDM2/P53 Inhibitor, in Combination with Pembrolizumab for Treatment of Patients with Malignant Peripheral Nerve Sheath Tumor (MPNST). JCO 2023, 41, e14627. [Google Scholar] [CrossRef]
Figure 1. Summary of key findings from recent sequencing technologies (created by Biorender).
Figure 1. Summary of key findings from recent sequencing technologies (created by Biorender).
Cancers 17 00180 g001
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

Xiao, K.; Yang, K.; Hirbe, A.C. A Sequencing Overview of Malignant Peripheral Nerve Sheath Tumors: Findings and Implications for Treatment. Cancers 2025, 17, 180. https://doi.org/10.3390/cancers17020180

AMA Style

Xiao K, Yang K, Hirbe AC. A Sequencing Overview of Malignant Peripheral Nerve Sheath Tumors: Findings and Implications for Treatment. Cancers. 2025; 17(2):180. https://doi.org/10.3390/cancers17020180

Chicago/Turabian Style

Xiao, Kangwen, Kuangying Yang, and Angela C. Hirbe. 2025. "A Sequencing Overview of Malignant Peripheral Nerve Sheath Tumors: Findings and Implications for Treatment" Cancers 17, no. 2: 180. https://doi.org/10.3390/cancers17020180

APA Style

Xiao, K., Yang, K., & Hirbe, A. C. (2025). A Sequencing Overview of Malignant Peripheral Nerve Sheath Tumors: Findings and Implications for Treatment. Cancers, 17(2), 180. https://doi.org/10.3390/cancers17020180

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